Next Article in Journal
5-Bromoprotocatechualdehyde Combats against Palmitate Toxicity by Inhibiting Parkin Degradation and Reducing ROS-Induced Mitochondrial Damage in Pancreatic β-Cells
Next Article in Special Issue
Antioxidant and In Vitro Preliminary Anti-Inflammatory Activity of Castanea sativa (Italian Cultivar “Marrone di Roccadaspide” PGI) Burs, Leaves, and Chestnuts Extracts and Their Metabolite Profiles by LC-ESI/LTQOrbitrap/MS/MS
Previous Article in Journal
Plant-Based Phenolic Molecules as Natural Preservatives in Comminuted Meats: A Review
Previous Article in Special Issue
Antioxidant, Pancreatic Lipase Inhibitory, and Tyrosinase Inhibitory Activities of Extracts of the Invasive Plant Spartina anglica (Cord-Grass)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Emerging Natural-Product-Based Treatments for the Management of Osteoarthritis

by
Maria-Luisa Pérez-Lozano
1,2,
Annabelle Cesaro
1,2,
Marija Mazor
3,
Eric Esteve
4,
Sabine Berteina-Raboin
5,
Thomas M. Best
6,
Eric Lespessailles
1,2,7 and
Hechmi Toumi
1,2,7,*
1
Laboratory I3MTO, EA 4708, Université d’Orléans, CEDEX 2, 45067 Orléans, France
2
Plateforme Recherche Innovation Médicale Mutualisée d’Orléans, Centre Hospitalier Régional d’Orléans, 14 Avenue de l’Hôpital, 45100 Orléans, France
3
Center for Proteomics, Department for Histology and Embryology, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51000 Rijeka, Croatia
4
Service de Dermatologie, Centre Hospitalier Régional d′Orléans, 14 Avenue de l’Hôpital, 45100 Orléans, France
5
Institut de Chimie Organique et Analytique ICOA, Université d’Orléans-Pôle de Chimie, UMR CNRS 7311, Rue de Chartres-BP 6759, CEDEX 2, 45067 Orléans, France
6
Department of Orthopedics, Division of Sports Medicine, Health Sports Medicine Institute, University of Miami, Coral Gables, FL 33146, USA
7
Centre Hospitalier Régional d’Orléans, Institut Département de Rhumatologie, 45067 Orléans, France
*
Author to whom correspondence should be addressed.
Antioxidants 2021, 10(2), 265; https://doi.org/10.3390/antiox10020265
Submission received: 13 January 2021 / Revised: 1 February 2021 / Accepted: 4 February 2021 / Published: 9 February 2021
(This article belongs to the Special Issue Antioxidant and Biological Properties of Plant Extracts)

Abstract

:
Osteoarthritis (OA) is a complex degenerative disease in which joint homeostasis is disrupted, leading to synovial inflammation, cartilage degradation, subchondral bone remodeling, and resulting in pain and joint disability. Yet, the development of new treatment strategies to restore the equilibrium of the osteoarthritic joint remains a challenge. Numerous studies have revealed that dietary components and/or natural products have anti-inflammatory, antioxidant, anti-bone-resorption, and anabolic potential and have received much attention toward the development of new therapeutic strategies for OA treatment. In the present review, we provide an overview of current and emerging natural-product-based research treatments for OA management by drawing attention to experimental, pre-clinical, and clinical models. Herein, we review current and emerging natural-product-based research treatments for OA management.

1. Introduction

Osteoarthritis (OA) is the most common degenerative musculoskeletal disease and is a leading cause of disability in the adult population [1]. OA is a whole-joint disease that is characterized by irreversible cartilage degradation; disruption of the tidemark, accompanied by angiogenesis and cartilage calcification; subchondral bone remodeling; osteophyte formation; mild-to-moderate inflammation of the synovial lining [2,3,4]. The most common risk factors for OA include age, prior joint injury, obesity, muscle atrophy, metabolic disorders, and mechanical stress [5,6]. The disease evolution is typically slow and can take years to develop, with resultant joint pain and stiffness, mobility limitations, and compromised quality of life. Despite the tremendous personal and societal burden of OA, there are no curative treatments available and most conventional therapies (medications, physiotherapy, mechanical devices) provide relatively short-term, unsustained relief of the symptoms [7,8,9,10,11].
Promise exists for emerging disease-modifying drugs in the management of OA patients that regulate cartilage metabolism, subchondral bone remodeling, synovial inflammation, and angiogenesis. Recently, the use of plant-derived natural products has increased because of their therapeutic value in bone health, which is attributable to their chondroprotective and osteoprotective properties [12,13]. Many of these natural products have been reported to have anti-inflammatory and antioxidant properties, anti-catabolic effects on chondrocytes, and inhibitory effects on osteoclast differentiation [14,15,16]. Accordingly, this review of natural-derived compounds that have shown promise in the treatment of OA highlights our current thinking for this novel approach.

2. Natural-Compound-Based Treatments for OA Therapy

Conventional pharmaceutical agents (steroids or non-steroids anti-inflammatory (NSAIDs) drugs) have small-to-moderate effects in patients with OA [7,9,11,17,18,19]. Accordingly, there is an increasing interest in identifying novel approaches, including the use of natural bioactive components that could promote joint health, and mitigate and/or reverse OA [20].

2.1. Alkaloids

Berberine

Berberine is an alkaloid (benzylisoquinoline) that is found in medicinal plants of the genera Berberis, such as Berberis vulgaris, and is usually found in the roots, rhizomes, and stems (Table 1) [21]. It has been reported that berberine has anti-osteoarthritic effects [21]. In vivo studies in two different OA animal models (collagenase- and surgically induced OA) have demonstrated that berberine has chondroprotective effects, which ameliorates cartilage degradation while inducing chondrocyte proliferation [22,23]. It has been shown that berberine inhibits chondrocyte apoptosis and cartilage degradation via activating AMPK signaling and suppressing p38 MAPK activity [24,25]. Berberine also decreases inflammation and cartilage degradation by modulating the host immune response through the inhibition of TLR4/NF-κB signaling [26]. Moreover, berberine has been associated with bone formation by promoting osteogenic differentiation via activation of Runx-2 and p38 MAPK and reducing osteoclast differentiation [27,28].

2.2. Flavonoids

2.2.1. Apigenin

Apigenin is a flavonoid (4′,5,7-trihydroxyflavone) that is found in herbs (chamomile, thyme), fruits (orange), vegetable oils (extra virgin olive oil), and in plant-based beverages (tea, beer, and wine) (Table 2) [29]. This bioactive agent has already been used as therapeutic therapy against diabetes, cancer, Alzheimer’s disease, and OA [30,31]. Apigenin has anti-inflammatory properties through inhibiting IL-1β/NF-κB and TGFβ/Smad2/3 pathways in chondrocytes [32]. Park et al. have demonstrated that apigenin blocks cartilage degradation in in vitro and in vivo OA mouse models through Hif-2α inhibition and the consequent downregulation of MMP-3, MMP-13, ADAMTS-5, and ADAMTS-4 in articular chondrocytes [33]. Furthermore, apigenin has shown bone protective effects via modulating the gene expression of TGF-β1 and its receptors, BMP-2, BMP-7, ALP, and collagen type I in MG63 osteoblasts [34]. Apigenin also promotes osteogenic differentiation of human mesenchymal stem cells through the JNK and p38 MAPK pathways [35].

2.2.2. Astragalin

Astragalin is a natural flavonoid (kaempferol 3-glucoside) found in various traditional medicinal plants, such as Cuscuta chinensis. Its antioxidant and anti-inflammatory therapeutic properties have led some to consider its potential as a therapeutic agent for OA patients [36,37]. According to Ma et al. [38], astragalin inhibits the IL-1β-stimulated activation of NF-κB and MAPK in the chondrocytes of patients with OA while suppressing inflammation and bone destruction in a mouse model of OA [38,39].

2.2.3. Baicalein

Baicalein is a flavonoid (5,6,7-trihydroxyflavone) that is isolated from the roots of Scutellaria baicalensis and Scutellaria lateriflora and has medicinal properties, including neuroprotective, anti-oxidant, anti-fibrosis, and anti-cancer properties [40,41]. Recently, it has been demonstrated that baicalein has anti-catabolic and anti-apoptotic effects through inhibiting IL-1β induction in chondrocytes [42,43]. Another study showed that the intra-articular injection of medium and high doses of baicalein alleviated OA progression in a rabbit OA model, diminishing cartilage degradation, and showing a lower Mankin score [44]. Similarly, positive results were obtained on bone through the induction of osteoblast differentiation and inhibiting osteoclast differentiation [45,46].

2.2.4. Chrysin

Chrysin is a flavonoid (5,7-dihydroxyflavone) that is found in various medicinal plants, such as Scutellaria baicalensis and Passiflora caerulea, but also in honey and propolis [47]. In human osteoarthritic chondrocytes, chrysin showed a suppressive effect on the IL-1β-induced inflammatory response, including the expression of inducible nitrous oxide synthase (iNOS), COX-2, MMP-1, MMP-3, MMP-13, ADAMTS-4, and ADAMTS-5 via the inhibition of NF-κB signaling and decreases in the concentrations of nitrous oxide (NO) and PGE2. Chrysin also inhibits the degradation of aggrecan and collagen-II [48]. In addition, chrysin attenuates the apoptosis and inflammation of stimulated human OA chondrocytes via the suppression of high-mobility group box chromosomal protein (HMGB-1) [49]. An osteoprotective effect was also observed under chrysin treatment via ERK/MAPK activation and the upregulating of Runx-2 and Osx expression [50,51].

2.2.5. Genistein

Genistein is a flavonoid (isoflavone) and a phytoestrogen that is extracted from Genista tinctoria. It has been reported to have promising benefits in the treatment of several pathologies [52,53,54]. The anti-osteoarthritic activity of genistein is suggested to be due to the relationship between OA and altered estrogen metabolism [55]. Phytoestrogens have some estrogen activity and ameliorate menopausal symptoms, bone loss, and symptoms of OA [56,57]. In vitro, genistein suppresses catabolic effects of IL-1β-induced in human OA chondrocytes by targeting the Nrf2/HO-1 pathway, decreasing the expression of MMPs, nitric oxide synthase 2 (NOS2), and COX-2 [58]. In vivo, genistein attenuated cartilage degradation in two different OA animal models [58,59]. Furthermore, a positive effect on bone was obtained through enhanced osteoblastic differentiation and maturation via the activation of ER (estrogen receptor), p38 MAPK–Runx2, and NO/cGMP pathways [60,61,62]. It also inhibited osteoclast formation and bone resorption by inducing the osteoclastogenic inhibitor osteoprotegerin (OPG) and by blocking NF-κB signaling [60,63].

