Next Article in Journal
Insights into Immune Exhaustion in Chronic Hepatitis B: A Review of Checkpoint Receptor Expression
Previous Article in Journal
Transmission-Blocking Strategies for Malaria Eradication: Recent Advances in Small-Molecule Drug Development
Previous Article in Special Issue
E-Stilbenes: General Chemical and Biological Aspects, Potential Pharmacological Activity Based on the Nrf2 Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Galangin: A Promising Flavonoid for the Treatment of Rheumatoid Arthritis—Mechanisms, Evidence, and Therapeutic Potential

1
Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut 11-5020, Lebanon
2
Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 11-0236, Lebanon
3
Health Sciences Division, Abu Dhabi Women’s College, Higher Colleges of Technology, Abu Dhabi P.O. Box 25026, United Arab Emirates
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2024, 17(7), 963; https://doi.org/10.3390/ph17070963
Submission received: 2 June 2024 / Revised: 24 June 2024 / Accepted: 3 July 2024 / Published: 19 July 2024
(This article belongs to the Special Issue Bioactive Substances, Oxidative Stress, and Inflammation)

Abstract

:
Rheumatoid Arthritis (RA) is a chronic autoimmune disease characterized by progressive joint inflammation and damage. Oxidative stress plays a critical role in the onset and progression of RA, significantly contributing to the disease’s symptoms. The complex nature of RA and the role of oxidative stress make it particularly challenging to treat effectively. This article presents a comprehensive review of RA’s development, progression, and the emergence of novel treatments, introducing Galangin (GAL), a natural flavonoid compound sourced from various plants, as a promising candidate. The bioactive properties of GAL, including its anti-inflammatory, antioxidant, and immunomodulatory effects, are discussed in detail. The review elucidates GAL’s mechanisms of action, focusing on its interactions with key targets such as inflammatory cytokines (e.g., TNF-α, IL-6), enzymes (e.g., SOD, MMPs), and signaling pathways (e.g., NF-κB, MAPK), which impact inflammatory responses, immune cell activation, and joint damage. The review also addresses the lack of comprehensive understanding of potential treatment options for RA, particularly in relation to the role of GAL as a therapeutic candidate. It highlights the need for further research and clinical studies to ascertain the effectiveness of GAL in RA treatment and to elucidate its mechanisms of action. Overall, this review provides valuable insights into the potential of GAL as a therapeutic option for RA, shedding light on its multifaceted pharmacological properties and mechanisms of action, while suggesting avenues for future research and clinical applications.

1. Introduction

1.1. Rheumatoid Arthritis (RA)

In rheumatoid arthritis, “arthr-” refers to joints, “-itis” means inflammation, and “rheumatoid” comes from rheumatism, which broadly refers to various musculoskeletal conditions. Therefore, rheumatoid arthritis (RA) is a chronic, inflammatory disorder that mostly affects the joints, but can also involve other organ systems like the skin and lungs as well [1,2]. RA is a chronic autoimmune disorder that impacts around 1% of the global population. While the disease can manifest at any age, it is most commonly diagnosed in people between the ages of 30 and 60 [3]. Interestingly, women are more than twice as likely to develop RA compared to men. This gender disparity may be attributed to the role of estrogen in modulating the immune response, potentially increasing women’s susceptibility to the disease [4,5].

1.1.1. Development of RA

Rheumatoid arthritis is an autoimmune process that is typically triggered by an interaction between genetic and environmental factors [6]. A deficiency in a gene that encodes an immune protein, such as human leukocyte antigen (HLA-DR1 and HLA-DR4), may result in rheumatoid arthritis when combined with environmental factors like cigarette smoke or specific infections [7,8,9]. These environmental factors can activate peptidyl arginine deiminases in mucosal cells, enhancing the post-translational conversion of arginine to citrulline in the presence of intracellular or matrix proteins, a process known as citrullination [10,11].
This process creates neo-epitopes, which are novel antigenic determinants formed from alterations in the protein sequence, that can be recognized by the adaptive immune system. These modified peptides are presented on major histocompatibility complex (MHC) proteins by antigen-presenting cells (APCs), activating T cells in lymphoid tissues. This, in turn, stimulates B cells to produce a variety of autoantibodies that recognize self-proteins, including rheumatoid factor (RF), which targets IgGs and anti-citrullinated protein antibodies (ACPAs), which target citrullinated proteins [12,13].

1.1.2. Pathogenesis of RA

The presence of ACPAs and RF is associated with a more aggressive disease progression and can serve as a diagnostic and prognostic indicator [14,15]. Upon binding to their targets, these antibodies form immune complexes that accumulate in the synovial fluid. Subsequently, these complexes activate the complement system, which operates in an enzymatic cascade to facilitate joint inflammation and injury. Initially, the classical pathway is activated when the C1 complex binds to the Fc region of the antibodies in the immune complexes. This triggers a sequential activation of complement proteins C2, C4, and C3, ultimately forming the C3 convertase. The convertase cleaves C3 into C3a and C3b, where C3b binds to the immune complex, leading to the formation of the C5 convertase. This convertase cleaves C5 into C5a and C5b.
C5a acts as a potent anaphylatoxin, promoting inflammation, and C5b initiates the assembly of the membrane attack complex (MAC) composed of C5b, C6, C7, C8, and C9. The MAC creates pores in cell membranes, leading to cell lysis and contributing to tissue damage. Additionally, the activation of complement components C3a and C5a recruits and activates neutrophils, macrophages, and other immune cells to the site, further amplifying the inflammatory response and resulting in joint inflammation and injury [16,17].
In RA, T-helper cells and antibodies move into the bloodstream and travel to the joints [18,19]. Once there, T cells release cytokines like interferon-gamma (IFN-γ) and interleukin-17 (IL-17), which attract more inflammatory cells such as macrophages into the joint space [20,21]. Macrophages also produce inflammatory cytokines like tumor necrosis factor-alpha (TNF-α), interleukin-1 (IL-1), and interleukin-6 (IL-6), which, along with T cell cytokines, stimulate the growth of synovial cells [22,23]. This process leads to the formation of pannus, a thickened synovial membrane containing fibroblasts, myofibroblasts, and inflammatory cells [24]. Over time, the pannus can damage cartilage and other soft tissues and erode bone. Synovial cells also release enzymes that break down proteins in the cartilage [25,26]. Without protective cartilage, bones can rub against each other. Additionally, inflammatory cytokines increase the production of RANKL, a protein that allows T cells to bind to osteoclasts, triggering bone breakdown [27,28]. Chronic inflammation also promotes the formation of new blood vessels around the joint, bringing more inflammatory cells [29].
As the disease progresses, inflammation and damage occur in multiple joints on both sides of the body [30]. These inflammatory cytokines do not remain limited to the joints; instead, they spread through the bloodstream, affecting other organs. For instance, IL-1 or IL-6 can travel to the brain, causing fever [31]. When these cytokines reach the brain, they stimulate the production of prostaglandins, especially in the hypothalamus. Prostaglandins act on the hypothalamus, specifically in the preoptic area, which is the body’s thermoregulatory center. This release of prostaglandins leads to an increase in the hypothalamic set point for body temperature, causing the body to generate and retain heat, resulting in fever [32]. The systemic effects of these cytokines extend beyond fever. In muscles, they promote protein breakdown. In the skin and body organs, they lead to the formation of round collections of immune cells called rheumatoid nodules [33,34]. The liver responds by producing high amounts of hepcidin, a protein that decreases iron levels in the blood [35,36]. In the lungs, fibroblasts become overactive, leading to scar tissue formation that impairs gas exchange. The pleural cavities around the lungs can also become inflamed, filling with fluid (pleural effusion), which can affect lung expansion [37,38].
Hence, as Rheumatoid Arthritis progresses, the systemic spread of inflammatory cytokines leads to a wide range of extra-articular manifestations. These affect multiple organs and tissues throughout the body, resulting in diverse complications.

1.1.3. Role of NF-κB, MAPK, JAK/STAT, and PI3K/Akt in the Pathogenesis of RA

RA is characterized by an intricate interplay of inflammatory pathways, including NF-κB, MAPK, JAK/STAT, and PI3K/Akt. All of these contribute to the chronic inflammatory state and joint damage observed in the disease [39,40]. The NF-κB pathway, a pivotal regulator of inflammation, is aberrantly activated in RA synovial cells, leading to the transcription of pro-inflammatory genes such as TNF-α, IL-1β, and IL-6 [41]. These cytokines, in turn, activate the MAPK pathway, comprising p38 MAPK, ERK, and JNK subfamilies, which further promotes cytokine production and matrix metalloproteinase (MMP) expression, contributing to inflammation and tissue damage in RA [42].
Concurrently, the JAK/STAT pathway, activated by cytokines like IL-6 and IL-17 abundant in RA synovium, plays a crucial role in RA pathogenesis [43]. This pathway not only drives the differentiation of pro-inflammatory T-helper 17 (Th17) cells but also enhances the activation and recruitment of immune cells, including macrophages and neutrophils, to the inflamed synovium, perpetuating the inflammatory cascade. Additionally, the PI3K/Akt pathway, activated by growth factors and cytokines, promotes the survival and proliferation of synovial fibroblasts and immune cells, contributing to synovial hyperplasia and inflammation [44]. Akt activation also stimulates the expression of MMPs, notably MMP-9, further exacerbating tissue damage in RA joints.
Overall, the dysregulated activation of these signaling pathways in RA leads to a vicious cycle of inflammation, immune cell infiltration, and tissue destruction. Targeting these pathways individually or in combination presents a promising therapeutic approach for RA, aiming to alleviate inflammation, preserve joint function, and improve patient outcomes.

1.1.4. Oxidative Stress in RA

The development of RA is closely associated with oxidative stress, a condition characterized by an imbalance between the excessive production of reactive oxygen species (ROS) and the body’s insufficient antioxidant defenses. This condition contributes to inflammation, immune dysregulation, and joint damage, all of which are characteristic of RA [45]. During the inflammatory process in RA, various cells such as neutrophils, macrophages, and synoviocytes produce ROS, including superoxide anions, hydrogen peroxide, and hydroxyl radicals [46]. These ROS are known to cause damage to lipids, proteins, and DNA, leading to cell dysfunction and death [47], and activate redox-sensitive signaling pathways like NF-κB, and MAPKs, which in turn promote the production of pro-inflammatory cytokines, chemokines, and MMPs. This chain of events further worsens the inflammation and joint damage experienced by RA patients [48,49,50]. The human body combats oxidative stress through a range of antioxidant mechanisms [51]. Enzymatic antioxidants, such as superoxide dismutase (SOD), catalase, and glutathione peroxidase, play a crucial role in detoxifying ROS [52]. Non-enzymatic antioxidants, including vitamins C and E, also help neutralize free radicals [53]. Additionally, the activation of transcription factors like NRF2 stimulates the production of antioxidant proteins, enhancing the body’s resilience to oxidative stress [54]. By understanding these defense systems, we can better appreciate the therapeutic potential of promising compounds, that not only reduce inflammation but also bolster these antioxidant defenses, thereby protecting joint tissues from oxidative damage and inflammation associated with RA.
Several studies have shown that RA patients have increased oxidative stress markers. For instance, the levels of lipid peroxidation products, such as malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE), were observed to be higher in the synovial fluid and serum of RA patients [55]. Furthermore, the levels of antioxidants, such as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx), were found to be decreased in RA patients, further exacerbating oxidative stress [56].
Additionally, three transcription factors—activator protein 1 (AP-1), hypoxia-inducible factor (HIF), and nuclear factor 2 (Nrf2)—play crucial roles in disease progression [57]. AP-1, a complex of proteins from the Fos and Jun families, regulates the expression of genes involved in cell proliferation, differentiation, and inflammation. In RA, AP-1 is activated by pro-inflammatory cytokines such as TNF-α and IL-1β, as well as by other stimuli like ROS and mechanical stress. Activated AP-1 in RA synovial fibroblasts promotes the expression of MMPs, contributing to the degradation of cartilage and bone [58]. Additionally, AP-1 regulates the expression of pro-inflammatory cytokines and chemokines, further amplifying the inflammatory response in RA.
In the hypoxic microenvironment of the RA synovium, HIFs play a critical role in regulating genes involved in angiogenesis, inflammation, and cell survival [59]. HIF-1α, a subunit of the HIF complex, stabilizes under hypoxic conditions and promotes the expression of vascular endothelial growth factor (VEGF) and other angiogenic factors, leading to excessive blood vessel formation (angiogenesis) in the synovium. This aberrant angiogenesis contributes to the chronic inflammatory state and pannus formation in RA. Moreover, HIF-1α regulates the expression of genes encoding pro-inflammatory cytokines and MMPs, further exacerbating joint inflammation and damage in RA.
Nrf2 regulates the expression of genes involved in antioxidant defense and detoxification of ROS. In RA, oxidative stress is a prominent feature, leading to increased ROS production and oxidative damage in the joints [60]. Nrf2 activation in response to oxidative stress is impaired in RA, contributing to the accumulation of ROS and oxidative damage. Restoring Nrf2 activity was proposed as a potential therapeutic strategy to alleviate oxidative stress and inflammation in RA.
In summary, AP-1 promotes joint inflammation and destruction by regulating the expression of MMPs and pro-inflammatory mediators. HIFs contribute to RA pathogenesis by promoting angiogenesis, inflammation, and cell survival in the hypoxic synovial microenvironment. Nrf2 dysfunction leads to increased oxidative stress and inflammation in RA joints. Therefore, chronic inflammation and oxidative stress are pivotal players in the aggravation of chronic inflammatory joint disease. In this regard, anti-inflammatory and antioxidant therapy may offer novel adjuvant/complementary treatment options aimed at enhancing disease control.

1.1.5. Novel Treatments for RA

The ultimate goal of RA treatment is to reduce inflammation, minimize joint damage, and enhance the patient’s physical function and quality of life [61,62]. Treatment is expected to improve the clinical outcomes of RA by reducing synovitis, thereby decreasing the likelihood of further joint damage [63].
There are three general classes of drugs commonly used in RA therapy [61]: non-steroidal anti-inflammatory drugs or NSAIDs, corticosteroids, and disease-modifying antirheumatic drugs or DMARDs. Although NSAIDs and corticosteroids are effective at providing symptom relief for patients with RA, only DMARDs were demonstrated to modify the course of the disease and improve radiologic findings [64]. DMARDs are divided into two types: biological and synthetic small molecules (Figure 1) [65,66].
Biologics represent a novel approach to treating RA based on recent advances in understanding the immune and inflammatory processes associated with the disease [67,68]. TNF-α inhibitors were the first biological DMARDs approved for arthritis treatment [69]. They function by binding to TNF-α, thereby reducing its activity and minimizing inflammation. Blocking agents are another type of biological DMARD that interfere with the interaction between certain immune cells [70]. B-cell depleting agents are also used, reducing the number of B cells in the joints and lowering T cell activation and plasma cell count [71].
Synthetic small molecule DMARDs are also effective treatments for RA, acting by modifying the immune response within cells. Methotrexate (MTX), a non-biologic DMARD, is a commonly prescribed medication for RA treatment [72]. Initially developed as a cancer drug, MTX has proven beneficial in treating other inflammatory diseases like psoriasis [73]. MTX inhibits dihydrofolate reductase, an enzyme necessary for cell division and growth [74]. This disruption in nucleic acid production leads to decreased cell proliferation and suppression of the immune response. However, prolonged use of MTX can lead to toxicity in various tissues, including bone marrow suppression, liver damage, gastrointestinal irritation, pulmonary inflammation or fibrosis, and in high doses, kidney damage [75].
Many current treatments for RA can cause significant side effects, which may limit their long-term use. Treatment failure is also a concern, as some patients do not respond to existing therapies, leading to continued disease progression and joint damage. While many treatments can alleviate symptoms, achieving complete disease remission remains a challenge for numerous patients. With the development of new therapies, more options will become available to reduce inflammation in RA and potentially achieve disease remission. To avoid side effects and treatment failure, researchers are exploring natural sources of drugs, specifically bioactive compounds obtained from natural products.
Numerous studies were conducted to investigate the potential of natural alternatives for treating rheumatoid arthritis (RA) and preventing the side effects of Methotrexate (MTX), commonly used in RA treatment. Synthetic trans-Δ9-tetrahydrocannabinol (Δ9-THC) [76], Indole-3-Carbinol (I3C) found in cruciferous vegetables [77], an Aloe species’ leaf extract [78] Boswellia extract rich in Acetyl Keto Boswellic Acid (AKBA) [79], glabridin found in Glycyrrhiza glabra roots [80], and a natural agent named Silibinin [81] have shown anti-inflammatory and anti-arthritic effects. These natural agents were also shown to be effective in protecting the liver from MTX-induced damage. The combination therapy of these natural agents with MTX was found to be more effective in inhibiting arthritis and reducing MTX-induced hepatotoxicity in arthritic animals, highlighting the potential of natural compounds as adjuncts to conventional RA therapy.

