Next Article in Journal
Change in Neurocognitive Function in Patients Who Receive CAR-T Cell Therapies: A Steep Hill to Climb
Previous Article in Journal
Glycyrrhizic Acid Nanoparticles Subside the Activity of Methicillin-Resistant Staphylococcus aureus by Suppressing PBP2a
Previous Article in Special Issue
Medical Ozone: A Redox Regulator with Selectivity for Rheumatoid Arthritis Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

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

by
Ana C. Gonçalves
1,2,3,
Sofia Rodrigues
4,
Rafael Fonseca
5 and
Luís R. Silva
1,3,6,*
1
CICS-UBI—Health Sciences Research Center, University of Beira Interior, 6201-001 Covilhã, Portugal
2
CIBIT—Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, 3000-548 Coimbra, Portugal
3
SPRINT Sport Physical Activity and Health Research & Innovation Center, Instituto Politécnico da Guarda, 6300-559 Guarda, Portugal
4
Health Superior School, Polytechnic Institute of Viseu, 3500-843 Viseu, Portugal
5
Faculty of Medicine, University of Lisbon, 1649-028 Lisbon, Portugal
6
CERES, Department of Chemical Engineering, University of Coimbra, 3030-790 Coimbra, Portugal
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2024, 17(5), 590; https://doi.org/10.3390/ph17050590
Submission received: 8 April 2024 / Revised: 30 April 2024 / Accepted: 1 May 2024 / Published: 6 May 2024

Abstract

:
Rheumatoid arthritis (RA) is a complex illness with both hereditary and environmental components. Globally, in 2019, 18 million people had RA. RA is characterized 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 actions that benefit functional meals and nutraceuticals. These compounds have received a lot of attention recently because they have antioxidant, anti-inflammatory, immunomodulatory, and anti-rheumatoid activity by modulating tumor necrosis factor, mitogen-activated protein kinase, nuclear 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.

1. Introduction

There exist more than 100 conditions of arthritis known, and among them, rheumatoid arthritis (RA) is one of the most common forms observed in the 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 develop the disease between 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 the 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 the hands, wrists, feet, ankles, knees, shoulders, and elbows are most often affected [6]. The lining of joints 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. To 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, have been largely used to attenuate RA; however, most of them are expensive, rarely effective or 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, polyphenols, since their chemical structure with catechol methoxy and pyrogallol groups confer them notable health benefits, like antioxidant, anti-inflammatory, anti-bacterial, and antiproliferative activities, as well as the capacity to prevent neurological and cardiovascular pathologies, promote gastrointestinal health and vision without, it is believed, side-effects, and enhance wound healing [14,15,16,17,18,19,20,21,22,23]. In addition, polyphenols 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 signaling pathways [24,25,26,27,28]. Particularly, curcumin displayed the capability to relieve rheumatoid arthritis progression by suppressing inflammatory responses, synovial hyperplasia, and protein expression levels of phosphorylated JAK2 and STAT3 in mice with the collagen-induced arthritis model that were treated at a dose of 100 µM curcumin/day for almost three months [29], while in docking studies, salvianolic acid C exhibited a great binding affinity for JAK (10.7 kcal/mol) [24]. 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 [30], while tea polyphenols showed the capacity to enhance the expression of growth factor VEGF-A, which, in turn, promotes vascular endothelial cell division, induces angiogenesis, and improves wound morphology, restoring the type III/I collagen ratio during wound healing in mice model [23].
Furthermore, in addition to acting as nutraceuticals and functional foods, evidence shows that those polyphenols can also present many industrial applications, being used for water treatment and as natural food additives, natural dyes for textiles, smart food packaging and edible biofilms, anti-browning and food improvement agents, food enrichment and colorant, anti-aging creams and sunscreens, and hair products and nail remedies [31,32].
Keeping these facts in mind, the main goal of this work is to review and discuss the dietary polyphenols commonly found in the human diet, their rich sources in preventing and/or attenuating RA, and the most advanced strategies and also suggest future research.

2. Data Collection

The data collection was performed by searching, from the time of their establishment to 24 March 2024, scientific databases including National Center for Biotechnology Information (NCBI), Google Scholar, PubMed, ResearchGate, Science Direct, Scopus, SpringerLink, Web of Science, and trusted abstracts. The free terms, keywords, and MeSH terms used were polyphenols, phenolics, total phenolic 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 total, 369 papers were cited in this review.

3. Bioavailability and Metabolism of Polyphenols

After daily intake, polyphenols suffer many modifications along the gastrointestinal tract, which greatly influence their nutritional value and consequent health benefits. Therefore, exploring their bioavailability and metabolism is crucial to reveal the full potential of these compounds, of which the majority are considered non-toxic and safe for people [33,34].
Nowadays, it is well-established that polyphenolics are subject to extensive metabolization after consumption, and hence, it is not surprising that absorption is higher than expected, and some of them can be almost totally assimilated. In general, the maximum plasma concentration of polyphenolics is reached 1.5–2 h after ingestion, and excretion rates around 30%, varying between 12 h (flavanols) and 24 h (anthocyanins and hydroxycinnamics) [35,36].
Within different subclasses, isoflavones seem to exhibit the highest absorption rates (33.0–100.0%), followed by hydroxycinnamics (8.0–72.0%), anthocyanins (2.40–55.0%), flavonols and flavones (12.0–41.0%), flavanones, (11.0–16.0%), lignans (2.70–12.20%), and flavan-3-ols (2.0–8.0%) [34,37].
As far as we know, both the bioavailability and metabolism of polyphenol parameters differ significantly between different polyphenol compounds, and sometimes, the most dominant in our diet are not necessarily those that show the highest concentrations of active derivatives in tissues and organs. Indeed, molecular weight, glycosylation and/or acylation and/or polymerization degrees, conjugations, and/or combinations with other molecules impact bioavailability [38]. In addition, different pH values also influence these characteristics, as well as solubility, digestibility, the capacity to cross cell membranes, cooking, agronomic practices, food maturity index, and if they are consumed alone, and/or after an overnight fasting and/or accompanied with other foods or alone [39]. It is also important to take into account individual genetics, the existence of food intolerances, gender, age, sex, diet, lifestyle, and physiological and/or pathological states, which also interfere with different activities of gut microbiota, enterocytes, related enzymes, and biliary acids, also impact the bioavailability, metabolism and consequent biological effects of these phytochemicals [38].
Figure 1 summarizes the main steps involved in the absorption, metabolism, distribution, and excretion of polyphenols.
As expected, and before reaching systemic circulation, there is in the mouth that the degradation of polyphenolics initiates. Here, saliva enzymes remove glycoside groups, and then the compounds reach the stomach, where simpler aglycones, like isoflavones and phenolic acids, such as gallic acid, can be readily absorbed by bilitranslocase (bioaccessibility) or reach intestinal epithelium by passive diffusion thanks to their high lipophilicity or by using active sodium-dependent glucose cotransporters, including glycosides, polymers, and esters, becoming available for absorption [42]. On the other hand, the most complex polyphenols go to the small intestine. Here, they suffer glucuronidation, methylation, sulfation, or hydroxylation by cytosolic β-glucosidase and lactase-phlorizin hydrolase enzymes [41]. Next, simpler compounds reach the liver via the portal vein and the non-absorbed ones go to the colon to be transformed into simpler units, undergoing decarboxylation, dihydroxylation, demethylation, and/or hydrolysis thanks to the activity of bacterial esterases, including α-rhamnosidases, to suffer a new attempt at absorption by the liver [43]. Indeed, evidence reports that most polyphenols are only absorbed after being metabolized in the colon (90.0–95.0%) [44]. This event also leads to pH diminution by forming short-chain fatty acids, which, in turn, creates favorable conditions for the proliferation of Actinobacteria, Lactobacilli, and Bifidobacteria probiotic bacteria, hence exerting protective effects against digestive and gastrointestinal disorders, allergies, eczema, and others [38].
Therefore, microbiota causes hydrolysis of glycosidic and ester bonds and cleavage of aglycones into smaller molecules, while hepatocytes produce mainly phase I and II metabolites (conjugates) from intestinal metabolites such as aglycones, phenolic acids, and simple phenols [45]. Phase II also occurs in the small intestine and kidneys [46]. In addition, the non-absorbed polyphenols in the liver can go again, via bile, to the small intestine to be metabolized again [47]. The non-absorbed polyphenols are excreted in urine and feces [47].
Even so, to enhance the absorption, metabolism, and stability of polyphenolics, several strategies have been conducted to improve these topics. Among the most promising ones, nano-encapsulation with biopolymers, chitosan, pectin, whey protein, and liposomal micelle have been hot topics in scientific communities and medicine, pharmaceutical, and other related industries [48,49].

4. Major Sources of Dietary Polyphenols

Nowadays, the search for more effective, safer, and cheaper bioactive compounds than chemical drugs assumes great importance. Indeed, because most chemical drugs possess several unwanted effects and are little active, many people prefer to use natural molecules [33]. Among the most promising molecules, phenolic compounds, including simple phenols (e.g., catechol, pyrogallol, phloroglucinol, and resorcinol) and polyphenols (e.g., coumarins, anthocyanins, lignans, and phenolic acids) seem to be promising therapeutic and/or adjuvant approaches (Figure 2) [50,51,52]. Simple phenolics have a low molecular weight, and most of them (except resorcinol) are rapidly absorbed by the human skin, while polyphenols are largely widespread and ubiquitous in nature, being largely found in several types of algae, beans, fruits, bark, herbs and spices, legumes, leaves, nuts, whole grains, oilseeds, and roots (Table 1) [53,54]. In a general way, polyphenols can occur in free forms or conjugated with sugar moieties, acids, and other biomolecules that are soluble or insoluble in water, and, of course, these substitutions can interfere with the biological potential of these compounds [55]. Indeed, depending on the number and spatial position of hydroxyl groups, they can confer polyphenols with a great capacity to act as electron and hydrogen donors [56]. In addition, the CH=CH–COOH group and the double bond found in hydroxycinnamic acids make them more effective in reducing oxidative stress than hydroxybenzoics [57]. On the other hand, the presence of an o-diphenolic group and catechol hydroxyl groups, a double bond, and the conjugation between the double bond and the oxo group on the C ring enhance the antioxidant properties of flavonoids [58]. It is also important to note the obstruction of the O-methylation at the catechol group can diminish the biological potential of these phytochemicals [59].
Noticeably, 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 (TPCs) of 9.3–75.3, 94.4–228.4, and 100–2300 mg GAE per 100 g fresh weight (fw), respectively [60,61,62,63]. 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 [64,65]; in addition, flaxseeds also present considerable quantities of phenolic acids (800–1000 mg per 100 g) [66].
Focusing on phenolic acids, hydroxybenzoic derivatives are mainly found in black radish, onions, tea, and wine [67,68,69,70]. Particularly, gallic acid is found in considerable amounts in guava [4.21 mg/100 g dried weight (dw)], white mulberry (7.33–23.90 mg/100 g dw), mango (94.6–98.7 mg/100 g dw), avocado (0.56–2.54 mg/100 g dw), raspberry (0.73–3.75 mg/100 g dw), grape skins and seeds (3 and 10 mg/100 g dw, respectively), and blackcurrant leaves (17.98–22.85 mg/100 g dw) [71]. On the other hand, hydroxycinnamics are largely present in cereals, fruits, vegetables, tea, wine, and coffee [71,72,73,74]. Particularly, plums and sweet and tart cherries are richer in 3-O-caffeolquinic acid (amounts of 8.8–77.1, 7.3–62.8, and 8.2–53.6 mg/100 g fw, respectively) [67,75,76]. On the other hand, 5-O-caffeolquinic acid is largely present in apples and pears (both around 0.2 mg/100 g fw), while 3-feruloyquinic acid is mostly found in plums and apricots (quantities around 1.3 and 0.7 mg/100 g fw, respectively), caffeoylquinic acids in black currants (amounts between 4.5–5.2 mg/100 g fw), and p-coumaroylquinic derivatives in blueberries (186.0–208.0 mg/100 g fw) [67,77]. Regarding vegetables, 3-O-caffeolquinic acid is predominant in red cabbage (1.9–11.0 mg/100 g fw), Brussel sprouts (7.0–12.0 mg/100 g fw), broccoli (5.8 mg/100 g fw), and kale stalk leaves (0.61–7110.7 mg/100 g fw). Kale stalk leaves also present notable amounts of feruloyl glucose acid derivative (31.71 mg/kg) [67]. On the other hand, eggplant is richer in 4-O-caffeolquinic acid (57,563.2 mg/100 g fw) [67].
Focusing on anthocyanins, they are widely found in red and purple fruits and vegetables and are considered as being mainly responsible for their vibrant colors and health-promoting properties. Specifically, cyanidin 3-O-rutinoside is mainly found in sweet cherries, while cyanidin 3-O-glucosyl-rutinoside is the predominant anthocyanin in sour cherries [75,78]. Regarding TPC values, sour cherries present higher values than sweet cherries (275.3–652.27 versus 72.9–493.6 mg gallic acid equivalent (GAE) per 100 g fw for sweet cherries) [78,79,80]. On the other hand, the TPC value of blueberries fluctuates between 2.7 and 585.3 mg GAE per 100 g fw, being richer in peonidin derivatives, whereas for black elderberries, the TPC value is around 537.9 mg GAE per 100 g fw, and cyanidin 3-O-sambubioside is the main anthocyanin [77,81,82,83]. Blackberries also present considerable amounts of TPC (292.2–446.4 mg GAE per 100 g fw), where cyanidin 3-O-glucoside is the major anthocyanin [84,85]. For strawberries, the TPC value varies from 36.5 to 116.3 mg GAE per 100 g fw, where the most abundant is pelargonidin 3-O-glucoside [86,87,88], while for grapevines (TPC 9.3–75.3 mg per 100 g fw), malvidin 3-O-glucoside is the predominant anthocyanin [63,89,90]. Additionally, mulberries and chokeberries also present considerable amounts of TPCs (424–485 and 1022.4–1705.9 mg GAE per 100 g fw, respectively), where cyanidin 3-O-sophoroside and cyanidin 3-O-galactoside are the predominant ones, respectively [91,92,93], whereas black currants are richer in delphinidin 3-O-rutinoside and present TPC values fluctuating from 1930.0 to 3410.0 mg GAE per 100 g fw [94,95]. Red cabbage also presents considerable TPC scores (115.31 mg per 100 g fw), where the most prevalent anthocyanin is cyanidin 3-O-diglucoside-5-O-glucoside [96,97].
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) [98]. Regarding coumarins, marmelosin is found in high quantities in Aegle quince fruits (290 g per 100 g for TPC values varying from 905.0 to 4900.0 mg GAE per 100 g) [99,100], while esculin is predominantly found in horse-chestnut bark, accounting for around 7.9% of their total composition (33–50 mg/100 g) [101,102].
Regarding flavan-3-ols, the presence of (+)-catechin was reported in peaches (30–150 mg/100 g fw), apricots (2.1–3.2 mg/100 g fw), apples (0.4–2.1 mg/100 g fw), and green tea (50–100 mg in 250 mL), whereas (−)-epicatechin was reported in apricots (2.01–6.50 mg/100 g fw), sour cherries (1.1–9.6 mg/100 g), apples (3.1–7.8 mg/100 g fw), and cocoa (34.65–43.27 mg/g fw) [103,104,105,106,107,108,109,110,111]. On the other hand, epigallocatechin gallate is the major flavan-3-ol found in green tea (68–69%), followed by (−)-epigallocatechin (15–18%), (−)-epicatechin (5–6%), and epicatechin (2–5%) [112,113,114,115].
Flavonols, especially kaempferol derivatives, are largely present in onion leaves (amounts around 83.2 mg/100 g), papaya shoots (amounts around 45.3 mg/100 g fw), pumpkins (amounts around 37.1 mg/100 g), carrots (amounts around 14.0 mg/100 g), and black tea (amounts around 11.8 mg/100 g) [116,117,118,119]. On the other hand, Mentha pulegium L. is rich in quercetin aglycone (8.48 mg/100 g) [120,121], while dill (79.0 mg/100 g fw), onions (45.0 mg/100 g fw), oregano (42.0 mg/100 g fw), spinach (27.2 mg/100 g fw), and cranberries (25.0 mg/100 g fw) are rich in quercetin derivatives [122]. Myricetin derivatives are largely found in cranberries (6600 mg/100 g fw), dock (5700 mg/100 g fw), broad beans (2600 mg/100 g fw), rutabagas (2100 mg/100 g fw), blueberries (1300 mg/100 g fw), peppers (1200 mg/100 g fw), and blackberries (700 mg/100 g fw) [123]. Finally, fisetin derivatives are principally reported in strawberries (16 mg/100 g fw), apples (2.7 mg/100 g fw), persimmon (1.1 mg/100 g fw), lotus root (0.6 mg/100 g fw), onions (0.5 mg/100 g fw), grapes (0.4 mg/100 g fw), kiwi (0.2 mg/100 g fw), peaches (0.06 mg/100 g fw), and cucumber (0.01 mg/100 g fw) [124].
Relative to flavanone compounds, higher levels of hesperetin are found in dried peppermint (480.85 mg/100 g), honeybush tea (11.82 mg/100 g), onions (0.02 mg/100 g fw), clementine (39.9 mg/100 mL), sweet oranges (28.6 mg/100 mL), tangerines (24.3 mg/100 mL), lemons (20.5 mg/100 mL), limes (1.77 mg/100 mL), grapefruit pure juice (0.92 mg/100 g), and mandarins (0.15 mg/100 mL) [125,126]. On the other hand, naringin is mainly present in grapefruit pure juice (1.56 mg/100 mL), red wine (0.07 mg/100 mL), Mexican oregano (372 mg//100 g), rosemary (55.1 mg/100 g), sweet oranges (11.22 mg/100 g fw), tomatoes (0.96–3.84 mg/100 g fw), limes (3.40 mg/100 g fw), lemons (0.55 mg/100 g fw), and almonds (0.09 mg/100 g) [126,127]. Eriodictyol is present in Yerba Santa (23–795 mg/100 g) [128].
Focusing on flavones, apigenin is the most predominant compound found in nature, being largely reported in celery seeds (78.65 mg/100 g), spinach (62.0 mg/100 g), parsley (45.04 mg/100 g), marjoram (4.40 mg/100 g), oregano (3.50 mg/100 g), sage (2.40 mg/100 g), extra-virgin oil (1.17 mg/100 g), rosemary (0.55 mg/100 g), chamomile (3–5 mg/100 g), and pistachios (0.03 mg/100 g) [127].
Regarding flavonones, hesperidin derivatives are also predominant in citrus-based foods (0.026 mg/100 mL) [129,130], while honey and propolis present higher levels of pinocembrin (1.60–1.85 mg/100 g and 0.079–0.166 mg/100 mL, respectively) [131,132,133], and Glycyrrhiza glabra L. plant leaves and roots of liquiritin (1821 mg/100 g and 3336 mg/100 g, respectively) [132,134].
Focusing on ellagitannins, they were found to be only present in berries from the Rosaceae family, including strawberries and raspberries, rose hip, cloudberries, and sea buckthorn (amounts varying from 21.7 to 83.2 mg/100 g fw) [135]. Particularly, punicalagin is predominantly found in pomegranate peels (2803–10,414 mg/100 g) [136].
Regarding curcuminoid compounds, turmeric presents higher amounts of curcumin (877–1648 mg/100 g), 2-demethoxycurcumin (364–760 mg/100 g), and bisdemethoxycurcumin (276–431 mg/100 g fw) [137].
Of course, quantity values are greatly influenced by genotype, origin, climate, time of harvest, agricultural and processing techniques, and so on [138].
Table 1. Main polyphenols found in nature and their phenolic-rich sources.
Table 1. Main polyphenols found in nature and their phenolic-rich sources.
PolyphenolsSourcesReferences
Non-flavonoids
   LignansFlaxseeds[139]
   Phenolic acids
      Hydroxybenzoic acid derivatives
      Hydroxycinnamic acid derivatives