2.2.6. Icariin

Icariin is a flavonoid (flavonoid glycoside) obtained from the genus Epimedium. The therapeutic potential of this natural compound in cartilage regeneration has been shown in both in vitro and in vivo studies [64,65]. In vitro, Icariin increases the secretion of extracellular matrix proteins, such as collagen type II and the expression of SOX-9, while decreasing the expression of MMPs via the activation of HIF-1α. In vivo, icariin enhances articular cartilage repair in mouse osteochondral-defective models [65]. It has been reported that icariin protects chondrocytes from lipopolysaccharide (LPS)-, IL-1β-, or TNF-α-induced inflammation. Apoptosis and extracellular matrix degradation was also observed via diminishing the expression of MMP-1, 3, 9, 13, COX-2, and iNOS, suppressing NF-κB signaling and activating the Nrf2/ARE pathway [66,67,68]. Icariin also demonstrates protective effects in bone metabolism. This compound can induce osteoblast proliferation, differentiation, and mineralization through estrogen-receptor-mediated ERK and JNK signal activation in the MC3T3-E1 osteoblastic cell line, resulting in an increased expression of differentiation markers, alkaline phosphatase (ALP), and collagen type I [69]. It has been demonstrated that icariin induces the miR-153/Runx2 pathway, which is involved in osteoblast differentiation [70]. Icariin also attenuates hypoxia-induced oxidative stress and apoptosis in osteoblasts [71]. In an in vivo OA mouse model, it was shown that icariin enhanced bone remodeling with a positive effect on subchondral bone and hyaline cartilage [72].

2.2.7. Kaempferol

Kaempferol is a flavonoid (3,4′,5,7-tetrahydroxyflavone) that is derived from the rhizome Kaempferia galanga L. and can also be found in numerous common vegetables and fruits, including beans, broccoli, cabbage, grapes, strawberries, tomatoes, citrus fruits, and apples [73]. Kaempferol alleviates IL-1β-stimulated inflammation in rat OA chondrocytes by decreasing the production of PGE2 and NO and downregulating the expression of MMPs, ADAMTS-5, iNOS, and COX-2. These effects were all mediated through the inhibition of the MAPK p38 and NF-κB pathways [74,75]. It has been shown that kaempferol increased the osteoblast differentiation and mineralization, and increasing the expression of BMP-2, Runx-2, Osx, and collagen type I by activating Wnt/β-catenin signaling [76,77]. Another study revealed that kaempferol stimulated bone formation in part via the mTOR signaling pathway [78]. Kaempferol prevents osteoclast formation through MAPKs, c-Fos, and NFATc1 [76,79]. In addition, in vivo studies have reported that kaempferol decreased bone loss in ovariectomized mice [80,81].

2.2.8. Luteolin

Luteolin is a flavonoid (3′,4′,5,7-tetrahydroxyflavone) that is present in herb vegetables and fruits, including Salvia tomentosa, Chrysanthemum indicum, Artemisia asiatica, broccoli, carrots, peppers, cabbages, parsley, thyme, peppermint, basil, and celery [82]. Luteolin has shown anti-inflammatory and anti-catabolic effects in chondrocytes through the inhibition of NF-κB signaling [83]. It diminishes the IL-1β-induced production of NO, PGE2, TNF-α, MMP-2, MMP-3, MMP-8, and MMP-9; downregulates the expression of COX-2, iNOS, MMP-1, MMP-3, MM-13, ADAMTS-4, and ADAMTS-5; inhibits the degradation of collagen type II [83,84,85]. Luteolin also protects chondrocytes from apoptosis by increasing Foxo3a expression via regulating the IRE1α pathway and miR-29a/Wnt/β-catenin signaling [86,87]. Its administration can also attenuate cartilage degradation and increase collagen type II expression in OA rats in vivo [83]. In vitro studies have demonstrated that luteolin upregulates the expression of osteoblastic differentiation markers, including TGF-β1, BMP7, Runx-2, ALP, Osc, Osx, and collagen type I [34,88,89]. It also has anti-oxidative and anti-apoptotic effects on osteoblasts, in part via the regulation of the ERK/Lrp-5/GSK-3β signaling pathway [90,91,92]. Furthermore, luteolin diminishes osteoclastic differentiation and function in vitro and in vivo, increasing the bone mineral density and content of trabecular and cortical bones in ovariectomized rats [93,94].

2.2.9. Naringin

Naringin is a flavonoid (flavanone-7-O-glycoside) that is formed from the flavanone naringenin and the disaccharide neohesperidose, and is found in citrus fruits, such as grapefruit. Naringenin inhibited TNFα-, LPS-, and IL-1β-induced catabolic effects, diminishing the expression of MMPs, ADAMTS-4, and ADAMTS-5 via the suppression of the NF-κB pathway and caveolin–p38 MAPK signaling [95,96,97,98]. In vivo, naringin attenuates cartilage destruction via the suppression of inflammatory cytokines. Naringin also promotes bone formation via increased osteoblast proliferation and differentiation [99,100,101]. Mechanistically, this occurs through the increased expression and secretion of bone-formation-related genes including Osc, Runx-2, Osx, OPN, BMP-2, and collagen type I [102]. Naringin also inhibits osteoclast differentiation and maturation, therefore preventing bone loss [103].

2.2.10. Puerarin

Puerarin is a flavonoid (isoflavone) that is found in several plants and herbs, such as the root of Pueraria lobate [104]. It reduces OA progression by inhibiting the pro-catabolic responses in chondrocytes [105]. It also has a negative effect on monocyte recruitment [106] and promotes bone formation through the estrogen receptor, p38 MAPK, ERK1/2–Runx2, and Wnt/β–catenin pathways [107,108]. Oral administration of puerarin in ovariectomized rats protected against a reduction in bone mineral density and content while improving femur trabecular bone structure [108]. The effects of puerarin on osteoblastic proliferation and differentiation are mediated by the inhibition of TRPM3/miR-204 expression and the activation of Runx-2 [109,110,111]. In ovariectomized rats, puerarin was shown to inhibit osteoblastogenesis through the downregulation of TRAP and RANKL [112]. The inhibition of RANKL osteoclastogenesis is mediated by the downregulation of CREB/PGC1β/c-Fos/NFATc1 signaling [113]. Furthermore, another study showed that puerarin inhibits osteoclastogenesis by suppressing RANKL-dependent and -independent autophagic responses [114].

2.2.11. Silibinin/Silymarin

Silibinin, also known as silybin, is the major active flavonoid constituent of silymarin, which is an extract of milk thistle seeds (Silybum marianum), comprising approximately 50–70% of the extract [115,116]. It is also a phytoestrogen. Other flavonolignans, such as silychristin, isosilychristin, silydianin, and silimonin, are also present in silymarin. The anti-inflammatory properties of silymarin for OA treatment have been demonstrated using several protocols [115,116,117,118]. A study employing MIA-induced OA rats showed that silymarin exerts anti-inflammatory and antioxidant effects by diminishing the NO and IL-1β content in synovial tissue and attenuating cartilage degradation [119]. Another study demonstrated that silibinin inhibits the IL-1β-induced production of NO, PGE2, TNF-α, and IL-6; downregulates the expression of COX-2, iNOS, MMP-1, MMP-3, MMP-13, ADAMTS-4, and ADAMTS-5; diminishes the degradation of aggrecan and collagen type II in human OA chondrocytes through the suppression of PI3K/Akt and NF-κB signaling pathways [120]. Furthermore, treatment with silibinin prevented cartilage degradation and synovitis in an in vivo mice OA model. Silibinin also has osteoprotective properties, promoting osteoblastogenesis and inhibiting osteoclastogenesis [121,122]. In vitro experiments have shown that silibinin and silymarin induce osteoblast differentiation in MC3T3-E1 osteoblasts by increasing the expression of ALP, collagen type I, Runx-2, and BMP-2 [122,123]. It also promotes the osteogenic differentiation of human bone marrow stem cells via BMP signaling [124]. In addition, silibinin has antioxidant and anti-apoptotic effects in osteoblasts [125]. It has been reported that silibinin and silymarin suppress osteoclastic differentiation in RAW 264.7 osteoclasts, decreasing TRAP and cathepsin K induction induced by RANKL via disturbing TRAF6-c-Src signaling pathways and inhibiting femoral bone loss in ovariectomized mice [121,126].

2.2.12. Wogonin

Wogonin is a flavonoid (O-methylated flavone) that is found in Scutellaria baicalensis as baicalein [127,128]. It has been reported that wogonin has chondroprotective effects, inhibiting IL-1β-induced catabolic markers, such as IL-6, COX-2, iNOS, MMP-3, MMP-9, MMP-13, and ADAMTS-4, while increasing the anabolic markers aggrecan and collagen type II in chondrocytes and cartilage explants [129,130,131]. These wogonin effects are mediated through the suppression of c-Fos/AP-1 and JAK/STAT signaling pathways and the activation of ROS/ERK/Nrf2 signaling pathways [129,131,132]. A recent study has shown that the utilization of tetrahedral framework nucleic acid/wogonin complexes alleviated inflammation in in vitro and in vivo OA models, preventing cartilage destruction and increasing bone mineral density [133]. Wogonin has also been shown to attenuate intervertebral disc degeneration [134].

2.3. Phenols

2.3.1. Curcumin

Curcumin is a natural phenol (diferuloylmethane) that is responsible for turmeric’s yellow color and comes from the Curcuma longa root (Table 3). Anti-inflammatory, anti-oxidant, anti-apoptotic, and anti-catabolic effects were observed on chondrocytes under curcumin treatment. It inhibited the expression of the inflammation mediators IL-6, iNOS, and COX-2. It also blocked the expression of proteinases MMP-1, MMP-3, MMP-9, MMP-13, ADAMTS-4, and ADAMTS-5, and increased the expression of SOX-9 and production of collagen II, attenuating cartilage degradation [12,135,136,137,138]. These effects occur through the direct inhibition of 5-LOX and NF-κB, indirect inhibition of phospholipase A2 and COX-2, and activation of the Nrf2/ARE signaling pathway [135,136,137,138]. Chen et al. showed that curcumin also inhibited osteoblast apoptosis and promoted osteoblast differentiation, both in vitro and in vivo [139]. It increased the gene expression of Runx2, Osx, Osc, and collagen type I via the regulation of Wnt signaling [140,141]. The bioavailability of curcumin is a major challenge because it is inherently low in humans, but new formulations have enhanced the therapeutic efficacy of curcumin [142,143]. Furthermore, the use of curcumin in combination with other natural products, such as Boswellia serrate, gingerly, and pipeline, are being studied in several clinical trials to investigate whether their therapeutically synergy enhances their performance in OA treatment but the results showed no significant difference between each component separately or in combination [144,145,146].