1.2. Introduction of GAL as a Potential Therapeutic Agent and Its Sources

Flavonoids are polyphenols found in honey and various parts of plants, including barks, roots, rhizomes, stems, leaves, flowers, seeds, grains, fruits, and vegetables [82]. Flavonols, a subclass of flavonoids, possess a 3-hydroxyflavone backbone. Both flavonoids and flavonols exhibit a range of health benefits, including antioxidant, anti-inflammatory, and immune-modulatory properties, potentially offering relief from various ailments [83,84]. Galangin (GAL), a flavonol, is a promising contender that has demonstrated favorable outcomes in various disease models [85,86,87,88]. GAL is extracted from honey [89], propolis [90], and bee pollen [91,92], and is found in a wide variety of plants. Its sources are summarized in Table 1.

2. Bioactive Properties of GAL

2.1. Molecular Structure of GAL

GAL, a bioactive compound, is classified as a 3,5,7-trihydroxyflavone, a subclass of flavonoids. This molecule possesses a distinct chemical structure characterized by a chromone ring (also known as 1-benzopyran-4-one) substituted with hydroxyl groups at specific positions 3, 5, and 7. Additionally, a phenyl ring is linked to the chromone ring (see Figure 2) [117]. The molecular formula of GAL is C15H10O5, and its molecular weight is 270.24 g/mol. A summary of the chemical and physical properties of galangin can be found in the Supplementary Data, Table S1.

2.2. Pharmacological Properties of GAL

Using the ultra-high-performance liquid chromatography–tandem mass spectrometry (UHPLC–MS/MS) method, researchers studied the pharmacokinetic properties of GAL. They discovered that when GAL was administered orally or via intraperitoneal injection to a rat model, it was quickly absorbed (tmax = 0.25 h) and eliminated (t1/2 < 1.1 h) with an absolute bioavailability of 7.6%. GAL was found in high amounts in the kidney, liver, and spleen, and it was also able to cross the blood–brain barrier to reach the brain, although in lower concentrations [118]. GAL undergoes metabolism through glucuronidation reaction, producing metabolites such as galangin-3-O-β-D-glucuronic acid (GG-1) and galangin-7-O-β-D-glucuronic acid (GG-2), which were detected in the urine and plasma of rats [119,120].
Due to limitations in absorption and solubility that hinder its effectiveness, researchers have investigated methods to improve these properties of GAL for optimal delivery [121,122,123,124,125]. These methods include liposomes, which act as carriers to deliver GAL to targeted sites within the body [121]. Micelles, on the other hand, can significantly enhance the water solubility of GAL, making it more readily absorbed [122]. Additionally, nanostructured lipid carriers (NLCs) and nanoparticles (NPs) offer advantages like increased drug-loading capacity and controlled release [123,124]. Finally, β-cyclodextrin inclusion complexes can enhance both the solubility and stability of GAL, improving its therapeutic potential [125].
Given its interaction with CYP450 enzymes, GAL has the potential to alter the metabolism of other drugs. It was shown to inhibit several CYP450 enzymes, including CYP2A6, CYP2C8, CYP2C13, and CYP3A1, while activating CYP1A2 and CYP2B3 [126,127]. Therefore, caution should be exercised when co-administering GAL with other pharmacological agents. GAL administration, both in vitro and in vivo, exhibited no cytotoxicity on normal cells, suggesting a favorable safety profile [128,129]. Studies demonstrated good tolerance at concentrations up to 30 µM in microglial BV2 cells and at high oral doses of 320 mg/kg in rats [128,129]. Remarkably, this non-toxic effect on normal cells aligns with GAL’s ability to destroy cancer cells, suggesting potential avenues for further research into its therapeutic applications [130].
In vitro and in vivo studies have revealed that GAL possesses a broad spectrum of biological activities, including anti-inflammatory, antioxidant, antifibrotic, antihypertensive, antimicrobial, and anticancer properties [131,132,133,134,135]. These diverse pharmacological effects have led researchers to investigate its potential therapeutic applications in various disease models, as illustrated in Figure 3.

2.2.1. Anti-Inflammatory Effects of GAL

GAL exhibited anti-inflammatory properties in various disease models, both in vivo and in vitro. These diseases include cardiovascular, renal, neural, skin, pulmonary, hepatic, gastrointestinal, pancreatic, foot, and retinal illnesses [86,132,136,137,138,139,140,141,142,143]. GAL works by modulating several signaling pathways that participate in the production of both proinflammatory and anti-inflammatory mediators, thereby alleviating inflammation. Detailed insights into these pathways are provided in Table 2.
Multiple studies in various disease models have consistently shown that GAL can inhibit the activation of NF-κB [136,137,138,140]. This inhibition occurs by targeting specific proteins in the NF-κB signaling pathway. These proteins, IκBα, IKKβ, and p65, undergo phosphorylation, a crucial step for NF-κB activation [146]. GAL inhibits this phosphorylation process, thereby preventing NF-κB from dissociating from its natural inhibitor, IκBα [146]. This effectively suppresses NF-κB activity, thereby hindering its ability to initiate the inflammatory cascade [146]. Consequently, GAL-mediated inhibition of NF-κB results in reduced levels of various pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-6 [136,137,148,151,164]. Additionally, GAL reduces the expression of proinflammatory enzymes such as inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) [132,136,137,142,148,149,156], which are responsible for the production of nitric oxide (NO) and prostaglandin E2 (PGE2), respectively, both contributing to inflammation [129,149]. Furthermore, GAL modulates NF-κB by regulating other factors such as Toll-like receptor 4 (TLR4) and Peroxisome proliferator-activated receptor gamma (PPARγ) [139,140,151,153,160,165]. In studies using lipopolysaccharide-stimulated microglia models and in vivo models of pulmonary inflammation in vivo study, GAL demonstrated its ability to activate PPARγ, a molecule that downregulates NF-κB and inflammatory cytokine production [151,160]. GAL also disrupted the TLR4-NF-κB p65 signaling axis, further reducing inflammation in lipopolysaccharide (LPS)-injured rat intestinal epithelial (IEC-6) cells and sulphate sodium-induced ulcerative colitis in mice [140,165].
GAL’s influence extends beyond NF-κB. It exerts its anti-inflammatory effects through additional pathways, including the PI3K/AKT, MAPK, and NOD-like receptor family pyrin domain containing three (NLRP3 inflammasome) pathways [144,145,146,151,152,153]. GAL may also help control MMPs, enzymes that contribute to inflammation in RA as discussed previously. Notably, GAL inhibits MMP expression in activated microglia, human neuroblastoma cells, and an animal model of induced pulmonary inflammation [151,152,157]. Furthermore, in lipopolysaccharide-induced neuroinflammation in rats, GAL inhibited the Dipeptidyl peptidase 4 (DPP-4) activity, ultimately increasing levels of the anti-inflammatory molecule GLP-1 [153]. Furthermore, GAL activates Nrf2, a molecule known to be compromised in RA. This activation of Nrf2, as demonstrated in various in vivo models of liver, gastrointestinal, and skin inflammation, leads to the restoration of the Nrf2/HO-1 pathway and the production of the potent anti-inflammatory enzyme HO-1 [156,161,162,164].
Overall, GAL’s multifaceted anti-inflammatory actions, including NF-κB inhibition, cytokine reduction, MMP control, and Nrf2 activation, position it as a promising therapy for RA. By addressing inflammation, immune dysfunction, and joint degradation, GAL offers a comprehensive approach to combating RA’s complex pathogenesis.

2.2.2. Antioxidant Effect of GAL

Due to its structure, particularly the presence of hydroxyl groups, GAL (3,5,7-trihydroxyflavone) effectively functions as an antioxidant. This functionality arises from its ability to readily donate a hydrogen atom, specifically from its 3-hydroxyl group. This donation transforms a portion of the GAL molecule into a 3-flavonoid phenoxyl radical. The phenoxyl radical acts as a free radical scavenger, neutralizing free radicals [166].
Endogenous antioxidants, including superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx), serve as the primary shield against cellular damage caused by reactive oxygen species (ROS). These enzymes facilitate the conversion of ROS into less reactive molecules. SOD specifically catalyzes the dismutation of superoxide radicals into hydrogen peroxide, a less harmful species. Furthermore, CAT offers additional protection by catalyzing the reduction of hydrogen peroxide into water and oxygen. Reduced glutathione (GSH) scavenges free radicals and acts as a substrate for several detoxifying enzymes. Therefore, a compromised antioxidant defense system can lead to the breakdown of hydrogen peroxide into highly reactive hydroxyl radicals, which can disrupt various cellular components [167].
Numerous studies have shown that GAL exhibits protective effects against oxidative stress. These studies revealed that GAL upregulated the activity of various antioxidant enzymes and molecules, including SOD, CAT, glutathione S-transferase (GST), GPx, and GSH [128,140,141,144,145,156,167,168,169,170,171,172,173]. Additionally, it increased the levels of antioxidant molecules like ascorbic acid (vitamin C) and α-tocopherol (vitamin E) [128,168,170]. Moreover, GAL suppressed lipid peroxidation, leading to a reduction in thiobarbituric acid reactive substances (TBARS) and MDA levels [133,140,144,145,156,167,170,173]. GAL’s ability to reduce ROS levels [144,174] may also be explained by its effect on NADPH oxidase-1 (NOX-1). This enzyme is a major source of ROS production in cells, and studies using a neurotoxicity-induced rat model have shown that GAL treatment downregulates NOX-1 [175].
Additionally, GAL administration inhibited protein oxidation and nitration, resulting in a decrease in protein carbonyls (PCO) and nitrotyrosine levels across various disease models. These models include induced nephrotoxicity, fructose feeding to induce metabolic syndrome, and a hepatorenal disease model [144,170,173]. It also downregulated Acyl-CoA Synthetase Long-Chain Family Member 4 (ACSL4) and 4-Hydroxynonenal (4-HNE) levels, attenuating oxidative stress in AR42J pancreatic cells [141].
Finally, GAL treatment restored and upregulated the Nrf2/HO-1 pathway, leading to an increase in heme oxygenase-1 (HO-1) expression. HO-1 is a potent antioxidant enzyme that helps eliminate harmful free radicals and alleviate oxidative stress. This, in turn, protects dermal fibroblasts from senescence following H2O2 exposure and mitigates the severity of acute pancreatitis, psoriasis-like skin inflammation, and cognitive alterations in rodent models [131,141,156,175].
Therefore, GAL’s antioxidant potential is derived from its direct scavenging of free radicals and reinforcement of the cellular antioxidant defense system. By enhancing the activity of antioxidant enzymes and molecules, it effectively shields cells from oxidative stress damage. This multifaceted antioxidant activity holds significant promise as oxidative stress mediators are pivotal in the development of joint damage in RA. Given the direct contribution of oxidative stress to the progression of RA, there is a compelling rationale to further explore GAL as a potential therapeutic strategy for safeguarding joints and managing RA.

2.2.3. Immunomodulatory Effect of GAL

The immunomodulatory effects of GAL were shown in several in vivo and in vitro studies, as summarized in Table 3.
These effects contribute to a reduced inflammatory response through multiple mechanisms. First, GAL suppresses the infiltration of inflammatory cells, including neutrophils, lymphocytes, macrophages, eosinophils, and mast cells [154,158,178]. Notably, mast cells are key mediators of inflammatory reactions, releasing signaling molecules like histamine that cause redness and swelling (edema) [154]. Interestingly, GAL treatment directly suppresses histamine release in human mast cells (HMC)-1 by decreasing intracellular calcium levels [177]. Furthermore, GAL’s anti-inflammatory properties extend to its effects on macrophages. In LPS-stimulated RAW 264.7 macrophages, GAL downregulates COX-2, an enzyme crucial for the production of inflammatory prostaglandins [94]. This finding aligns with an in vivo study where GAL treatment attenuated myocardial ischemia-reperfusion injury in mice. This protective effect was associated with an increase in anti-inflammatory M2 macrophages and a decrease in proinflammatory M1 macrophages following GAL administration [181].
GAL’s anti-inflammatory effects extend beyond just suppressing immune cell infiltration. It also acts by downregulating key signaling pathways within the very cells it targets. This included inhibiting the activation of JNK, p38, ERK MAPKs, and NF-κB pathways in LPS-stimulated RAW 264.7 macrophages and human mast cells (HMC)-1, all of which are known contributors to the production of proinflammatory cytokines [94,177]. Furthermore, GAL disrupts the maturation process of these proinflammatory cytokines in HMC-1 by reducing caspase-1 activity [177]. Similarly, in LPS-stimulated RAW 264.7 macrophages, GAL targets upstream mediators in inflammatory signaling [94]. It inhibits interleukin-1 receptor-associated kinase 1 (IRAK-1), a key player in TLR signaling that triggers proinflammatory cytokine production [94]. Additionally, GAL modulates the JAK/STAT pathway by suppressing JAK-1, another crucial component of proinflammatory cytokine signaling within these macrophages [94]. This multi-pronged approach is evident in the reduced levels of pro-inflammatory mediators observed in various models. Treatment with GAL in LPS-stimulated macrophages and a concanavalin A-induced hepatitis model significantly decreased the production of cytokines (TNF-α, IFN-γ, and IL-12), chemokines (CXCL10 and MIP-1α), and adhesion molecules (ICAM-1) [94,178].
Beyond its influence on innate immune cells like macrophages and mast cells, GAL also modulates the adaptive immune response by affecting T lymphocytes. Following treatment with GAL in the bleomycin-induced pulmonary fibrosis model, atopic dermatitis model, and LPS-induced dendritic cell model, T cell activation and expansion were reduced. These studies revealed that GAL downregulated CD8+ T cells and CD4+ T cells (Th1, Th2, and Th17 cells), and the levels of cytokines associated with both Th1 (IFN-γ) and Th2 (IL-4, IL-5, IL-13, IL-31, and IL-32) immune responses [154,179,180]. GAL further dampens the inflammatory response by suppressing the production of immunoglobulins involved in allergic and inflammatory responses [154,158]. In models of ovalbumin-induced airway inflammation and atopic dermatitis, GAL treatment inhibited the production of immunoglobulin E (IgE) and decreased serum levels of IgG2a [154,158].
GAL’s influence extends to antigen-presenting cells. Research has investigated the potential role of GAL’s immunomodulatory effect on dendritic cells (DCs) [180]. GAL treatment of LPS-stimulated DCs promoted the development of tolerogenic dendritic cells (tolDCs) [180]. Compared to bone marrow-derived DCs (BMDCs), GAL-treated DCs (Gal-DCs) showed lower levels of CD86, a costimulatory molecule, and reduced expression of major histocompatibility complex class II (MHC-II) molecules, indicating diminished antigen presentation capabilities. Gal-DCs displayed an increase in the production of the anti-inflammatory cytokine IL-10 and programmed death ligand 1 (PD-L1) expression, which was associated with the activation of MAPKs such as ERK, JNK, and p38. Moreover, Gal-DCs induced allogeneic CD4 T cells differentiation into regulatory T cells [180]. This suggests that GAL skews DC towards a tolerogenic phenotype, leading to suppressed inflammatory T cell responses.
In conclusion, GAL’s immunomodulatory effects align well with the pathogenesis of rheumatoid arthritis (RA), making it a promising treatment option. GAL’s ability to suppress inflammatory cell infiltration, modulate macrophage and mast cell activity, and reduce proinflammatory cytokine production directly addresses these key aspects of RA pathogenesis. Additionally, its effects on T lymphocytes and antigen-presenting cells suggest a broader impact on the adaptive immune response, which is often dysregulated in RA. By targeting multiple components of the immune response implicated in RA, GAL offers a comprehensive approach to mitigating inflammation and potentially slowing down joint damage in RA patients.