Black radish, onions, tea, guava, white mulberry, mango, avocado, raspberry, grape skins and seeds, and blackcurrant leaves
Cereals, coffee, fruits, tea, vegetables, and wine
[67,68,69,70,71]
   StilbenesGrapevines, peanuts and sorghums[140]
Flavonoids
   Anthocyanins
      Cyanidin derivatives
      Delphinidin derivatives
      Pelargonidin derivatives
      Peonidin derivatives
      Petunidin derivatives
      Malvidin derivatives

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
[75,77,88,94,141,142,143,144,145,146,147,148,149,150,151,152]
   Coumarins
      Umbelliferone
      Esculin

Aegle marmelo
Horse-chestnut bark
[100,101,153]
   Flavan-3-ols
      (+)-Catechin
      (−)-Epicatechin
      Epigallocatechin gallate

Peaches, apricots, apples, and green tea
Apricots, sour cherries, apples, cholate, and cocoa
Green tea
[67,103,154,155,156]
   Flavonols
      Quercetin
      Kaempferol derivatives
      Quercetin derivatives
      Myricetin derivatives
      Fisetin derivatives

Mentha pulegium L.
Onion leaves, papaya shoots, pumpkins, carrots, and black tea
Dill, onions, spinach, and cranberries
Cranberries, dock, broad beans, rutabagas, blueberries, peppers, and blackberries
Strawberries, apples, persimmons, grapes, onions, and cucumbers
[116,118,119,120,121,122,123,157,158]
   Flavanones
      Hesperetin
      Naringin
      Eriodictyol

Dried peppermint, honeybush tea, onions, clementine, sweet orange, tangerine, lemon, lime, grapefruit pure juice, and mandarins
Grapefruit pure juice, red wine, Mexican oregano, rosemary, sweet oranges, tomatoes, limes, lemons, and almonds
Yerba Santa
[127,159,160,161,162]
   Flavonones
      Hesperidin
      Liquiritin
      Pinocembrin

Citrus fruits
Glycyrrhiza glabra L. leaves and roots
Honey and propolis
[163,164,165,166]
   Flavones
      Apigenin

Celery seeds, spinaches, parsley, marjoram, oregano, sage, extra virgin oil, rosemary, chamomile, and pistachio
[127]
   IsoflavonesSoybeans[98,167,168,169,170,171]
   Ellagitannins
      Punicalagin
Strawberries and raspberries, rose hip, cloudberries, and sea buckthorn
Pomegranate
[135]
[136]
   Curcuminoids
      Curcumin

Turmeric
[137]

5. Pharmacological Properties of Dietary Polyphenols

Based on 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 break chains, by giving an electron or hydrogen atom, and interfere with inflammatory pathways, thus contributing to increase immunity defenses and promoting a healthy status. In addition, evidence also shows that they are effective in acting as anti-aging, anti-microbial, and antiproliferative agents and possess abilities to increase insulin sensitivity and digestion, lower blood sugar levels, and ameliorate brain and heart functions.
As expected, their benefits are closely linked to their chemical structure and quantities. Indeed, several studies already reported that, in most cases, an increment in phenolics enhances biological potential [65,75,79,172,173,174,175,176,177]. Therefore, it is not surprising that their interest is increasing worldwide, especially in preventing and/or attenuating several diseases, especially those without medical cures, like AR (Figure 3). In this disease, there is a verified 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 the nuclear factor erythroid 2–related factor 2 (Nrf2) pathway (discussed below). Phenolics have shown potential to counteract oxidative stress levels and downregulate 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, of the 247 anticancer drugs available, 200 were from natural products while 38 were synthetic drugs and nine were vaccines [33].

5.1. Antioxidant Capacity

Reactive oxygen and nitrogen species, namely, superoxide and nitric oxide radicals, play a vital part in the human body, which are signaling molecules that interfere with immune responses and signal transduction pathways, cell 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, thus playing a pathogenic role in chronic inflammatory diseases. Consequently, inflammation signaling cascades and oxidative stress components are triggered, which in turn enhance the risk of neuropathologies and chronic diseases, such as atherosclerosis, cancer, metabolic syndrome, and, of course, RA [179,180].
Regarding RA, its pathophysiology has been linked to oxidative stress, principally with the Nrf-2 pathway. This is the major pathway involved in the maintenance of homeostatic responses by raising intracellular defense mechanisms and regulating the heme oxygenase-1 axis, controlling macrophage activation and the NF-κB pathway, and reducing stress oxidative and inflammation [181]. 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 enters the nucleus, where the transcription of antioxidant enzymes occurs [182,183,184].
Among the most promising therapeutic and adjuvant approaches, polyphenols assume a prominent interest since they have already been shown to possess notable capabilities to restore oxidative stress and inflammation near basal levels by interacting with Keap1 protein, avoiding its linkage with its binding site in Nrf-2. This subsequently, causes the dissociation of Keap1 from Nrf2, leading to the transcriptional activation of Nrf-2 and, hence, stimulating intracellular antioxidant enzyme (glutathione, catalase, and superoxide dismutase) activity, and, of course, neutralizing oxidative stress [182,183,184].
In general, and in order to establish the antioxidant properties of phenolics, three statements were recently proposed, known as Bors criteria (Figure 4). The first one is the most significant, and it states that because of hydrogen bonding, the presence of a catechol group on the 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 the C ring, which in turn, facilitates electron delocalization and allows for better aryloxyl radical stabilization, while the third one is related to the presence of OH groups at positions 3 and 5 on the A and C rings combined with a 4-oxo group on the C ring, allowing electron delocalization via hydrogen bonds [185]. Thus, these characteristics make phenolics capable of acting as electron and hydrogen donors [55,186]. Among phenolics, quercetin and myricetin complete all Bors criteria, and hence, they are well-known because of their notable capacity to reduce radicals [187]. Moreover, the existence of the catechol group in quercetin and its derivatives enhances their biological potential [185].
Regarding non-flavonoid compounds, hydroxycinnamics showed better antioxidant capacities than hydroxybenzoic acids because of 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 [188,189,190,191]. For lignans, it is well-established that their capacity to eliminate radicals and free species is mainly because of the presence of catechol groups [192], while the hydroxylation at position 4 and hydroxyl groups at the ortho position are the main contributors to the antioxidant potential attributed to stilbenes [193].
Compared with flavonoids, these last ones are more effective in eliminating free species and radicals than non-flavonoids, not only because of the presence of more hydroxyl groups but also because of the double bond in positions 2 and 3, the o-diphenolic group and 3′,4′-catechol hydroxyl groups on the B ring, and the conjugation between the double-bond and the 4-oxo group on the C ring [191,194].
Even so, it is important not to forget that the 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 behavior [195,196,197,198]. Furthermore, pH also influences the antioxidant properties of phenolics; in fact, although lower pH levels increase iron-reducing activity, they diminish iron catalytic activity and chelate activities [199].
Focusing on their potential to interact with the Nrf-2 pathway, Diniyah and colleagues [200] 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, and are therefore promising new Nrf2 activators. Similar results were found involving hesperetin, hesperidin, naringenin, naringin, narirutin, neohesperidin, neohesperidin dihydrochalcone, and nobiletin citrus-derived flavonoids [201] and sesamol isolated from Sesamum indicum L. seed oil [183]. Mice treated with 37.5, 75, and 150 mg/kg green tea every 8 h by intragastric administration, which were then sacrificed 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 in 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 [184]. In accordance with the mentioned results, the daily intake of 25 mg/kg/body weight (bw) epigallocatechin gallate by rats with lung injury and oxidative stress induced by fluoride displayed the capability to increase antioxidant status and Nrf2 gene expression and reduce inflammatory cytokine [202]. Similar results were obtained using endothelial cells pre-treated with polychlorinated biphenyls 126 [203]. Furthermore, Li et al. [182] 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. It was also demonstrated that 10–100 µM 5-caffeoylquinic acid was a potent Nrf-2 activator in human hepatocellular carcinoma HepG2 cells [204]. 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 counteracting oxidative stress in hypothyroid rats by interfering with NF-κB/AKT/mTOR/KEAP1 [205].
Moreover, positive correlations were already reported between polyphenol amounts and biological potential. Particularly, positive correlations were already reported between sweet cherry anthocyanin-rich fractions and nitric oxide, superoxide radicals (r > 0.9013) [73], and ferric species (r = 0.739) and between TPCs and non-colored and colored fractions from sweet cherry fruits and hydrogen peroxide (r > 0.940) [79]. Additionally, a high correlation (r = 0.7581) between honey TPC and the elimination of nitric oxide radicals was also found [180,206], and between total flavonoid amounts in sweet cherry vegetal parts and the inhibition of hemoglobin oxidation (r > 0.9636) [207].
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) [206], whereas a mild correlation (r = 0.64) between the quercetin content of pollen extracts and protective effects against oxidative injury induced by AAPH on Hepa1-6 hepatic cells was also reported [208]. 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 in sweet cherries and the nitric oxide scavenging test, and also involving this last hydroxycinnamic and superoxide radical (r = 0.9958) [180,209] as well as between malvidin, delphinidin 3-O-arabinoside, and 5-O-caffeoylquinic acid and the antioxidant potential shown by blueberry fruits (r > 0.8689) [209].
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 the Keap1-Nrf2 complex, thus 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 [210,211,212]. Thus, several attempts have been made to reduce Nrf-2 levels [213,214]. Indeed, chrysin, luteolin, resveratrol, clofarabine, and 3′,4′,5′,5,7-pentamethoxyflavone have already been shown 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 the ability to reduce heme-oxygenase 1 [215,216,217,218]. 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), and neferine isolated from the 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 cell proliferation [152,212,214,219,220]. Finally, Castanea crenata Siebold and Zucc, Cinnamomi cortex, together with procyanidins isolated from it, and rosemary extracts, Chrysanthemum zawadskii Herbich and licorice (Glycyrrhiza uralensis Fisch. ex DC.), Bergenia ligulate (Wall.) Engl. and Rhododendron luteum sweet extracts, and strawberry tree honey also showed similar effects [210,211,213,221,222,223,224,225,226], as well as pterostilbene, a natural dimethoxylated analog of resveratrol [227].

5.2. 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 [73]. 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, hence stopping reactive species accumulation [228,229]. However, although inflammatory processes are directly linked to individual development, when these responses are exaggerated, carbohydrate damage, lipid peroxidation protein, and nucleic acid oxidation happen, contributing to the onset of many disorders, including AR [230,231,232]. Once again, given that phenolics can counteract oxidative stress and normalize immune responses, it is not surprising that phenolics are a target of extensive research. In fact, they have already been shown to possess capabilities to diminish the activity of COXs, lipoxygenase, and iNOS by interfering with different stages of the inflammatory cascade, namely, by down-regulating NF-κB and activating protein-1 and stimulating Nrf-2, mitogen-activated protein kinase (MAPK), a protein complex responsible for modulating a group of protein kinases that play an essential role in signal transduction by modulating gene transcription in the nucleus, and protein kinase-C [233,234,235,236]. In fact, the inhibition of NF-κB is crucial to attenuate inflammatory responses once this complex of proteins is 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 the severity of inflammation [237,238,239].
Within polyphenols, apigenin, catechin, ellagic acid, epigallocatechin gallate, epigallocatechin gallate, homoplantaginin, luteoloside, quercetin aglycone, quercetin 3-O-rutinoside, allopurinol, resveratrol, and tangeretin have already shown notable anti-inflammatory effects [203,240,241,242,243,244,245]. 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 [246,247,248,249]. Resveratrol can downregulate the inflammatory pathway activated by TNF-α in articular chondrocytes [250].
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) have already been shown to possess capabilities to suppress MAPK, JNK1 phosphorylation, and NF-κB and hence, downregulate the activity of COX-2 and inducible nitric oxide synthase and, consequently, diminish the production of prostaglandins and nitric oxide radicals, respectively [251,252]. In addition, caffeic acid phenethyl ester can also avoid the activation of toll-like receptor (TLR)-4 activation and liposaccharide-mediated NF-κB in macrophages [253].
Catechin and epicatechin (1.7–17.2 µM) can also modulate phorbol 12-myristate 13-acetate-induced NF-κB activation in Jurkat T cells [254]. Particularly, quercetin can inhibit the biosynthesis of leukotrienes in human polymorphonuclear leukocytes and the activation of NF-κB and p38 MAPK in human mast cells by preventing the degradation of Iκ Bα and the nuclear translocation of p65. Hence, it can reduce IL-1β, IL-6, IL-8, and TNF-α expression [255] as well as avoid iNOS gene expression by blocking the activation of IkK kinases, NF-κB, and STAT1 in mouse BV-2 microglia [256]. It 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 [257]. On the other hand, kaempferol and galangin display identic properties concerning epicatechin effects in mouse BV-2 microglia [258,259].
Similar to epicatechin, epigallocatechin gallate can also avoid IkK kinase activation, avoiding the degradation of Ik-Bα in culture respiratory epithelial cells and in rat models [260] 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 [233,261]. Furthermore, 10, 25, 50, and 100 µM epigallocatechin gallate also showed the capacity to attenuate COX-2 expression without affecting COX-1 in colon cancer PC-3 cells [233], as well as the ability to block NF-κB activation in human epithelial cells and reduce iNOS expression and, consequently, nitric oxide radical levels in macrophages at doses of 5 and 10 µM [233]. Furthermore, like genistein [262], luteolin can also suppress the activation of NF-κB 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 [263,264].
On the other hand, 25, 50, and 100 µM chebulanim, a natural polyphenol acid isolated from Terminalia chebula Retz inhibited the nuclear translocation of p38 and p65 in liposaccharide-stimulated macrophages in a dose-dependent manner [231]. Additionally, aqueous birch leaf extract of Betula pendula Roth inhibits the growth and cell division of inflammatory lymphocytes in a dose-dependent manner because of apoptosis induction [265].
Furthermore, hydroxytyrosol and resveratrol, two polyphenols largely abundant in olive oil and red wine, can also downregulate NF-κB and the expression of vascular cell adhesion molecule-1 in umbilical veins stimulated with liposaccharide at doses varying between 1 and 100 µM/L [266]. 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) [267]. 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 [194]. Furthermore, homoplantaginin, luteoloside, quercetin, quercetin 3-O-rutinoside, allopurinol, and resveratrol already showed the potential ability, both in vitro and in vivo, to suppress NLRP3 and/or TLR4 inflammasome activation [240,242,243,244,245].
Relative to in vivo studies, the daily ingestion of cherries (141 g) for 10 days by rats and ringdoves downregulated IL-1β and TNF-α pro-inflammatory cytokine levels and raised IL-4 and IL-2 anti-inflammatory cytokines [268]. In addition, gingerenone A (10 mg/kg/bw), a polyphenol largely present in ginger, also displayed positive effects in suppressing obesity and adipose tissue inflammation by avoiding macrophage infiltration and enhancing adiponectin, in high-fat diet-fed mice that were treated for 15 weeks [269], as well as quercetin [270]. 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-κB [241]. Apigenin, as well as other polyphenol compounds extracted from chamomile, meadowsweet, and willow bark, including quercetin and salicylic acid (0–100 μM), also revealed the ability to decrease IL-1β, IL-6, and TNF-α in THP-1 macrophages and also protect them against oxidative damage [271]. Tannic acid-based nanogel is also an efficient anti-inflammatory agent, showing a notable potential to reduce neutrophil recruitment and pro-inflammatory cytokines, indicating the successful alleviation of inflammation [272].
Moreover, curcumin can easily interfere with NF-κB, as well as with STAT3, thus 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 [273,274].
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 [30]. 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 [28]. Moreover, some Brazilian plants also showed potential to reduce TNF-α and CCL2 levels in lipopolysaccharide-stimulated human monocytic THP-1 cells [275]. Regarding individual compounds, ferulic and coumaric acids, which are largely found in Chinese propolis, Lonicera japonica Thunb and Kalanchoe gracilis showed similar capacities [276,277,278]. In addition, 1–1000 µg/mL quince (Cydonia oblonga Miller) peel polyphenols modulated liposaccharide-induced inflammation in human THP-1-derived macrophages, reducing IL-6 and increasing IL-10 expression by inhibiting NF-κB, p38MAPK and Akt pathways [279].
Focusing on clinical trials, it 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 [280].