2.3.2. Gingerly/Ginger

Ginger is the rhizome of the Zingier officinal plant and has been commonly consumed as a spice and herbal medicine due to its anti-inflammatory properties. The major active component is the phenolic gingerly (6-gingerol) [147]. The efficacy and safety of ginger were evaluated in various studies [148]. Ginger extract has shown anti-inflammatory, antioxidant, and anti-apoptotic effects in IL-β-treated human chondrocytes via the activation of Nrf2 [149,150]. It also stimulated osteoblasts differentiation and inhibited IL-1β-induced osteoclasts differentiation in in vitro studies [151,152]. Randomized clinical trials have demonstrated that ginger extracts improved pain and mobility and reduced osteoarthritis inflammation in OA individuals [153,154]. The local application of ginger was also found to be effective at reducing symptoms of knee OA [155]. In addition, the synergistic effects of ginger with other natural products were also studied in patients with chronic OA, but the results did not show any significant enhanced effects [145,156,157].

2.3.3. Oleuropein

Oleuropein is a phenolic compound (secoiridoid glycoside) that is present in green olive (Olea europea) and argan oil [158]. It has been reported that olive oil extract has beneficial effects in OA treatment [159]. An in vivo study demonstrated that oleuropein decreases the spontaneous development of OA in guinea pigs, reducing cartilage, osteophytes, and synovial OA scores [160]. It also inhibited IL-1β-induced inflammatory response in human OA chondrocytes in vitro by suppressing NF-κB and MAPK signaling pathways [161]. It suppresses the production of NO and PGE2 and decreased the expression of COX-2, iNOS, MMP-1, MMP-13, and ADAMTS-5. Furthermore, it has been shown that oleuropein does not stimulate osteoblast proliferation but increases the deposition of calcium and suppresses osteoclast formation and differentiation [162,163,164]. It also protected against bone loss in ovariectomized rats [165]. Yet, there is no clinical trial with this natural compound for OA; however, a randomized clinical trial with postmenopausal women showed that the consumption of a polyphenol extract from olive increases serum osteocalcin levels and improves serum lipid profiles [166].

2.3.4. Resveratrol

Resveratrol is a stilbenoid (3,5,4′-trihydroxy-trans-stilbene), which is a type of natural phenol that is produced by several plants in response to injury and in fruits, such as red grapes, blueberries, raspberries, and mulberries [167,168]. Since it prevents degeneration and apoptosis, resveratrol has been strongly suggested to be a potential therapeutic agent for OA [169,170]. Resveratrol was demonstrated to inhibit IL-1β-induced catabolic effects in chondrocytes. It suppressed the expression of iNOS, MMP-3, MMP-1, MMP-13, ADAMTS-4, ADAMTS-5, and NO production by inducing SIRT-1 expression and inhibiting NF-κB signaling [171,172,173]. It also prevented IL-1β-mediated inflammation via TLR4 inhibition [174,175]. In vitro studies have demonstrated that these inhibitory effects of resveratrol are mediated via the activation of SIRT-1 by suppressing HIF-2 expression and inducing autophagy via the AMPK/mTOR pathway [171,176,177,178]. Preclinical models have shown that resveratrol treatment prevented OA progression, maintaining the structural homeostasis in cartilage and subchondral bone [173,178,179,180]. Resveratrol was demonstrated to exert bone protection through the suppression of osteoclast functions and the induction and differentiation of osteoblasts in both in vivo and in vitro studies. Resveratrol induced osteoblast differentiation by regulating autophagy and modulating the Sirt1/Runx-2/Fox-O1 and PI3K/AKT/mTOR signaling pathways, therefore, ameliorating bone loss in osteoporotic animal models [181,182,183,184]. It was also shown to induce osteoblastic MC3T3-E1 cells differentiation via the induction of the calcineurin/NFATc1 signaling pathway [185]. Resveratrol also inhibited RANKL-induced osteoclastogenesis via SIRT1 and FoxOs activation [186,187,188]. A clinical study on postmenopausal women showed that resveratrol supplementation reduces pain experience; thus, it was proposed as a potential treatment to reduce chronic pain in age-related osteoarthritic individuals [189]. Another pilot study demonstrated that the co-administration of resveratrol with meloxicam in patients with knee OA improves pain, functions, and associated symptoms compared with a placebo, yet it was superior in terms of safety and efficacy compared to meloxicam alone [190].

2.3.5. Salvianolic Acid B

Salvianolic acid B (Sal B) is a major polyphenol constituent of the plant Radix salvia miltiorrhiza, which is commonly used in traditional Chinese medicine to cure pain [191]. It has been recently proposed as a potential therapeutic agent against OA that acts through the regulation of gene expression and the viability of chondrocytes [192]. It has been demonstrated that the pre-treatment of chondrocytes with Sal B followed by induction with IL-1β inhibited the overproduction NO and PGE2 and downregulated the expression of iNOS, COX-2, MMP-13, and ADAMTS-5 via the suppression of NF-κB [193]. This study also revealed that Sal B reduced cartilage degradation in an OA mouse model. Sal B was also found to stimulate osteoblastic differentiation in bone marrow stromal cells, upregulating the expression of Runx2, OPN, and Osx and stimulating mineralization through the activation of ERK signaling pathways [194]. In vivo, Sal B inhibited glucocorticoid-induced osteopenia. It enhanced bone thickness and bone mass by increasing the expression of BMPs, ALP activity, and collagen type I [194,195]. A pilot study in a rat tibia fracture model revealed that treatment with Sal B led to an enhancement in callus growth, histological scores, and post-fracture ALP activity, thus, accelerating early-stage fracture [196]. Furthermore, Sal B facilitates osteogenesis by targeting adipose tissue, reducing adipogenesis, and activating the MEK–ERK signaling pathway [197].

2.4. Polysaccharides

Achyranthes bidentata Extracts

Achyranthes bidentata is one of the most commonly used Chinese herbal medicines that is currently considered for the treatment of osteoarthritis (Table 4) [198]. This extract has shown chondroprotective effects in vitro, inducing chondrocyte proliferation via Wnt/β-catenin pathway activation and inhibiting apoptosis via the MAPK/Akt signaling axis [199,200]. Plant polysaccharides also have osteoprotective properties, suppressing osteoclastogenesis and bone resorption by inhibiting RANKL and promoting bone formation [201,202,203,204].

2.5. Terpenoids

2.5.1. Andrographolide

This terpenoid (diterpenoid) is a natural component from Andrographis paniculate, a plant with medicinal properties, such as antioxidant, anti-inflammatory, and anti-arthritic properties (Table 5) [205,206,207,208]. A recent study showed the effectiveness and safety of andrographolide in reducing pain in individuals suffering from mild-to-moderate knee osteoarthritis [209]. It has been reported to inhibit the expression of MMPs and reduces oxidative stress injury in chondrocytes [210,211]. An in vivo mouse OA model study revealed that this compound alleviates cartilage damage via the miR-27-3p/MMP13 signaling axis [212]. It also exerts a pro-osteogenic effect via inducing bone formation by inhibiting NF-κB signaling, with this bioactive compound being a potential therapeutic target in OA [213,214].

2.5.2. Astaxanthin

Astaxanthin is a carotenoid (tetraterpenoid) that is produced naturally in the microalgae Haematococcus pluvialis and can be found in animals who feed on the algae, such as salmon, red trout, and crustaceans [215]. It has therapeutic properties against rheumatoid arthritis and osteoarthritis [216,217,218,219,220]. In OA, astaxanthin has shown potent antioxidant and anti-inflammatory activities on cartilage due to the activation of Nrf2–ARE signaling in chondrocytes [221]. Astaxanthin also attenuated cartilage degradation in vitro and in vivo via blockade MAPK signaling [222]. Despite the fact that the effects of astaxanthin’s properties on OA bone remodeling have not yet been examined, it could be a good therapeutic target due to its effects on the suppression of bone loss in periodontitis and osteoporotic models [223,224].

2.5.3. Aucubin

Aucubin is a terpenoid (iridoid glycoside) that is derived from diverse medicinal plants, including Aucuba japonica and Eucommia ulmoides. It has recently received increasing attention due to its pharmacological properties, including antioxidation, anti-inflammation, and osteoprotection [225]. In vitro studies showed that aucubin suppressed IL-1β-induced inflammation and matrix degradation and reduced oxidative stress by decreasing iNOS expression and the production of NO [226,227]. It has been reported that aucubin prevented OA progression in an in vivo mouse model and that the co-treatment with hyaluronic acid (HA) enhanced the anti-catabolic and anti-inflammatory effects of HA on OA chondrocytes [228,229].

2.5.4. Boswellia serrata

Boswellia serrata is a plant that produces Indian frankincense and has two mains active terpenoid compounds, 11-keto-β-boswellic acid and acetyl-11-keto-β-boswellic acid [230]. The extracts of this plant have been clinically studied for osteoarthritis treatment, exerting anti-inflammatory activity and resulting in decreased pain and increased joint functionality [231]. B. serrata has been reported to have anti-inflammatory properties by inhibiting 5-LOX and TNF-α [232]. An in vitro model of cartilage degeneration showed that B. serrata diminished the catabolic effects mediated by IL-1α and oncostatin-M through inhibiting MMP-9 and MMP-13 transcription and reducing the levels of NO, PGE2, and COX-2 [233]. Its chondroprotective properties were confirmed in a mouse model of OA, showing antioxidative and anti-inflammatory effects [15,234]. Additionally, it has been reported that boswellic acids promoted osteoblast differentiation and suppressed osteoclastogenesis by inhibiting TNF-α and NF-κB signaling [235,236].