2.2.4. GAL and Autoimmunity

In the context of autoimmune diseases, GAL was shown to inhibit the activation of immune complex-stimulated neutrophils by FcγRs (Fc gamma receptors) and CRs (complement receptors). This effect reduces the activity of myeloperoxidase and horseradish peroxidase enzymes, which are involved in the production of reactive oxygen species (ROS) by neutrophils [176]. Consequently, GAL helps to reduce the overall ROS production, potentially mitigating tissue damage associated with autoimmune conditions.
Building on its ability to modulate immune cell activity and ROS production, a study using an experimental autoimmune encephalomyelitis (EAE) mouse model showed that GAL treatment ameliorated demyelination, inhibited the infiltration of mononuclear cells (MNCs) into the spinal cord, and reduced T cell proliferation and differentiation. Notably, GAL led to a decrease in TH1 and TH17 cells in the spinal cords of EAE mice. Furthermore, GAL treatment impaired the function of dendritic cells (DCs), including their ability to present antigens and produce cytokines (IL-6, IL-12, and IL-23). These combined effects contributed to the alleviation of clinical symptoms of EAE [87]. This suggests that GAL holds promise as a therapeutic strategy for multiple sclerosis and other neuroinflammatory diseases.
GAL’s therapeutic potential extends beyond neuroinflammatory conditions to other autoimmune diseases. In a study on psoriasis, an autoimmune skin disease causing chronic inflammation, GAL demonstrated its antioxidant and anti-inflammatory properties in an induced psoriasis-like skin inflammation model. This effect was mediated by the recovery of the Nrf2/HO-1 pathway and the upregulation of antioxidant enzymes and markers, including SOD, CAT, GST, GSH, glutathione reductase (GR), and vitamin C [156]. These findings suggest that GAL may offer therapeutic benefits for various autoimmune diseases by alleviating inflammation and oxidative stress.
While GAL exhibits therapeutic potential, it is important to consider its compatibility with existing medications used for autoimmune diseases. MTX is a well-established medication for managing autoimmune diseases such as rheumatoid arthritis and psoriatic dermatomyositis. Like all drugs, MTX has several side effects including the development of hepatotoxicity. GAL exerted its hepatoprotective effects in a rat model of MTX-induced hepatotoxicity by suppressing inflammation through the downregulation of NF-κB p65, iNOS, and the proinflammatory cytokines TNF-α, IL-1β, and IL-6. Furthermore, GAL alleviated oxidative stress induced by MTX by reducing the levels of ROS, NO, and MDA, and by upregulating antioxidant enzymes and molecules (GSH, SOD, and CAT), thereby mitigating MTX-induced hepatotoxicity [86]. The therapeutic potential of combining MTX with GAL for rheumatoid arthritis remains unexplored. Further research is warranted to investigate their efficacy and safety in this context.

3. Mechanism of Action of GAL against Arthritis: In Vivo and In Vitro Studies

GAL was studied for its potential therapeutic properties against bone disorders. Its anti-proliferative properties were shown to inhibit the growth of cancer cells and promote apoptosis in osteosarcoma [182]. GAL also exhibits immune protective effects by enhancing the immune system’s response to infections and reducing inflammation. These properties make GAL a promising candidate for the treatment of various bone disorders, such as osteoporosis and rheumatoid arthritis.

3.1. Effect of GAL on Osteoarthritis (OA)

Osteoarthritis (OA) is a prevalent form of arthritis characterized by joint degeneration [183]. Several studies suggest that GAL could be a potential treatment for OA, as shown in Table 4.
In vivo, GAL was found to prevent cartilage destruction, slow down the development and progression of OA, and protect articular cartilage [184,185,186,187,188,189]. In vitro, studies have demonstrated that treatment with GAL can decrease the expression of catabolic factors in chondrocytes, ameliorate the loss of extracellular matrix (ECM) components, and promote osteogenic differentiation [184,185,186,188,189,190]. Mechanistically, Huang et al. identified GAL as a potent substance with remarkable anti-inflammatory properties. It effectively reverses the inflammatory response caused by exposure to IL-1β in rat chondrocytes, reduces the degradation of collagen II and aggrecan, and inhibits Akt phosphorylation and NF-κB activation [185]. Treatment with GAL significantly reduces levels of ROS, lipid peroxidation IL-1β, IL-6, and TNF-a, while increasing levels of CAT, SOD, Gpx, and GSH.
Su et al. also observed that GAL significantly reduces the mRNA and protein expression levels of C-terminal cross-linked telopeptide of type II (CTX-II) [187]. Furthermore, Wang et al. showed that GAL has the ability to suppress the inflammatory response that is induced by IL-1β and ameliorate African Caribbean Leukaemia Trust (ACLT)-induced cartilage degeneration by effectively inhibiting the NF-κB pathway, as well as the JNK and ERK pathways [186]. Lin et al. demonstrated that GAL prevents ECM degradation by activating Proline and Arginine Rich End Leucine Rich Repeat Protein (PRELP) and inhibiting the PI3K-AKT signaling pathway. This helps in safeguarding chondrocytes from oxidative stress by inhibiting the expression of ROS and MDA while promoting the expression of SOD and CAT [188]. Moreover, Li et al. found that GAL suppresses RANKL-induced ERK-MAPK, p38-MAPK, and NF-κB signaling pathways in bone marrow macrophages by attenuating phosphor-ERK, phosphor-p38, and phosphor-p65, while stabilizing IκB-α expression [190]. Recently, Zeng et al. explored the effects of GAL on autophagy and osteogenesis in bone marrow stromal cells (BMSCs). The results showed that GAL enhanced the PKA/CREB signaling pathway in BMSCs, inducing autophagy and increasing osteogenic differentiation [189]. This suggests that GAL directly inhibits osteoclastogenesis via the NF-κB and MAPK signaling pathways, offering a potential avenue for preventing bone loss associated with OA.

3.2. Effect of GAL on Rheumatoid Arthritis (RA)

The antioxidant and immunomodulatory properties of GAL in RA were investigated in several studies [191,192,193,194]. GAL was found to prevent osteoclastic bone destruction and inhibit osteoclastogenesis by reducing RANKL-induced JNK, p38, and NF-κB activation in osteoclast precursors and in mice with collagen-induced arthritis [191]. Santos et al. demonstrated that GAL effectively reduces the production of ROS in neutrophils and inhibits myeloperoxidase (MPO) activity [194]. Furthermore, GAL can inhibit the expression of inflammatory mediators such as IL-1β, TNF-α, IL-18, PGE2, and NO in a dose-dependent manner in RA fibroblast-like synovial cells (RAFSCs), as well as the expression of iNOS and COX-2. Additionally, GAL can increase the activity of SOD and decrease MDA content in a dose-dependent manner [192]. Fu et al. discovered that GAL could suppress pro-inflammatory signaling in synoviocytes through the inhibition of the NF-κB/NLRP3 pathway, making it a potential therapeutic agent for RA [192]. Finally, GAL can also suppress inflammation, cell proliferation, migration, and invasion, while promoting apoptosis in rheumatoid arthritis fibroblast-like synoviocytes (RAFLSs) by modulating the PI3K/AKT pathway [193]. All these studies are summarized in Table 5.

4. Conclusions and Future Perspectives

The encouraging findings from the mentioned studies suggest that GAL, a natural flavonoid compound found in various plants such as Alpinia officinarum and Alpinia galanga, holds potential as a therapeutic agent for treating rheumatoid arthritis (RA). The search strategy involved querying PubMed using MeSH terms, covering publications from 1985 to 2024. GAL was shown to possess significant anti-inflammatory and antioxidative properties, which could be beneficial in reducing the inflammation and joint damage associated with RA. The compound functions by inhibiting the production of inflammatory mediators and reducing oxidative stress within the joints.
However, further research is necessary to determine the synergistic effects of GAL compared to the first-line drug for treating RA, such as methotrexate. Additionally, its potential additive and protective effects against the side effects of conventional RA drugs need a thorough evaluation. Understanding the precise mechanisms of action and potential interactions with other medications is crucial.
Clinical studies are also required to assess the safety and efficacy of GAL in humans before considering its widespread use. This involves investigating the optimal dosage, the potential adverse effects, and understanding how GAL may interact with other medications commonly used in RA treatment.
If proven effective and safe, GAL could provide a natural alternative to conventional anti-inflammatory drugs, such as NSAIDs and DMARDs, which often have side effects with prolonged use. Moreover, these study findings could pave the way for the development of other natural compounds with similar therapeutic potential, offering new avenues for the treatment of inflammatory diseases beyond RA.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph17070963/s1, Table S1: Summary of the Chemical and Physical Properties of Galangin.

Author Contributions

G.K. and Y.E.-O.: Conceptualization and design; Y.E.-O. and A.S.: Investigation; Writing—original draft preparation; G.K. and S.A.N.: Writing—review and editing; G.K.: Supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AKT: AKT serine/threonine kinase; ACSL4: Acyl-CoA Synthetase Long-Chain Family Member 4; AD: Atopic Dermatitis; AP-1: Activator protein 1; CAT: Catalase; CCL17: Chemokine (C-C motif) ligand 17; CD8+ T cells: Cytotoxic T cells; CIH: Concanavalin A-induced Hepatitis; COX-2: Cyclooxygenase-2; CRs: Complement receptors; CXCL10: C-X-C motif chemokine ligand 10; DCs: Dendritic cells; DNMT3A: DNA methyltransferase 3A; DPP-4: Dipeptidyl peptidase 4; Egr-1: Early growth response protein 1; ERK: Mitogen-activated protein kinase; FcγRs: Fc gamma receptors; GAL: GAL; GSH: Reduced glutathione; GPx: Glutathione peroxidase; HMGB1: High mobility group box protein 1; HO-1: Heme oxygenase-1; HRP: Horseradish peroxidase; IFN-γ: Interferon gamma; IgE: Immunoglobulin E; IgG2a: Immunoglobulin G subclass 2a; IKBa: Inhibitor of kappa B alpha; IKKβ: I kappa B kinase beta; IL-1β: Interleukin 1 beta; IL-4: Interleukin 4; IL-5: Interleukin 5; IL-6: Interleukin 6; IL-12: Interleukin 12; IL-13: Interleukin 13; IL-31: Interleukin 31; IL-32: Interleukin 32; IRAK-1: Interleukin-1 receptor-associated kinase 1; JAK: Janus kinase; JNK: c-Jun N-terminal kinase; LPS: Lipopolysaccharide; M1 macrophages: Macrophage subtype with pro-inflammatory properties; M2 macrophages: Macrophage subtype with anti-inflammatory properties; MAPK: Mitogen-activated protein kinase; MCP-1: Monocyte chemoattractant protein 1; MDA: Malondialdehyde; MHC-II: Major histocompatibility complex class II; MIP-1α: Macrophage inflammatory protein 1 alpha; MMPs: Matrix metalloproteinases; MPO: Myeloperoxidase; N/D: Not determined; NF-κB: Nuclear factor kappa-light-chain-enhancer of activated B cells; NLRP3: NOD-like receptor family pyrin domain containing 3; NO: Nitric oxide; NOX-1: NADPH oxidase-1; Nrf2: Nuclear factor (erythroid-derived 2)-like 2; OVA: Ovalbumin; p65: RelA protein p65 subunit of NF-κB; PCO: Protein carbonyls; PD-L1: Programmed death ligand 1; PI3K: Phosphatidylinositol-3-kinase; PGE2: Prostaglandin E2; PPARγ: Peroxisome proliferator-activated receptor gamma; RA: Rheumatoid arthritis; RAS: Renin-angiotensin system; ROS: Reactive oxygen species; SOD: Superoxide dismutase; TBARS: Thiobarbituric Acid Reactive Substances.