5.2.1. Anti-Rheumatoid Effects

As expected, 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 previously mentioned results, it is not surprising that polyphenols can be considered promising molecules in preventing and/or attenuating rheumatoid arthritis.
So far, focusing on in vitro assays, it has been already reported that slibinin, a natural flavonoid extracted from milk thistle (Silybum marianum L. Gaertner), shows effectiveness in inhibiting Th17 cell differentiation and inducing macrophage M2 polarization in RAW 264.7 cells; it also promotes apoptotic events and inhibits NF-κB, SIRT1, and autophagy in fibroblast-like synoviocytes at doses of 50, 100, and 200 μM [281]. 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 to CD4+CD25+FoxP3 Tregs and induce the production of IL-10 and TGF-β [282]. 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 the MAPK and NF-κB signaling pathways [283].
Moreover, 5 and 10 µM curcumin showed effectiveness in reducing survivability and decreasing levels of MMP1 and TNF-α in synovial sarcoma SW982 cells, which is considered the best in vitro approach to study RA [284]. Additionally, 25–100 µM curcumin was shown to 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 [285]. In addition, at 12.5–50 µM, this compound also showed capacity to reduce IL-1β, PMA-induced IL-6, and VEGF-A expression by inhibiting NF-κB and induce dephosphorylation of ERK1/2 and enhance apoptosis in both in MH7A cells and RA-fibroblast-like synoviocytes [286].
On the other hand, 1, 5, and 25 µM quercetin diminish IL-17-stimulated RANKL production in RA fibroblast-like synoviocytes, IL-17-stimulated osteoclast formation, and Th17 differentiation, and hence, modulate bone destructive processes in RA [287]. Furthermore, 12.5–100 µM Punicalagin, a natural polyphenol extracted from pomegranate juice, also showed the capability to reduce IL-1β, IL-6, IL-8, IL-17A, MMP-1, and MMP-13 in fibroblast-like synoviocytes [288].
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 by inhibiting the MAPK/NF-κB signaling pathways [289].
Table 2. Main effects attributed to polyphenolic exposition and daily treatment.
Table 2. Main effects attributed to polyphenolic exposition and daily treatment.
Polyphenolic/PlantModelDoseEffectsReferences
In vitro studies
SilibininRAW 264.7 cells50, 100, and 200 μMTh17 cell differentiation inhibition
NF-κB, SIRT1, and autophagy inhibition
Macrophage M2 polarization induction
Apoptotic event promotion
[281]
OleuropeinPeripheral blood mononuclear cells and of cells of patient with RA50, 100, and 200 µg/mL↑ IL-10 and TGF-β
Shift CD4+ T cells from peripheral blood mononuclear cells of patient with RA to CD4+CD25+FoxP3 Tregs
[282]
Extra virgin olive oilSynovial fibroblasts12.5–50 µg/mL↓ IL-1β, TNF-α, IL-6, COX-2, and microsomal PGE synthase-1, and the MAPK and NF-κB signaling pathways[283]
CurcuminSynovial sarcoma SW982 cells5 and 10 µM↓ MMP1 and TNF-α[284]
QuercetinRA-fibroblasts-like synoviocytes1, 5, and 25 µM↓ IL-17-stimulated RANKL production
IL-17-stimulated osteoclast formation
Th17 differentiation
Modulate bone destructive processes in RA
[287]
PunicalaginFibroblast-like synoviocytes12.5–100 µM↓ IL-1β, IL-6, IL-8, IL-17A, MMP-1, and MMP-13[288]
SyringaldehydeLymphoblastic leukemia T lymphocytes25, 50, 100, 250, and 500 µM↓ CD86, CD40, MHC II, and IL-23
↑ IL-10 and antigen phagocytosis
Inhibition of the MAPK/NF-κB signaling pathways
[289]
ResveratrolFibroblast-like synoviocytes1, 3, and 10 µg/mL↓ Sirt1 protein, MMP1, and MMP13[290]
ResveratrolFibroblast-like synoviocytes20 µMInhibition of phosphorylation and acetylation of p65, c-Jun, and Fos
↓ COX-2 expression
[291]
ResveratrolFibroblast-like synoviocytes1–40 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
[292,293]
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
[294]
Resveratrol U251 glioma cells 1–100 µM Interference with the PI3K/Akt/BAD signaling pathway Inhibition of cells growth and apoptosis[295,296]
ResveratrolHuman umbilical vein endothelial cells20 µMInterference with PI3K/AKT and MEK/ERK
Induce FOXO transcriptional activity
Inhibition of cell migration and capillary tube formation
Prevent angiogenesis
[297]
Resveratrol Fibroblast-like synoviocytes 50 µMBlock cells at the G2/M stage
↓ TNF-α and S phase cell ratio
Promote serine–threonine kinase-p53 axis and autophagy
Cell apoptosis
[298]
Resveratrol Human RA synovial MH7A cells 100 and 200 µM ↓ Cell viability
Stimulate H2A.X phosphorylation and apoptosis events
Mitochondrial membrane potential disruption
Stimulate cytochrome c release from the mitochondria to the cytosol
Caspase-3 and caspase-9 activation
Upregulate the expression of NAD-dependent deacetylase SIRT1 mRNA
Downregulate the expression of Bcl-X(L) mRNA
Hyperplasia suppression
[299]
Resveratrol Fibroblast-like synoviocytes 200 µM/L Caspase-3 activation
Inhibition of cell proliferation
Induces cell apoptosis
[300]
Resveratrol Fibroblast-like synoviocytes 25–200 µM ↓ ROS and Bax
↑ Bcl-2 levels and apoptotic cells
Regulate the expression of mitochondrial superoxide dismutase
[301]
Resveratrol Fibroblast-like synoviocytes 100 µM ↓ MMP-1, MMP-3, MMP-9, RANKL, and osteoprotegrin[302]
Resveratrol Fibroblast-like synoviocytes 100 µM ↓ TNF-α by interfering with the SIRT1/cortistatin pathway[303]
Resveratrol Fibroblast-like synoviocytes 100 µM ↑ The expression of genes involved in mitosis, the cell cycle, chromosome segregation, and apoptosis [304]
ResveratrolFibroblast-like synoviocytes5, 15, and 45 mg/kg↓ IL-1, IL-6, IL-8, and TNF-α
↑ IL-10 and apoptosis
[305]
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
[306]
Resveratrol Mouse preosteoblastic MC3T3-E1 cells 1, 2, 3, and 5 µM Mediate SIRT-1 interactions with p300
Modulate NF-κB signaling activation
Inhibition of osteoclastogenesis
Prevent bone loss in bone-derived cells
[307]
Resveratrol + methotrexate Synovial mononuclear cells from patients with RA 25 µM resveratrol with 0.5 μg/mL methotrexate ↓ Monocyte chemoattractant protein 1 levels[308]
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
[285]
Curcumin Fibroblast-like synoviocytes and MH7A cells12.5–50 µM↓ IL-1β, PMA-induced IL-6 and VEGF-A expression, and cell viability
Inhibition of NF-κB and induced dephosphorylation of ERK1/2
↑ Apoptosis
[286]
Purified grape-derived compounds 1, 10, and 100 µM Human peripheral blood mononuclear cells↓ TNF-α, IL1, IL-6, and iNOS genes[150]
GallotanninsHuman mast cells1, 1, and 10 µg/mLDownregulate NF-κB expression[309]
Ellagic acidFibroblast-like synoviocytes10, 25, 50, and 100 µM↓ IL-6, IL-1β, MDA, and TNF-α
↑ Superoxide dismutase and apoptosis
[310]
Gallic acidFibroblast-like synoviocytes0.1 and 1 µM↑ Caspase-3 activity
Regulate Bcl-2, Bax, p53, and pAkt production
↓ IL-1β, IL-6, CCL-2/MCP-1, CCL-7/MCP-3, COX-2, and MMP-9
[311]
Rosmarinic acid nanoparticlesMacrophagesNot mentioned↓ RONS and pro-inflammatory cytokines[237]
ρ-Coumaric acid encapsulated with mannosylated liposomesMacrophagesNot mentioned↓ RONS and pro-inflammatory cytokines
Inhibition of osteoclast differentiation
Downregulate the expression of MMP-9 and NFATc1
[312]
Ferulic acidFibroblast-like synoviocytes25–300 µM↓ IL-17-levels
Inhibition of the IL-17/IL-17RA/STAT-3 signaling cascade
[313]
Ferulic acidRAW 264.7 macrophages25, 50, and 100 µMAttenuate RANKL-induced osteoclast differentiation
↓ Bone resorption activity
Downregulate NFATc1, c-Fos, TRAP, Cathepsin K, and MMP-9 levels
[314]
Chlorogenic acidT cells c110–50 µg/mLInhibit osteoclast differentiation and bone resorption
Downregulate RANKL
Suppress mRNA expression of NFATc1, TRAP, and OSCAR
[315]
Tea polyphenol carrier-enhanced dexamethasoneUmbilical vein endothelial, murine fibroblast cells L929, and murine macrophage RAW 264.7 cellsNot mentioned↓ Inflammation[316]
Tinospora cordifoliaRAW 264.7 cells100, 250, and 500 µg/mL↓ IL-6, TNF-α, PGE2, and NO, and iNOS and COX
Modulate JAK/STAT pathway
[317]
Blueberry polyphenolsHIG-82 rabbit synoviocytes100–200 µM↓ TNF-α, IL-1β, MMP3, and NF-κB levels[318]
Cocoa polyphenolsMouse epidermal cells10 and 20 µg/mL↓ TNF-α-induced vascular endothelial growth factor expression
Inhibition PI3K and MEK1
[319]
Catechin-7,4′-O-digallate from Woodfordia unifloraMouse macrophages5–80 µM↓ IL-6 and IL-1β levels
Regulate the NF-κB signaling pathway
[320]
Salacia reticulata leavesMTS-C H7 cellsIC50 score of ~850 μg/mLInhibition of cell proliferation[321]
In vivo studies
SilibininRats with induced RA50, 100, and 150 mg/kg↓ IL-1β, IL-6, and TNF-α levels and joint inflammation[281]
ResveratrolRats with induced RA5 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, glutathione peroxidase, and the glutathione reductase ratio
[322]
ResveratrolRats with induced RA10 mg/kg↓ Progression of periodontitis and rheumatoid factor amount[228]
ResveratrolRats with induced RA10 mg/kg↓ Wnt5a, MAPK3, Src kinase, and STAT3 levels[323]
ResveratrolRats with induced RA10 mg/kg↓ IL-6 and TNF-α levels, atrial apoptosis and fibrosis, and activate the AMPK/PGC-1α pathway[324]
ResveratrolRats with induced RA10 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
[325]
ResveratrolRats with induced RA50 mg/kg↓ Paw swelling, TNF-α, IL-1β, TBARs, and NOx
Suppress NF-κB p65 expression
[326]
ResveratrolRabbit inflammatory RA model10 µMol/kg↓ Inflammatory responses
Prevent the loss of matrix proteoglycan content in the cartilage
[327]
ResveratrolMurine collagen-induced arthritis15 and 20 mg/kgInhibition of Th17 and B-cell function[328]
ResveratrolRats with bovine type-II collagen-induced arthritis400 g/kg/bw↓ Oxidative stress, inflammation, and MDA levels
↑ Serum superoxide dismutase
Suppress MAPK signaling pathways and angiogenesis
[294]
ResveratrolAdjuvant arthritis rat model45 mg/kg↓ Store-operated Ca2+ entry
↑ Apoptosis
Interference with the ORAI1-STIM1 complex
[329]
ResveratrolRats with induced RA12.5 mg/kgInduce the noncanonical autophagy pathway
↓ p62 expression, caspase-3 expression, poly(ADP-ribose) polymerase, IL-1β, C-reactive protein, prostaglandin E2, and NF-κB synovial tissue expression
[330]
ResveratrolRats with induced RA12.5 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[331]
ResveratrolCollagen-induced arthritis rat model2.5 and 10 mg/kgSuppress MMP1 and MMP13 amounts[290]
ResveratrolAdjuvant arthritis rats10 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
[332]
ResveratrolRats with induced RA10 mg/kgAlleviates adjuvant arthritis-interstitial lung disease[333]
ResveratrolRats with induced RA10 mg/kgPrevent the production of pro-inflammatory by modulating JAK/STAT/RANKL signaling pathway
Ameliorate fibrosis via the autophagy lysosome pathway
[232]
Resveratrol combined with methotrexate loaded-nanoemulsionRats with induced RANot mentioned↓ Inflammation
Better anti-arthritic effects potentiated by resveratrol
[334]
QRu-PLGA-DS nanoparticles carried resveratrolArthritic ratsNot mentionedImproves the water solubility and targets the effectiveness of this compound
Ameliorate anti-inflammatory effects
↑ M2-type macrophage transformation
↓ The recruitment of M1-type macrophages
[335]
Ellagic acidArthritic rats5, 50, and 100 mg/kg↓ Oxidative stress and inflammation
↑ Serum superoxide dismutase
Suppresses MAPK signaling pathways, angiogenesis, and MTA1/HDAC1-mediated Nur77 deacetylation
[310]
Ellagic acidArthritic rats25 mg/kg↓ Articular edema, NF-κB, neutrophil elastase, and neutrophil extracellular traps
Interference with TLR-4, peptidyl arginine deiminase 4 enzyme, and COX-2
[336]
Epigallocatechin gallateRats with induced RA10 mg/kgAmeliorate RA symptoms
↓ Histological scores in arthritic mice, as well as reduces 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-κB, Nrf-2, and heme oxygenase-1
[337]
Epigallocatechin gallateCollagen-induced arthritis rat model50 mg/kg↓ TNF-α, IL-17, Nrf-2, and MDA levels
↑ Heme oxygenase-1, superoxide dismutase, catalase, and glutathione peroxidase levels
[234]
Epigallocatechin gallateRats with induced RA10 mg/kg↓ Neuroinflammation, namely, by activating caspase-3[338]
Epigallocatechin gallateMice with collagen-induced arthritis50 mg/kg↓ The arthritis index
Protective effects against joint destruction
Inhibition of osteoclastogenesis and TH17 cell activation
↑ The number of Treg cells
[339]
Extracellular vesicles-encapsulated epigallocatechin gallateRats with induced RANot mentionedDownregulate the expression of hypoxia-inducible factor 1-α
Inhibition apoptosis of chondrocytes
Promote the recovery of type II collagen
↓ Joint swelling
[340]
EpigallocatechinArthritic ratsNot mentioned↑ Reduced elastic modulus, hardness, and stiffness in cartilage[341]
EpigallocatechinRats with induced RA10 mg/kgPrevent cartilage destruction in at by imbibing myeloperoxidase activity [342]
Green teaRats with induced RA t2–12 g/L↓ RA severity and IL-17 levels
↑ IL-10 levels
Suppress the anti-Bhsp65 antibody response
[343]
Tinospora cordifoliaRats with induced RA150 mg/kg↓ Erythema, paw edema, hyperplasia, IL-6, TNF-α, IL-17, NO, and PGE2 levels, phosphorylation of STAT3, and the expression of VEGF[317]
KalpaamruthaaRats with induced RA150 mg/kg↓ Oxidative stress, myeloperoxidase and lipid peroxide, and increase the activity of enzymic and non-enzymic antioxidants[344]