2.5.5. Celastrol

Celastrol is a terpenoid (triterpenoid) that is isolated from the root extracts of Tripterygium wilfordii and Celastrus regelii [237]. Celastrol is an inhibitor of heat shock protein (HSP) 90β, which has chondroprotective effects. It has been reported that diminished IL-1β-induced catabolic effects in human osteoarthritic chondrocytes, such as the decrease expression of MMP1, MMP-3, MMP-13, iNOS, and COX-2 [238]. Using an in vivo OA rat model, it has been shown that celastrol suppresses apoptosis through the inhibition of the NF-κB signaling pathway and alleviates pain and cartilage damage via SDF-1/CXCR4 signaling [239,240]. Celastrol also has therapeutic effects on bone structure, where it prevented bone loss and bone microarchitecture degradation in a rat model of arthritis [241]. It has been shown that celastrol reduced the RANKL-induced expression of osteoclastic genes (TRAP, CTSK, CTR, and MMP-9) and transcriptional factors (c-Fos, c-Jun, and NFATc1), as well as the phosphorylation of NF-κB and MAPK in RAW 264.7 cells [242].

2.5.6. Ginsenoside

Ginsenosides are a class of natural product triterpene saponins (terpenoid glycoside) that are found almost exclusively in the plant genus Panax (ginseng), which is used in traditional medicine [243]. Ginsenosides exhibit a large variety of subtypes with different chemical profiles and biological effects. It has been reported that ginsenosides Rg1, Rg3, Rg5, Rk1, Rf, Rd, Rc, and F4 have chondroprotective effects [244]. Ginsenoside Rb1 has antioxidative and anti-apoptotic effects in chondrocytes in vitro, stabilizing mitochondria and inhibiting caspase-3 through PI3K/Akt signaling [245,246,247]. It also suppresses IL-1β-induced effects on chondrocytes, decreasing MMP-1, MMP-13, iNOS, and COX-2 expressions and the concentration of PGE2, and promoting the expression of ACAN and collagen type II [248,249]. Ginsenosides, such as Rb1, Rg1, and Rg5 have alleviated inflammation and cartilage degradation in in vivo OA rat models [250,251,252]. Recent studies have reported the chondroprotective effect of different Panax plant extracts in vivo OA rat models, protecting chondrocytes from inflammation, senescence, and apoptosis, thus, attenuating OA progression [253,254]. Ginsenosides also have osteoprotective properties. Several studies have demonstrated that Rb1, Rh1, Rg3, and Rg5 stimulated osteoblast differentiation in vitro [255,256,257,258]. Furthermore, Rb1 and Rg3 inhibited osteoclastogenesis by suppressing RANKL-induced activation via modulating MAPKs and NF-κB pathways in vitro, but only Rg3 was able to alleviate bone mineral density loss in vivo [259,260].

2.5.7. Harpagophytum procumbens

Harpagophytum procumbens, also known as devil’s claw, is a medicinal plant native to Africa that has been used as an analgesic for the treatment of degenerative diseases of the musculoskeletal system [261]. The bioactive components responsible for the anti-osteoarthritic effect are the iridoid glycosides (harpagoside, harpagide, and procumbide), which are found in a higher amount in the tubers and root [262]. An in vitro study showed that the pre-treatment of IL-1β-induced OA chondrocytes with harpagoside exerted some anti-inflammatory effects, inhibiting IL-6 and MMP-13 expression via the suppressing c-Fos/AP-1 activity [263]. Another study showed that harpagide improved bone properties, stimulating the differentiation of osteoblasts and suppressing the RANKL-induced differentiation of osteoclasts in an ovariectomized mouse model, thus, improving the recovery of bone mineral density and trabecular bone volume [264]. Furthermore, some human clinical studies showed that various H. procumbens tuber extracts improved clinical pain and movement limitation in individuals with knee and hip OA [265]. However, more studies are required to elucidate the therapeutic properties of H. procumbens in OA.

3. Conclusions

Osteoarthritis is a disease that is becoming more prevalent with the increase in the aging population. There are few conventional therapies that are available for the systematic treatment of OA and no treatment to prevent it. Unfortunately, all these therapies have significant adverse effects and are not adequate for long-term OA management. Therefore, the protective effects shown by natural products could be a potential alternative to conventional therapy. This review shows that natural compound supplementation plays an important role in the prevention of osteoarthritis. Various natural products have shown similar mechanistic properties, such as anti-inflammatory and antioxidant effects, on chondrocytes, inhibiting the cytokine-induced expression and catabolic activity of MMPs by inhibiting the NF-κB signaling pathway. Some phytochemicals have been shown to protect against cartilage degradation in preclinical studies. Natural products have also shown osteoprotective effects, upregulating the expression of various factors, such as Runx2, OPN, and Osx, in addition to the upregulation of the MAPK pathway and OPG/RANKL ratio. These regulations decreased bone resorption and enhanced osteoblastic activity and downregulation of the osteoclastic activity. Furthermore, some phytochemicals showed synergistic effects when explored in combination with other natural products or standard therapies. Although there are several bibliographical studies that show that some natural compounds are of interest in terms of fighting against inflammation or oxidation processes, as far as we know, there is no natural product that can prevent osteoarthritis or reverse it. The studies of these natural products from human clinical trials are still too few to be able to confirm their therapeutic effect at present. Therefore, the optimization of the formulation of natural products, and/or the combination of them, to combat and prevent osteoarthritis is a challenge.

Author Contributions

Conceptualization, M.-L.P.-L., A.C., E.E., E.L., T.M.B., M.M., S.B.-R. and H.T.; methodology, M.-L.P.-L., A.C., E.E., E.L., T.M.B., M.M., S.B.-R. and H.T.; validation, M.-L.P.-L., A.C., E.E., E.L., T.M.B., M.M., S.B.-R. and H.T.; resources, E.L., S.B.-R. and H.T.; writing—original draft preparation, M.-L.P.-L.; writing—review and editing, T.M.B., S.B.-R. and H.T.; supervision, E.L., S.B.-R. and H.T.; project administration, E.L., S.B.-R. and H.T; funding acquisition, E.L., S.B.-R. and H.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported and funded by the FEDER (Fonds Européen pour le Développement Régional) dedicated to PRIMMO (Plateforme Recherche Innovation Médicale Mutualisée d’Orléans), the Orleans Mutualized Medical Innovation Research Platform for translational research.

Conflicts of Interest

To the best of our knowledge, no conflict of interest, financial or otherwise, exists.