References

  1. Yang, S.; Zhao, M.; Jia, S. Macrophage: Key player in the pathogenesis of autoimmune diseases. Front. Immunol. 2023, 14, 1080310. [Google Scholar] [CrossRef] [PubMed]
  2. Di Matteo, A.; Bathon, J.M.; Emery, P. Rheumatoid arthritis. Lancet 2023, 402, 2019–2033. [Google Scholar] [CrossRef] [PubMed]
  3. Venetsanopoulou, A.I.; Alamanos, Y.; Voulgari, P.V.; Drosos, A.A. Epidemiology of rheumatoid arthritis: Genetic and environmental influences. Expert. Rev. Clin. Immunol. 2022, 18, 923–931. [Google Scholar] [CrossRef] [PubMed]
  4. Cutolo, M.; Straub, R.H. Sex steroids and autoimmune rheumatic diseases: State of the art. Nat. Rev. Rheumatol. 2020, 16, 628–644. [Google Scholar] [CrossRef] [PubMed]
  5. Gravallese, E.M.; Firestein, G.S. Rheumatoid Arthritis—Common Origins, Divergent Mechanisms. N. Engl. J. Med. 2023, 388, 529–542. [Google Scholar] [CrossRef]
  6. Deane, K.D.; Demoruelle, M.K.; Kelmenson, L.B.; Kuhn, K.A.; Norris, J.M.; Holers, V.M. Genetic and environmental risk factors for rheumatoid arthritis. Best Pract. Res. Clin. Rheumatol. 2017, 31, 3–18. [Google Scholar] [CrossRef] [PubMed]
  7. Liu, X.; Tedeschi, S.K.; Barbhaiya, M.; Leatherwood, C.L.; Speyer, C.B.; Lu, B.; Costenbader, K.H.; Karlson, E.W.; Sparks, J.A. Impact and Timing of Smoking Cessation on Reducing Risk of Rheumatoid Arthritis among Women in the Nurses’ Health Studies. Arthritis Care Res. 2019, 71, 914–924. [Google Scholar] [CrossRef] [PubMed]
  8. Möller, B.; Kollert, F.; Sculean, A.; Villiger, P.M. Infectious Triggers in Periodontitis and the Gut in Rheumatoid Arthritis (RA): A Complex Story About Association and Causality. Front. Immunol. 2020, 11, 1108. [Google Scholar] [CrossRef]
  9. Ishigaki, K.; Lagattuta, K.A.; Luo, Y.; James, E.A.; Buckner, J.H.; Raychaudhuri, S. HLA autoimmune risk alleles restrict the hypervariable region of T cell receptors. Nat. Genet. 2022, 54, 393–402. [Google Scholar] [CrossRef] [PubMed]
  10. Kwon, E.J.; Ju, J.H. Impact of Posttranslational Modification in Pathogenesis of Rheumatoid Arthritis: Focusing on Citrullination, Carbamylation, and Acetylation. Int. J. Mol. Sci. 2021, 22, 10576. [Google Scholar] [CrossRef]
  11. Kissel, T.; Ge, C.; Hafkenscheid, L.; Kwekkeboom, J.C.; Slot, L.M.; Cavallari, M.; He, Y.; van Schie, K.A.; Vergroesen, R.D.; Kampstra, A.S.B.; et al. Surface Ig variable domain glycosylation affects autoantigen binding and acts as threshold for human autoreactive B cell activation. Sci. Adv. 2022, 8, eabm1759. [Google Scholar] [CrossRef] [PubMed]
  12. Wehr, P.; Purvis, H.; Law, S.C.; Thomas, R. Dendritic cells, T cells and their interaction in rheumatoid arthritis. Clin. Exp. Immunol. 2019, 196, 12–27. [Google Scholar] [CrossRef] [PubMed]
  13. Wu, F.; Gao, J.; Kang, J.; Wang, X.; Niu, Q.; Liu, J.; Zhang, L. B Cells in Rheumatoid Arthritis: Pathogenic Mechanisms and Treatment Prospects. Front. Immunol. 2021, 12, 750753. [Google Scholar] [CrossRef] [PubMed]
  14. Pruijn, G.J.; Wiik, A.; van Venrooij, W.J. The use of citrullinated peptides and proteins for the diagnosis of rheumatoid arthritis. Arthritis Res. Ther. 2010, 12, 203. [Google Scholar] [CrossRef] [PubMed]
  15. 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]
  16. Rönnelid, J.; Turesson, C.; Kastbom, A. Autoantibodies in Rheumatoid Arthritis—Laboratory and Clinical Perspectives. Front. Immunol. 2021, 12, 685312. [Google Scholar] [CrossRef] [PubMed]
  17. Kongpachith, S.; Lingampalli, N.; Ju, C.H.; Blum, L.K.; Lu, D.R.; Elliott, S.E.; Mao, R.; Robinson, W.H. Affinity Maturation of the Anti-Citrullinated Protein Antibody Paratope Drives Epitope Spreading and Polyreactivity in Rheumatoid Arthritis. Arthritis Rheumatol. 2019, 71, 507–517. [Google Scholar] [CrossRef] [PubMed]
  18. Jiang, Q.; Yang, G.; Liu, Q.; Wang, S.; Cui, D. Function and Role of Regulatory T Cells in Rheumatoid Arthritis. Front. Immunol. 2021, 12, 626193. [Google Scholar] [CrossRef] [PubMed]
  19. Cao, G.; Chi, S.; Wang, X.; Sun, J.; Zhang, Y. CD4+CXCR5+PD-1+ T Follicular Helper Cells Play a Pivotal Role in the Development of Rheumatoid Arthritis. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2019, 25, 3032–3040. [Google Scholar] [CrossRef]
  20. Onishi, R.M.; Gaffen, S.L. Interleukin-17 and its target genes: Mechanisms of interleukin-17 function in disease. Immunology 2010, 129, 311–321. [Google Scholar] [CrossRef]
  21. Alivernini, S.; MacDonald, L.; Elmesmari, A.; Finlay, S.; Tolusso, B.; Gigante, M.R.; Petricca, L.; Di Mario, C.; Bui, L.; Perniola, S.; et al. Distinct synovial tissue macrophage subsets regulate inflammation and remission in rheumatoid arthritis. Nat. Med. 2020, 26, 1295–1306. [Google Scholar] [CrossRef] [PubMed]
  22. Takaki-Kuwahara, A.; Arinobu, Y.; Miyawaki, K.; Yamada, H.; Tsuzuki, H.; Irino, K.; Ayano, M.; Kimoto, Y.; Mitoma, H.; Akahoshi, M.; et al. CCR6+ group 3 innate lymphoid cells accumulate in inflamed joints in rheumatoid arthritis and produce Th17 cytokines. Arthritis Res. Ther. 2019, 21, 198. [Google Scholar] [CrossRef] [PubMed]
  23. Culemann, S.; Grüneboom, A.; Nicolás-Ávila, J.; Weidner, D.; Lämmle, K.F.; Rothe, T.; Quintana, J.A.; Kirchner, P.; Krljanac, B.; Eberhardt, M.; et al. Locally renewing resident synovial macrophages provide a protective barrier for the joint. Nature 2019, 572, 670–675. [Google Scholar] [CrossRef] [PubMed]
  24. Kuo, D.; Ding, J.; Cohn, I.S.; Zhang, F.; Wei, K.; Rao, D.A.; Rozo, C.; Sokhi, U.K.; Shanaj, S.; Oliver, D.J.; et al. HBEGF(+) macrophages in rheumatoid arthritis induce fibroblast invasiveness. Sci. Transl. Med. 2019, 11, eaau8587. [Google Scholar] [CrossRef] [PubMed]
  25. Müller-Ladner, U.; Pap, T.; Gay, R.E.; Neidhart, M.; Gay, S. Mechanisms of Disease: The molecular and cellular basis of joint destruction in rheumatoid arthritis. Nat. Clin. Pract. Rheumatol. 2005, 1, 102–110. [Google Scholar] [CrossRef] [PubMed]
  26. Komatsu, N.; Win, S.; Yan, M.; Huynh, N.C.; Sawa, S.; Tsukasaki, M.; Terashima, A.; Pluemsakunthai, W.; Kollias, G.; Nakashima, T.; et al. Plasma cells promote osteoclastogenesis and periarticular bone loss in autoimmune arthritis. J. Clin. Investig. 2021, 131, e143060. [Google Scholar] [CrossRef] [PubMed]
  27. Danks, L.; Komatsu, N.; Guerrini, M.M.; Sawa, S.; Armaka, M.; Kollias, G.; Nakashima, T.; Takayanagi, H. RANKL expressed on synovial fibroblasts is primarily responsible for bone erosions during joint inflammation. Ann. Rheum. Dis. 2016, 75, 1187–1195. [Google Scholar] [CrossRef] [PubMed]
  28. O’Brien, W.; Fissel, B.M.; Maeda, Y.; Yan, J.; Ge, X.; Gravallese, E.M.; Aliprantis, A.O.; Charles, J.F. RANK-Independent Osteoclast Formation and Bone Erosion in Inflammatory Arthritis. Arthritis Rheumatol. 2016, 68, 2889–2900. [Google Scholar] [CrossRef] [PubMed]
  29. Guo, D.; Lin, C.; Lu, Y.; Guan, H.; Qi, W.; Zhang, H.; Shao, Y.; Zeng, C.; Zhang, R.; Zhang, H.; et al. FABP4 secreted by M1-polarized macrophages promotes synovitis and angiogenesis to exacerbate rheumatoid arthritis. Bone Res. 2022, 10, 45. [Google Scholar] [CrossRef]
  30. Conforti, A.; Di Cola, I.; Pavlych, V.; Ruscitti, P.; Berardicurti, O.; Ursini, F.; Giacomelli, R.; Cipriani, P. Beyond the joints, the extra-articular manifestations in rheumatoid arthritis. Autoimmun. Rev. 2021, 20, 102735. [Google Scholar] [CrossRef]
  31. Gao, D.; Gao, X.; Yang, F.; Wang, Q. Neuroimmune Crosstalk in Rheumatoid Arthritis. Int. J. Mol. Sci. 2022, 23, 8158. [Google Scholar] [CrossRef] [PubMed]
  32. Roth, J.; Blatteis, C.M. Mechanisms of fever production and lysis: Lessons from experimental LPS fever. Compr. Physiol. 2014, 4, 1563–1604. [Google Scholar] [CrossRef] [PubMed]
  33. Lora, V.; Cerroni, L.; Cota, C. Skin manifestations of rheumatoid arthritis. G. Ital. Dermatol. Venereol. 2018, 153, 243–255. [Google Scholar] [CrossRef] [PubMed]
  34. Tilstra, J.S.; Lienesch, D.W. Rheumatoid Nodules. Dermatol. Clin. 2015, 33, 361–371. [Google Scholar] [CrossRef] [PubMed]
  35. Chen, Y.; Xu, W.; Yang, H.; Shao, M.; Xu, S.; Deng, J.; Gao, X.; Liu, H.; Shuai, Z.; Xu, S.; et al. Serum Levels of Hepcidin in Rheumatoid Arthritis and Its Correlation with Disease Activity and Anemia: A Meta-analysis. Immunol. Investig. 2021, 50, 243–258. [Google Scholar] [CrossRef] [PubMed]
  36. Sato, H.; Takai, C.; Kazama, J.J.; Wakamatsu, A.; Hasegawa, E.; Kobayashi, D.; Kondo, N.; Nakatsue, T.; Abe, A.; Ito, S.; et al. Serum hepcidin level, iron metabolism and osteoporosis in patients with rheumatoid arthritis. Sci. Rep. 2020, 10, 9882. [Google Scholar] [CrossRef] [PubMed]
  37. Kadura, S.; Raghu, G. Rheumatoid arthritis-interstitial lung disease: Manifestations and current concepts in pathogenesis and management. Eur. Respir. Rev. Off. J. Eur. Respir. Soc. 2021, 30, 210011. [Google Scholar] [CrossRef] [PubMed]
  38. England, B.R.; Duryee, M.J.; Roul, P.; Mahajan, T.D.; Singh, N.; Poole, J.A.; Ascherman, D.P.; Caplan, L.; Demoruelle, M.K.; Deane, K.D.; et al. Malondialdehyde-Acetaldehyde Adducts and Antibody Responses in Rheumatoid Arthritis-Associated Interstitial Lung Disease. Arthritis Rheumatol. 2019, 71, 1483–1493. [Google Scholar] [CrossRef] [PubMed]
  39. Liu, S.; Ma, H.; Zhang, H.; Deng, C.; Xin, P. Recent advances on signaling pathways and their inhibitors in rheumatoid arthritis. Clin. Immunol. 2021, 230, 108793. [Google Scholar] [CrossRef]
  40. Malemud, C.J. Negative Regulators of JAK/STAT Signaling in Rheumatoid Arthritis and Osteoarthritis. Int. J. Mol. Sci. 2017, 18, 484. [Google Scholar] [CrossRef]
  41. Ilchovska, D.D.; Barrow, D.M. An Overview of the NF-kB mechanism of pathophysiology in rheumatoid arthritis, investigation of the NF-kB ligand RANKL and related nutritional interventions. Autoimmun. Rev. 2021, 20, 102741. [Google Scholar] [CrossRef] [PubMed]
  42. Du, H.; Zhang, X.; Zeng, Y.; Huang, X.; Chen, H.; Wang, S.; Wu, J.; Li, Q.; Zhu, W.; Li, H.; et al. A Novel Phytochemical, DIM, Inhibits Proliferation, Migration, Invasion and TNF-α Induced Inflammatory Cytokine Production of Synovial Fibroblasts from Rheumatoid Arthritis Patients by Targeting MAPK and AKT/mTOR Signal Pathway. Front. Immunol. 2019, 10, 1620. [Google Scholar] [CrossRef] [PubMed]
  43. Hu, L.; Liu, R.; Zhang, L. Advance in bone destruction participated by JAK/STAT in rheumatoid arthritis and therapeutic effect of JAK/STAT inhibitors. Int. Immunopharmacol. 2022, 111, 109095. [Google Scholar] [CrossRef] [PubMed]
  44. Ba, X.; Huang, Y.; Shen, P.; Huang, Y.; Wang, H.; Han, L.; Lin, W.J.; Yan, H.J.; Xu, L.J.; Qin, K.; et al. WTD Attenuating Rheumatoid Arthritis via Suppressing Angiogenesis and Modulating the PI3K/AKT/mTOR/HIF-1α Pathway. Front. Pharmacol. 2021, 12, 696802. [Google Scholar] [CrossRef] [PubMed]
  45. Fonseca, L.J.S.d.; Nunes-Souza, V.; Goulart, M.O.F.; Rabelo, L.A. Oxidative Stress in Rheumatoid Arthritis: What the Future Might Hold regarding Novel Biomarkers and Add-On Therapies. Oxidative Med. Cell. Longev. 2019, 2019, 7536805. [Google Scholar] [CrossRef] [PubMed]
  46. Han, Z.; Gao, X.; Wang, Y.; Cheng, S.; Zhong, X.; Xu, Y.; Zhou, X.; Zhang, Z.; Liu, Z.; Cheng, L. Ultrasmall iron-quercetin metal natural product nanocomplex with antioxidant and macrophage regulation in rheumatoid arthritis. Acta Pharm. Sin. B 2023, 13, 1726–1739. [Google Scholar] [CrossRef] [PubMed]
  47. López-Armada, M.J.; Fernández-Rodríguez, J.A.; Blanco, F.J. Mitochondrial Dysfunction and Oxidative Stress in Rheumatoid Arthritis. Antioxidants 2022, 11, 1151. [Google Scholar] [CrossRef] [PubMed]
  48. Behl, T.; Upadhyay, T.; Singh, S.; Chigurupati, S.; Alsubayiel, A.M.; Mani, V.; Vargas-De-La-Cruz, C.; Uivarosan, D.; Bustea, C.; Sava, C.; et al. Polyphenols Targeting MAPK Mediated Oxidative Stress and Inflammation in Rheumatoid Arthritis. Molecules 2021, 26, 6570. [Google Scholar] [CrossRef]
  49. Huang, Y.; Guo, L.; Chitti, R.; Sreeharsha, N.; Mishra, A.; Gubbiyappa, S.K.; Singh, Y. Wogonin ameliorate complete Freund’s adjuvant induced rheumatoid arthritis via targeting NF-κB/MAPK signaling pathway. BioFactors 2020, 46, 283–291. [Google Scholar] [CrossRef]
  50. Puppala, E.R.; Jain, S.; Saha, P.; Rachamalla, M.; Np, S.; Yalamarthi, S.S.; Abubakar, M.; Chaudhary, A.; Chamundeswari, D.; Usn, M.; et al. Perillyl alcohol attenuates rheumatoid arthritis via regulating TLR4/NF-κB and Keap1/Nrf2 signaling pathways: A comprehensive study onin-vitro and in-vivo experimental models. Phytomed. Int. J. Phytother. Phytopharm. 2022, 97, 153926. [Google Scholar] [CrossRef]
  51. Sies, H. Oxidative stress: A concept in redox biology and medicine. Redox Biol. 2015, 4, 180–183. [Google Scholar] [CrossRef] [PubMed]
  52. Forrester, S.J.; Kikuchi, D.S.; Hernandes, M.S.; Xu, Q.; Griendling, K.K. Reactive Oxygen Species in Metabolic and Inflammatory Signaling. Circ. Res. 2018, 122, 877–902. [Google Scholar] [CrossRef] [PubMed]
  53. Carr, A.C.; Maggini, S. Vitamin C and Immune Function. Nutrients 2017, 9, 1211. [Google Scholar] [CrossRef] [PubMed]