Ribes orientale Def.Sprague Dawley rats with induced RA50, 100, and 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
[229]
ChebulaninCollagen-induced arthritis mouse model80 mg/kgSuppress the progression and development of RA
↓ Arthritis severity scores, paw swelling and joint destruction, IL-6 and TNF-α amounts and excised phosphorylated (p)-p38 and p-p65, phosphorylated-c-JUN N-terminal kinase, and phosphorylated NF-κB and inhibitor alpha
[231]
PunicalaginRats with induced RA50 mg/kg/Prevent the translocation of p-65
Avoid the phosphorylation of IkK and Ik Bα
Modulate the NF-κB pathway
↓ TNFα, IL-6, CD86, CCR7, CD40, and MHC II expression, Th1, Th17, and Th17/Th1-like
↑ IL-10 expression
Suppress dendritic cell migration
Promote the generation of Tregs via the regulation of dendritic cells maturation
[288]
SyringaldehydeRats with induced RA10, 25, and 50 mg/kgAlleviate paw and joint edema
↓ TNF-α and IL-6 levels
↑ IL-10
[289]
SyringaldehydeRats with induced RA100 and 200 mg/kg↓ IL-6 and TNF-α levels[345]
Clitoria ternatea L. flower petals and its major compound, quercetin-3ß-D-glucosideRats with induced RA50 mg/kg Clitoria ternatea L. 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[346]
Berberis orthobotrys Bien ex AitchRats with induced RA150 mg/kgProtected against arthritic lesions, oxidative damage, and body weight alterations
Ameliorated altered hematological parameters and the rheumatoid factor
Contributed to positively modified radiographic and histopathological changes
[347]
Diospyros malabarica (Desr.) Kostel fruitsRats with induced RA250, 500, and 750 mg/kg↑ Anti-inflammatory enzymes
↓ Anti-inflammatory enzymes
[348]
ρ-Coumaric acidRats with induced RA100 mg/kgSuppress paw edema and body weight loss
↓ cartel
[316]
ρ-Coumaric acidRats with induced RA100 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 RANKL/OPG imbalance
Inhibit RANKL-induced NFATc-1 and c-Fos expression
[238,349]
Chlorogenic acidRats with induced RA10 mg/kgAttenuate liposaccharide-induced bone loss of rat femurs[315]
Theaflavin-3,3′-digallateCollagen-induced RA mouse model10 mg/kg↓ IL-1β, TNF-α, IL-6, as well as MMP-1, MMP-2, and MMP-3 amounts
Inhibition the activation of NF-κB and the phosphorylation of P38, JNK2, and ERK
[230]
Cinnamtannin D1Rats with induced RA50 mg/kgAlleviate the severity of RA
↓ Clinical scores and paw swelling, inflammatory cell infiltration, cartilage damage in joints, 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
[350]
Cinnamon barksMice with induced RA200 mg/kg↓ Paw volume, weight loss, and IL-2, IL-4, and IFNγ levels[351]
N-feruloylserotoninRats with induced RA3 mg/kg↓ C-reactive protein, the activity of LOX, as well as mRNA transcription of TNF-α, iNOS IL-1β, and IL-1β mRNA expression[352]
Extra virgin olive oilMice with collagen-induced RA100 and 200 mg/kg↓ Inflammatory markers, joint edema, cell migration, cartilage degradation and bone erosion, and also reduces COX-2 and microsomal prostaglandin E synthase-1 expression
Inhibition c-Jun N-terminal kinase, p38, signal transducer, and activator of transcription-3
[353]
Hydroxytyrosol acetateMice with collagen-induced RA0.05%↓ 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
[354]
MangiferinMice with induced RA50, 100, and 400 mg/kgInhibition of mRNA expression of cytokine genes in the thymus and spleen, and also NF-κB and activating ERK1/2
↓ IL-1β, IL-6, TNF-α, and RANKL
[355]
Sarcococca salignaRats with induced RA250 mg/kg↓ IL-1β, IL-6, COX-2, prostaglandin E2, TNF-α, and NF-κB levels, the arthritic index, and paw inflammation
↑ IL-4 and IL-10 levels
[236]
CurcuminRats with induced RA10 mg/kg↓ TNF-α and IL-1β[356]
Dichrostachys cinerea Wight et Arn. fruitsRats with induced RA75.48 mg↓ IL-1β, IL-6, TNF-α, and cortisol levels, lipid peroxidation, and NOx production[357]
Circaea mollis Sieb. and Zucc. plantFreund’s complete adjuvant-induced arthritis model in rats170–1350 mg/kg↓ Paw and inflammatory swelling, the arthritis index, TNF-α, and IL-1β levels
↑ IL-10 levels
[358]
Opuntia littoralisRats with induced RA10 and 20 mg/100 g bw↓ Joint inflammation, paw swelling, edemas, MDA, and IL-1β, IL-6R, IL-6, IL-17, and IL-23,
Ameliorate COX-2, NF-κB, STAT-3, PTEN, and RANKL expression
Upregulate the expression of miR-28 and miR-199a
[239]
Antrocaryon micraster A. Chev. and Guillaumin seedsRats with induced RA25 and 100 mg/kg↓ Cachexia, paw edema, infiltration of inflammatory cells, pannus formation, and synovium damage[359]
Dried plumsTransgenic mice with induced RA+ 20% dried plums in the normal dietProtect 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
[360]
Opuntia monacanthaRats with induced RA750 mg/kg↓ Paw edema, the arthritic score, the rheumatoid factor, inflammation, COX-2, IL-6, TNF-α, IL-1, NF-κB, bone erosion, and pannus formation
Restore hemoglobin, white blood count, and platelet parameters
↑ Catalase and superoxide dismutase, IL-4 and IL-10 levels
Inhibition of glutaminase 1 activity
[361]
Solanum nigrumRats with induced RA800 mg/kg↓ Paw edema
Restore body weight, hematologic parameters, and radiographic and histopathologic alterations
[362]
Quercetin and quercetin-loaded chitosanRats with induced RA15 mg/kg quercetin and 10 and 20 mg/kg quercetin-loaded chitosan↓ TNF-α and IL-6
The nanoencapsulation of quercetin enhances its efficacy
[363]
Grape polyphenols + propolisFemale rats with induced RA1.25 g/kg grape polyphenols mixed with 1.25 g/kg propolis↓ The intensity of cachexia and alleviate RA scores[364]
Malvidin 3-O-β glucosideChronic rat adjuvant-induced arthritis with125 mg/kg↓ Cachexia and arthritic paw scores[150]
Phoenix dactylifera L. seedsRats with induced RA30 mg/kg↓ IL-1β levels, paw edema, the erythrocyte sedimentation rate, and C-reactive protein[365]
Liposomal drug delivery system for morinRats with induced RANot mentioned↓ TNF-α, IL-1β, IL-6, IL-17, RANKL, STAT-3, p-STAT-3, VEGF, iNOS, and NF-κB-p65
↑ Osteoprotegerin and murin uptake by rats synovial and spleen macrophages
[235]
Clinical trials
Low-calorie cranberry juice500 mL/dayWomen with RA↓ Anti-cyclic citrullinated peptide antibodies levels, pain intensity, and swollen joints[366]
Low-calorie cranberry juice + fish oil ω-3 fatty acids500 mL/day of low-calorie cranberry juice with 3 g of fish oil ω-3 fatty acidsPeople with rheumatoid arthritis↓ C-reactive protein, the erythrocyte sedimentation rate, and related pain[367]
Pomegranate extract250 mgPatients with RA↓ Swollen, pain intensity and tender joints, the erythrocyte sedimentation rate, and morning stiffness
↑ Glutathione peroxidase
[368]
Resveratrol1 gPatients with↓ Joint swelling, tenderness, TNF-α, IL-6, protein C-reactive, MMP-3, the erythrocyte sedimentation rate, and undercarboxylated osteocalcin[369]
↑: 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: receptor activator of nuclear factor kappa beta; RA: rheumatoid arthritis; NF-κB: nuclear factor kappa B; MPO: myeloperoxidase; COX: cyclooxygenase; JNK: c-Jun N-terminal kinase; BAX: Bcl-2 associated X protein; Bcl-2: B-cell lymphoma 2; MAPK: mitogen-activated protein kinase; ERK: extracellular signal-regulated kinase.
Furthermore, 1, 3, and 10 µg/mL resveratrol also showed ability to reduce Sirt1 protein, MMP1 and MMP13 expression [290]. In addition at 20 µM, it inhibited the phosphorylation and acetylation of p65, c-Jun, and Fos and reduced binding to the COX-2 promoter, thereby attenuating COX-2 expression in fibroblast-like synoviocytes [291]. In addition, at 1–40 µM, this compound also showed an ability to activate Nrf2-2, heme oxygenase-1, and Bcl-2/Bax; induce apoptosis; reduce Keap1 expression and reactive oxygen species and malondialdehyde levels; block NF-κB p6 translocation; inhibit cell proliferation; and migration on fibroblast-like synoviocytes in a dose-dependent manner [292,293]. Moreover, Yang and co-workers [294] demonstrated that 25 and 50 µmol/L resveratrol can reduce hypoxia-inducible factor-1α and activate phosphorylation of p38 MAPK and c-Jun N-terminal kinase in IL-1β-stimulated RSC-364 cells, as well as arrest these cells at the G0/G1 cell cycle and enhance their apoptosis. Similar data were reported by Tian et al. [295,296], who also verified that resveratrol can interfere with the PI3K/Akt/BAD signaling 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 the MEK/ERK pathway, and thus induce FOXO transcriptional activity and inhibit cell migration and capillary tube formation in human umbilical vein endothelial cells, preventing angiogenesis [297]. On the other hand, Li et al. [298] reported that 50 μM resveratrol can block fibroblast-like synoviocytes in RA cells at the G2/M stage and reduce TNF-α levels and the S phase cell ratio by promoting the serine–threonine kinase-p53 axis, and autophagy, which, subsequently, leads to cells apoptosis. Moreover, 100 and 200 µM resveratrol also showed the ability to reduce the viability of human RA synovial MH7A cells by stimulating H2A.X phosphorylation and consequent apoptosis events, disrupt mitochondrial membrane potentials and stimulat cytochrome c release from the mitochondria to the cytosol, activate caspase-3 and caspase-9 but not caspase-8, upregulate the expression of the NAD-dependent deacetylase sirtuin (SIRT) 1 mRNA and downregulate the expression of the Bcl-X(L) mRNA, hence suppressing synovial cells hyperplasia [299]. The capacity of resveratrol to activate caspase-3, and consequently inhibit the proliferation of synoviocytes and induce cell apoptosis in synoviocytes in RA, was already reported by Tiang et al. [300]. Furthermore, Wang and colleagues revealed that 25–200 µM of this compound can also reduce mitochondrial reactive oxygen species and 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 [301]. On the other hand, a dose of 100 µM resveratrol reduces the expression of MMP-1, MMP-3, MMP-9, RANKL, osteoprotegrin [302], and TNF-α by interfering with the sirtuin1/cortistatin pathway [303], as well as increasing the expression of genes involved in mitosis, the cell cycle, chromosome segregation, and apoptosis in RA fibroblast-like synoviocytes [304]. Similar data were obtained by Lu and co-workers [305], who also verified that resveratrol can diminish the expression of IL-1, IL-6, IL-8, and TNF-α and raise the expression of IL-10; in addition, the administration of resveratrol together with 5 µM hydrogen peroxide can induce fibroblast-like synoviocyte 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 [306]. Finally, 1, 2, 3, and 5 µM resveratrol can mediated SIRT-1 interactions with p300, modulating RANKL activation of NF-κB signaling, inhibiting osteoclastogenesis, and thus, preventing bone loss in bone-derived cells [307].
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 patients with RA [308].
It was previously shown that 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 [150].
Furthermore, 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 the potential to downregulate NF-κB expression in a dose dependent in human mast cells [309].
Focusing on phenolic acids, 10, 25, 50, and 100 µM ellagic acid 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-κB signaling and a upstream modulator of inflammation and immunologic responses [310]. On the other hand, 0.1 and 1 µM gallic acid increase caspase-3 activity, regulate Bcl-2, Bax, p53, and pAkt production and reduce IL-1β, IL-6, CCL-2/MCP-1, CCL-7/MCP-3, COX-2, and MMP-9 on fibroblast-like synoviocytes from patients with RA [311]. More recently, it was reported that rosmarinic acid nanoparticles showed a favorable capability in scavenging reactive oxygen and nitrogen species and pro-inflammatory cytokines produced by macrophages [237]. ρ-Coumaric acid encapsulated with mannosylated liposomes showed similar effects, as well as an ability to inhibit osteoclast differentiation and downregulate the expression of MMP-9 and NFATc1 [312]. Moreover, 25–300 µM ferulic acid showed ability to reduce IL-17-levels in RA fibroblast-like synoviocytes thanks to its ability to inhibit the IL-17/IL-17RA/STAT-3 signaling cascade [313]. 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 [314]. On the other hand, 10–50 µg/mL chlorogenic acid can inhibit osteoclast differentiation and bone resorption by downregulating RANKL on activated T cells c1, as well as by suppressing mRNA expression of NFATc1, TRAP, and OSCAR [315].
Tea polyphenol carrier-enhanced dexamethasone showed an ability to reduce inflammatory mediators in LPS/INF-γ-induced inflammatory cell models, including umbilical vein endothelial, murine fibroblast cells L929, and murine macrophage RAW 264.7 cells [316]. Tinospora cordifolia (Willd.) Hook. f. and Thomson 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 the JAK/STAT pathway [317]. On the other hand, 100–200 µM blueberry polyphenols can reduce TNF-α, IL-1β, MMP3, and NF-κB levels on HIG-82 rabbit synoviocytes previous stimulated with TNF-α [318]. 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 [319]. Another study showed that 5–80 µM Catechin-7,4′-O-digallate from Woodfordia uniflora can also regulate the NF-κB signaling pathway in mouse macrophages and reduce IL-6 and IL-1β levels [320]. On the other hand, Salacia reticulata leaves showed the capacity to inhibit MTS-CH7 cell proliferation (IC50 score of ~850 µg/mL) [321].