References

  1. Brennan-Olsen, S.L.; Cook, S.; Leech, M.T.; Bowe, S.J.; Kowal, P.; Ackerman, N.; Page, R.S.; Hosking, S.M.; Pasco, J.A.; Mohebbi, M. Prevalence of arthritis according to age, sex and socioeconomic status in six low and middle income countries: Analysis of data from the World Health Organization study on global AGEing and adult health (SAGE) Wave 1. BMC Musculoskelet. Disord. 2017, 18, 271. [Google Scholar] [CrossRef] [Green Version]
  2. Loeser, R.F.; Goldring, S.R.; Scanzello, C.R.; Goldring, M.B. Osteoarthritis: A disease of the joint as an organ. Arthritis Rheum. 2012, 64, 1697–1707. [Google Scholar] [CrossRef] [Green Version]
  3. Goldring, S.R.; Goldring, M.B. Changes in the osteochondral unit during osteoarthritis: Structure, function and cartilage–bone crosstalk. Nat. Rev. Rheumatol. 2016, 12, 632–644. [Google Scholar] [CrossRef]
  4. Burr, D.B.; Gallant, M.A. Bone remodelling in osteoarthritis. Nat. Rev. Rheumatol. 2012, 8, 665–673. [Google Scholar] [CrossRef] [PubMed]
  5. Berenbaum, F.; Wallace, I.J.; Lieberman, D.E.; Felson, D.T. Modern-day environmental factors in the pathogenesis of osteoarthritis. Nat. Rev. Rheumatol. 2018, 14, 674–681. [Google Scholar] [CrossRef] [PubMed]
  6. Palazzo, C.; Nguyen, C.; Lefevre-Colau, M.-M.; Rannou, F.; Poiraudeau, S. Risk factors and burden of osteoarthritis. Ann. Phys. Rehabil. Med. 2016, 59, 134–138. [Google Scholar] [CrossRef]
  7. Da Costa, B.R.; Reichenbach, S.; Keller, N.; Nartey, L.; Wandel, S.; Jüni, P.; Trelle, S. Effectiveness of non-steroidal anti-inflammatory drugs for the treatment of pain in knee and hip osteoarthritis: A network meta-analysis. Lancet 2017, 390, e21–e33. [Google Scholar] [CrossRef]
  8. Spetea, M. Opioid Receptors and Their Ligands in the Musculoskeletal System and Relevance for Pain Control. Curr. Pharm. Des. 2014, 19, 7382–7390. [Google Scholar] [CrossRef]
  9. Osani, M.C.; Bannuru, R.R. Efficacy and safety of duloxetine in osteoarthritis: A systematic review and meta-analysis. Korean J. Intern. Med. 2019, 34, 966–973. [Google Scholar] [CrossRef] [Green Version]
  10. Oo, W.M.; Liu, X.; Hunter, D.J. Pharmacodynamics, efficacy, safety and administration of intra-articular therapies for knee osteoarthritis. Expert Opin. Drug Metab. Toxicol. 2019, 15, 1021–1032. [Google Scholar] [CrossRef]
  11. Bannuru, R.R.; Osani, M.C.; Vaysbrot, E.E.; Arden, N.K.; Bennell, K.; Bierma-Zeinstra, S.M.A.; Kraus, V.B.; Lohmander, L.S.; Abbott, J.H.; Bhandari, M.; et al. OARSI guidelines for the non-surgical management of knee, hip, and polyarticular osteoarthritis. Osteoarthr. Cartil. 2019, 27, 1578–1589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Henrotin, Y.; Clutterbuck, A.L.; Allaway, D.; Lodwig, E.M.; Harris, P.; Mathy-Hartert, M.; Shakibaei, M.; Mobasheri, A. Biological actions of curcumin on articular chondrocytes. Osteoarthr. Cartil. 2010, 18, 141–149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Bu, S.Y.; Lerner, M.; Stoecker, B.J.; Boldrin, E.; Brackett, D.J.; Lucas, E.A.; Smith, B.J. Dried Plum Polyphenols Inhibit Osteoclastogenesis by Downregulating NFATc1 and Inflammatory Mediators. Calcif. Tissue Int. 2008, 82, 475–488. [Google Scholar] [CrossRef] [PubMed]
  14. Mathy-Hartert, M.; Jacquemond-Collet, I.; Priem, F.; Sanchez, C.; Lambert, C.; Henrotin, Y. Curcumin inhibits pro-inflammatory mediators and metalloproteinase-3 production by chondrocytes. Inflamm. Res. 2009, 58, 899–908. [Google Scholar] [CrossRef]
  15. Umar, S.; Umar, K.; Sarwar, A.H.M.G.; Khan, A.; Ahmad, N.; Ahmad, S.; Katiyar, C.K.; Husain, S.A.; Khan, H.A. Boswellia serrata extract attenuates inflammatory mediators and oxidative stress in collagen induced arthritis. Phytomedicine 2014, 21, 847–856. [Google Scholar] [CrossRef] [PubMed]
  16. Wu, X.; Li, Z.; Yang, Z.; Zheng, C.; Jing, J.; Chen, Y.; Ye, X.; Lian, X.; Qiu, W.; Yang, F.; et al. Caffeic acid 3,4-dihydroxy-phenethyl ester suppresses receptor activator of NF-κB ligand-induced osteoclastogenesis and prevents ovariectomy-induced bone loss through inhibition of mitogen-activated protein kinase/activator protein 1 and Ca2+-nuclear fact. J. Bone Miner. Res. 2012, 27, 1298–1308. [Google Scholar] [CrossRef]
  17. Crofford, L.J. Use of NSAIDs in treating patients with arthritis. Arthritis Res. Ther. 2013, 15, S2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Benyamin, R.; Trescot, A.M.; Datta, S.; Buenaventura, R.; Adlaka, R.; Sehgal, N.; Glaser, S.E.; Vallejo, R. Opioid complications and side effects. Pain Physician 2008, 11, S105–S120. [Google Scholar] [PubMed]
  19. Smith, C.; Patel, R.; Vannabouathong, C.; Sales, B.; Rabinovich, A.; McCormack, R.; Belzile, E.L.; Bhandari, M. Combined intra-articular injection of corticosteroid and hyaluronic acid reduces pain compared to hyaluronic acid alone in the treatment of knee osteoarthritis. Knee Surg. Sports Traumatol. Arthrosc. 2019, 27, 1974–1983. [Google Scholar] [CrossRef]
  20. Cao, P.; Li, Y.; Tang, Y.; Ding, C.; Hunter, D.J. Pharmacotherapy for knee osteoarthritis: Current and emerging therapies. Expert Opin. Pharmacother. 2020, 21, 1–13. [Google Scholar] [CrossRef]
  21. Wong, S.K.; Chin, K.-Y.; Ima-Nirwana, S. Berberine and musculoskeletal disorders: The therapeutic potential and underlying molecular mechanisms. Phytomedicine 2019, 152892. [Google Scholar] [CrossRef] [PubMed]
  22. Liu, S.C.; Lee, H.P.; Hung, C.Y.; Tsai, C.H.; Li, T.M.; Tang, C.H. Berberine attenuates CCN2-induced IL-1β expression and prevents cartilage degradation in a rat model of osteoarthritis. Toxicol. Appl. Pharmacol. 2015, 289, 20–29. [Google Scholar] [CrossRef]
  23. Zhou, Y.; Tao, H.; Li, Y.; Deng, M.; He, B.; Xia, S.; Zhang, C.; Liu, S. Berberine promotes proliferation of sodium nitroprusside-stimulated rat chondrocytes and osteoarthritic rat cartilage via Wnt/β-catenin pathway. Eur. J. Pharmacol. 2016, 789, 109–118. [Google Scholar] [CrossRef] [PubMed]
  24. Zhou, Y.; Liu, S.-Q.; Yu, L.; He, B.; Wu, S.-H.; Zhao, Q.; Xia, S.-Q.; Mei, H.-J. Berberine prevents nitric oxide-induced rat chondrocyte apoptosis and cartilage degeneration in a rat osteoarthritis model via AMPK and p38 MAPK signaling. Apoptosis 2015, 20, 1187–1199. [Google Scholar] [CrossRef] [PubMed]
  25. Zhou, Y.; Liu, S.-Q.; Peng, H.; Yu, L.; He, B.; Zhao, Q. In vivo anti-apoptosis activity of novel berberine-loaded chitosan nanoparticles effectively ameliorates osteoarthritis. Int. Immunopharmacol. 2015, 28, 34–43. [Google Scholar] [CrossRef] [PubMed]
  26. Zhou, Y.; Ming, J.; Deng, M.; Li, Y.; Li, B.; Li, J.; Ma, Y.; Chen, Z.; Liu, S. Berberine-mediated up-regulation of surfactant protein D facilitates cartilage repair by modulating immune responses via the inhibition of TLR4/NF-ĸB signaling. Pharmacol. Res. 2020, 155, 104690. [Google Scholar] [CrossRef]
  27. Lee, H.W.; Suh, J.H.; Kim, H.N.; Kim, A.Y.; Park, S.Y.; Shin, C.S.; Choi, J.-Y.; Kim, J.B. Berberine Promotes Osteoblast Differentiation by Runx2 Activation With p38 MAPK. J. Bone Miner. Res. 2008, 23, 1227–1237. [Google Scholar] [CrossRef]
  28. Wei, P.; Jiao, L.; Qin, L.-P.; Yan, F.; Han, T.; Zhang, Q.-Y. Effects of berberine on differentiation and bone resorption of osteoclasts derived from rat bone marrow cells. J. Chin. Integr. Med. 2009, 7, 342–348. [Google Scholar] [CrossRef]
  29. Hostetler, G.L.; Ralston, R.A.; Schwartz, S.J. Flavones: Food Sources, Bioavailability, Metabolism, and Bioactivity. Adv. Nutr. Int. Rev. J. 2017, 8, 423–435. [Google Scholar] [CrossRef] [Green Version]
  30. Salehi, B.; Venditti, A.; Sharifi-Rad, M.; Kregiel, D.; Sharifi-Rad, J.; Durazzo, A.; Lucarini, M.; Santini, A.; Souto, E.B.; Novellino, E.; et al. The Therapeutic Potential of Apigenin. Int. J. Mol. Sci. 2019, 20, 1305. [Google Scholar] [CrossRef] [Green Version]
  31. Shoara, R.; Hashempur, M.H.; Ashraf, A.; Salehi, A.; Dehshahri, S.; Habibagahi, Z. Efficacy and safety of topical Matricaria chamomilla L. (chamomile) oil for knee osteoarthritis: A randomized controlled clinical trial. Complement. Ther. Clin. Pract. 2015, 21, 181–187. [Google Scholar] [CrossRef]
  32. Davidson, R.K.; Green, J.; Gardner, S.; Bao, Y.; Cassidy, A.; Clark, I.M. Identifying chondroprotective diet-derived bioactives and investigating their synergism. Sci. Rep. 2018, 8, 17173. [Google Scholar] [CrossRef] [PubMed]