  54. Katsuoka, F.; Yamamoto, M. Small Maf proteins (MafF, MafG, MafK): History, structure and function. Gene 2016, 586, 197–205. [Google Scholar] [CrossRef] [PubMed]
  55. Esalatmanesh, K.; Loghman, A.; Esalatmanesh, R.; Soleimani, Z.; Khabbazi, A.; Mahdavi, A.M.; Mousavi, S.G.A. Effects of melatonin supplementation on disease activity, oxidative stress, inflammatory, and metabolic parameters in patients with rheumatoid arthritis: A randomized double-blind placebo-controlled trial. Clin. Rheumatol. 2021, 40, 3591–3597. [Google Scholar] [CrossRef] [PubMed]
  56. Zamani, B.; Taghvaee, F.; Akbari, H.; Mohtashamian, A.; Sharifi, N. Effects of Selenium Supplementation on the Indices of Disease Activity, Inflammation and Oxidative Stress in Patients with Rheumatoid Arthritis: A Randomized Clinical Trial. Biol. Trace Elem. Res. 2024, 202, 1457–1467. [Google Scholar] [CrossRef] [PubMed]
  57. Liu, X.; Wang, Z.; Qian, H.; Tao, W.; Zhang, Y.; Hu, C.; Mao, W.; Guo, Q. Natural medicines of targeted rheumatoid arthritis and its action mechanism. Front. Immunol. 2022, 13, 945129. [Google Scholar] [CrossRef]
  58. Shiozawa, S.; Tsumiyama, K. Pathogenesis of rheumatoid arthritis and c-Fos/AP-1. Cell Cycle 2009, 8, 1539–1543. [Google Scholar] [CrossRef] [PubMed]
  59. Islam, S.M.T.; Won, J.; Khan, M.; Mannie, M.D.; Singh, I. Hypoxia-inducible factor-1 drives divergent immunomodulatory functions in the pathogenesis of autoimmune diseases. Immunology 2021, 164, 31–42. [Google Scholar] [CrossRef] [PubMed]
  60. Chadha, S.; Behl, T.; Kumar, A.; Khullar, G.; Arora, S. Role of Nrf2 in rheumatoid arthritis. Curr. Res. Transl. Med. 2020, 68, 171–181. [Google Scholar] [CrossRef]
  61. Fraenkel, L.; Bathon, J.M.; England, B.R.; St Clair, E.W.; Arayssi, T.; Carandang, K.; Deane, K.D.; Genovese, M.; Huston, K.K.; Kerr, G.; et al. 2021 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis. Arthritis Care Res. 2021, 73, 924–939. [Google Scholar] [CrossRef] [PubMed]
  62. Singh, J.A. Treatment Guidelines in Rheumatoid Arthritis. Rheum. Dis. Clin. N. Am. 2022, 48, 679–689. [Google Scholar] [CrossRef] [PubMed]
  63. Prasad, P.; Verma, S.; Surbhi; Ganguly, N.K.; Chaturvedi, V.; Mittal, S.A. Rheumatoid arthritis: Advances in treatment strategies. Mol. Cell Biochem. 2023, 478, 69–88. [Google Scholar] [CrossRef] [PubMed]
  64. Radu, A.F.; Bungau, S.G. Management of Rheumatoid Arthritis: An Overview. Cells 2021, 10, 2857. [Google Scholar] [CrossRef] [PubMed]
  65. Lin, Y.J.; Anzaghe, M.; Schulke, S. Update on the Pathomechanism, Diagnosis, and Treatment Options for Rheumatoid Arthritis. Cells 2020, 9, 880. [Google Scholar] [CrossRef] [PubMed]
  66. Kerschbaumer, A.; Sepriano, A.; Bergstra, S.A.; Smolen, J.S.; van der Heijde, D.; Caporali, R.; Edwards, C.J.; Verschueren, P.; de Souza, S.; Pope, J.E.; et al. Efficacy of synthetic and biological DMARDs: A systematic literature review informing the 2022 update of the EULAR recommendations for the management of rheumatoid arthritis. Ann. Rheum. Dis. 2023, 82, 95–106. [Google Scholar] [CrossRef] [PubMed]
  67. Zhang, B.; Wang, Y.; Yuan, Y.; Sun, J.; Liu, L.; Huang, D.; Hu, J.; Wang, M.; Li, S.; Song, W.; et al. In vitro elimination of autoreactive B cells from rheumatoid arthritis patients by universal chimeric antigen receptor T cells. Ann. Rheum. Dis. 2021, 80, 176–184. [Google Scholar] [CrossRef] [PubMed]
  68. Arneson, L.C.; Carroll, K.J.; Ruderman, E.M. Bruton’s Tyrosine Kinase Inhibition for the Treatment of Rheumatoid Arthritis. ImmunoTargets Ther. 2021, 10, 333–342. [Google Scholar] [CrossRef] [PubMed]
  69. Zamri, F.; de Vries, T.J. Use of TNF Inhibitors in Rheumatoid Arthritis and Implications for the Periodontal Status: For the Benefit of Both? Front. Immunol. 2020, 11, 591365. [Google Scholar] [CrossRef]
  70. Lucas, C.; Perdriger, A.; Amé, P. Definition of B cell helper T cells in rheumatoid arthritis and their behavior during treatment. Semin. Arthritis Rheum. 2020, 50, 867–872. [Google Scholar] [CrossRef]
  71. Humby, F.; Durez, P.; Buch, M.H.; Lewis, M.J.; Rizvi, H.; Rivellese, F.; Nerviani, A.; Giorli, G.; Mahto, A.; Montecucco, C.; et al. Rituximab versus tocilizumab in anti-TNF inadequate responder patients with rheumatoid arthritis (R4RA): 16-week outcomes of a stratified, biopsy-driven, multicentre, open-label, phase 4 randomised controlled trial. Lancet 2021, 397, 305–317. [Google Scholar] [CrossRef] [PubMed]
  72. Mysler, E.; Caubet, M.; Lizarraga, A. Current and Emerging DMARDs for the Treatment of Rheumatoid Arthritis. Open Access Rheumatol. Res. Rev. 2021, 13, 139–152. [Google Scholar] [CrossRef]
  73. Sbidian, E.; Chaimani, A.; Garcia-Doval, I.; Doney, L.; Dressler, C.; Hua, C.; Hughes, C.; Naldi, L.; Afach, S.; Le Cleach, L. Systemic pharmacological treatments for chronic plaque psoriasis: A network meta-analysis. Cochrane Database Syst. Rev. 2022, 5, Cd011535. [Google Scholar] [CrossRef]
  74. Friedman, B.; Cronstein, B. Methotrexate mechanism in treatment of rheumatoid arthritis. Jt. Bone Spine 2019, 86, 301–307. [Google Scholar] [CrossRef]
  75. Hamed, K.M.; Dighriri, I.M.; Baomar, A.F.; Alharthy, B.T.; Alenazi, F.E.; Alali, G.H.; Alenazy, R.H.; Alhumaidi, N.T.; Alhulayfi, D.H.; Alotaibi, Y.B.; et al. Overview of Methotrexate Toxicity: A Comprehensive Literature Review. Cureus 2022, 14, e29518. [Google Scholar] [CrossRef] [PubMed]
  76. Ismail, M.; Hasan, H.; El-Orfali, Y.; Ismail, H.; Khawaja, G. Anti-Inflammatory, Antioxidative, and Hepatoprotective Effects of Trans Delta9-Tetrahydrocannabinol/Sesame Oil on Adjuvant-Induced Arthritis in Rats. Evid.-Based Complement. Altern. Med. 2018, 2018, 9365464. [Google Scholar] [CrossRef]
  77. Hasan, H.; Ismail, H.; El-Orfali, Y.; Khawaja, G. Therapeutic benefits of Indole-3-Carbinol in adjuvant-induced arthritis and its protective effect against methotrexate induced-hepatic toxicity. BMC Complement. Altern. Med. 2018, 18, 337. [Google Scholar] [CrossRef]
  78. Kamal, R.M.; Sabry, M.M.; Aly, Z.Y.; Hifnawy, M.S. Phytochemical and In-Vivo Anti-Arthritic Significance of Aloe thraskii Baker in Combined Therapy with Methotrexate in Adjuvant-Induced Arthritis in Rats. Molecules 2021, 26, 3660. [Google Scholar] [CrossRef] [PubMed]
  79. Banji, D.; Banji, O.J.F.; Rashida, S.; Alshahrani, S.; Alqahtani, S.S. Bioavailability, anti-inflammatory and anti-arthritic effect of Acetyl Keto Boswellic acid and its combination with methotrexate in an arthritic animal model. J. Ethnopharmacol. 2022, 292, 115200. [Google Scholar] [CrossRef]
  80. Dogra, A.; Kour, D.; Bhardwaj, M.; Dhiman, S.; Kumar, A.; Vij, B.; Kumar, A.; Nandi, U. Glabridin Plays Dual Action to Augment the Efficacy and Attenuate the Hepatotoxicity of Methotrexate in Arthritic Rats. ACS Omega 2022, 7, 34341–34351. [Google Scholar] [CrossRef]
  81. Khawaja, G.; El-Orfali, Y. Silibinin’s Effects against Methotrexate-Induced Hepatotoxicity in Adjuvant-Induced Arthritis Rat Model. Pharmaceuticals 2024, 17, 431. [Google Scholar] [CrossRef] [PubMed]
  82. Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef] [PubMed]
  83. Tungmunnithum, D.; Drouet, S.; Garros, L.; Hano, C. Differential Flavonoid and Other Phenolic Accumulations and Antioxidant Activities of Nymphaea lotus L. Populations throughout Thailand. Molecules 2022, 27, 3590. [Google Scholar] [CrossRef] [PubMed]
  84. Othman, M.S.; Khaled, A.M.; Al-Bagawi, A.H.; Fareid, M.A.; Hameed, R.A.; Zahra, F.A.A.; Moneim, A.E.A. Echinops spinosus effect against diabetes and its hepatorenal complications: Total extract and flavonoids fraction. Environ. Sci. Pollut. Res. Int. 2022, 29, 38606–38617. [Google Scholar] [CrossRef] [PubMed]
  85. Liu, D.; You, P.; Luo, Y.; Yang, M.; Liu, Y. Galangin Induces Apoptosis in MCF-7 Human Breast Cancer Cells Through Mitochondrial Pathway and Phosphatidylinositol 3-Kinase/Akt Inhibition. Pharmacology 2018, 102, 58–66. [Google Scholar] [CrossRef]
  86. Alfwuaires, M.A. Galangin mitigates oxidative stress, inflammation, and apoptosis in a rat model of methotrexate hepatotoxicity. Environ. Sci. Pollut. Res. Int. 2022, 29, 20279–20288. [Google Scholar] [CrossRef]
  87. Tan, K.T.; Li, S.; Panny, L.; Lin, C.C.; Lin, S.C. Galangin ameliorates experimental autoimmune encephalomyelitis in mice via modulation of cellular immunity. J. Immunotoxicol. 2021, 18, 50–60. [Google Scholar] [CrossRef]
  88. Wang, D.; Chen, J.; Pu, L.; Yu, L.; Xiong, F.; Sun, L.; Yu, Q.; Cao, X.; Chen, Y.; Peng, F.; et al. Galangin: A food-derived flavonoid with therapeutic potential against a wide spectrum of diseases. Phytother. Res. 2023, 37, 5700–5723. [Google Scholar] [CrossRef] [PubMed]
  89. Shen, S.; Wang, J.; Chen, X.; Liu, T.; Zhuo, Q.; Zhang, S.Q. Evaluation of cellular antioxidant components of honeys using UPLC-MS/MS and HPLC-FLD based on the quantitative composition-activity relationship. Food Chem. 2019, 293, 169–177. [Google Scholar] [CrossRef]
  90. Xu, W.; Lu, H.; Yuan, Y.; Deng, Z.; Zheng, L.; Li, H. The Antioxidant and Anti-Inflammatory Effects of Flavonoids from Propolis via Nrf2 and NF-kappaB Pathways. Foods 2022, 11, 2439. [Google Scholar] [CrossRef]
  91. Sahin, S.; Karkar, B. The antioxidant properties of the chestnut bee pollen extract and its preventive action against oxidatively induced damage in DNA bases. J. Food Biochem. 2019, 43, e12888. [Google Scholar] [CrossRef] [PubMed]
  92. Saric, A.; Balog, T.; Sobocanec, S.; Kusic, B.; Sverko, V.; Rusak, G.; Likic, S.; Bubalo, D.; Pinto, B.; Reali, D.; et al. Antioxidant effects of flavonoid from Croatian Cystus incanus L. rich bee pollen. Food Chem. Toxicol. 2009, 47, 547–554. [Google Scholar] [CrossRef] [PubMed]
  93. Song, H.K.; Park, S.H.; Kim, H.J.; Jang, S.; Kim, T. Alpinia officinarum water extract inhibits the atopic dermatitis-like responses in NC/Nga mice by regulation of inflammatory chemokine production. Biomed. Pharmacother. 2021, 144, 112322. [Google Scholar] [CrossRef] [PubMed]
  94. Erusappan, T.; Paramasivam, S.; Ekambaram, S.P. Identification of galangin as the bioactive compound from Alpinia calcarata (Haw.) Roscoe rhizomes to inhibit IRAK-1/ MAPK/ NF-kappaB p65 and JAK-1 signaling in LPS stimulated RAW 264.7 cells. J. Ethnopharmacol. 2022, 288, 114975. [Google Scholar] [CrossRef] [PubMed]
  95. Upadhye, A.S.; Rajopadhye, A.; Dias, L. Development and validation of HPTLC fingerprints of three species of Alpinia with biomarker Galangin. BMC Complement. Altern. Med. 2018, 18, 16. [Google Scholar] [CrossRef] [PubMed]
  96. Mohamed Azar, K.A.H.; Ezhilarasan, D.; Shree Harini, K. Coleus vettiveroides ethanolic root extract induces cytotoxicity by intrinsic apoptosis in HepG2 cells. J. Appl. Toxicol. 2024, 44, 245–259. [Google Scholar] [CrossRef] [PubMed]
  97. Lin, L.-Z.; Mukhopadhyay, S.; Robbins, R.J.; Harnly, J.M. Identification and quantification of flavonoids of Mexican oregano (Lippia graveolens) by LC-DAD-ESI/MS analysis. J. Food Compos. Anal. 2007, 20, 361–369. [Google Scholar] [CrossRef] [PubMed]
  98. Cortes-Chitala, M.D.C.; Flores-Martinez, H.; Orozco-Avila, I.; Leon-Campos, C.; Suarez-Jacobo, A.; Estarron-Espinosa, M.; Lopez-Muraira, I. Identification and Quantification of Phenolic Compounds from Mexican Oregano (Lippia graveolens HBK) Hydroethanolic Extracts and Evaluation of Its Antioxidant Capacity. Molecules 2021, 26, 702. [Google Scholar] [CrossRef] [PubMed]
  99. Gyeltshen, T.; Jordan, G.J.; Smith, J.A.; Bissember, A.C. Natural products isolation studies of the paleoendemic plant species Nothofagus gunnii and Nothofagus cunninghamii. Fitoterapia 2022, 156, 105088. [Google Scholar] [CrossRef]
  100. Barauna, S.C.; Delwing-Dal Magro, D.; Brueckheimer, M.B.; Maia, T.P.; Sala, G.; Dohler, A.W.; Harger, M.C.; de Melo, D.F.M.; de Gasper, A.L.; Alberton, M.D.; et al. Antioxidant and antidepressant-like effects of Eugenia catharinensis D. Legrand in an animal model of depression induced by corticosterone. Metab. Brain Dis. 2018, 33, 1985–1994. [Google Scholar] [CrossRef]
  101. Basile, A.; Sorbo, S.; Giordano, S.; Ricciardi, L.; Ferrara, S.; Montesano, D.; Castaldo Cobianchi, R.; Vuotto, M.L.; Ferrara, L. Antibacterial and allelopathic activity of extract from Castanea sativa leaves. Fitoterapia 2000, 71 (Suppl. S1), S110–S116. [Google Scholar] [CrossRef]
  102. de Zorzi, V.N.; Haupenthal, F.; Cardoso, A.S.; Cassol, G.; Facundo, V.A.; Balico, L.J.; Lima, D.K.S.; Santos, A.R.S.; Furian, A.F.; Oliveira, M.S.; et al. Galangin Prevents Increased Susceptibility to Pentylenetetrazol-Stimulated Seizures by Prostaglandin E2. Neuroscience 2019, 413, 154–168. [Google Scholar] [CrossRef]
  103. Ludwiczuk, A.; Saha, A.; Kuzuhara, T.; Asakawa, Y. Bioactivity guided isolation of anticancer constituents from leaves of Alnus sieboldiana (Betulaceae). Phytomedicine 2011, 18, 491–498. [Google Scholar] [CrossRef] [PubMed]
  104. Jokubaite, M.; Pukenaite, G.; Marksa, M.; Ramanauskiene, K. Balsam Poplar Buds Extracts-Loaded Gels and Emulgels: Development, Biopharmaceutical Evaluation, and Biological Activity In Vitro. Gels 2023, 9, 821. [Google Scholar] [CrossRef] [PubMed]