In Vivo Studies

On the other hand, in RA-mice models, 50, 100, and 150 mg/kg/day silibinin showed an ability to diminish IL-1β, IL-6, and TNF-α levels and joint inflammation [281].
Resveratrol was shown to be a promising strategy for attenuating RA in a 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, glutathione peroxidase, and the glutathione reductase ratio [322]. Furthermore, at 10 mg/kg, this compound showed an ability to reduce the progression of periodontitis and the rheumatoid factor amount [228], Wnt5a, MAPK3, Src kinase, and STAT3 levels [323], diminish IL-6 and TNF-α levels and atrial apoptosis and fibrosis, and activate the AMPK/PGC-1α pathway [324], as well as decrease the serum rheumatoid factor, MMP-3, cartilage oligomeric matrix protein, IgG, antinuclear antibody, TNF-α, myeloperoxidase, C-reactive protein and malondialdehyde to about 37, 59, 44, 70, 5, 30, 23, 33, and 28%, respectively, and enhance IL-10 and glutathione to about 225 and 273%, respectively, in RA-rats [325]. Furthermore, 50 mg/kg resveratrol can diminish paw swelling, TNF-α, IL-1β, TBARs, and NOx, namely, by suppressing NF-κB p65 expression [326], 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 [327]. Moreover, a 10-day treatment with 15 and 20 mg/kg resveratrol modulates murine collagen-induced arthritis by inhibiting Th17 and B-cell function [328]. In accordance with other studies, Yang et al. [294] also showed that 400 g/kg/bw resveratrol diminishes oxidative stress and inflammation, by raising serum superoxide dismutase and reducing malonaldehyde, and suppresses the MAPK signaling 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 a capacity to suppress MTA1/HDAC1-mediated Nur77 deacetylation [310]. Furthermore, 25 mg/kg of this compound can also decrease articular edema, NF-κB, and neutrophil elastase, and hence, diminish neutrophil extracellular traps, in RA rats, namely, by interfering with TLR-4, peptidyl arginine deiminase 4 enzyme and COX-2 [336]. Resveratrol at 45 mg/kg can also reduce store-operated Ca2+ entry and enhance the apoptosis in an adjuvant arthritis rat model by targeting the ORAI1-STIM1 complex [329]. Additionally, Fernández-Rodríguez et al. [330] reported that 12.5 mg/kg/day resveratrol can also induce the noncanonical autophagy pathway, reduce 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 [330]. It can also diminish PCNA, CD68, CD3, monocyte chemoattractant protein-1 staining, cytokine-induced neutrophil chemoattractant-1 and the level of the marker of DNA damage, and 8-oxo-7,8-dihydro-2′-deoxyguanine in RA rats [331]. Resveratrol at 2.5 and 10 mg/kg/day can also display an ability to suppress MMP1 and MMP13 amounts in a collagen-induced arthritis rat model [290], 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 [332]. Resveratrol at 10 mg/kg/day alleviates adjuvant arthritis-interstitial lung disease in rats with induced RA, namely, by preventing the production of pro-inflammatory by modulating the JAK/STAT/RANKL signaling pathway [333], as well as ameliorate fibrosis in rats with induced-RA via the autophagy-lysosome pathway [232].
More recently, 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 in inflammation by 78.76% and a better anti-arthritic effects [334]. Moreover, the nanocarrier QRu-PLGA-DS nanoparticles effectively improved the water solubility and targeting ability of this compound to reverse M1 to 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 raises the transformation of M2 type macrophages and decreases the recruitment of M1 type macrophages [335].
Regarding epigallocatechin gallate, 10 mg/kg 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-κB, Nrf-2, and heme oxygenase-1 [337]. On the other hand, in a collagen-induced arthritis rat model, 50 mg/kg/day epigallocatechin gallate also showed the capacity to diminish TNF-α, IL-17, Nrf-2, and malondialdehyde levels and improve heme oxygenase-1, superoxide dismutase, catalase, and glutathione peroxidase levels [234]. Moreover, it can also reduce neuroinflammation in RA rats after 2 months of treatment, namely, by activating caspase-3 [338]. Moreover, 50 mg/kg epigallocatechin gallate showed the ability to decrease the arthritis index, provide protective effects against joint destruction in mice with collagen-induced arthritis, inhibit osteoclastogenesis and TH17 cell activation, and increase the number of Treg cells [339]. On the other hand, the administration of extracellular vesicle-encapsulated epigallocatechin gallate for cartilage repair in rat arthritis downregulated the expression of hypoxia-inducible factor 1-α, inhibited apoptosis of chondrocytes, promoted the recovery of type II collagen, and reduced joint swelling in rheumatoid rats by approximately 39.5% [340]. In addition, the nanoindentation of epigallocatechin significantly increased reduced elastic modulus (57.5%), hardness (83.2%), and stiffness (17.6%) in the cartilage of arthritic rats [341]. It also showed the capability of preventing cartilage destruction in rats with induced-RA at 10 mg/kg by imbibing myeloperoxidase activity [342].
Regarding green tea, it was also reported that rats with induced RA that ingested 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. This is probably due to the capacity of green tea polyphenols to suppress the anti-Bhsp65 antibody response [343].
Another study showed that 150 mg/kg Tinospora cordifolia (Willd.) Hook. f. and Thomson 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 [317].
Furthermore, 150 mg/kg/bw Kalpaamruthaa, a modified indigenous Siddha preparation constituting Semecarpus anacardium nut milk extract, Emblica officinalis, and honey, also showed the potential to reduce oxidative stress, myeloperoxidase, and lipid peroxide and increase the activity of enzymic and non-enzymic antioxidants in arthritic rats [344].
In addition, 50, 100, and 200 mg/kg doses of Ribes orientale Desf. aqueous ethanolic extract (30:70) showed potential to reduce paw volume/diameter and PGE2, COX-2, IL-1β, IL-6, NF-κB, and TNF-α levels and enhance IL-4 and IL-10 contents in Sprague Dawley rats with induced RA [229]. Additionally, 80 mg/kg daily of chebulanin for 3 weeks significantly suppressed the progression and development of RA in a collagen-induced arthritis mouse model by decreasing the arthritis severity score, 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 [231].
Punicalagin at 50 mg/kg/d also showed the capacity to prevent the translocation of p-65 and avoid the phosphorylation of IkK and Ik Bα by modulating NF-κB pathway in arthritic rats. It also reduced TNFα, IL-6, CD86, CCR7, CD40, and MHC II expression, raised IL-10 expression, and suppressed dendritic cells migration, which, in turn, diminished the differentiation of Th1, Th17, and Th17/Th1-like and promoted the generation of Tregs via the regulation of dendritic cell maturation [288]. Syringaldehyde at 10, 25, and 50 mg/kg also showed the ability to alleviate paw and joint edema, reduce TNF-α and IL-6 levels, and increase the level of IL-10 in arthritic rat serum [289]. 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 [345]. Furthermore, 50 mg/kg Clitoria ternatea L. flower petals and 2.5 mg/kg of its major compound, which is quercetin-3ß-D-glucoside, showed the ability to reduce MPO activity and lower pro-inflammatory cytokines, chemokines, reactive oxygen and nitrogen species, and TNFR1, TLR2, iNOS, COX-2, and MMP-2 expression levels in rats with RA [346]. Similar effects were observed with 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, the rheumatoid factor, and contributed to positively modified radiographic and histopathological changes [347]. Furthermore, 250, 500 and 750 mg/kg Diospyros malabarica (Desr.) Kostel fruits also showed the capacity to raise anti-inflammatory enzymes and diminish the anti-inflammatory ones in RA rat model [348].
On the other hand, the daily administration of 100 mg/kg ρ-coumaric acid for 2 weeks showed remarkable capacity to suppress paw edema and body weight loss and curtail cartel [316], age, bone erosion, TNF-α, IL-1β, IL-6, IL-17, and MCP-1 quantities in serum and ankle joints 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 RANKL/OPG imbalance and inhibiting RANKL-induced NFATc-1 and c-Fos expression [238]. Similar data were reported by Pragasam and colleagues [349]. Focusing on chlorogenic acid, at 10 mg/kg, it can attenuate liposaccharide-induced bone loss based on micro-computed tomography and histologic analysis of femurs from arthritic rats [315].
Theaflavin-3,3′-digallate administrated at 10 mg/kg three times per week for 9 continuous weeks also showed the capability of reducing the expression of IL-1β, TNF-α, and IL-6, as well as MMP-1, MMP-2, and MMP-3 amounts in the synovium of a collagen-induced arthritis mouse model, chiefly by inhibiting the activation of NF-κB and the phosphorylation of P38, JNK2, and ERK [230]. 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; reduces inflammatory cell infiltration, cartilage damage in the joints, and IL-17, IL-6, and IL-1β; enhances TGF-β and IL-10 levels; reduces the frequency of Th17 cells; and enhances the frequency of Treg cells, namely by its potential to inhibit aryl hydrocarbon receptor expression and phospho-STAT3/RORγt [350]. 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 [351].
N-feruloylserotonin, a natural polyphenol that belongs to indole hydroxycinnamic acid amides, administered at 3 mg/kg/day orally reduced C-reactive protein in plasma and the activity of LOX in the liver rats. It also reduced 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 [352].
Moreover, 100 and 200 mg/kg extra virgin olive oil showed the capability to decrease inflammatory markers, joint edema, cell migration, cartilage degradation, and bone erosion in a 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 [353]. Furthermore, 0.05% hydroxytyrosol acetate, a polyphenol present in extra virgin olive oil, showed the ability to diminish IgG1 and IgG2a, COMP, MMP-3, TNF-α, IFN-γ, IL-1β, IL-6, IL-17A, and the MAPKs JAK/STAT and NF-κB pathways and enhance Nrf-2 and heme oxygenase-1 in mice with collagen-induced arthritis [354].
In addition, 50, 100, and 400 mg/kg mangiferin inhibits mRNA expression of cytokine genes in the 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 [355].
Sarcococca saligna plant at 250 mg/kg also showed ability to diminish IL-1β, IL-6, COX-2, prostaglandin E2, TNF-α and NF-κB levels, and the arthritic index and paw inflammation and enhance the expression of IL-4 and IL-10 [236]. On the other hand, 10 mg/kg curcumin showed similar effects as the methotrexate (0.5 mg/kg) positive control in reducing TNF-α and IL-1β in both the synovial fluid and blood serum of arthritic rats [356]. Aqueous extracts of Dichrostachys cinerea Wight et Arn. 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 [357]. In addition, 170–1350 mg/kg of the Circaea mollis Sieb. and Zucc. plant can also reduce paw and inflammatory swelling, the arthritis index, and TNF-α and IL-1β and increase the production of serum IL-10 in Freund’s complete adjuvant-induced arthritis model in rats [358]. Opuntia littoralis at 10 and 20 mg/100 g bw also revealed the potential to reduce joint inflammation, paw swelling, edema, MDA, and IL-1β, IL-6R, IL-6, IL-17, and IL-23 and ameliorated COX-2, NF-κB, STAT-3, PTEN, and RANKL expression, namely, by upregulating the expression of miR-28 and miR-199a [239]. Furthermore, 25 and 100 mg/kg Antrocaryon micraster A. Chev. and Guillaumin seed extract also showed capacity to diminish cachexia, paw edema, infiltration of inflammatory cells, pannus formation, and synovium damage in rats with induced RA [359]. Moreover, the addition of 20% dried plums into 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 [360]. On the other hand, 750 mg/kg Opuntia monacantha showed effectiveness in reducing paw edema, the arthritic score, the rheumatoid factor, inflammation, COX-2, IL-6, TNF-α, IL-1, NF-κB, bone erosion, and pannus formation, restoring hemoglobin, white blood count, and platelet parameters, increasing catalase and superoxide dismutase levels, and enhancing IL-4 and IL-10, namely, by its potential to inhibit glutaminase 1 activity in a RA rat model [361]. In addition, 800 mg/kg Solanum nigrum also showed effectiveness in reducing paw edem, and restoring body weight, hematologic parameters, and radiographic and histopathologic alterations towards normal in different RA rat models [362].
In addition, 15 mg/kg quercetin and 10 and 20 mg/kg quercetin-loaded chitosan showed the capacity to reduce TNF-α and IL-6, as it was observed that the nanoencapsulation of quercetin is an added value to improve its efficacy in rats with induced RA [363]. In another study, 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 [364]. Reduced cachexia and arthritic paw scores were also reported by Decendit [150], 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, the erythrocyte sedimentation rate, and C-reactive protein in rats [365].
Finally, the liposomal drug delivery system for morin, a dietary polyphenol, showed the capacity to ameliorate RA in rats, mainly by increasing its uptake by synovial and spleen macrophages, reducing mRNA expression and consequent production of TNF-α, IL-1β, IL-6, IL-17, RANKL, STAT-3, p-STAT-3, VEGF, iNOS, and NF-κB-p65, and increasing the expression of osteoprotegerin [235].

Clinical Trials

Regarding clinical trials, it was found 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 [366]. 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, the erythrocyte sedimentation rate, and related-pain [367]. On the other hand, pomegranate extract (250 mg for 8 weeks), in addition to alleviating swollen, pain intensity, and tender joints in patients with, can also diminish the erythrocyte sedimentation rate and morning stiffness and enhance glutathione peroxidase. No alterations were found in CRP, MDA, or MMP3 levels [368]. Finally, a 1 g daily dose of resveratrol for 3 months [369] also showed effectiveness in reducing joint swelling and tenderness, as well as TNF-α, IL-6, protein C-reactive, MMP-3, the erythrocyte sedimentation rate, and undercarboxylated osteocalcin in patients with RA of both genders [369].

6. Conclusions

In general, the present review provides evidence that supports the therapeutic effect of polyphenols. Polyphenols are largley present in nature and seem to be a promising approach to the boost immune system and contribute to a healthy status. Contrary to chemical pharmaceutics that originate several side effects and their loss of efficacy over time, it is largely belief that phenolics are more effective and a safer option. This is mainly due to their capacity to counteract oxidative stress and interact with inflammatory pathways. In this way, it is not surprising that their use as an adjuvant to conventional antirheumatic drugs is considered an added value. However, although phenolics are easy to obtain and economical, most of them present poor bioavailability and stability and rapid metabolism. Therefore, their encapsulation is beneficial to increase their efficacy. In addition, their safety profile and clinical trials need to be more explored.

Author Contributions

Conceptualization, A.C.G., S.R. and R.F.; methodology, A.C.G. and 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. and L.R.S.; investigation, A.C.G., S.R., R.F. and L.R.S.; resources, L.R.S.; data curation, A.C.G., S.R., R.F. and 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 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).