  33. Park, J.S.; Kim, D.K.; Shin, H.-D.; Lee, H.J.; Jo, H.S.; Jeong, J.H.; Choi, Y.L.; Lee, C.J.; Hwang, S.-C. Apigenin Regulates Interleukin-1β-Induced Production of Matrix Metalloproteinase Both in the Knee Joint of Rat and in Primary Cultured Articular Chondrocytes. Biomol. Ther. 2016, 24, 163–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Melguizo-Rodríguez, L.; Manzano-Moreno, F.J.; Illescas-Montes, R.; Ramos-Torrecillas, J.; De luna-Bertos, E.; Ruiz, C.; Garcia-Martinez, O. Bone Protective Effect of Extra-Virgin Olive Oil Phenolic Compounds by Modulating Osteoblast Gene Expression. Nutrients 2019, 11, 1722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Zhang, X.; Zhou, C.; Zha, X.; Xu, Z.; Li, L.; Liu, Y.; Xu, L.; Cui, L.; Xu, D.; Zhu, B. Apigenin promotes osteogenic differentiation of human mesenchymal stem cells through JNK and p38 MAPK pathways. Mol. Cell. Biochem. 2015, 407, 41–50. [Google Scholar] [CrossRef] [PubMed]
  36. Riaz, A.; Rasul, A.; Hussain, G.; Zahoor, M.K.; Jabeen, F.; Subhani, Z.; Younis, T.; Ali, M.; Sarfraz, I.; Selamoglu, Z. Astragalin: A Bioactive Phytochemical with Potential Therapeutic Activities. Adv. Pharmacol. Sci. 2018, 1–15. [Google Scholar] [CrossRef] [PubMed]
  37. Liu, L.; Wang, D.; Qin, Y.; Xu, M.; Zhou, L.; Xu, W.; Liu, X.; Ye, L.; Yue, S.; Zheng, Q.; et al. Astragalin Promotes Osteoblastic Differentiation in MC3T3-E1 Cells and Bone Formation in vivo. Front. Endocrinol. 2019, 10, 228. [Google Scholar] [CrossRef]
  38. Ma, Z.; Piao, T.; Wang, Y.; Liu, J. Astragalin inhibits IL-1β-induced inflammatory mediators production in human osteoarthritis chondrocyte by inhibiting NF-κB and MAPK activation. Int. Immunopharmacol. 2015, 25, 83–87. [Google Scholar] [CrossRef]
  39. Jia, Q.; Wang, T.; Wang, X.; Xu, H.; Liu, Y.; Wang, Y.; Shi, Q.; Liang, Q. Astragalin Suppresses Inflammatory Responses and Bone Destruction in Mice With Collagen-Induced Arthritis and in Human Fibroblast-Like Synoviocytes. Front. Pharmacol. 2019, 10, 94. [Google Scholar] [CrossRef] [Green Version]
  40. Sowndhararajan, K.; Deepa, P.; Kim, M.; Park, S.J.; Kim, S. Baicalein as a potent neuroprotective agent: A review. Biomed. Pharmacother. 2017, 95, 1021–1032. [Google Scholar] [CrossRef]
  41. Bie, B.; Sun, J.; Guo, Y.; Li, J.; Jiang, W.; Yang, J.; Huang, C.; Li, Z. Baicalein: A review of its anti-cancer effects and mechanisms in Hepatocellular Carcinoma. Biomed. Pharmacother. 2017, 93, 1285–1291. [Google Scholar] [CrossRef]
  42. Zhang, X.; Zhu, Y.; Chen, X.; Zhang, Y.; Zhang, Y.; Jia, Y.; Wang, H.; Liu, Y.; Xiao, L. Baicalein ameliorates inflammatory-related apoptotic and catabolic phenotypes in human chondrocytes. Int. Immunopharmacol. 2014, 21, 301–308. [Google Scholar] [CrossRef] [PubMed]
  43. Li, Y.; Wang, J.; Song, X.; Bai, H.; Ma, T.; Zhang, Z.; Li, X.; Jiang, R.; Wang, G.; Fan, X.; et al. Effects of baicalein on IL-1β-induced inflammation and apoptosis in rat articular chondrocytes. Oncotarget 2017, 8, 90781–90795. [Google Scholar] [CrossRef]
  44. Chen, W.-P.; Xiong, Y.; Hu, P.-F.; Bao, J.-P.; Wu, L.-D. Baicalein Inhibits MMPs Expression via a MAPK-Dependent Mechanism in Chondrocytes. Cell. Physiol. Biochem. 2015, 36, 325–333. [Google Scholar] [CrossRef] [PubMed]
  45. Kim, M.H.; Ryu, S.Y.; Bae, M.A.; Choi, J.-S.; Min, Y.K.; Kim, S.H. Baicalein inhibits osteoclast differentiation and induces mature osteoclast apoptosis. Food Chem. Toxicol. 2008, 46, 3375–3382. [Google Scholar] [CrossRef]
  46. Li, S.; Tang, J.-J.; Chen, J.; Zhang, P.; Wang, T.; Chen, T.-Y.; Yan, B.; Huang, B.; Wang, L.; Huang, M.-J.; et al. Regulation of bone formation by baicalein via the mTORC1 pathway. Drug Des. Devel. Ther. 2015, 9, 5169–5183. [Google Scholar] [CrossRef] [PubMed]
  47. Samarghandian, S.; Farkhondeh, T.; Azimi-Nezhad, M. Protective Effects of Chrysin Against Drugs and Toxic Agents. Dose Response 2017, 15, 1559325817711782. [Google Scholar] [CrossRef]
  48. Zheng, W.; Tao, Z.; Cai, L.; Chen, C.; Zhang, C.; Wang, Q.; Ying, X.; Hu, W.; Chen, H. Chrysin Attenuates IL-1β-Induced Expression of Inflammatory Mediators by Suppressing NF-κB in Human Osteoarthritis Chondrocytes. Inflammation 2017, 40, 1143–1154. [Google Scholar] [CrossRef]
  49. Zhang, C.; Yu, W.; Huang, C.; Ding, Q.; Liang, C.; Wang, L.; Hou, Z.; Zhang, Z. Chrysin protects human osteoarthritis chondrocytes by inhibiting inflammatory mediator expression via HMGB1 suppression. Mol. Med. Rep. 2018, 19, 1222–1229. [Google Scholar] [CrossRef] [Green Version]
  50. Zeng, W.; Yan, Y.; Zhang, F.; Zhang, C.; Liang, W. Chrysin promotes osteogenic differentiation via ERK/MAPK activation. Protein Cell 2013, 4, 539–547. [Google Scholar] [CrossRef] [Green Version]
  51. Menon, A.H.; Soundarya, S.P.; Sanjay, V.; Chandran, S.V.; Balagangadharan, K.; Selvamurugan, N. Sustained release of chrysin from chitosan-based scaffolds promotes mesenchymal stem cell proliferation and osteoblast differentiation. Carbohydr. Polym. 2018, 195, 356–367. [Google Scholar] [CrossRef]
  52. Mukund, V.; Mukund, D.; Sharma, V.; Mannarapu, M.; Alam, A. Genistein: Its role in metabolic diseases and cancer. Crit. Rev. Oncol. Hematol. 2017, 119, 13–22. [Google Scholar] [CrossRef]
  53. Oliviero, F.; Scanu, A.; Zamudio-Cuevas, Y.; Punzi, L.; Spinella, P. Anti-inflammatory effects of polyphenols in arthritis. J. Sci. Food Agric. 2018, 98, 1653–1659. [Google Scholar] [CrossRef] [PubMed]
  54. Spagnuolo, C.; Moccia, S.; Russo, G.L. Anti-inflammatory effects of flavonoids in neurodegenerative disorders. Eur. J. Med. Chem. 2018, 153, 105–115. [Google Scholar] [CrossRef] [PubMed]
  55. Claassen, H.; Briese, V.; Manapov, F.; Nebe, B.; Schünke, M.; Kurz, B. The phytoestrogens daidzein and genistein enhance the insulin-stimulated sulfate uptake in articular chondrocytes. Cell Tissue Res. 2008, 333, 71–79. [Google Scholar] [CrossRef]
  56. Tanamas, S.K.; Wijethilake, P.; Wluka, A.E.; Davies-Tuck, M.L.; Urquhart, D.M.; Wang, Y.; Cicuttini, F.M. Sex hormones and structural changes in osteoarthritis: A systematic review. Maturitas 2011, 69, 141–156. [Google Scholar] [CrossRef] [PubMed]
  57. Thangavel, P.; Puga-Olguín, A.; Rodríguez-Landa, J.F.; Zepeda, R.C. Genistein as Potential Therapeutic Candidate for Menopausal Symptoms and Other Related Diseases. Molecules 2019, 24, 3892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Liu, F.-C.; Wang, C.-C.; Lu, J.-W.; Lee, C.-H.; Chen, S.-C.; Ho, Y.-J.; Peng, Y.-J. Chondroprotective Effects of Genistein against Osteoarthritis Induced Joint Inflammation. Nutrients 2019, 11, 1180. [Google Scholar] [CrossRef] [Green Version]
  59. Yuan, J.; Ding, W.; Wu, N.; Jiang, S.; Li, W. Protective Effect of Genistein on Condylar Cartilage through Downregulating NF-κB Expression in Experimentally Created Osteoarthritis Rats. BioMed Res. Int. 2019, 3, 1–6. [Google Scholar] [CrossRef] [Green Version]
  60. Ming, L.-G.; Chen, K.-M.; Xian, C.J. Functions and action mechanisms of flavonoids genistein and icariin in regulating bone remodeling. J. Cell. Physiol. 2013, 228, 513–521. [Google Scholar] [CrossRef]
  61. Kim, M.; Lim, J.; Lee, J.-H.; Lee, K.-M.; Kim, S.; Park, K.W.; Nho, C.W.; Cho, Y.S. Understanding the functional role of genistein in the bone differentiation in mouse osteoblastic cell line MC3T3-E1 by RNA-seq analysis. Sci. Rep. 2018, 8, 3257. [Google Scholar] [CrossRef]
  62. Cepeda, S.B.; Sandoval, M.J.; Crescitelli, M.C.; Rauschemberger, M.B.; Massheimer, V.L. The isoflavone genistein enhances osteoblastogenesis: Signaling pathways involved. J. Physiol. Biochem. 2020, 76, 99–110. [Google Scholar] [CrossRef]
  63. Yamaguchi, M.; Levy, R.M. Combination of alendronate and genistein synergistically suppresses osteoclastic differentiation of RAW267.4 cells in vitro. Exp. Ther. Med. 2017, 14, 1769–1774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Li, D.; Yuan, T.; Zhang, X.; Xiao, Y.; Wang, R.; Fan, Y.; Zhang, X. Icariin: A potential promoting compound for cartilage tissue engineering. Osteoarthr. Cartil. 2012, 20, 1647–1656. [Google Scholar] [CrossRef] [Green Version]
  65. Wang, P.; Zhang, F.; He, Q.; Wang, J.; Shiu, H.T.; Shu, Y.; Tsang, W.P.; Liang, S.; Zhao, K.; Wan, C. Flavonoid Compound Icariin Activates Hypoxia Inducible Factor-1α in Chondrocytes and Promotes Articular Cartilage Repair. PLoS ONE 2016, 11, e0148372. [Google Scholar] [CrossRef] [Green Version]
  66. Liu, M.-H.; Sun, J.-S.; Tsai, S.-W.; Sheu, S.-Y.; Chen, M.-H. Icariin protects murine chondrocytes from lipopolysaccharide-induced inflammatory responses and extracellular matrix degradation. Nutr. Res. 2010, 30, 57–65. [Google Scholar] [CrossRef] [PubMed]
  67. Mi, B.; Wang, J.; Liu, Y.; Liu, J.; Hu, L.; Panayi, A.C.; Liu, G.; Zhou, W. Icariin Activates Autophagy via Down-Regulation of the NF-κB Signaling-Mediated Apoptosis in Chondrocytes. Front. Pharmacol. 2018, 9, 605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Zuo, S.; Zou, W.; Wu, R.M.; Yang, J.; Fan, J.N.; Zhao, X.K.; Li, H.Y. Icariin Alleviates IL-1β-Induced Matrix Degradation By Activating The Nrf2/ARE Pathway In Human Chondrocytes. Drug Des. Devel. Ther. 2019, 13, 3949–3961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Song, L.; Zhao, J.; Zhang, X.; Li, H.; Zhou, Y. Icariin induces osteoblast proliferation, differentiation and mineralization through estrogen receptor-mediated ERK and JNK signal activation. Eur. J. Pharmacol. 2013, 714, 15–22. [Google Scholar] [CrossRef]