  105. Yang, S.; Zhou, Y.; Ye, J.; Fan, G.; Peng, L.; Pan, S. Effects of poplar buds as an alternative to propolis on postharvest diseases control of strawberry fruits. J. Sci. Food Agric. 2016, 96, 2136–2141. [Google Scholar] [CrossRef] [PubMed]
  106. Hage, S.; Morlock, G.E. Bioprofiling of Salicaceae bud extracts through high-performance thin-layer chromatography hyphenated to biochemical, microbiological and chemical detections. J. Chromatogr. A 2017, 1490, 201–211. [Google Scholar] [CrossRef] [PubMed]
  107. Cui-ping, Z.; Shuai, H.; Wen-ting, W.; Shun, P.; Xiao-ge, S.; Ya-jing, L.; Fu-liang, H. Development of high-performance liquid chromatographic for quality and authenticity control of Chinese propolis. J. Food Sci. 2014, 79, C1315–C1322. [Google Scholar] [CrossRef]
  108. Aguiniga-Sanchez, I.; Soto-Hernandez, M.; Cadena-Iniguez, J.; Suwalsky, M.; Colina, J.R.; Castillo, I.; Rosado-Perez, J.; Mendoza-Nunez, V.M.; Santiago-Osorio, E. Phytochemical Analysis and Antioxidant and Anti-Inflammatory Capacity of the Extracts of Fruits of the Sechium Hybrid. Molecules 2020, 25, 4637. [Google Scholar] [CrossRef]
  109. Salazar-Aguilar, S.; Ruiz-Posadas, L.D.M.; Cadena-Iniguez, J.; Soto-Hernandez, M.; Santiago-Osorio, E.; Aguiniga-Sanchez, I.; Rivera-Martinez, A.R.; Aguirre-Medina, J.F. Sechium edule (Jacq.) Swartz, a New Cultivar with Antiproliferative Potential in a Human Cervical Cancer HeLa Cell Line. Nutrients 2017, 9, 798. [Google Scholar] [CrossRef]
  110. Grutzmann Arcari, S.; Arena, K.; Kolling, J.; Rocha, P.; Dugo, P.; Mondello, L.; Cacciola, F. Polyphenolic compounds with biological activity in guabiroba fruits (Campomanesia xanthocarpa Berg.) by comprehensive two-dimensional liquid chromatography. Electrophoresis 2020, 41, 1784–1792. [Google Scholar] [CrossRef]
  111. Alrgei, H.O.S.; Dabić, D.Č.; Natić, M.M.; Rakonjac, V.S.; Milojković-Opsenica, D.; Tešić, Ž.L.; Fotirić Akšić, M.M. Chemical profile of major taste- and health-related compounds of Oblačinska sour cherry. J. Sci. Food Agric. 2015, 96, 1241–1251. [Google Scholar] [CrossRef] [PubMed]
  112. Seraglio, S.K.T.; Schulz, M.; Silva, B.; Pasini Deolindo, C.T.; Hoff, R.B.; Gonzaga, L.V.; Fett, R.; Costa, A.C.O. Chemical Constituents and Antioxidant Potential of Red Guava (Psidium cattleianum Sabine) from Southern Brazil in Different Edible Ripening Stages. Plant Foods Hum. Nutr. 2024, 79, 166–172. [Google Scholar] [CrossRef] [PubMed]
  113. Deng, G.F.; Shen, C.; Xu, X.R.; Kuang, R.D.; Guo, Y.J.; Zeng, L.S.; Gao, L.L.; Lin, X.; Xie, J.F.; Xia, E.Q.; et al. Potential of fruit wastes as natural resources of bioactive compounds. Int. J. Mol. Sci. 2012, 13, 8308–8323. [Google Scholar] [CrossRef]
  114. Wang, B.; Hui, Y.; Liu, L.; Zhao, A.; Chiou, Y.S.; Zhang, F.; Pan, M.H. Optimized Extraction of Phenolics from Jujube Peel and Their Anti-inflammatory Effects in LPS-Stimulated Murine Macrophages. J. Agric. Food Chem. 2019, 67, 1666–1673. [Google Scholar] [CrossRef] [PubMed]
  115. Kubo, I.; Kinst-Hori, I. Flavonols from saffron flower: Tyrosinase inhibitory activity and inhibition mechanism. J. Agric. Food Chem. 1999, 47, 4121–4125. [Google Scholar] [CrossRef]
  116. Kamat, S.; Kumari, M.; Sajna, K.V.; Singh, S.K.; Kaushalendra; Kumar, A.; Jayabaskaran, C. Improved Chrysin Production by a Combination of Fermentation Factors and Elicitation from Chaetomium globosum. Microorganisms 2023, 11, 999. [Google Scholar] [CrossRef]
  117. Ouyang, X.; Li, X.; Lu, W.; Zhao, X.; Chen, D. A Null B-Ring Improves the Antioxidant Levels of Flavonol: A Comparative Study between Galangin and 3,5,7-Trihydroxychromone. Molecules 2018, 23, 3083. [Google Scholar] [CrossRef] [PubMed]
  118. Ye, W.; Sun, W.; Chen, R.; Wang, Z.; Cui, X.; Zhang, H.; Qian, S.; Zheng, Q.; Zhou, Y.; Wan, J.; et al. Pharmacokinetics in rat plasma and tissue distribution in mice of galangin determined by UHPLC–MS/MS. Acta Chromatogr. 2019, 31, 120–125. [Google Scholar] [CrossRef]
  119. Liu, R.; Li, H.; Wei, N.; Tan, Y. Simultaneous determination of two galangin metabolites from Alpinia Officinarum Hance in rat plasma by UF LC-MS/MS and its application in pharmacokinetics study. PeerJ 2021, 9, e11041. [Google Scholar] [CrossRef] [PubMed]
  120. Zhang, X.; Cheng, S.; Li, H.; Zhang, X.; Chen, F.; Li, Y.; Zhang, J.; Tan, Y. Isolation and identification of two galangin metabolites from rat urine and determination of their in vitro hypolipidemic activity. Trop. J. Pharm. Res. 2016, 15, 1235–1241. [Google Scholar] [CrossRef]
  121. Yao, H.; Lu, H.; Zhang, J.; Xue, X.; Yin, C.; Hu, J.; Zou, R.; Wang, L.; Xu, H. Preparation of Prolonged-Circulating Galangin-Loaded Liposomes and Evaluation of Antitumor Efficacy In Vitro and Pharmacokinetics In Vivo. J. Nanomater. 2019, 2019, 7236895. [Google Scholar] [CrossRef]
  122. Patil, S.; Ujalambkar, V.; Rathore, A.; Rojatkar, S.; Pokharkar, V. Galangin loaded galactosylated pluronic F68 polymeric micelles for liver targeting. Biomed. Pharmacother. 2019, 112, 108691. [Google Scholar] [CrossRef] [PubMed]
  123. Hajipour, H.; Nouri, M.; Ghorbani, M.; Bahramifar, A.; Emameh, R.Z.; Taheri, R.A. Targeted nanostructured lipid carrier containing galangin as a promising adjuvant for improving cytotoxic effects of chemotherapeutic agents. Naunyn Schmiedeberg’s Arch. Pharmacol. 2021, 394, 2353–2362. [Google Scholar] [CrossRef] [PubMed]
  124. Xiong, Y.; Wu, B.; Guo, X.; Shi, D.; Xia, H.; Xu, H.; Liu, X. Galangin delivered by retinoic acid-modified nanoparticles targeted hepatic stellate cells for the treatment of hepatic fibrosis. RSC Adv. 2023, 13, 10987–11001. [Google Scholar] [CrossRef] [PubMed]
  125. Abbas, Z.S.; Sulaiman, G.M.; Jabir, M.S.; Mohammed, S.A.A.; Khan, R.A.; Mohammed, H.A.; Al-Subaiyel, A. Galangin/beta-Cyclodextrin Inclusion Complex as a Drug-Delivery System for Improved Solubility and Biocompatibility in Breast Cancer Treatment. Molecules 2022, 27, 4521. [Google Scholar] [CrossRef] [PubMed]
  126. Bojic, M.; Kondza, M.; Rimac, H.; Benkovic, G.; Males, Z. The Effect of Flavonoid Aglycones on the CYP1A2, CYP2A6, CYP2C8 and CYP2D6 Enzymes Activity. Molecules 2019, 24, 3174. [Google Scholar] [CrossRef] [PubMed]
  127. Ma, Y.L.; Zhao, F.; Yin, J.T.; Liang, C.J.; Niu, X.L.; Qiu, Z.H.; Zhang, L.T. Two Approaches for Evaluating the Effects of Galangin on the Activities and mRNA Expression of Seven CYP450. Molecules 2019, 24, 1171. [Google Scholar] [CrossRef]
  128. Aloud, A.A.; Veeramani, C.; Govindasamy, C.; Alsaif, M.A.; El Newehy, A.S.; Al-Numair, K.S. Galangin, a dietary flavonoid, improves antioxidant status and reduces hyperglycemia-mediated oxidative stress in streptozotocin-induced diabetic rats. Redox Rep. 2017, 22, 290–300. [Google Scholar] [CrossRef] [PubMed]
  129. Kim, M.E.; Park, P.R.; Na, J.Y.; Jung, I.; Cho, J.H.; Lee, J.S. Anti-neuroinflammatory effects of galangin in LPS-stimulated BV-2 microglia through regulation of IL-1beta production and the NF-kappaB signaling pathways. Mol. Cell. Biochem. 2019, 451, 145–153. [Google Scholar] [CrossRef]
  130. Rampogu, S.; Gajula, R.G.; Lee, K.W. A comprehensive review on chemotherapeutic potential of galangin. Biomed. Pharmacother. 2021, 141, 111808. [Google Scholar] [CrossRef]
  131. Lee, J.J.; Ng, S.C.; Hsu, J.Y.; Liu, H.; Chen, C.J.; Huang, C.Y.; Kuo, W.W. Galangin Reverses H2O2-Induced Dermal Fibroblast Senescence via SIRT1-PGC-1alpha/Nrf2 Signaling. Int. J. Mol. Sci. 2022, 23, 1387. [Google Scholar] [CrossRef]
  132. Thangaiyan, R.; Arjunan, S.; Govindasamy, K.; Khan, H.A.; Alhomida, A.S.; Prasad, N.R. Galangin Attenuates Isoproterenol-Induced Inflammation and Fibrosis in the Cardiac Tissue of Albino Wistar Rats. Front. Pharmacol. 2020, 11, 585163. [Google Scholar] [CrossRef]
  133. Chaihongsa, N.; Maneesai, P.; Sangartit, W.; Potue, P.; Bunbupha, S.; Pakdeechote, P. Galangin alleviates vascular dysfunction and remodelling through modulation of the TNF-R1, p-NF-kappaB and VCAM-1 pathways in hypertensive rats. Life Sci. 2021, 285, 119965. [Google Scholar] [CrossRef] [PubMed]
  134. Huang, H.; Chen, A.Y.; Ye, X.; Guan, R.; Rankin, G.O.; Chen, Y.C. Galangin, a Flavonoid from Lesser Galangal, Induced Apoptosis via p53-Dependent Pathway in Ovarian Cancer Cells. Molecules 2020, 25, 1579. [Google Scholar] [CrossRef] [PubMed]
  135. Pepeljnjak, S.; Kosalec, I. Galangin expresses bactericidal activity against multiple-resistant bacteria: MRSA, Enterococcus spp. and Pseudomonas aeruginosa. FEMS Microbiol. Lett. 2004, 240, 111–116. [Google Scholar] [CrossRef] [PubMed]
  136. Aladaileh, S.H.; Al-Swailmi, F.K.; Abukhalil, M.H.; Shalayel, M.H. Galangin protects against oxidative damage and attenuates inflammation and apoptosis via modulation of NF-kappaB p65 and caspase-3 signaling molecules in a rat model of diabetic nephropathy. J. Physiol. Pharmacol. 2021, 72, 35–44. [Google Scholar] [CrossRef]
  137. Chen, G.; Liu, J.; Jiang, L.; Ran, X.; He, D.; Li, Y.; Huang, B.; Wang, W.; Fu, S. Galangin Reduces the Loss of Dopaminergic Neurons in an LPS-Evoked Model of Parkinson’s Disease in Rats. Int. J. Mol. Sci. 2017, 19, 12. [Google Scholar] [CrossRef]
  138. Wen, S.Y.; Chen, J.Y.; Weng, Y.S.; Aneja, R.; Chen, C.J.; Huang, C.Y.; Kuo, W.W. Galangin suppresses H2O2-induced aging in human dermal fibroblasts. Environ. Toxicol. 2017, 32, 2419–2427. [Google Scholar] [CrossRef] [PubMed]
  139. Henry, L.J.K.; Ramar, M.K.; Palanisamy, S.; Natesan, S.; Kandasamy, R. Mechanistic investigation of PPARgamma-facilitated anti-asthmatic effects of Galangin (Norizalpinin): Insights from in silico and in vivo analyses. Biochem. Biophys. Res. Commun. 2020, 526, 833–840. [Google Scholar] [CrossRef]
  140. Gerges, S.H.; Tolba, M.F.; Elsherbiny, D.A.; El-Demerdash, E. The natural flavonoid galangin ameliorates dextran sulphate sodium-induced ulcerative colitis in mice: Effect on Toll-like receptor 4, inflammation and oxidative stress. Basic Clin. Pharmacol. Toxicol. 2020, 127, 10–20. [Google Scholar] [CrossRef]
  141. Song, Y.-D.; Liu, Y.-Y.; Li, D.-J.; Yang, S.-J.; Wang, Q.-F.; Liu, Y.-N.; Li, M.-K.; Mei, C.-P.; Cui, H.-N.; Chen, S.-Y.; et al. Galangin ameliorates severe acute pancreatitis in mice by activating the nuclear factor E2-related factor 2/heme oxygenase 1 pathway. Biomed. Pharmacother. 2021, 144, 112293. [Google Scholar] [CrossRef] [PubMed]
  142. Lin, K.; Fu, D.; Wang, Z.; Zhang, X.; Zhu, C. Analgesic and anti-inflammatory effects of galangin: A potential pathway to inhibit transient receptor potential vanilloid 1 receptor activation. Korean J. Pain 2024, 37, 151–163. [Google Scholar] [CrossRef] [PubMed]
  143. Zhang, T.; Mei, X.; Ouyang, H.; Lu, B.; Yu, Z.; Wang, Z.; Ji, L. Natural flavonoid galangin alleviates microglia-trigged blood-retinal barrier dysfunction during the development of diabetic retinopathy. J. Nutr. Biochem. 2019, 65, 1–14. [Google Scholar] [CrossRef] [PubMed]
  144. Huang, Y.C.; Tsai, M.S.; Hsieh, P.C.; Shih, J.H.; Wang, T.S.; Wang, Y.C.; Lin, T.H.; Wang, S.H. Galangin ameliorates cisplatin-induced nephrotoxicity by attenuating oxidative stress, inflammation and cell death in mice through inhibition of ERK and NF-kappaB signaling. Toxicol. Appl. Pharmacol. 2017, 329, 128–139. [Google Scholar] [CrossRef] [PubMed]
  145. Tomar, A.; Vasisth, S.; Khan, S.I.; Malik, S.; Nag, T.C.; Arya, D.S.; Bhatia, J. Galangin ameliorates cisplatin induced nephrotoxicity in vivo by modulation of oxidative stress, apoptosis and inflammation through interplay of MAPK signaling cascade. Phytomedicine 2017, 34, 154–161. [Google Scholar] [CrossRef] [PubMed]
  146. Lu, H.; Yao, H.; Zou, R.; Chen, X.; Xu, H. Galangin Suppresses Renal Inflammation via the Inhibition of NF-kappaB, PI3K/AKT and NLRP3 in Uric Acid Treated NRK-52E Tubular Epithelial Cells. Biomed. Res. Int. 2019, 2019, 3018357. [Google Scholar] [CrossRef] [PubMed]
  147. Wang, H.B.; Huang, S.H.; Xu, M.; Yang, J.; Yang, J.; Liu, M.X.; Wan, C.X.; Liao, H.H.; Fan, D.; Tang, Q.Z. Galangin ameliorates cardiac remodeling via the MEK1/2-ERK1/2 and PI3K-AKT pathways. J. Cell. Physiol. 2019, 234, 15654–15667. [Google Scholar] [CrossRef] [PubMed]
  148. Abukhalil, M.H.; Althunibat, O.Y.; Aladaileh, S.H.; Al-Amarat, W.; Obeidat, H.M.; Al-Khawalde, A.A.A.; Hussein, O.E.; Alfwuaires, M.A.; Algefare, A.I.; Alanazi, K.M.; et al. Galangin attenuates diabetic cardiomyopathy through modulating oxidative stress, inflammation and apoptosis in rats. Biomed. Pharmacother. 2021, 138, 111410. [Google Scholar] [CrossRef] [PubMed]
  149. Prasatthong, P.; Meephat, S.; Rattanakanokchai, S.; Khamseekaew, J.; Bunbupha, S.; Prachaney, P.; Maneesai, P.; Pakdeechote, P. Galangin Resolves Cardiometabolic Disorders through Modulation of AdipoR1, COX-2, and NF-kappaB Expression in Rats Fed a High-Fat Diet. Antioxidants 2021, 10, 769. [Google Scholar] [CrossRef]
  150. Pakdeechote, P.; Poasakate, A.; Prasatthong, P.; Potue, P.; Khamseekaew, J.; Maneesai, P. Mitigation effect of galangin against aortic dysfunction and hypertrophy in rats with metabolic syndrome. Heliyon 2023, 9, e16500. [Google Scholar] [CrossRef]