Conflicts of Interest

The authors declare no conflicts 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. Rheumatol. Int. 2021, 41, 863–877. [Google Scholar] [CrossRef]
  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]
  5. Silman, A.J.; Pearson, J.E. Epidemiology and genetics of rheumatoid arthritis. Arthritis Res. Ther. 2002, 4, S265. [Google Scholar] [CrossRef]
  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 Rheumatol. 2022, 74, 1172–1183. [Google Scholar] [CrossRef]
  7. Guo, Q.; Wang, Y.; Xu, D.; Nossent, J.; Pavlos, N.J.; Xu, J. Rheumatoid arthritis: Pathological mechanisms and modern pharmacologic therapies. Bone Res. 2018, 6, 15. [Google Scholar] [CrossRef]
  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 2004, 364, 665–674. [Google Scholar] [CrossRef] [PubMed]
  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. Inform. Med. Unlocked 2024, 45, 101464. [Google Scholar] [CrossRef]
  10. Kumar, P.; Banik, S. Pharmacotherapy Options in Rheumatoid Arthritis. Clin. Med. Insights Arthritis Musculoskelet. Disord. 2013, 6, CMAMD.S5558. [Google Scholar] [CrossRef]
  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. J. Cell. Physiol. 2019, 234, 10018–10031. [Google Scholar] [CrossRef] [PubMed]
  12. Donahue, K.E.; Gartlehner, G.; Jonas, D.E.; Lux, L.J.; Thieda, P.; Jonas, B.L.; Hansen, R.A.; Morgan, L.C.; Lohr, K.N. Systematic Review: Comparative Effectiveness and Harms of Disease-Modifying Medications for Rheumatoid Arthritis. Ann. Intern. Med. 2008, 148, 124–134. [Google Scholar] [CrossRef]
  13. Wang, W.; Zhou, H.; Liu, L. Side effects of methotrexate therapy for rheumatoid arthritis: A systematic review. Eur. J. Med. Chem. 2018, 158, 502–516. [Google Scholar] [CrossRef]
  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. Proc. Natl. Acad. Sci. USA 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. Br. J. Nutr. 2020, 124, 1179–1189. [Google Scholar] [CrossRef]
  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, 1071. [Google Scholar] [CrossRef]
  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 2021, 26, 3604. [Google Scholar] [CrossRef]
  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 Funct. 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. Clin. Nutr. 2023, 42, 1389–1398. [Google Scholar] [CrossRef]
  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. J. Food Biochem. 2022, 46, e14287. [Google Scholar] [CrossRef]
  22. Van de Velde, F.; Esposito, D.; Grace, M.H.; Pirovani, M.E.; Lila, M.A. Anti-inflammatory and wound healing properties of polyphenolic extracts from strawberry and blackberry fruits. Food Res. Int. 2019, 121, 453–462. [Google Scholar] [CrossRef] [PubMed]
  23. Zhao, H.; Lou, Z.; Chen, Y.; Cheng, J.; Wu, Y.; Li, B.; He, P.; Tu, Y.; Liu, J. Tea polyphenols (TPP) as a promising wound healing agent: TPP exerts multiple and distinct mechanisms at different phases of wound healing in a mouse model. Biomed. Pharmacother. 2023, 166, 115437. [Google Scholar] [CrossRef] [PubMed]
  24. 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. Int. J. Mol. Sci. 2022, 23, 4033. [Google Scholar] [CrossRef]
  25. 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 2021, 26, 2824. [Google Scholar] [CrossRef]
  26. Nunes, C.; Almeida, L.; Barbosa, R.M.; Laranjinha, J. Luteolin suppresses the JAK/STAT pathway in a cellular model of intestinal inflammation. Food Funct. 2017, 8, 387–396. [Google Scholar] [CrossRef] [PubMed]
  27. 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? Biochem. Pharmacol. 2022, 197, 114929. [Google Scholar] [CrossRef] [PubMed]
  28. 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] [PubMed]
  29. Zhao, L.; Wu, Q.; Long, Y.; Qu, Q.; Qi, F.; Liu, L.; Zhang, L.; Ai, K. microRNAs: Critical targets for treating rheumatoid arthritis angiogenesis. J. Drug Target. 2024, 32, 1–20. [Google Scholar] [CrossRef]
  30. 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, 268. [Google Scholar] [CrossRef]
  31. Sridhar, A.; Ponnuchamy, M.; Kumar, P.S.; Kapoor, A.; Vo, D.N.; Prabhakar, S. Techniques and modeling of polyphenol extraction from food: A review. Environ. Chem. Lett. 2021, 19, 3409–3443. [Google Scholar] [CrossRef]
  32. Câmara, J.S.; Albuquerque, B.R.; Aguiar, J.; Corrêa, R.C.G.; Gonçalves, J.L.; Granato, D.; Pereira, J.A.M.; Barros, L.; Ferreira, I.C.F.R. Food Bioactive Compounds and Emerging Techniques for Their Extraction: Polyphenols as a Case Study. Foods 2021, 10, 37. [Google Scholar] [CrossRef] [PubMed]
  33. Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef]
  34. Hollman, P.C.H.; Cassidy, A.; Comte, B.; Heinonen, M.; Richelle, M.; Richling, E.; Serafini, M.; Scalbert, A.; Sies, H.; Vidry, S. The Biological Relevance of Direct Antioxidant Effects of Polyphenols for Cardiovascular Health in Humans Is Not Established1–4. J. Nutr. 2011, 141, 989S–1009S. [Google Scholar] [CrossRef] [PubMed]
  35. Zhong, S.; Sandhu, A.; Edirisinghe, I.; Burton-Freeman, B. Characterization of Wild Blueberry Polyphenols Bioavailability and Kinetic Profile in Plasma over 24-h Period in Human Subjects. Mol. Nutr. Food Res. 2017, 61, 1700405. [Google Scholar] [CrossRef]
  36. Serra, D.; Henriques, J.F.; Serra, T.; Bento Silva, A.; Bronze, M.R.; Dinis, T.C.P.; Almeida, L.M. An Anthocyanin-Rich Extract Obtained from Portuguese Blueberries Maintains Its Efficacy in Reducing Microglia-Driven Neuroinflammation after Simulated Digestion. Nutrients 2020, 12, 3670. [Google Scholar] [CrossRef]
  37. Martini, S.; Conte, A.; Tagliazucchi, D. Bioactivity and cell metabolism of in vitro digested sweet cherry (Prunus avium) phenolic compounds. Int. J. Food Sci. Nutr. 2019, 70, 335–348. [Google Scholar] [CrossRef]
  38. Tabasco, R.; Sánchez-Patán, F.; Monagas, M.; Bartolomé, B.; Victoria Moreno-Arribas, M.; Peláez, C.; Requena, T. Effect of grape polyphenols on lactic acid bacteria and bifidobacteria growth: Resistance and metabolism. Food Microbiol. 2011, 28, 1345–1352. [Google Scholar] [CrossRef]
  39. D’Archivio, M.; Filesi, C.; Varì, R.; Scazzocchio, B.; Masella, R. Bioavailability of the polyphenols: Status and controversies. Int. J. Mol. Sci. 2010, 11, 1321–1342. [Google Scholar] [CrossRef]
  40. Olivero-David, R.; Ruiz-Roso, M.B.; Caporaso, N.; Perez-Olleros, L.; De las Heras, N.; Lahera, V.; Ruiz-Roso, B. In vivo bioavailability of polyphenols from grape by-product extracts, and effect on lipemia of normocholesterolemic Wistar rats. J. Sci. Food Agric. 2018, 98, 5581–5590. [Google Scholar] [CrossRef] [PubMed]
  41. Ray, S.K.; Mukherjee, S. Evolving Interplay Between Dietary Polyphenols and Gut Microbiota—An Emerging Importance in Healthcare. Front. Nutr. 2021, 8, 634944. [Google Scholar] [CrossRef] [PubMed]
  42. Coelho, M.C.; Ribeiro, T.B.; Oliveira, C.; Batista, P.; Castro, P.; Monforte, A.R.; Rodrigues, A.S.; Teixeira, J.; Pintado, M. In Vitro Gastrointestinal Digestion Impact on the Bioaccessibility and Antioxidant Capacity of Bioactive Compounds from Tomato Flours Obtained after Conventional and Ohmic Heating Extraction. Foods 2021, 10, 554. [Google Scholar] [CrossRef] [PubMed]
  43. Zhang, W.; Qi, S.; Xue, X.; Al Naggar, Y.; Wu, L.; Wang, K. Understanding the Gastrointestinal Protective Effects of Polyphenols using Foodomics-Based Approaches. Front. Immunol. 2021, 12, 671150. [Google Scholar] [CrossRef]
  44. Fazzari, M.; Fukumoto, L.; Mazza, G.; Livrea, M.A.; Tesoriere, L.; Marco, L.D. In Vitro Bioavailability of Phenolic Compounds from Five Cultivars of Frozen Sweet Cherries (Prunus avium L.). J. Agric. Food Chem. 2008, 56, 3561–3568. [Google Scholar] [CrossRef]
  45. Rocchetti, G.; Bhumireddy, S.R.; Giuberti, G.; Mandal, R.; Lucini, L.; Wishart, D.S. Edible nuts deliver polyphenols and their transformation products to the large intestine: An in vitro fermentation model combining targeted/untargeted metabolomics. Food Res. Int. 2019, 116, 786–794. [Google Scholar] [CrossRef]
  46. Guerreiro, Í.; Ferreira-Pêgo, C.; Carregosa, D.; Santos, C.N.; Menezes, R.; Fernandes, A.S.; Costa, J.G. Polyphenols and Their Metabolites in Renal Diseases: An Overview. Foods 2022, 11, 1060. [Google Scholar] [CrossRef] [PubMed]
  47. Wojtunik-Kulesza, K.; Oniszczuk, A.; Oniszczuk, T.; Combrzyński, M.; Nowakowska, D.; Matwijczuk, A. Influence of In Vitro Digestion on Composition, Bioaccessibility and Antioxidant Activity of Food Polyphenols—A Non-Systematic Review. Nutrients 2020, 12, 1401. [Google Scholar] [CrossRef] [PubMed]
  48. Polia, F.; Pastor-Belda, M.; Martínez-Blázquez, A.; Horcajada, M.N.; Tomás-Barberán, F.A.; García-Villalba, R. Technological and Biotechnological Processes to Enhance the Bioavailability of Dietary (Poly)phenols in Humans. J. Agric. Food Chem. 2022, 70, 2092–2107. [Google Scholar] [CrossRef]
  49. Trindade, L.R.; da Silva, D.V.T.; Baião, D.D.S.; Paschoalin, V.M.F. Increasing the Power of Polyphenols through Nanoencapsulation for Adjuvant Therapy against Cardiovascular Diseases. Molecules 2021, 26, 4621. [Google Scholar] [CrossRef]
  50. Hamad, H.A.M. Phenolic Compounds: Classification, Chemistry, and Updated Techniques of Analysis and Synthesis. In Phenolic Compounds; Farid, A.B., Ed.; IntechOpen: Rijeka, Croatia, 2021; Chapter 4. [Google Scholar]
  51. 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. Int. J. Mol. Sci. 2022, 23, 4652. [Google Scholar] [CrossRef]
  52. 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]
  53. Carregosa, D.; Carecho, R.; Figueira, I.; N Santos, C. Low-Molecular Weight Metabolites from Polyphenols as Effectors for Attenuating Neuroinflammation. J. Agric. Food Chem. 2020, 68, 1790–1807. [Google Scholar] [CrossRef] [PubMed]
  54. Capanoglu, E.; Chen, F. The interaction between food components and gut microbiota. Food Chem. 2024, 442, 138117. [Google Scholar] [CrossRef] [PubMed]
  55. Soobrattee, M.A.; Neergheen, V.S.; Luximon-Ramma, A.; Aruoma, O.I.; Bahorun, T. Phenolics as potential antioxidant therapeutic agents: Mechanism and actions. Mutat. Res./Fundam. Mol. Mech. Mutagen. 2005, 579, 200–213. [Google Scholar] [CrossRef]
  56. Charlton, N.C.; Mastyugin, M.; Török, B.; Török, M. Structural Features of Small Molecule Antioxidants and Strategic Modifications to Improve Potential Bioactivity. Molecules 2023, 28, 1057. [Google Scholar] [CrossRef] [PubMed]
  57. Wu, H.; Bak, K.H.; Goran, G.V.; Tatiyaborworntham, N. Inhibitory mechanisms of polyphenols on heme protein-mediated lipid oxidation in muscle food: New insights and advances. Crit. Rev. Food Sci. Nutr. 2022, 1–19. [Google Scholar] [CrossRef]
  58. Moazzen, A.; Öztinen, N.; Ak-Sakalli, E.; Koşar, M. Structure-antiradical activity relationships of 25 natural antioxidant phenolic compounds from different classes. Heliyon 2022, 8, e10467. [Google Scholar] [CrossRef]
  59. Lemańska, K.; van der Woude, H.; Szymusiak, H.; Boersma, M.G.; Gliszczyńska-Świgło, A.; Rietjens, I.M.C.M.; Tyrakowska, B. The Effect of Catechol O-methylation on Radical Scavenging Characteristics of Quercetin and Luteolin—A Mechanistic Insight. Free Radic. Res. 2004, 38, 639–647. [Google Scholar] [CrossRef]
  60. Chukwumah, Y.; Walker, L.T.; Verghese, M. Peanut skin color: A biomarker for total polyphenolic content and antioxidative capacities of peanut cultivars. Int. J. Mol. Sci. 2009, 10, 4941–4952. [Google Scholar] [CrossRef]
  61. 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]
  62. Sobolev, V.S.; Cole, R.J. trans-Resveratrol Content in Commercial Peanuts and Peanut Products. J. Agric. Food Chem. 1999, 47, 1435–1439. [Google Scholar] [CrossRef]
  63. 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). J. Food Process. Preserv. 2015, 39, 1682–1691. [Google Scholar] [CrossRef]
  64. De Silva, S.F.; Alcorn, J. Flaxseed Lignans as Important Dietary Polyphenols for Cancer Prevention and Treatment: Chemistry, Pharmacokinetics, and Molecular Targets. Pharmaceuticals 2019, 12, 68. [Google Scholar] [CrossRef] [PubMed]
  65. 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 2023, 28, 1807. [Google Scholar] [CrossRef]
  66. Oomah, B.D.; Kenaschuk, E.O.; Mazza, G. Phenolic Acids in Flaxseed. J. Agric. Food Chem. 1995, 43, 2016–2019. [Google Scholar] [CrossRef]
  67. Herrmann, K.; Nagel, C.W. Occurrence and content of hydroxycinnamic and hydroxybenzoic acid compounds in foods. Crit. Rev. Food Sci. Nutr. 1989, 28, 315–347. [Google Scholar] [CrossRef]
  68. Ramli, N.; Yatim, A.M.; Said, M.; Hok, H.C. HPLC determination of methylxanthines and polyphenols levels In cocoa and chocolate products. Malays. J. Anal. Sci. 2001, 7, 377–386. [Google Scholar]
  69. Sova, M.; Saso, L. Natural Sources, Pharmacokinetics, Biological Activities and Health Benefits of Hydroxycinnamic Acids and Their Metabolites. Nutrients 2020, 12, 2190. [Google Scholar] [CrossRef] [PubMed]
  70. Inglett, G.E.; Chen, D.; Liu, S.X. Antioxidant Activities of Selective Gluten Free Ancient Grains. Food Nutr. Sci. 2015, 6, 67065. [Google Scholar] [CrossRef]
  71. 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]
  72. 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. Jt. Bone Spine 2013, 80, 274–279. [Google Scholar] [CrossRef]
  73. 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]
  74. 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. Curr. Issues Mol. Biol. 2024, 46, 81–95. [Google Scholar] [CrossRef]
  75. 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]
  76. Picariello, G.; Ferranti, P.; De Cunzo, F.; Sacco, E.; Volpe, M.G. Polyphenol patterns to trace sweet (Prunus avium) and tart (Prunus cerasus) varieties in cherry jam. J. Food Sci. Technol. 2017, 54, 2316–2323. [Google Scholar] [CrossRef] [PubMed]
  77. 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. Microbiol. Res. 2024, 278, 127544. [Google Scholar] [CrossRef]
  78. 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. J. Food Sci. Technol. 2019, 56, 4177–4188. [Google Scholar] [CrossRef] [PubMed]
  79. 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. J. Food Sci. 2022, 87, 450–465. [Google Scholar] [CrossRef] [PubMed]
  80. Nowicka, P.; Wojdyło, A.; Samoticha, J. Evaluation of phytochemicals, antioxidant capacity, and antidiabetic activity of novel smoothies from selected Prunus fruits. J. Funct. Foods 2016, 25, 397–407. [Google Scholar] [CrossRef]
  81. 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]
  82. 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]
  83. Młynarczyk, K.; Walkowiak-Tomczak, D.; Łysiak, G.P. Bioactive properties of Sambucus nigra L. as a functional ingredient for food and pharmaceutical industry. J. Funct. Foods 2018, 40, 377–390. [Google Scholar] [CrossRef] [PubMed]
  84. 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, 2902. [Google Scholar] [CrossRef]
  85. 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. Appl. Sci. 2022, 12, 4004. [Google Scholar] [CrossRef]
  86. 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. Int. J. Food Prop. 2011, 14, 59–67. [Google Scholar] [CrossRef]
  87. Sondheimer, E.; Karash, C.B. The Major Anthocyanin Pigments of the Wild Strawberry (Fragaria vesca). Nature 1956, 178, 648–649. [Google Scholar] [CrossRef]
  88. 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, 1072. [Google Scholar] [CrossRef]
  89. Orak, H.H. Total antioxidant activities, phenolics, anthocyanins, polyphenoloxidase activities of selected red grape cultivars and their correlations. Sci. Hortic. 2007, 111, 235–241. [Google Scholar] [CrossRef]
  90. 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]
  91. Negro, C.; Aprile, A.; De Bellis, L.; Miceli, A. Nutraceutical Properties of Mulberries Grown in Southern Italy (Apulia). Antioxidants 2019, 8, 223. [Google Scholar] [CrossRef]
  92. Denev, P.; Kratchanova, M.; Petrova, I.; Klisurova, D.; Georgiev, Y.; Ognyanov, M.; Yanakieva, I. Black Chokeberry (Aronia melanocarpa (Michx.) Elliot) Fruits and Functional Drinks Differ Significantly in Their Chemical Composition and Antioxidant Activity. J. Chem. 2018, 2018, 9574587. [Google Scholar] [CrossRef]
  93. 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. Int. J. Mol. Sci. 2021, 22, 6541. [Google Scholar] [CrossRef] [PubMed]
  94. 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, 1745. [Google Scholar] [CrossRef] [PubMed]
  95. 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]
  96. Wiczkowski, W.; Szawara-Nowak, D.; Topolska, J. Red cabbage anthocyanins: Profile, isolation, identification, and antioxidant activity. Food Res. Int. 2013, 51, 303–309. [Google Scholar] [CrossRef]
  97. 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]
  98. Mujić, I.; Šertović, E.; Jokić, S.; Sarić, Z.; Alibabić, V.; Vidovic, S.S.; Zivkovic, J.V. Isoflavone content and antioxidant properties of soybean seeds. Croat. J. Food Sci. Technol. 2011, 3, 16–20. [Google Scholar]
  99. 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. J. Pharmacogn. Phytochem. 2020, 9, 1192–1198. [Google Scholar]
  100. 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. J. Food Sci. Technol. 2014, 51, 2251–2255. [Google Scholar] [CrossRef]
  101. Stanic, G.; Jurisic, B.; Brkic, D. HPLC Analysis of Esculin and Fraxin in Horse-Chestnut Bark (Aesculus hippocastanum L.). Croat. Chem. Acta 1999, 72, 827–834. [Google Scholar]
  102. Matysik, G.; Glowniak, K.; Soczewiński, E.; Garbacka, M. Chromatography of esculin from stems and bark of Aesculus hippocastanum L. for consecutive vegetative periods. Chromatographia 1994, 38, 766–770. [Google Scholar] [CrossRef]
  103. 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. J. Funct. Foods 2018, 43, 224–233. [Google Scholar] [CrossRef]
  104. 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. J. Agric. Food Chem. 2000, 48, 1752–1757. [Google Scholar] [CrossRef] [PubMed]
  105. 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]
  106. Dragovic-Uzelac, V.; Levaj, B.; Mrkic, V.; Bursac, D.; Boras, M. The content of polyphenols and carotenoids in three apricot cultivars depending on stage of maturity and geographical region. Food Chem. 2007, 102, 966–975. [Google Scholar] [CrossRef]
  107. Mokrani, A.; Krisa, S.; Cluzet, S.; Da Costa, G.; Temsamani, H.; Renouf, E.; Mérillon, J.-M.; Madani, K.; Mesnil, M.; Monvoisin, A.; et al. Phenolic contents and bioactive potential of peach fruit extracts. Food Chem. 2016, 202, 212–220. [Google Scholar] [CrossRef]
  108. Shafi, W.; Mansoor, S.; Jan, S.; Singh, D.B.; Kazi, M.; Raish, M.; Alwadei, M.; Mir, J.I.; Ahmad, P. Variability in Catechin and Rutin Contents and Their Antioxidant Potential in Diverse Apple Genotypes. Molecules 2019, 24, 943. [Google Scholar] [CrossRef]
  109. Podsędek, A.; Wilska-Jeszka, J.; Anders, B.; Markowski, J. Compositional characterisation of some apple varieties. Eur. Food Res. Technol. 2000, 210, 268–272. [Google Scholar] [CrossRef]
  110. Tsanova-Savova, S.; Ribarova, F.; Gerova, M. (+)-Catechin and (−)-epicatechin in Bulgarian fruits. J. Food Compos. Anal. 2005, 18, 691–698. [Google Scholar] [CrossRef]
  111. Jówko, E. Antioxidants in Sport Nutrition. In Green Tea Catechins and Sport Performance; Lamprecht, M., Ed.; CRC Press/Taylor & Francis: Boca Raton, FL, USA, 2015. [Google Scholar]
  112. 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 2021, 26, 6052. [Google Scholar] [CrossRef]
  113. Kris-Etherton, P.M.; Keen, C.L. Evidence that the antioxidant flavonoids in tea and cocoa are beneficial for cardiovascular health. Curr. Opin. Lipidol. 2002, 13, 41–49. [Google Scholar] [CrossRef] [PubMed]