  70. Huang, Z.; Cheng, C.; Wang, J.; Liu, X.; Wei, H.; Han, Y.; Yang, S.; Wang, X. Icariin regulates the osteoblast differentiation and cell proliferation of MC3T3-E1 cells through microRNA-153 by targeting Runt-related transcription factor 2. Exp. Ther. Med. 2018, 15, 5159–5166. [Google Scholar] [CrossRef] [Green Version]
  71. Ma, H.-P.; Ma, X.-N.; Ge, B.-F.; Zhen, P.; Zhou, J.; Gao, Y.-H.; Xian, C.J.; Chen, K.-M. Icariin attenuates hypoxia-induced oxidative stress and apoptosis in osteoblasts and preserves their osteogenic differentiation potential in vitro. Cell Prolif. 2014, 47, 527–539. [Google Scholar] [CrossRef] [PubMed]
  72. Gao, K.; Wang, S.; Wang, Q. Effect of icariin on serum bone turnover markers expressions and histology changes in mouse osteoarthritis model. Chin. J. Reparative Reconstr. Surg. 2017, 31, 963–969. [Google Scholar] [CrossRef]
  73. Ren, J.; Lu, Y.; Qian, Y.; Chen, B.; Wu, T.; Ji, G. Recent progress regarding kaempferol for the treatment of various diseases (Review). Exp. Ther. Med. 2019, 18, 2759–2776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zhuang, Z.; Ye, G.; Huang, B. Kaempferol Alleviates the Interleukin-1β-Induced Inflammation in Rat Osteoarthritis Chondrocytes via Suppression of NF-κB. Med. Sci. Monit. 2017, 23, 3925–3931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Huang, X.; Pan, Q.; Mao, Z.; Wang, P.; Zhang, R.; Ma, X.; Chen, J.; You, H. Kaempferol inhibits interleukin-1β stimulated matrix metalloproteinases by suppressing the MAPK-associated ERK and P38 signaling pathways. Mol. Med. Rep. 2018, 18, 2697–2704. [Google Scholar] [CrossRef] [Green Version]
  76. Kim, I.-R.; Kim, S.-E.; Baek, H.-S.; Kim, B.-J.; Kim, C.-H.; Chung, I.-K.; Park, B.-S.; Shin, S.-H. The role of kaempferol-induced autophagy on differentiation and mineralization of osteoblastic MC3T3-E1 cells. BMC Complement. Altern. Med. 2016, 16, 333. [Google Scholar] [CrossRef] [Green Version]
  77. Sharma, A.R.; Nam, J.-S. Kaempferol stimulates WNT/β-catenin signaling pathway to induce differentiation of osteoblasts. J. Nutr. Biochem. 2019, 74, 108228. [Google Scholar] [CrossRef]
  78. Zhao, J.; Wu, J.; Xu, B.; Yuan, Z.; Leng, Y.; Min, J.; Lan, X.; Luo, J. Kaempferol promotes bone formation in part via the mTOR signaling pathway. Mol. Med. Rep. 2019, 20, 5197–5207. [Google Scholar] [CrossRef]
  79. Kim, C.-J.; Shin, S.-H.; Kim, B.-J.; Kim, C.-H.; Kim, J.-H.; Kang, H.-M.; Park, B.-S.; Kim, I.-R. The Effects of Kaempferol-Inhibited Autophagy on Osteoclast Formation. Int. J. Mol. Sci. 2018, 19, 125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Ma, X.-Q.; Han, T.; Zhang, X.; Wu, J.-Z.; Rahman, K.; Qin, L.-P.; Zheng, C.-J. Kaempferitrin prevents bone lost in ovariectomized rats. Phytomed. Int. J. Phytother. Phytopharm. 2015, 22, 1159–1162. [Google Scholar] [CrossRef]
  81. Adhikary, S.; Choudhary, D.; Ahmad, N.; Karvande, A.; Kumar, A.; Banala, V.T.; Mishra, P.R.; Trivedi, R. Dietary flavonoid kaempferol inhibits glucocorticoid-induced bone loss by promoting osteoblast survival. Nutrition 2018, 53, 64–76. [Google Scholar] [CrossRef]
  82. Aziz, N.; Kim, M.-Y.; Cho, J.Y. Anti-inflammatory effects of luteolin: A review of in vitro, in vivo, and in silico studies. J. Ethnopharmacol. 2018, 225, 342–358. [Google Scholar] [CrossRef]
  83. Fei, J.; Liang, B.; Jiang, C.; Ni, H.; Wang, L. Luteolin inhibits IL-1β-induced inflammation in rat chondrocytes and attenuates osteoarthritis progression in a rat model. Biomed. Pharmacother. 2019, 109, 1586–1592. [Google Scholar] [CrossRef] [PubMed]
  84. Moncada-Pazos, A.; Obaya, A.J.; Viloria, C.G.; López-Otín, C.; Cal, S. The nutraceutical flavonoid luteolin inhibits ADAMTS-4 and ADAMTS-5 aggrecanase activities. J. Mol. Med. 2011, 89, 611–619. [Google Scholar] [CrossRef] [PubMed]
  85. Kang, B.-J.; Ryu, J.; Lee, C.J.; Hwang, S.-C. Luteolin Inhibits the Activity, Secretion and Gene Expression of MMP-3 in Cultured Articular Chondrocytes and Production of MMP-3 in the Rat Knee. Biomol. Ther. 2014, 22, 239–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Wu, L.; Liu, H.; Li, L.; Xu, D.; Gao, Y.; Guan, Y.; Chen, Q. 5,7,3′,4′-Tetramethoxyflavone protects chondrocytes from ER stress-induced apoptosis through regulation of the IRE1α pathway. Connect. Tissue Res. 2018, 59, 157–166. [Google Scholar] [CrossRef]
  87. Huang, X.; Chen, Z.; Shi, W.; Zhang, R.; Li, L.; Liu, H.; Wu, L. TMF inhibits miR-29a/Wnt/β-catenin signaling through upregulating Foxo3a activity in osteoarthritis chondrocytes. Drug Des. Dev. Ther. 2019, 13, 2009–2019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Choi, E.-M. Modulatory effects of luteolin on osteoblastic function and inflammatory mediators in osteoblastic MC3T3-E1 cells. Cell Biol. Int. 2007, 31, 870–877. [Google Scholar] [CrossRef]
  89. Nash, L.A.; Sullivan, P.J.; Peters, S.J.; Ward, W.E. Rooibos flavonoids, orientin and luteolin, stimulate mineralization in human osteoblasts through the Wnt pathway. Mol. Nutr. Food Res. 2015, 59, 443–453. [Google Scholar] [CrossRef] [PubMed]
  90. Yang, H.; Liu, Q.; Ahn, J.H.; Kim, S.B.; Kim, Y.C.; Sung, S.H.; Hwang, B.Y.; Lee, M.K. Luteolin downregulates IL-1β-induced MMP-9 and -13 expressions in osteoblasts via inhibition of ERK signalling pathway. J. Enzyme Inhib. Med. Chem. 2012, 27, 261–266. [Google Scholar] [CrossRef] [PubMed]
  91. Abbasi, N.; Khosravi, A.; Aidy, A.; Shafiei, M. Biphasic Response to Luteolin in MG-63 Osteoblast-Like Cells under High Glucose-Induced Oxidative Stress. Iran. J. Med. Sci. 2016, 41, 118–125. [Google Scholar]
  92. Jing, Z.; Wang, C.; Yang, Q.; Wei, X.; Jin, Y.; Meng, Q.; Liu, Q.; Liu, Z.; Ma, X.; Liu, K.; et al. Luteolin attenuates glucocorticoid-induced osteoporosis by regulating ERK/Lrp-5/GSK-3β signaling pathway in vivo and in vitro. J. Cell. Physiol. 2019, 234, 4472–4490. [Google Scholar] [CrossRef]
  93. Kim, T.-H.; Jung, J.W.; Ha, B.G.; Hong, J.M.; Park, E.K.; Kim, H.-J.; Kim, S.-Y. The effects of luteolin on osteoclast differentiation, function in vitro and ovariectomy-induced bone loss. J. Nutr. Biochem. 2011, 22, 8–15. [Google Scholar] [CrossRef]
  94. Song, F.; Wei, C.; Zhou, L.; Qin, A.; Yang, M.; Tickner, J.; Huang, Y.; Zhao, J.; Xu, J. Luteoloside prevents lipopolysaccharide-induced osteolysis and suppresses RANKL-induced osteoclastogenesis through attenuating RANKL signaling cascades. J. Cell. Physiol. 2017, 233, 1723–1735. [Google Scholar] [CrossRef] [Green Version]
  95. Su, Y.-X.; Yan, H.; Chen, B.-J.; Zahn, Q.; Wang, Y.-R.; Lu, M.L.; Wang, W.-T.; He, Z.; Sheng, L. Effect of naringin of Drynaria Rhizome, a Chinese medical component of Zhuanggu Jianxi Recipe containing serum on caveolin-p38MAPK signal pathway in IL-1β induced rabbit degenerated chondrocytes. Chin. J. Integr. Tradit. West. Med. 2014, 34, 1492–1498. [Google Scholar]
  96. Zhao, Y.; Li, Z.; Wang, W.; Zhang, H.; Chen, J.; Su, P.; Liu, L.; Li, W. Naringin Protects Against Cartilage Destruction in Osteoarthritis Through Repression of NF-κB Signaling Pathway. Inflammation 2016, 39, 385–392. [Google Scholar] [CrossRef] [PubMed]
  97. Wang, C.C.; Guo, L.; Tian, F.D.; An, N.; Luo, L.; Hao, R.H.; Wang, B.; Zhou, Z.H. Naringenin regulates production of matrix metalloproteinases in the knee-joint and primary cultured articular chondrocytes and alleviates pain in rat osteoarthritis model. Braz. J. Med. Biol. Res. 2017, 50, e5714. [Google Scholar] [CrossRef] [Green Version]
  98. Xu, Q.; Zhang, Z.-F.; Sun, W.-X. Effect of Naringin on Monosodium Iodoacetate-Induced Osteoarthritis Pain in Rats. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2017, 23, 3746–3751. [Google Scholar] [CrossRef] [Green Version]
  99. Li, N.; Jiang, Y.; Wooley, P.H.; Xu, Z.; Yang, S.-Y. Naringin promotes osteoblast differentiation and effectively reverses ovariectomy-associated osteoporosis. J. Orthop. Sci. Off. J. Jpn. Orthop. Assoc. 2013, 18, 478–485. [Google Scholar] [CrossRef]
  100. Wang, D.; Ma, W.; Wang, F.; Dong, J.; Wang, D.; Sun, B.; Wang, B. Stimulation of Wnt/β-Catenin Signaling to Improve Bone Development by Naringin via Interacting with AMPK and Akt. Cell. Physiol. Biochem. 2015, 36, 1563–1576. [Google Scholar] [CrossRef] [PubMed]