  151. Choi, M.J.; Lee, E.J.; Park, J.S.; Kim, S.N.; Park, E.M.; Kim, H.S. Anti-inflammatory mechanism of galangin in lipopolysaccharide-stimulated microglia: Critical role of PPAR-gamma signaling pathway. Biochem. Pharmacol. 2017, 144, 120–131. [Google Scholar] [CrossRef] [PubMed]
  152. Yang, C.C.; Lin, C.C.; Hsiao, L.D.; Yang, C.M. Galangin Inhibits Thrombin-Induced MMP-9 Expression in SK-N-SH Cells via Protein Kinase-Dependent NF-kappaB Phosphorylation. Int. J. Mol. Sci. 2018, 19, 4084. [Google Scholar] [CrossRef] [PubMed]
  153. Kabel, A.M.; Arab, H.H.; Atef, A.; Estfanous, R.S. Omarigliptin/galangin combination mitigates lipopolysaccharide-induced neuroinflammation in rats: Involvement of glucagon-like peptide-1, toll-like receptor-4, apoptosis and Akt/GSK-3beta signaling. Life Sci. 2022, 295, 120396. [Google Scholar] [CrossRef] [PubMed]
  154. Choi, J.K.; Kim, S.H. Inhibitory effect of galangin on atopic dermatitis-like skin lesions. Food Chem. Toxicol. 2014, 68, 135–141. [Google Scholar] [CrossRef] [PubMed]
  155. Lee, H.N.; Shin, S.A.; Choo, G.S.; Kim, H.J.; Park, Y.S.; Kim, B.S.; Kim, S.K.; Cho, S.D.; Nam, J.S.; Choi, C.S.; et al. Anti-inflammatory effect of quercetin and galangin in LPS-stimulated RAW264.7 macrophages and DNCB-induced atopic dermatitis animal models. Int. J. Mol. Med. 2018, 41, 888–898. [Google Scholar] [CrossRef] [PubMed]
  156. Sangaraju, R.; Alavala, S.; Nalban, N.; Jerald, M.K.; Sistla, R. Galangin ameliorates Imiquimod-Induced psoriasis-like skin inflammation in BALB/c mice via down regulating NF-kappaB and activation of Nrf2 signaling pathways. Int. Immunopharmacol. 2021, 96, 107754. [Google Scholar] [CrossRef] [PubMed]
  157. Liu, Y.N.; Zha, W.J.; Ma, Y.; Chen, F.F.; Zhu, W.; Ge, A.; Zeng, X.N.; Huang, M. Galangin attenuates airway remodelling by inhibiting TGF-beta1-mediated ROS generation and MAPK/Akt phosphorylation in asthma. Sci. Rep. 2015, 5, 11758. [Google Scholar] [CrossRef] [PubMed]
  158. Zha, W.J.; Qian, Y.; Shen, Y.; Du, Q.; Chen, F.F.; Wu, Z.Z.; Li, X.; Huang, M. Galangin Abrogates Ovalbumin-Induced Airway Inflammation via Negative Regulation of NF-kappaB. Evid. Based Complement. Altern. Med. 2013, 2013, 767689. [Google Scholar] [CrossRef] [PubMed]
  159. Shu, Y.S.; Tao, W.; Miao, Q.B.; Lu, S.C.; Zhu, Y.B. Galangin dampens mice lipopolysaccharide-induced acute lung injury. Inflammation 2014, 37, 1661–1668. [Google Scholar] [CrossRef]
  160. Hu, K.; Li, Y.; Jin, L.; Chen, Y.; Chen, L.; Zhang, Y.; Huang, M.; Ding, Y.; Yin, H.; Liang, M.; et al. Protective Effect of Galangin Methylation Modification Based on Cell Imaging on Inflammatory Lung Injury and Its Molecular Mechanism. Contrast Media Mol. Imaging 2022, 2022, 7511345. [Google Scholar] [CrossRef]
  161. Aladaileh, S.H.; Abukhalil, M.H.; Saghir, S.A.M.; Hanieh, H.; Alfwuaires, M.A.; Almaiman, A.A.; Bin-Jumah, M.; Mahmoud, A.M. Galangin Activates Nrf2 Signaling and Attenuates Oxidative Damage, Inflammation, and Apoptosis in a Rat Model of Cyclophosphamide-Induced Hepatotoxicity. Biomolecules 2019, 9, 346. [Google Scholar] [CrossRef] [PubMed]
  162. Salama, S.A.; Elshafey, M.M. Galangin mitigates iron overload-triggered liver injury: Up-regulation of PPARgamma and Nrf2 signaling, and abrogation of the inflammatory responses. Life Sci. 2021, 283, 119856. [Google Scholar] [CrossRef] [PubMed]
  163. Skiba, M.A.; Szendzielorz, K.; Mazur, B.; Krol, W. The inhibitory effect of flavonoids on interleukin-8 release by human gastric adenocarcinoma (AGS) cells infected with cag PAI (+) Helicobacter pylori. Cent. Eur. J. Immunol. 2016, 41, 229–235. [Google Scholar] [CrossRef] [PubMed]
  164. Sangaraju, R.; Nalban, N.; Alavala, S.; Rajendran, V.; Jerald, M.K.; Sistla, R. Protective effect of galangin against dextran sulfate sodium (DSS)-induced ulcerative colitis in Balb/c mice. Inflamm. Res. 2019, 68, 691–704. [Google Scholar] [CrossRef] [PubMed]
  165. Cai, S.Q.; Zhang, Q.; Zhao, X.H.; Shi, J. The In Vitro Anti-Inflammatory Activities of Galangin and Quercetin towards the LPS-Injured Rat Intestinal Epithelial (IEC-6) Cells as Affected by Heat Treatment. Molecules 2021, 26, 7495. [Google Scholar] [CrossRef] [PubMed]
  166. Seyoum, A.; Asres, K.; El-Fiky, F.K. Structure-radical scavenging activity relationships of flavonoids. Phytochemistry 2006, 67, 2058–2070. [Google Scholar] [CrossRef] [PubMed]
  167. Li, Y.; Tong, L.; Zhang, J.; Zhang, Y.; Zhang, F. Galangin Alleviates Liver Ischemia-Reperfusion Injury in a Rat Model by Mediating the PI3K/AKT Pathway. Cell. Physiol. Biochem. 2018, 51, 1354–1363. [Google Scholar] [CrossRef] [PubMed]
  168. Devadoss, D.; Ramar, M.; Chinnasamy, A. Galangin, a dietary flavonol inhibits tumor initiation during experimental pulmonary tumorigenesis by modulating xenobiotic enzymes and antioxidant status. Arch. Pharm. Res. 2018, 41, 265–275. [Google Scholar] [CrossRef] [PubMed]
  169. Sivakumar, A.S.; Viswanathan, P.; Anuradha, C.V. Dose-dependent effect of galangin on fructose-mediated insulin resistance and oxidative events in rat kidney. Redox Rep. 2010, 15, 224–232. [Google Scholar] [CrossRef]
  170. Sivakumar, A.S.; Anuradha, C.V. Effect of galangin supplementation on oxidative damage and inflammatory changes in fructose-fed rat liver. Chem. Biol. Interact. 2011, 193, 141–148. [Google Scholar] [CrossRef]
  171. Aloud, A.A.; Veeramani, C.; Govindasamy, C.; Alsaif, M.A.; Al-Numair, K.S. Galangin, a natural flavonoid reduces mitochondrial oxidative damage in streptozotocin-induced diabetic rats. Redox Rep. 2018, 23, 29–34. [Google Scholar] [CrossRef] [PubMed]
  172. Kumar, S.; Alagawadi, K.R. Anti-obesity effects of galangin, a pancreatic lipase inhibitor in cafeteria diet fed female rats. Pharm. Biol. 2013, 51, 607–613. [Google Scholar] [CrossRef] [PubMed]
  173. Tsai, M.S.; Chien, C.C.; Lin, T.H.; Liu, C.C.; Liu, R.H.; Su, H.L.; Chiu, Y.T.; Wang, S.H. Galangin Prevents Acute Hepatorenal Toxicity in Novel Propacetamol-Induced Acetaminophen-Overdosed Mice. J. Med. Food 2015, 18, 1187–1197. [Google Scholar] [CrossRef] [PubMed]
  174. Kim, Y.R.; Kim, M.A.; Cho, H.J.; Oh, S.K.; Lee, I.K.; Kim, U.K.; Lee, K.Y. Galangin prevents aminoglycoside-induced ototoxicity by decreasing mitochondrial production of reactive oxygen species in mouse cochlear cultures. Toxicol. Lett. 2016, 245, 78–85. [Google Scholar] [CrossRef] [PubMed]
  175. Abd El-Aal, S.A.; AbdElrahman, M.; Reda, A.M.; Afify, H.; Ragab, G.M.; El-Gazar, A.A.; Ibrahim, S.S.A. Galangin mitigates DOX-induced cognitive impairment in rats: Implication of NOX-1/Nrf-2/HMGB1/TLR4 and TNF-alpha/MAPKs/RIPK/MLKL/BDNF. Neurotoxicology 2022, 92, 77–90. [Google Scholar] [CrossRef] [PubMed]
  176. Moreira, M.R.; Kanashiro, A.; Kabeya, L.M.; Polizello, A.C.; Azzolini, A.E.; Curti, C.; Oliveira, C.A.; Amaral, T.d.A.; Lucisano-Valim, Y.M. Neutrophil effector functions triggered by Fc-gamma and/or complement receptors are dependent on B-ring hydroxylation pattern and physicochemical properties of flavonols. Life Sci. 2007, 81, 317–326. [Google Scholar] [CrossRef] [PubMed]
  177. 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]
  178. Luo, Q.; Zhu, L.; Ding, J.; Zhuang, X.; Xu, L.; Chen, F. Protective effect of galangin in Concanavalin A-induced hepatitis in mice. Drug Des. Dev. Ther. 2015, 9, 2983–2992. [Google Scholar] [CrossRef]
  179. Wang, L.; Liu, H.; He, Q.; Gan, C.; Li, Y.; Zhang, Q.; Yao, Y.; He, F.; Ye, T.; Yin, W. Galangin ameliorated pulmonary fibrosis in vivo and in vitro by regulating epithelial-mesenchymal transition. Bioorg. Med. Chem. 2020, 28, 115663. [Google Scholar] [CrossRef]
  180. Song, H.Y.; Kim, W.S.; Han, J.M.; Seo, H.S.; Lim, S.T.; Byun, E.B. Galangin treatment during dendritic cell differentiation confers tolerogenic properties in response to lipopolysaccharide stimulation. J. Nutr. Biochem. 2021, 87, 108524. [Google Scholar] [CrossRef]
  181. Zhang, J.; Hu, S.; Gao, Y.; Wei, X.; Qu, Y.; Gao, R.; Lv, Y.; Wang, J.; Wang, Y.; Yang, J.; et al. Galangin alleviated myocardial ischemia-reperfusion injury by enhancing autophagic flux and inhibiting inflammation. Eur. J. Pharmacol. 2023, 945, 175621. [Google Scholar] [CrossRef] [PubMed]
  182. Zhang, F.; Yan, Y.; Zhang, L.M.; Li, D.X.; Li, L.; Lian, W.W.; Xia, C.Y.; He, J.; Xu, J.K.; Zhang, W.K. Pharmacological activities and therapeutic potential of galangin, a promising natural flavone, in age-related diseases. Phytomed. Int. J. Phytother. Phytopharm. 2023, 120, 155061. [Google Scholar] [CrossRef] [PubMed]
  183. Yao, Q.; Wu, X.; Tao, C.; Gong, W.; Chen, M.; Qu, M.; Zhong, Y.; He, T.; Chen, S.; Xiao, G. Osteoarthritis: Pathogenic signaling pathways and therapeutic targets. Signal Transduct. Target. Ther. 2023, 8, 56. [Google Scholar] [CrossRef] [PubMed]
  184. Su, Y.; Chen, Y.; Liu, Y.; Yang, Y.; Deng, Y.; Gong, Z.; Chen, J.; Wu, T.; Lin, S.; Cui, L. Antiosteoporotic effects of Alpinia officinarum Hance through stimulation of osteoblasts associated with antioxidant effects. J. Orthop. Transl. 2016, 4, 75–91. [Google Scholar] [CrossRef] [PubMed]
  185. Huang, X.; Pei, W.; Ni, B.; Zhang, R.; You, H. Chondroprotective and antiarthritic effects of galangin in osteoarthritis: An in vitro and in vivo study. Eur. J. Pharmacol. 2021, 906, 174232. [Google Scholar] [CrossRef] [PubMed]
  186. Wang, Y.; Zhang, T.; Yang, Q.; Jia, Y.; Sun, P.; Fu, Z.; Han, W.; Qian, Y. Galangin attenuates IL-1β-induced catabolism in mouse chondrocytes and ameliorates murine osteoarthritis. Am. J. Transl. Res. 2021, 13, 8742–8753. [Google Scholar] [PubMed]
  187. Su, Y.; Shen, L.; Xue, J.; Zou, J.; Wan, D.; Shi, Z. Therapeutic evaluation of galangin on cartilage protection and analgesic activity in a rat model of osteoarthritis. Electron. J. Biotechnol. 2021, 53, 8–13. [Google Scholar] [CrossRef]
  188. Lin, Q.; Zhang, Y.; Hong, W.; Miao, H.; Dai, J.; Sun, Y. Galangin ameliorates osteoarthritis progression by attenuating extracellular matrix degradation in chondrocytes via the activation of PRELP expression. Eur. J. Pharmacol. 2022, 936, 175347. [Google Scholar] [CrossRef] [PubMed]
  189. Zeng, C.; Wang, S.; Gu, H.; Chen, F.; Wang, Z.; Li, J.; Xie, Z.; Feng, P.; Shen, H.; Wu, Y. Galangin mitigates glucocorticoid-induced osteoporosis by activating autophagy of BMSCs via triggering the PKA/CREB signaling pathway. Acta Biochim. Biophys. Sin. 2023, 55, 1275–1287. [Google Scholar] [CrossRef]
  190. Li, X.; Jiang, J.; Yang, Z.; Jin, S.; Lu, X.; Qian, Y. Galangin suppresses RANKL-induced osteoclastogenesis via inhibiting MAPK and NF-kappaB signalling pathways. J. Cell. Mol. Med. 2021, 25, 4988–5000. [Google Scholar] [CrossRef]
  191. Huh, J.E.; Jung, I.T.; Choi, J.; Baek, Y.H.; Lee, J.D.; Park, D.S.; Choi, D.Y. The natural flavonoid galangin inhibits osteoclastic bone destruction and osteoclastogenesis by suppressing NF-kappaB in collagen-induced arthritis and bone marrow-derived macrophages. Eur. J. Pharmacol. 2013, 698, 57–66. [Google Scholar] [CrossRef] [PubMed]
  192. Fu, Q.; Gao, Y.; Zhao, H.; Wang, Z.; Wang, J. Galangin protects human rheumatoid arthritis fibroblast-like synoviocytes via suppression of the NF-kappaB/NLRP3 pathway. Mol. Med. Rep. 2018, 18, 3619–3624. [Google Scholar] [CrossRef] [PubMed]
  193. Deng, X.; Le, H.; Wan, T.; Weng, M.; Tan, Y. Galangin alleviates rheumatoid arthritis in rats by downregulating the phosphatidylinositol 3-kinase/protein kinase B signaling pathway. Bioengineered 2022, 13, 11192–11201. [Google Scholar] [CrossRef] [PubMed]
  194. Santos, E.O.; Kabeya, L.M.; Figueiredo-Rinhel, A.S.; Marchi, L.F.; Andrade, M.F.; Piatesi, F.; Paoliello-Paschoalato, A.B.; Azzolini, A.E.; Lucisano-Valim, Y.M. Flavonols modulate the effector functions of healthy individuals’ immune complex-stimulated neutrophils: A therapeutic perspective for rheumatoid arthritis. Int. Immunopharmacol. 2014, 21, 102–111. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of DMARDs used in RA treatment: mechanisms and effects.
Figure 1. Overview of DMARDs used in RA treatment: mechanisms and effects.
Pharmaceuticals 17 00963 g001
Figure 2. Chemical structure of GAL.
Figure 2. Chemical structure of GAL.
Pharmaceuticals 17 00963 g002
Figure 3. Different biological activities and numerous health benefits of GAL.
Figure 3. Different biological activities and numerous health benefits of GAL.
Pharmaceuticals 17 00963 g003
Table 1. Sources of GAL.
Table 1. Sources of GAL.
CategorySourceReferences
Honeybee ProductsPropolis, Bee Pollen[89,90,91,92]
Plants (Rhizomes)Alpinia officinarum, Alpinia calcarata, Alpinia galanga[93,94,95]
Plants (Roots)Coleus vettiveroides[96]
Plants (Leaves)Mexican Oregano (Lippia graveolens), Nothofagus gunnii, Eugenia catharinensis, Castanea sativa, Piper aleyreanum, Japanese Alnus sieboldiana[97,98,99,100,101,102,103]
Plants (Buds)Poplar (Populus nigra)[104,105,106,107]
Plants (Other)Poplar tree gum, Fruits (Sechium hybrid, Sechium edule Perla Negra, Campomanesia xanthocarpa, Prunus cerasus (Oblacinska), Psidium cattleianum), Plantain peel, Jujube peel and seed, Crocus sativus flower petals[108,109,110,111,112,113,114,115]