  114. Wang, S.; Moustaid-Moussa, N.; Chen, L.; Mo, H.; Shastri, A.; Su, R.; Bapat, P.; Kwun, I.; Shen, C.L. Novel insights of dietary polyphenols and obesity. J. Nutr. Biochem. 2014, 25, 1–18. [Google Scholar] [CrossRef]
  115. Balasundram, N.; Sundram, K.; Samman, S. Phenolic compounds in plants and agri-industrial by-products: Antioxidant activity, occurrence, and potential uses. Food Chem. 2006, 99, 191–203. [Google Scholar] [CrossRef]
  116. 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]
  117. Jin, S.; Zhang, L.; Wang, L. Kaempferol, a potential neuroprotective agent in neurodegenerative diseases: From chemistry to medicine. Biomed. Pharmacother. 2023, 165, 115215. [Google Scholar] [CrossRef]
  118. Fieschi, M.; Codignola, A.; Mosca, A.M.L. Mutagenic Flavonol Aglycones in Infusions and in Fresh and Pickled Vegetables. J. Food Sci. 1989, 54, 1492–1495. [Google Scholar] [CrossRef]
  119. 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]
  120. 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. S. Afr. J. Bot. 2022, 146, 196–204. [Google Scholar] [CrossRef]
  121. Alharbi, N.K.; Naghmouchi, S.; Al-Zaban, M. Evaluation of Antimicrobial Potential and Comparison of HPLC Composition, Secondary Metabolites Count, and Antioxidant Activity of Mentha rotundifolia and Mentha pulegium Extracts. Evid.-Based Complement. Altern. Med. eCAM 2021, 2021, 9081536. [Google Scholar] [CrossRef]
  122. Dabeek, W.M.; Marra, M.V. Dietary Quercetin and Kaempferol: Bioavailability and Potential Cardiovascular-Related Bioactivity in Humans. Nutrients 2019, 11, 2288. [Google Scholar] [CrossRef] [PubMed]
  123. 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 Sci. Nutr. 2021, 9, 5854–5868. [Google Scholar] [CrossRef] [PubMed]
  124. Kimira, M.; Arai, Y.; Shimoi, K.; Watanabe, S. Japanese intake of flavonoids and isoflavonoids from foods. J. Epidemiol. 1998, 8, 168–175. [Google Scholar] [CrossRef]
  125. 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. J. Nutr. 2010, 140, 1615–1620. [Google Scholar] [CrossRef]
  126. Rehman, M.F.u.; Batool, A.I.; Qadir, R.; Aslam, M. Chapter 18—Hesperidin and naringenin. In A Centum of Valuable Plant Bioactives; Mushtaq, M., Anwar, F., Eds.; Academic Press: Cambridge, MA, USA, 2021; pp. 403–444. [Google Scholar]
  127. Cannataro, R.; Fazio, A.; La Torre, C.; Caroleo, M.C.; Cione, E. Polyphenols in the Mediterranean Diet: From Dietary Sources to microRNA Modulation. Antioxidants 2021, 10, 328. [Google Scholar] [CrossRef]
  128. 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. J. Pharm. Biomed. Anal. 2023, 234, 115570. [Google Scholar] [CrossRef] [PubMed]
  129. García-Nicolás, M.; Ledesma-Escobar, C.A.; Priego-Capote, F. Spatial Distribution and Antioxidant Activity of Extracts from Citrus Fruits. Antioxidants 2023, 12, 781. [Google Scholar] [CrossRef] [PubMed]
  130. Mazzaferro, L.S.; Breccia, J.D. Quantification of hesperidin in citrus-based foods using a fungal diglycosidase. Food Chem. 2012, 134, 2338–2344. [Google Scholar] [CrossRef]
  131. 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]
  132. Jasicka-Misiak, I.; Gruyaert, S.; Poliwoda, A.; Kafarski, P. Chemical Profiling of Polyfloral Belgian Honey: Ellagic Acid and Pinocembrin as Antioxidants and Chemical Markers. J. Chem. 2017, 2017, 5393158. [Google Scholar] [CrossRef]
  133. Nayak, G.; Sahu, A.; Bhuyan, S.K.; Bhuyan, R.; Kar, D.; Kuanar, A. A comparative study on antioxidant activity of propolis ethanolic extract and oil from different agroclimatic regions of Eastern India. Biocatal. Agric. Biotechnol. 2023, 50, 102685. [Google Scholar] [CrossRef]
  134. 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 Chem. 2020, 324, 126854. [Google Scholar] [CrossRef] [PubMed]
  135. Landete, J.M. Ellagitannins, ellagic acid and their derived metabolites: A review about source, metabolism, functions and health. Food Res. Int. 2011, 44, 1150–1160. [Google Scholar] [CrossRef]
  136. Lu, J.; Wei, Y.; Yuan, Q. Preparative separation of punicalagin from pomegranate husk by high-speed countercurrent chromatography. J. Chromatogr. B 2007, 857, 175–179. [Google Scholar] [CrossRef]
  137. Chao, I.C.; Wang, C.M.; Li, S.P.; Lin, L.G.; Ye, W.C.; Zhang, Q.W. Simultaneous Quantification of Three Curcuminoids and Three Volatile Components of Curcuma longa Using Pressurized Liquid Extraction and High-Performance Liquid Chromatography. Molecules 2018, 23, 1568. [Google Scholar] [CrossRef] [PubMed]
  138. Guofang, X.; Xiaoyan, X.; Xiaoli, Z.; Yongling, L.; Zhibing, Z. Changes in phenolic profiles and antioxidant activity in rabbiteye blueberries during ripening. Int. J. Food Prop. 2019, 22, 320–329. [Google Scholar] [CrossRef]
  139. Popova, I.E.; Hall, C.; Kubátová, A. Determination of lignans in flaxseed using liquid chromatography with time-of-flight mass spectrometry. J. Chromatogr. A 2009, 1216, 217–229. [Google Scholar] [CrossRef] [PubMed]
  140. Reinisalo, M.; Kårlund, A.; Koskela, A.; Kaarniranta, K.; Karjalainen, R.O. Polyphenol Stilbenes: Molecular Mechanisms of Defence against Oxidative Stress and Aging-Related Diseases. Oxid. Med. Cell. Longev. 2015, 2015, 340520. [Google Scholar] [CrossRef]
  141. 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]
  142. Usenik, V.; Štampar, F.; Veberič, R. Anthocyanins and fruit colour in plums (Prunus domestica L.) during ripening. Food Chem. 2009, 114, 529–534. [Google Scholar] [CrossRef]
  143. 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]
  144. 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 Chem. 2018, 262, 184–190. [Google Scholar] [CrossRef] [PubMed]
  145. 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 2020, 25, 4587. [Google Scholar] [CrossRef] [PubMed]
  146. Santoni, A.; Amanda, H.; Darwis, D. Characterization of pelargonidin compound from raspberry fruit (Rubus rosifolius Sm) with mass spectroscopy method. J. Chem. Pharm. Res. 2015, 7, 804–808. [Google Scholar]
  147. 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 Res. Int. 2016, 2016, 9070537. [Google Scholar] [CrossRef]
  148. 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. Pharm. Biol. 2022, 60, 247–254. [Google Scholar] [CrossRef]
  149. 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. J. Food Compos. Anal. 2008, 21, 291–299. [Google Scholar] [CrossRef]
  150. 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]
  151. Silva, L.R.; Queiroz, M. Bioactive compounds of red grapes from Dão region (Portugal): Evaluation of phenolic and organic profile. Asian Pac. J. Trop. Biomed. 2016, 6, 315–321. [Google Scholar] [CrossRef]
  152. 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]
  153. 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, 645. [Google Scholar] [CrossRef]
  154. Wang, H.; Provan, G.J.; Helliwell, K. HPLC determination of catechins in tea leaves and tea extracts using relative response factors. Food Chem. 2003, 81, 307–312. [Google Scholar] [CrossRef]
  155. 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. Chem.-Biol. Interact. 2009, 180, 10–19. [Google Scholar] [CrossRef] [PubMed]
  156. 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 Chem. 2024, 433, 137321. [Google Scholar] [CrossRef]
  157. Alam, W.; Khan, H.; Shah, M.A.; Cauli, O.; Saso, L. Kaempferol as a Dietary Anti-Inflammatory Agent: Current Therapeutic Standing. Molecules 2020, 25, 4073. [Google Scholar] [CrossRef] [PubMed]
  158. Khan, N.; Syed, D.N.; Ahmad, N.; Mukhtar, H. Fisetin: A dietary antioxidant for health promotion. Antioxid. Redox Signal. 2013, 19, 151–162. [Google Scholar] [CrossRef] [PubMed]
  159. 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. J. Pharm. Biomed. Anal. 2003, 33, 243–249. [Google Scholar] [CrossRef]
  160. 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. Iran. J. Basic Med. Sci. 2020, 23, 140–153. [Google Scholar] [CrossRef] [PubMed]
  161. Mödinger, Y.; Schön, C.; Wilhelm, M.; Pickel, C.; Grothe, T. A Food Supplement with Antioxidative Santa Herba Extract Modulates Energy Metabolism and Contributes to Weight Management. J. Med. Food 2021, 24, 1235–1242. [Google Scholar] [CrossRef] [PubMed]
  162. Chun, O.K.; Chung, S.J.; Song, W.O. Estimated Dietary Flavonoid Intake and Major Food Sources of U.S. Adults. J. Nutr. 2007, 137, 1244–1252. [Google Scholar] [CrossRef] [PubMed]
  163. Bogdanov, S. Determination of Pinocembrin in Honey Using HPLC. J. Apic. Res. 1989, 28, 55–57. [Google Scholar] [CrossRef]
  164. 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 2014, 19, 9101–9113. [Google Scholar] [CrossRef]
  165. 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. Int. J. Pharm. Sci. Rev. Res. 2015, 33, 220–224. [Google Scholar]
  166. Volpi, N.; Bergonzini, G. Analysis of flavonoids from propolis by on-line HPLC–electrospray mass spectrometry. J. Pharm. Biomed. Anal. 2006, 42, 354–361. [Google Scholar] [CrossRef] [PubMed]
  167. Fukutake, M.; Takahashi, M.; Ishida, K.; Kawamura, H.; Sugimura, T.; Wakabayashi, K. Quantification of genistein and genistin in soybeans and soybean products. Food Chem. Toxicol. 1996, 34, 457–461. [Google Scholar] [CrossRef]
  168. 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. J. Chromatogr. A 2003, 986, 169–177. [Google Scholar] [CrossRef] [PubMed]
  169. Oshima, A.; Mine, W.; Nakada, M.; Yanase, E. Analysis of isoflavones and coumestrol in soybean sprouts. Biosci. Biotechnol. Biochem. 2016, 80, 2077–2079. [Google Scholar] [CrossRef] [PubMed]
  170. 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. Proc. Soc. Exp. Biol. Med. 1998, 217, 263–273. [Google Scholar] [CrossRef]
  171. Tepavčević, V.; Atanacković, M.; Miladinović, J.; Malenčić, D.; Popović, J.; Cvejić, J. Isoflavone Composition, Total Polyphenolic Content, and Antioxidant Activity in Soybeans of Different Origin. J. Med. Food 2010, 13, 657–664. [Google Scholar] [CrossRef]
  172. 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 Chem. 2021, 360, 129999. [Google Scholar] [CrossRef]
  173. Kumar, N.; Goel, N. Phenolic acids: Natural versatile molecules with promising therapeutic applications. Biotechnol. Rep. 2019, 24, e00370. [Google Scholar] [CrossRef]
  174. Debelo, H.; Li, M.; Ferruzzi, M.G. Processing influences on food polyphenol profiles and biological activity. Curr. Opin. Food Sci. 2020, 32, 90–102. [Google Scholar] [CrossRef]
  175. 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. Prev. Nutr. Food Sci. 2022, 27, 282–298. [Google Scholar] [CrossRef]
  176. Nurzyńska-Wierdak, R. Phenolic Compounds from New Natural Sources-Plant Genotype and Ontogenetic Variation. Molecules 2023, 28, 1731. [Google Scholar] [CrossRef] [PubMed]
  177. 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]
  178. Weyand, C.M.; Goronzy, J.J. The immunology of rheumatoid arthritis. Nat. Immunol. 2021, 22, 10–18. [Google Scholar] [CrossRef] [PubMed]
  179. 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. Ann. Rheum. Dis. 2011, 70, 844. [Google Scholar] [CrossRef]
  180. 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 Res. Int. 2017, 95, 91–100. [Google Scholar] [CrossRef]
  181. 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]
  182. 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 Chem. Toxicol. 2019, 133, 110758. [Google Scholar] [CrossRef]
  183. 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). Mol. Divers. 2023. [Google Scholar] [CrossRef]
  184. 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. Nat. Prod. Commun. 2022, 17, 1934578X221139409. [Google Scholar] [CrossRef]
  185. 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 2021, 10, 811. [Google Scholar] [CrossRef]
  186. Rice-Evans, C.A.; Miller, N.J.; Paganga, G. Structure-antioxidant activity relationships of flavonoids and phenolic acids. Free Radic. Biol. Med. 1996, 20, 933–956. [Google Scholar] [CrossRef] [PubMed]
  187. 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 2021, 26, 1244. [Google Scholar] [CrossRef] [PubMed]
  188. Zhu, F.; Yuan, Z.; Zhao, X.; Yin, Y.; Feng, L. Composition and Contents of Anthocyanins in Different Pomegranate Cultivars. Acta Hortic. 2015, 1089, 35–41. [Google Scholar] [CrossRef]
  189. 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 J. 2019, 16, 9–14. [Google Scholar] [CrossRef]
  190. Heim, K.E.; Tagliaferro, A.R.; Bobilya, D.J. Flavonoid antioxidants: Chemistry, metabolism and structure-activity relationships. J. Nutr. Biochem. 2002, 13, 572–584. [Google Scholar] [CrossRef]
  191. 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. Org. Biomol. Chem. 2005, 3, 3336–3347. [Google Scholar] [CrossRef] [PubMed]
  192. Rühlmann, A.; Antovic, D.; Müller, T.J.J.; Urlacher, V.B. Regioselective Hydroxylation of Stilbenes by Engineered Cytochrome P450 from Thermobifida fusca YX. Adv. Synth. Catal. 2017, 359, 984–994. [Google Scholar] [CrossRef]
  193. 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]
  194. Simić, A.; Manojlović, D.; Segan, D.; Todorović, M. Electrochemical behavior and antioxidant and prooxidant activity of natural phenolics. Molecules 2007, 12, 2327–2340. [Google Scholar] [CrossRef] [PubMed]
  195. Alov, P.; Tsakovska, I.; Pajeva, I. Computational studies of free radical-scavenging properties of phenolic compounds. Curr. Top. Med. Chem. 2015, 15, 85–104. [Google Scholar] [CrossRef]
  196. Bayliak, M.M.; Burdyliuk, N.I.; Lushchak, V.I. Effects of pH on antioxidant and prooxidant properties of common medicinal herbs. Open Life Sci. 2016, 11, 298–307. [Google Scholar] [CrossRef]
  197. Cao, G.; Sofic, E.; Prior, R.L. Antioxidant and Prooxidant Behavior of Flavonoids: Structure-Activity Relationships. Free Radic. Biol. Med. 1997, 22, 749–760. [Google Scholar] [CrossRef] [PubMed]
  198. 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 Radic. Biol. Med. 1997, 22, 861–870. [Google Scholar] [CrossRef]
  199. 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. Arab. J. Chem. 2023, 16, 104414. [Google Scholar] [CrossRef]
  200. Guan, T.; Bian, C.; Ma, Z. In vitro and in silico perspectives on the activation of antioxidant responsive element by citrus-derived flavonoids. Front. Nutr. 2023, 10, 1257172. [Google Scholar] [CrossRef] [PubMed]
  201. 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. Int. Immunopharmacol. 2016, 39, 128–139. [Google Scholar] [CrossRef] [PubMed]
  202. 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. Toxicol. Appl. Pharmacol. 2012, 261, 181–188. [Google Scholar] [CrossRef]
  203. 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. Pharm. Biol. 2020, 58, 999–1005. [Google Scholar] [CrossRef]
  204. 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. Sci. Rep. 2019, 9, 7408. [Google Scholar] [CrossRef]
  205. 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]
  206. 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]
  207. 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 2020, 26, 6. [Google Scholar] [CrossRef]
  208. 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. Appl. Sci. 2023, 13, 6348. [Google Scholar] [CrossRef]
  209. 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. Arch. Biochem. Biophys. 2017, 635, 66–73. [Google Scholar] [CrossRef] [PubMed]
  210. 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]
  211. 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 2022, 96, 153807. [Google Scholar] [CrossRef]
  212. 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. Sci. Rep. 2019, 9, 808. [Google Scholar] [CrossRef]
  213. 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 2020, 25, 1746. [Google Scholar] [CrossRef]
  214. 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. Chem.-Biol. Interact. 2013, 206, 100–108. [Google Scholar] [CrossRef]
  215. 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. Mol. Cells 2015, 38, 396–401. [Google Scholar] [CrossRef]
  216. Lee, Y.J.; Im, J.H.; Lee, D.M.; Park, J.S.; Won, S.Y.; Cho, M.K.; Nam, H.S.; Lee, Y.J.; Lee, S.H. Synergistic inhibition of mesothelioma cell growth by the combination of clofarabine and resveratrol involves Nrf2 downregulation. BMB Rep. 2012, 45, 647–652. [Google Scholar] [CrossRef] [PubMed]
  217. 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]
  218. 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. Biochem. Pharmacol. 2020, 173, 113673. [Google Scholar] [CrossRef] [PubMed]
  219. 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. Int. Immunopharmacol. 2015, 29, 648–655. [Google Scholar] [CrossRef]
  220. 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. Biochem. Biophys. Res. Commun. 2011, 413, 623–629. [Google Scholar] [CrossRef] [PubMed]
  221. 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. Arch. Biochem. Biophys. 2015, 585, 17–24. [Google Scholar] [CrossRef]
  222. 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. AAPS J. 2011, 13, 1–13. [Google Scholar] [CrossRef]
  223. 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-κB signaling pathways and decreases metabolic phenotypes and metastatic activity in colon cancer cells. J. Funct. Foods 2019, 57, 477–487. [Google Scholar] [CrossRef]
  224. 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 Radic. Biol. Med. 2021, 172, 136–151. [Google Scholar] [CrossRef] [PubMed]
  225. 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. Nutr. Cancer 2022, 74, 1882–1893. [Google Scholar] [CrossRef] [PubMed]
  226. Benlloch, M.; Obrador, E.; Valles, S.L.; Rodriguez, M.L.; Sirerol, J.A.; Alcácer, J.; Pellicer, J.A.; Salvador, R.; Cerdá, C.; Sáez, G.T.; et al. Pterostilbene Decreases the Antioxidant Defenses of Aggressive Cancer Cells In Vivo: A Physiological Glucocorticoids- and Nrf2-Dependent Mechanism. Antioxid. Redox Signal. 2016, 24, 974–990. [Google Scholar] [CrossRef]
  227. 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]
  228. 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. J. Ethnopharmacol. 2019, 237, 92–107. [Google Scholar] [CrossRef] [PubMed]
  229. Liu, W.; Li, J. Theaflavin-3,3′-Digallate Attenuates Rheumatoid Inflammation in Mice Through the Nuclear Factor-κB and MAPK Pathways. Arch. Immunol. Ther. Exp. 2019, 67, 153–160. [Google Scholar] [CrossRef]
  230. 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. Int. Immunopharmacol. 2020, 88, 106823. [Google Scholar] [CrossRef]
  231. 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 2022, 27, 8475. [Google Scholar] [CrossRef]
  232. 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. Mol. Pharmacol. 1997, 52, 465–472. [Google Scholar] [CrossRef]
  233. 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. Biotechnol. Appl. Biochem. 2020, 67, 317–322. [Google Scholar] [CrossRef]
  234. 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. Eur. J. Pharm. Biopharm. 2017, 115, 229–242. [Google Scholar] [CrossRef]
  235. 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]
  236. 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. J. Control Release 2023, 362, 631–646. [Google Scholar] [CrossRef]
  237. 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 2017, 43, 698–717. [Google Scholar] [CrossRef]
  238. 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, 5366. [Google Scholar] [CrossRef]
  239. 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. J. Cardiovasc. Pharmacol. 2016, 67, 93–101. [Google Scholar] [CrossRef]
  240. 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]
  241. 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]
  242. 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]
  243. Yang, S.J.; Lim, Y. Resveratrol ameliorates hepatic metaflammation and inhibits NLRP3 inflammasome activation. Metab. Clin. Exp. 2014, 63, 693–701. [Google Scholar] [CrossRef]
  244. Aruna, R.; Geetha, A.; Suguna, P. Rutin modulates ASC expression in NLRP3 inflammasome: A study in alcohol and cerulein-induced rat model of pancreatitis. Mol. Cell. Biochem. 2014, 396, 269–280. [Google Scholar] [CrossRef]
  245. Kim, T.H.; Ku, S.K.; Bae, J.S. Inhibitory effects of kaempferol-3-O-sophoroside on HMGB1-mediated proinflammatory responses. Food Chem. Toxicol. 2012, 50, 1118–1123. [Google Scholar] [CrossRef]
  246. 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 Chem. Toxicol. 2012, 50, 1288–1294. [Google Scholar] [CrossRef]
  247. 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. Exp. Ther. Med. 2017, 14, 1975–1982. [Google Scholar] [CrossRef]
  248. 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. Ann. Transl. Med. 2020, 8, 88. [Google Scholar] [CrossRef]
  249. 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]
  250. 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. Turk. J. Med. Sci. 2020, 50, 2040–2047. [Google Scholar] [CrossRef]
  251. Xiong, H.; Cheng, Y.; Zhang, X.; Zhang, X. Effects of taraxasterol on iNOS and COX-2 expression in LPS-induced RAW 264.7 macrophages. J. Ethnopharmacol. 2014, 155, 753–757. [Google Scholar] [CrossRef] [PubMed]
  252. 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]
  253. 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 J. 2004, 18, 167–169. [Google Scholar] [CrossRef]
  254. 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. Inflamm. Res. 2007, 56, 210–215. [Google Scholar] [CrossRef] [PubMed]
  255. 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. Eur. J. Pharmacol. 2005, 521, 9–20. [Google Scholar] [CrossRef] [PubMed]
  256. 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]
  257. 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. Br. J. Pharmacol. 2011, 164, 1008–1025. [Google Scholar] [CrossRef] [PubMed]
  258. Kim, H.H.; Bae, Y.; Kim, S.H. Galangin attenuates mast cell-mediated allergic inflammation. Food Chem. Toxicol. 2013, 57, 209–216. [Google Scholar] [CrossRef] [PubMed]
  259. 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]
  260. 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. Biol. Pharm. Bull. 2004, 27, 1353–1358. [Google Scholar] [CrossRef]
  261. 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. Int. J. Cancer 2005, 113, 660–669. [Google Scholar] [CrossRef] [PubMed]
  262. 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]
  263. 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 2013, 39, 522–533. [Google Scholar] [CrossRef]
  264. 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 Sci. 2007, 81, 1602–1614. [Google Scholar] [CrossRef]
  265. 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. J. Ethnopharmacol. 2011, 136, 444–451. [Google Scholar] [CrossRef]
  266. 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. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 622–629. [Google Scholar] [CrossRef] [PubMed]
  267. Seeram, N.P.; Momin, R.A.; Nair, M.G.; Bourquin, L.D. Cyclooxygenase inhibitory and antioxidant cyanidin glycosides in cherries and berries. Phytomedicine 2001, 8, 362–369. [Google Scholar] [CrossRef] [PubMed]
  268. 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. J. Appl. Biomed. 2012, 10, 41–50. [Google Scholar] [CrossRef]
  269. 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. Mol. Nutr. Food Res. 2017, 61, 1700139. [Google Scholar] [CrossRef]
  270. Pei, Y.; Parks, J.S.; Kang, H.W. Quercetin alleviates high-fat diet-induced inflammation in brown adipose tissue. J. Funct. Foods 2021, 85, 104614. [Google Scholar] [CrossRef]
  271. 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. Phytother. Res. PTR 2013, 27, 588–594. [Google Scholar] [CrossRef]
  272. Yeo, J.; Lee, J.; Yoon, S.; Kim, W.J. Tannic acid-based nanogel as an efficient anti-inflammatory agent. Biomater. Sci. 2020, 8, 1148–1159. [Google Scholar] [CrossRef]
  273. 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. Crit. Care Med. 2006, 34, 1874–1882. [Google Scholar] [CrossRef]
  274. Lee, T.-P.; Matteliano, M.L.; Middleton, E. Effect of quercetin on human polymorphonuclear leukocyte lysosomal enzyme release and phospholipid metabolism. Life Sci. 1982, 31, 2765–2774. [Google Scholar] [CrossRef] [PubMed]
  275. 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-α and CCL2 by THP-1 Cells. Evid.-Based Complement. Altern. Med. 2015, 2015, 497123. [Google Scholar] [CrossRef]
  276. 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). Evid.-Based Complement. Altern. Med. eCAM 2013, 2013, 127672. [Google Scholar] [CrossRef] [PubMed]
  277. 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. Am. J. Chin. Med. 2011, 39, 1275–1290. [Google Scholar] [CrossRef]
  278. 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. Evid.-Based Complement. Altern. Med. eCAM 2012, 2012, 828521. [Google Scholar] [CrossRef] [PubMed]
  279. 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]
  280. 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. J. Nutr. 2003, 133, 1826–1829. [Google Scholar] [CrossRef] [PubMed]
  281. 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. Sci. Rep. 2018, 8, 3241. [Google Scholar] [CrossRef]
  282. 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. Int. J. Immunopathol. Pharmacol. 2022, 36, 3946320221086084. [Google Scholar] [CrossRef]
  283. 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. Br. J. Nutr. 2018, 121, 55–62. [Google Scholar] [CrossRef]
  284. Więcek, K.; Kupczyk, P.; Chodaczek, G.; Woźniak, M. The Impact of Curcumin on the Inflammatory Profile of SW982 Cells in a Rheumatoid Arthritis Model. J. Immunol. Res. 2022, 2022, 1208970. [Google Scholar] [CrossRef]
  285. 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. Int. J. Mol. Med. 2007, 20, 365–372. [Google Scholar] [CrossRef] [PubMed]
  286. 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]
  287. 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]
  288. 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. J. Inflamm. Res. 2021, 14, 1901–1913. [Google Scholar] [CrossRef] [PubMed]
  289. 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]
  290. 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. Mol. Med. Rep. 2017, 16, 5099–5106. [Google Scholar] [CrossRef] [PubMed]
  291. 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]
  292. Zhang, Y.; Wang, G.; Wang, T.; Cao, W.; Zhang, L.; Chen, X. Nrf2-Keap1 pathway-mediated effects of resveratrol on oxidative stress and apoptosis in hydrogen peroxide-treated rheumatoid arthritis fibroblast-like synoviocytes. Ann. N. Y. Acad. Sci. 2019, 1457, 166–178. [Google Scholar] [CrossRef]
  293. Wang, G.; Xie, X.; Yuan, L.; Qiu, J.; Duan, W.; Xu, B.; Chen, X. Resveratrol ameliorates rheumatoid arthritis via activation of SIRT1-Nrf2 signaling pathway. BioFactors 2020, 46, 441–453. [Google Scholar] [CrossRef]
  294. 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]
  295. 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 J. Chin. Mater. Medica 2010, 35, 1878–1882. [Google Scholar] [CrossRef] [PubMed]
  296. 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]
  297. Srivastava, R.K.; Unterman, T.G.; Shankar, S. FOXO transcription factors and VEGF neutralizing antibody enhance antiangiogenic effects of resveratrol. Mol. Cell. Biochem. 2010, 337, 201–212. [Google Scholar] [CrossRef] [PubMed]
  298. 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. Arch. Med. Sci. AMS 2024, 20, 280–288. [Google Scholar] [CrossRef] [PubMed]
  299. 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] [PubMed]
  300. 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 = J. Cent. South Univ. Med. Sci. 2006, 31, 528–533. [Google Scholar]
  301. 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. Eur. J. Pharmacol. 2019, 844, 65–72. [Google Scholar] [CrossRef] [PubMed]
  302. 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. Z. Fur Naturforschung C J. Biosci. 2013, 68, 336–342. [Google Scholar] [CrossRef]
  303. 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. Biol. Pharm. Bull. 2023, 46, 1592–1600. [Google Scholar] [CrossRef]
  304. 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 2013, 20, 310–318. [Google Scholar] [CrossRef] [PubMed]
  305. 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. Mol. Med. Rep. 2019, 20, 463–472. [Google Scholar] [CrossRef] [PubMed]
  306. 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. Int. J. Biochem. Cell Biol. 2017, 88, 113–123. [Google Scholar] [CrossRef]
  307. 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. J. Biol. Chem. 2011, 286, 11492–11505. [Google Scholar] [CrossRef]
  308. 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 Rheumatol. 2018, 2, 27. [Google Scholar] [CrossRef] [PubMed]
  309. 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]
  310. 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. Mediat. Inflamm. 2021, 2021, 6359652. [Google Scholar] [CrossRef]
  311. 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 2023, 12, 337. [Google Scholar] [CrossRef] [PubMed]
  312. 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. Eur. J. Pharm. Biopharm. 2018, 133, 162–175. [Google Scholar] [CrossRef]
  313. Ganesan, R.; Rasool, M. Ferulic acid inhibits interleukin 17-dependent expression of nodal pathogenic mediators in fibroblast-like synoviocytes of rheumatoid arthritis. J. Cell. Biochem. 2019, 120, 1878–1893. [Google Scholar] [CrossRef]
  314. 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 Sci. 2018, 207, 284–295. [Google Scholar] [CrossRef]
  315. 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. Biol. Pharm. Bull. 2013, 36, 1779–1786. [Google Scholar] [CrossRef]
  316. 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. J. Mater. Chem. B 2023, 11, 11505–11518. [Google Scholar] [CrossRef]
  317. 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]
  318. 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]
  319. 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. Br. J. Nutr. 2010, 104, 957–964. [Google Scholar] [CrossRef] [PubMed]
  320. 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, 408. [Google Scholar] [CrossRef]
  321. 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 Res. Notes 2012, 5, 198. [Google Scholar] [CrossRef] [PubMed]
  322. 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. Sci. Rep. 2016, 6, 32928. [Google Scholar] [CrossRef]
  323. 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 2019, 45, 69–74. [Google Scholar] [CrossRef]
  324. 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 Arch. Pharmacol. 2018, 391, 1179–1190. [Google Scholar] [CrossRef]
  325. 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. Pharm. Biol. 2016, 54, 1705–1715. [Google Scholar] [CrossRef]
  326. 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. Int. J. Radiat. Biol. 2020, 96, 857–867. [Google Scholar] [CrossRef]
  327. 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]
  328. 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]
  329. 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. Biol. Res. 2019, 52, 45. [Google Scholar] [CrossRef]
  330. 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. Mol. Nutr. Food Res. 2021, 65, e2000377. [Google Scholar] [CrossRef] [PubMed]
  331. 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 2016, 55, 1889–1900. [Google Scholar] [CrossRef] [PubMed]
  332. 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. Mol. Med. Rep. 2014, 9, 2592–2598. [Google Scholar] [CrossRef]
  333. 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. Pulm. Pharmacol. Ther. 2019, 56, 69–74. [Google Scholar] [CrossRef]
  334. 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 Drug Dev. Technol. 2020, 18, 356–368. [Google Scholar] [CrossRef]
  335. 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]
  336. Chen, Z.; Xiao, G.; Ao, J. Resveratrol Attenuates Rheumatoid Arthritis Induce Neutrophil Extracellular Traps via TLR-4 Mediated Inflammation in C57BL/6 Mice. Physiol. Res. 2024, 73, 91–104. [Google Scholar] [CrossRef] [PubMed]
  337. 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. J. Inflamm. 2015, 12, 53. [Google Scholar] [CrossRef] [PubMed]
  338. 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. Neurotox. Res. 2022, 40, 1223–1234. [Google Scholar] [CrossRef]
  339. 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. J. Leukoc. Biol. 2016, 100, 559–568. [Google Scholar] [CrossRef] [PubMed]
  340. 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. Regen. Biomater. 2021, 8, rbab067. [Google Scholar] [CrossRef] [PubMed]
  341. 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]
  342. 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]
  343. 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]
  344. 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]
  345. 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. Oxid. Med. Cell. Longev. 2011, 2011, 703676. [Google Scholar] [CrossRef]
  346. Adhikary, R.; Sultana, S.; Bishayi, B. Clitoria ternatea flower petals: Effect on TNFR1 neutralization via downregulation of synovial matrix metalloproteases. J. Ethnopharmacol. 2018, 210, 209–222. [Google Scholar] [CrossRef]
  347. Alamgeer; Uttra, A.M.; Hasan, U.H. Anti-arthritic activity of aqueous-methanolic extract and various fractions of Berberis orthobotrys Bien ex Aitch. BMC Complement. Altern. Med. 2017, 17, 371. [Google Scholar] [CrossRef] [PubMed]
  348. 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]
  349. 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]
  350. 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. Pharmacol. Res. 2020, 151, 104513. [Google Scholar] [CrossRef]
  351. 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. Sci. Pharm. 2013, 81, 567–589. [Google Scholar] [CrossRef] [PubMed]
  352. 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. J. Immunol. Res. 2016, 2016, 7509653. [Google Scholar] [CrossRef]
  353. 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]
  354. 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. Mol. Nutr. Food Res. 2015, 59, 2537–2546. [Google Scholar] [CrossRef] [PubMed]
  355. 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. Am. J. Transl. Res. 2015, 7, 1371–1381. [Google Scholar]
  356. 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 Des. Dev. Ther. 2015, 9, 4931–4942. [Google Scholar] [CrossRef] [PubMed]
  357. 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 2022, 27, 5445. [Google Scholar] [CrossRef]
  358. 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. J. Ethnopharmacol. 2018, 225, 359–366. [Google Scholar] [CrossRef] [PubMed]
  359. Kumatia, E.K.; Baffour, P.K.; Bolah, P. Antiarthritic and Antioxidant Activities of Antrocaryon micraster Seed Extract and Its Fractions. BioMed Res. Int. 2024, 2024, 8838626. [Google Scholar] [CrossRef] [PubMed]
  360. Mirza, F.; Lorenzo, J.; Drissi, H.; Lee, F.Y.; Soung, D.Y. Dried plum alleviates symptoms of inflammatory arthritis in TNF transgenic mice. J. Nutr. Biochem. 2018, 52, 54–61. [Google Scholar] [CrossRef]
  361. 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. J. Ethnopharmacol. 2024, 326, 117884. [Google Scholar] [CrossRef] [PubMed]
  362. Alamgeer; Shanila, A.; Ambreen Malik, U.; Umme Habiba, H. Alkaloids, flavonoids, polyphenols might be responsible for potent antiarthritic effect of Solanum nigrum. J. Tradit. Chin. Med. = Chung i Tsa Chih Ying Wen Pan 2019, 39, 632–641. [Google Scholar] [PubMed]
  363. 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]
  364. 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 2014, 21, 290–297. [Google Scholar] [CrossRef] [PubMed]
  365. 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]
  366. 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 2019, 60, 112–117. [Google Scholar] [CrossRef] [PubMed]
  367. 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 2021, 86, 111183. [Google Scholar] [CrossRef] [PubMed]
  368. Ghavipour, M.; Sotoudeh, G.; Tavakoli, E.; Mowla, K.; Hasanzadeh, J.; Mazloom, Z. Pomegranate extract alleviates disease activity and some blood biomarkers of inflammation and oxidative stress in Rheumatoid Arthritis patients. Eur. J. Clin. Nutr. 2017, 71, 92–96. [Google Scholar] [CrossRef]
  369. 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. Clin. Rheumatol. 2018, 37, 2035–2042. [Google Scholar] [CrossRef]
Figure 1. Summary of polyphenol absorption, distribution, metabolism, and excretion in different segments of the gastrointestinal tract (adapted Olivero-David and co-workers [40] and Ray et al. [41].
Figure 1. Summary of polyphenol absorption, distribution, metabolism, and excretion in different segments of the gastrointestinal tract (adapted Olivero-David and co-workers [40] and Ray et al. [41].
Pharmaceuticals 17 00590 g001
Figure 2. Main dietary polyphenols and simple phenols including their most notable health-benefits (OH: hydroxyl group) (adapted from Mamari [50], Gonçalves et al. [51], and Matsumura et al. [52]).
Figure 2. Main dietary polyphenols and simple phenols including their most notable health-benefits (OH: hydroxyl group) (adapted from Mamari [50], Gonçalves et al. [51], and Matsumura et al. [52]).
Pharmaceuticals 17 00590 g002
Figure 3. 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 3 were drawn using images from BioRender.com (https://biorender.com/) (accessed on 8 March 2024)] (adapted from Weyand and Goronzy [178] and Long et al. [6]).
Figure 3. 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 3 were drawn using images from BioRender.com (https://biorender.com/) (accessed on 8 March 2024)] (adapted from Weyand and Goronzy [178] and Long et al. [6]).
Pharmaceuticals 17 00590 g003
Figure 4. Representation of phenolic 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 the 3 and 5 groups on the A and C rings combined with a 4-oxo group on the C ring (adapted from Platzer and co-workers [185] and created with ChemDraw Professional 16.0 (CambridgeSoft, Perkin Elmer Inc., Waltham, MA, USA)).
Figure 4. Representation of phenolic 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 the 3 and 5 groups on the A and C rings combined with a 4-oxo group on the C ring (adapted from Platzer and co-workers [185] and created with ChemDraw Professional 16.0 (CambridgeSoft, Perkin Elmer Inc., Waltham, MA, USA)).
Pharmaceuticals 17 00590 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gonçalves, A.C.; Rodrigues, S.; Fonseca, R.; Silva, L.R. Potential Role of Dietary Phenolic Compounds in the Prevention and Treatment of Rheumatoid Arthritis: Current Reports. Pharmaceuticals 2024, 17, 590. https://doi.org/10.3390/ph17050590

AMA Style

Gonçalves AC, Rodrigues S, Fonseca R, Silva LR. Potential Role of Dietary Phenolic Compounds in the Prevention and Treatment of Rheumatoid Arthritis: Current Reports. Pharmaceuticals. 2024; 17(5):590. https://doi.org/10.3390/ph17050590

Chicago/Turabian Style

Gonçalves, Ana C., Sofia Rodrigues, Rafael Fonseca, and Luís R. Silva. 2024. "Potential Role of Dietary Phenolic Compounds in the Prevention and Treatment of Rheumatoid Arthritis: Current Reports" Pharmaceuticals 17, no. 5: 590. https://doi.org/10.3390/ph17050590

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

Article Metrics

Back to TopTop