Marine FungusChaetomium globosum[116]
Table 2. Anti-inflammatory mechanisms of GAL.
Table 2. Anti-inflammatory mechanisms of GAL.
Type of InflammationGAL’s Mechanism of ActionModelReferences
RenalDecreased NF-κB p65, iNOS, and TNF-α, IL-1β, IL-6 levelsRat model of diabetic nephropathy[136]
Inhibited ERK and p38 MAPK signaling and NF-ĸB activation and the release of proinflammatory cytokinesRodent model of nephrotoxicity[144,145]
Suppressed PI3K/AKT, NF-κB, NLRP3 inflammasome, and the production of TNF-α, IL-1β, PGE2, and NORat kidney epithelial cells (NRK-52E)[146]
CardiovascularSuppressed TNF-α, IL-6, NF-κB, COX-2, and iNOS gene expressionAlbino Wistar rat model of cardiac inflammation and fibrosis[132]
Decreased VCAM-1, TNF-R1, TNF-α, and the activity of NF-κBRat model of hypertension[133]
Decreased NF-κB p65 activation and inhibited MEK1/2-ERK1/2-GATA4 and PI3K/AKT-GSK3β pathwaysMouse model of aortic banding mediated pressure overload and angiotensin II treated H9c2 cells[147]
Decreased NF-κB p65, iNOS, and TNF-α, IL-1β, IL-6 levelsRat model of diabetic cardiomyopathy[148]
Positively regulated adiponectin/AdipoR1 signaling pathway and COX-2 and reduced NF-κB activityRat model of cardiometabolic syndrome[149]
Downregulated renin-angiotensin system (RAS) and IL-6/TNF-α and upregulated endothelial nitric oxide synthase (eNOS) pathwayRat model of cardiometabolic syndrome[150]
NeuralReduced prostaglandin E2Pentylenetetrazole (PTZ) mouse epilepsy model[102]
Suppressed production of proinflammatory mediators (TNF-α, IL-1β, IL-6, COX-2, iNOS) and inhibited phosphorylation of JNK, p38, AKT, and NF-κB p65Rat model of Parkinson’s disease[137]
Suppressed phosphorylation of p38 MAPK, JNK, PI3K/Akt, and NF-κB, activated PPARγ, reduced proinflammatory mediators (TNF-α, IL-6, MMP-3, MMP-9, iNOS), and increased anti-inflammatory mediators (IL-10, HO-1)Lipopolysaccharide (LPS) injected mouse brains and LPS-stimulated microglial cell line (BV-2 cells)[151]
Suppressed MMP-9 expression and inhibited phosphorylation of multiple signaling molecules including c-Src, Pyk2, PKC isoforms, Akt, mTOR, MAPKs (ERK1/2, JNK1/2, p38), FoxO1, c-Jun, and p65Human neuroblastoma cell line (SK-N-SH cells)[152]
Inhibited proinflammatory mediators (iNOS, IL-1β, NO) and suppressed phosphorylation of JNK, p38 MAPK, and NF-κBLPS-stimulated microglial cell line (BV-2 cells)[129]
Reduced proinflammatory cytokines (IL-8, TNF-α), inhibited TLR4/NLRP3 inflammasome and DPP-4, and increased GLP-1 levelsRat model of LPS-induced neuroinflammation[153]
SkinAttenuated NF-κB p65 activation and proinflammatory cytokine release (TNF-α, IL-1β, IL-6)Human HS68 dermal fibroblasts[138]
Inhibited ERK, p38 MAPKs, and NF-κB pathways and suppressed TNF-α/IFN-γ-induced production of CCL17 and proinflammatory cytokines (IL-6, TNF-α, IL-1β)Mouse model of atopic dermatitis and human keratinocytes (HaCaT cells)[154]
Attenuated NF-κB/p65 activation, inhibited Erk1/2 and JNK activation, suppressed production of NO, iNOS, and IL-6, and downregulated serum IgE levelsMouse model of atopic dermatitis and RAW264.7 macrophages[155]
Activated Nrf2/HO-1 pathway, inhibited NF-ĸB pathway, downregulated proinflammatory mediators (COX-2, iNOS) and cytokines (IL-17, IL-23, IL-1β), and upregulated anti-inflammatory cytokine IL-10BALB/c mice model of induced psoriasis-like skin inflammation[156]
PulmonaryInhibited collagen deposition, lowered alpha-smooth muscle actin, disrupted TGF-β1 pathway, and reduced VEGF and MMP-9Induced chronic asthma mice model and airway smooth muscle cell culture (ASMC)[157]
Activated PPARγ, reduced IL-4, IL-5, IL-13, TNF-α, IL-17, NO, eosinophil peroxidase, and IgE, and increased IFN-γInduced allergic asthma mice model[139]
Suppressed activation of NF-κB p65 and TNF-α-mediated translocation, decreased production of inflammatory mediators including monocyte chemoattractant protein-1, eotaxin, CXCL10, and VCAM-1, IL-4, IL-5, IL-13, and iNOSMouse model of induced airway inflammation[158]
Inhibited NF-κB activation and reduced TNF-α and IL-6 levelsMouse model of induced acute lung injury[159]
Activated PPARγ and DNMT3A, leading to suppressed ERK, p65, and AP-1, and reduced proinflammatory cytokines (IL-6, TNF-α)Rat model of induced inflammatory lung injury[160]
HepaticSuppressed NF-κB p65, iNOS, and proinflammatory cytokines (TNF-α, IL-1β, IL-6)Rat model of induced hepatotoxicity[86]
Induced Nrf2/HO-1 pathway, elevated PPARγ, inhibited NF-κB activation, and decreased proinflammatory cytokines (TNF-α, IL-1β)Rat model of induced hepatotoxicity[161,162]
GastrointestinalReduced TLR4 expression, decreased HMGB1, inhibited NF-κB p65 activation, and decreased proinflammatory cytokines (IL-6, TNF-α)Mouse model of induced ulcerative colitis[140]
Downregulated IL-8 secretionHuman gastric adenocarcinoma (AGS) cells[163]
Activated the Nrf2/HO-1 pathway and suppressed the NF-κB pathway and its downstream proinflammatory mediators (COX-2, iNOS, and TNF-α/IL-6)Mouse model of induced ulcerative colitis[164]
Suppressed TLR4/NF-κB signaling pathway, increased anti-inflammatory cytokines (IL-10, TGF-β), and decreased proinflammatory cytokines (IL-1β, IL-6, TNF-α), and PGE2LPS-injured rat intestinal epithelial (IEC-6) cells[165]
PancreaticActivated Nrf2/HO-1 pathway and inhibited NF-κB p65 and proinflammatory mediators, such as TNF-α, IL-18, MCP-1, and CXCL10Mouse model of pancreatitis[141]
FootSuppressed the expression of NF-κB, TNF-α, COX-2, and PGE2Acute inflammation and pain rat model[142]
RetinalInhibited the ERK1/2-NF-κB/Egr1 pathway and proinflammatory cytokines (TNFα, IL-1β, and IL-6)D-glucose-induced microglia BV2 cells and retinal inflammatory injury in diabetic mice model[143]
Table 3. GAL’s immunomodulatory mechanisms.
Table 3. GAL’s immunomodulatory mechanisms.
Condition/Cell TypeMechanism of ActionKey FindingsReference
Experimental Autoimmune Encephalomyelitis (EAE) ModelInhibits mononuclear cell infiltration, reduces T cell proliferation and differentiation (Th1 and Th17 cells), and impairs dendritic cell functionAlleviated autoimmune encephalomyelitis[87]
LPS-stimulated RAW 264.7 Macrophage CellsDownregulates COX-2, reduces NO and proinflammatory cytokines (TNF-α, IL-1β, IL-6), and inhibits IRAK-1, JAK/STAT pathway, MAPK (p38 and ERK), and NF-κBReduced inflammation[94]
Atopic Dermatitis (AD) ModelReduces infiltration of inflammatory cells (eosinophils, mast cells), decreases histamine levels, inhibits Th1 and Th2 cytokines, and reduces serum IgE and IgG2aReduced inflammation[154]
Ovalbumin-(OVA-) Induced Airway Inflammation ModelReduced total leukocytes (eosinophils, neutrophils, and lymphocytes) and downregulated the production of IgEAlleviate induced airway hyperresponsiveness and inflammation[158]
NeutrophilsInhibits FcγRs and CRs activation, and reduces myeloperoxidase and horseradish peroxidase activityDecreased ROS production, reduced tissue damage[176]
Human Mast Cells (HMC)-1Downregulates JNK, p38, and NF-κB pathways, suppresses histamine release and reduces activation of Caspase-1Reduced inflammation[177]
Concanavalin A (ConA)-induced Hepatitis (CIH) ModelSuppresses infiltration of inflammatory cells (neutrophils, macrophages, T cells), inhibits T cell activation via STAT1 pathway, and reduces proinflammatory cytokines and chemokinesReduced inflammation[178]
Bleomycin-Induced Pulmonary Fibrosis ModelReduced the number of CD4+ and CD8+ T cells and dendritic cellsAlleviate pulmonary fibrosis[179]
Dendritic Cells (DCs)Promotes tolerogenic DCs (tolDCs) and stimulates regulatory T cells (Tregs)Skews DCs towards tolerogenic phenotype and suppresses inflammatory T cell responses[180]
Myocardial Ischemia-Reperfusion InjuryInduces autophagic flux via PI3K/AKT/mTOR pathway, increases anti-inflammatory M2 macrophages, and decreases proinflammatory M1 macrophagesReduced inflammation[181]
Table 4. A synopsis of the in vitro and in vivo studies assessing GAL’s impact on OA.
Table 4. A synopsis of the in vitro and in vivo studies assessing GAL’s impact on OA.
Model and GAL’s DoseEffect on Disease ProgressionSignaling Pathway and
Antioxidant Defense
Reference
In vivo
  • Rat osteoblasts (ROB)
  • 10−8 g/mL to 10−4 g/mL for 24 h
  • Promote osteoblast calcification
  • Decrease ALP activity
N/D[184]
  • Anterior Cruciate Ligament Transection (ACLT) in SD rats
  • 0.1 mL/day for 8 weeks (5 mg/mL)
  • Decrease OARSI score
N/D[185,186]
  • Monoiodoacetate (MIA) in SD rats
  • 10 or 100 mg/kg/day for 14 days
  • Reduce CTX-II, IL-1b, IL-6, and TNF-a levels
  • Disturbance of pro-oxidant/antioxidant.
  • Reduce ROS and lipid peroxidation levels
  • Increase catalase, SOD, Gpx, and GSH levels
[187]
  • Destabilization of the medial meniscus (DMM) in SD rats
  • 20, 40 and 60 mg/kg, twice per week for 4 weeks
  • Decrease OARSI score
  • Decrease MMP13 and ADAMTS5
  • Increase the levels of COL2A1 and ACAN
N/D[188]
  • Glucocorticoid-induced osteoporosis (GIOP) in C57BL/6 mice
  • 10 or 40 mg/mL/day for 8 weeks
  • Alleviate bone damage
  • PKA/CREB-mediated autophagy signaling.
  • Increase in LC3B and p-CREB levels
[189]
In vitro
  • IL-1β (10 ng/mL) treated chondrocytes
  • Primary mouse bone marrow macrophages (BMMs)
  • [5–20] µM for 24 h
  • Decrease iNOS and COX-2 expression
  • Decrease MMP1, MMP3, MMP13 and ADAMTS5
  • Attenuate collagen II and aggrecan degradation
  • Inhibit NF-κB, MAPK and PI3K/AKT pathways
  • Inhibit phosphorylation of ERK, JNK Akt, IKKαβ, IKBα and P65
[185,186,190]
  • Human OA primary chondrocytes
  • [0–100] µM for 24 h
  • Decrease MMP13 and ADAMTS5
  • Increase the levels of COL2A1 and ACAN
  • Inhibit PI3K-AKT pathway
  • Downstream oxidative stress
  • Inhibit phosphorylation of PI3K and AKT
  • Activate PRELP
  • Inhibit the expression of ROS and MDA
  • Promote the expression of SOD and CAT
[188]
  • Dexamethasone-treated human Bone marrow-derived mesenchymal stem cells (BMSCs)
  • [5–20] µM for 24 h
  • Promote osteogenic differentiation
  • Increase protein expression levels of Runx2, OCN, OPN
  • PKA/CREB, AKT/mTOR, PI3K/AKT and Wnt/β-catenin
  • Upregulate p-PKA/PKA and p-CREB/CREB
  • Downregulate p-mTOR/mTOR
[189]
Table 5. A synopsis of the in vitro and in vivo studies assessing GAL’s impact on RA.
Table 5. A synopsis of the in vitro and in vivo studies assessing GAL’s impact on RA.
Model and GAL’s DoseEffect on Disease ProgressionSignaling Pathway and
Antioxidant Defense
Reference
In vivo
  • Collagen-induced arthritis (CIA) in DBA/1J mice
  • 10, 50, or 100 mg/kg for 25 days
  • Reduce arthritis score and paw edema
  • Improve bone/cartilage destruction, synovial hyperplasia, and pannus formation
  • Reduce production of IL-1b, TNF-a, IL-17 and RANKL
N/D[175]
  • Collagen-induced arthritis (CIA) in SD rats
  • 10, 20, and 40 µg for 4 weeks
  • Improve bone/cartilage destruction, synovial hyperplasia, and pannus formation
  • Improve histological score
  • Increase body weight
  • Decrease TNF-α, IL-1β, and IL-6 levels
  • Downregulate PI3K/AKT/mTOR signaling pathway
  • Inhibit of pPI3K, pAKT, and pmTOR protein levels
[178]
In vitro
  • Co-cultures of bone marrow-derived macrophages and primary osteoblasts
  • 1, 10, and 20 mg/mL
  • Inhibit osteoclast formation
  • Decrease TNF-α, IL-1β, and IL-17 levels
  • Suppress phospho-JNK and phospho p38 MAPK
  • Inhibit NF-kB/p65 and IkBa phosphorylation level
  • Increase IkBa level
[175]
Human neutrophil-10 µMSuppress O2, ROS and MPON/D[176]
  • Primary human RA fibroblast-like synovium cells (RAFSCs)
  • 1, 5, and 10 ng/mL for 24 h
  • Decrease IL-1β, TNF-α, IL-18, PGE2 and NO levels
  • Inhibit iNOS and COX-2 expression levels
  • Downstream oxidative stress
  • Suppress NF-κB/NLRP3 signaling pathway
  • Decrease SOD activity and increase MDA content
  • Decrease ASC, pro-caspase-1/caspase-1, p-IκBα, p-NF-κB, IL-1β and NLRP3 expression
[177]
  • Rheumatoid arthritis fibroblast-like synoviocytes (RAFLSs)
  • [10–160] µM for 24, 48, and 72 h
  • Suppress inflammation, proliferation, migration and invasion
  • Promote apoptosis of RAFLSs
N/D[178]
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

Khawaja, G.; El-Orfali, Y.; Shoujaa, A.; Abou Najem, S. Galangin: A Promising Flavonoid for the Treatment of Rheumatoid Arthritis—Mechanisms, Evidence, and Therapeutic Potential. Pharmaceuticals 2024, 17, 963. https://doi.org/10.3390/ph17070963

AMA Style

Khawaja G, El-Orfali Y, Shoujaa A, Abou Najem S. Galangin: A Promising Flavonoid for the Treatment of Rheumatoid Arthritis—Mechanisms, Evidence, and Therapeutic Potential. Pharmaceuticals. 2024; 17(7):963. https://doi.org/10.3390/ph17070963

Chicago/Turabian Style

Khawaja, Ghada, Youmna El-Orfali, Aya Shoujaa, and Sonia Abou Najem. 2024. "Galangin: A Promising Flavonoid for the Treatment of Rheumatoid Arthritis—Mechanisms, Evidence, and Therapeutic Potential" Pharmaceuticals 17, no. 7: 963. https://doi.org/10.3390/ph17070963

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