  101. Fan, J.; Li, J.; Fan, Q. Naringin promotes differentiation of bone marrow stem cells into osteoblasts by upregulating the expression levels of microRNA-20a and downregulating the expression levels of PPARγ. Mol. Med. Rep. 2015, 12, 4759–4765. [Google Scholar] [CrossRef] [Green Version]
  102. Zhai, Y.-K.; Niu, Y.-B.; Pan, Y.-L.; Li, C.-R.; Wu, X.-L.; Mei, Q.-B. Effects of naringin on proliferation, differentiation and maturation of rat calvarial osteoblasts in vitro. Chin. J. Integr. Tradit. West. Med. 2013, 38, 105–111. [Google Scholar]
  103. Xu, T.; Wang, L.; Tao, Y.; Ji, Y.; Deng, F.; Wu, X.-H. The Function of Naringin in Inducing Secretion of Osteoprotegerin and Inhibiting Formation of Osteoclasts. Evid.-Based Complement. Altern. Med. 2016, 8981650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Zhou, Y.-X.; Zhang, H.; Peng, C. Puerarin: A Review of Pharmacological Effects: ACTIVITY OF PUERARIN. Phytother. Res. 2014, 28, 961–975. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, L.; Shan, H.; Wang, B.; Wang, N.; Zhou, Z.; Pan, C.; Wang, F. Puerarin Attenuates Osteoarthritis via Upregulating AMP-Activated Protein Kinase/Proliferator-Activated Receptor-γ Coactivator-1 Signaling Pathway in Osteoarthritis Rats. Pharmacology 2018, 102, 117–125. [Google Scholar] [CrossRef] [PubMed]
  106. Peng, L.; Xie, Z.; Pei, J.; Wang, B.; Gao, Y.; Qu, Y. Puerarin alters the function of monocytes/macrophages and exhibits chondroprotection in mice. Mol. Med. Rep. 2019, 19, 2876–2882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Wang, P.-P.; Zhu, X.-F.; Yang, L.; Liang, H.; Feng, S.-W.; Zhang, R.-H. Puerarin stimulates osteoblasts differentiation and bone formation through estrogen receptor, p38 MAPK, and Wnt/β-catenin pathways. J. Asian Nat. Prod. Res. 2012, 14, 897–905. [Google Scholar] [CrossRef]
  108. Yang, X.; Yang, Y.; Zhou, S.; Gong, X.; Dai, Q.; Zhang, P.; Jiang, L. Puerarin Stimulates Osteogenic Differentiation and Bone Formation Through the ERK1/2 and p38-MAPK Signaling Pathways. Curr. Mol. Med. 2018, 17, 488–496. [Google Scholar] [CrossRef]
  109. Zhan, X.-Q.; Zeng, W.-W.; Zhang, Y.-Y.; Feng, Q.; Zhao, F.-M.; Jiang, Z.-Q.; Sun, C. Puerarin promotes the viability and differentiation of MC3T3-E1 cells by miR-204-regulated Runx2 upregulation. Mol. Med. Rep. 2017, 16, 6262–6268. [Google Scholar] [CrossRef] [PubMed]
  110. Zeng, X.; Feng, Q.; Zhao, F.; Sun, C.; Zhou, T.; Yang, J.; Zhan, X. Puerarin inhibits TRPM3/miR-204 to promote MC3T3-E1 cells proliferation, differentiation and mineralization. Phytother. Res. 2018, 32, 996–1003. [Google Scholar] [CrossRef] [PubMed]
  111. Feng, Q.; Cheng, S.-Y.; Yang, R.; Zeng, X.-W.; Zhao, F.-M.; Zhan, X.-Q. Puerarin promotes the viability and differentiation of MC3T3-E1 cells by enhancing LC3B-mediated autophagy through downregulation of miR-204. Exp. Ther. Med. 2019, 19, 883–890. [Google Scholar] [CrossRef] [Green Version]
  112. Liu, H.; Li, W.; Jian, S.; Li, B. Puerarin and zinc additively prevent mandibular bone loss through inhibiting osteoclastogenesis in ovariectomized rats. Histol. Histopathol. 2017, 32, 851–860. [Google Scholar] [CrossRef]
  113. Park, K.H.; Gu, D.R.; Jin, S.H.; Yoon, C.-S.; Ko, W.; Kim, Y.C.; Lee, S.H. Pueraria lobate Inhibits RANKL-Mediated Osteoclastogenesis Via Downregulation of CREB/PGC1β/c-Fos/NFATc1 Signaling. Am. J. Chin. Med. 2017, 45, 1725–1744. [Google Scholar] [CrossRef] [PubMed]
  114. Zhang, Y.; Yan, M.; Yu, Q.; Yang, P.; Zhang, H.; Sun, Y.; Zhang, Z.; Gao, Y. Puearin prevents LPS-induced Osteoclast formation and bone loss via inhibition of akt activation. Biol. Pharm. Bull. 2016, 39, 2028–2035. [Google Scholar] [CrossRef] [Green Version]
  115. Soleimani, V.; Delghandi, P.S.; Moallem, S.A.; Karimi, G. Safety and toxicity of silymarin, the major constituent of milk thistle extract: An updated review. Phytother. Res. 2019, 33, 1627–1638. [Google Scholar] [CrossRef]
  116. Xie, Y.; Zhang, D.; Zhang, J.; Yuan, J. Metabolism, Transport and Drug-Drug Interactions of Silymarin. Molecules 2019, 24, 3693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Dupuis, M.L.; Conti, F.; Maselli, A.; Pagano, M.-T.; Ruggieri, A.; Anticoli, S.; Fragale, A.; Gabriele, L.; Gagliardi, M.C.; Sanchez, M.; et al. The Natural Agonist of Estrogen Receptor β Silibinin Plays an Immunosuppressive Role Representing a Potential Therapeutic Tool in Rheumatoid Arthritis. Front. Immunol. 2018, 9, 1903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Hussain, S.A.; Jassim, N.A.; Numan, I.T.; Al-Khalifa, I.I.; Abdullah, T.A. Anti-inflammatory activity of silymarin in patients with knee osteoarthritis. A comparative study with piroxicam and meloxicam. Saudi Med. J. 2009, 30, 98–103. [Google Scholar]
  119. Ashkavand, Z.; Malekinejad, H.; Amniattalab, A.; Rezaei-Golmisheh, A.; Vishwanath, B.S. Silymarin potentiates the anti-inflammatory effects of Celecoxib on chemically induced osteoarthritis in rats. Phytomedicine 2012, 19, 1200–1205. [Google Scholar] [CrossRef] [PubMed]
  120. Zheng, W.; Feng, Z.; Lou, Y.; Chen, C.; Zhang, C.; Tao, Z.; Li, H.; Cheng, L.; Ying, X. Silibinin protects against osteoarthritis through inhibiting the inflammatory response and cartilage matrix degradation in vitro and in vivo. Oncotarget 2017, 8, 99649–99665. [Google Scholar] [CrossRef] [Green Version]
  121. Kim, J.H.; Kim, K.; Jin, H.M.; Song, I.; Youn, B.U.; Lee, J.; Kim, N. Silibinin inhibits osteoclast differentiation mediated by TNF family members. Mol. Cells 2009, 28, 201–207. [Google Scholar] [CrossRef] [PubMed]
  122. Kim, J.-L.; Kang, S.-W.; Kang, M.-K.; Gong, J.-H.; Lee, E.-S.; Han, S.J.; Kang, Y.-H. Osteoblastogenesis and osteoprotection enhanced by flavonolignan silibinin in osteoblasts and osteoclasts. J. Cell. Biochem. 2012, 113, 247–259. [Google Scholar] [CrossRef]
  123. Kim, J.-L.; Park, S.-H.; Jeong, D.; Nam, J.-S.; Kang, Y.-H. Osteogenic activity of silymarin through enhancement of alkaline phosphatase and osteocalcin in osteoblasts and tibia-fractured mice. Exp. Biol. Med. 2012, 237, 417–428. [Google Scholar] [CrossRef] [PubMed]
  124. Ying, X.; Sun, L.; Chen, X.; Xu, H.; Guo, X.; Chen, H.; Hong, J.; Cheng, S.; Peng, L. Silibinin promotes osteoblast differentiation of human bone marrow stromal cells via bone morphogenetic protein signaling. Eur. J. Pharmacol. 2013, 721, 225–230. [Google Scholar] [CrossRef] [PubMed]
  125. Mao, Y.X.; Cai, W.J.; Sun, X.Y.; Dai, P.P.; Li, X.M.; Wang, Q.; Huang, X.L.; He, B.; Wang, P.P.; Wu, G.; et al. RAGE-dependent mitochondria pathway: A novel target of silibinin against apoptosis of osteoblastic cells induced by advanced glycation end products. Cell Death Dis. 2018, 9, 674. [Google Scholar] [CrossRef]
  126. Kim, J.-L.; Kim, Y.-H.; Kang, M.-K.; Gong, J.-H.; Han, S.-J.; Kang, Y.-H. Antiosteoclastic activity of milk thistle extract after ovariectomy to suppress estrogen deficiency-induced osteoporosis. BioMed Res. Int. 2013, 919374. [Google Scholar] [CrossRef] [Green Version]
  127. Wu, X.; Zhang, H.; Salmani, J.M.M.; Fu, R.; Chen, B. Advances of wogonin, an extract from Scutellaria baicalensis, for the treatment of multiple tumors. OncoTargets Ther. 2016, 9, 2935–2943. [Google Scholar] [CrossRef] [Green Version]
  128. Tai, M.C.; Tsang, S.Y.; Chang, L.Y.F.; Xue, H. Therapeutic potential of wogonin: A naturally occurring flavonoid. CNS Drug Rev. 2005, 11, 141–150. [Google Scholar] [CrossRef]
  129. Lim, H.; Park, H.; Kim, H.P. Effects of flavonoids on matrix metalloproteinase-13 expression of interleukin-1β-treated articular chondrocytes and their cellular mechanisms: Inhibition of c-Fos/AP-1 and JAK/STAT signaling pathways. J. Pharmacol. Sci. 2011, 116, 221–231. [Google Scholar] [CrossRef] [Green Version]
  130. Khan, N.M.; Haseeb, A.; Ansari, M.Y.; Haqqi, T.M. A wogonin-rich-fraction of Scutellaria baicalensis root extract exerts chondroprotective effects by suppressing IL-1β-induced activation of AP-1 in human OA chondrocytes. Sci. Rep. 2017, 7, 43789. [Google Scholar] [CrossRef] [Green Version]
  131. Park, J.S.; Lee, H.J.; Lee, D.Y.; Jo, H.S.; Jeong, J.H.; Kim, D.H.; Nam, D.C.; Lee, C.J.; Hwang, S.-C. Chondroprotective Effects of Wogonin in Experimental Models of Osteoarthritis in vitro and in vivo. Biomol. Ther. 2015, 23, 442–448. [Google Scholar] [CrossRef] [Green Version]
  132. Khan, N.M.; Haseeb, A.; Ansari, M.Y.; Devarapalli, P.; Haynie, S.; Haqqi, T.M. Wogonin, a plant derived small molecule, exerts potent anti-inflammatory and chondroprotective effects through the activation of ROS/ERK/Nrf2 signaling pathways in human Osteoarthritis chondrocytes. Free Radic. Biol. Med. 2017, 106, 288–301. [Google Scholar] [CrossRef] [Green Version]
  133. Sirong, S.; Yang, C.; Taoran, T.; Songhang, L.; Shiyu, L.; Yuxin, Z.; Xiaoru, S.; Tao, Z.; Yunfeng, L.; Xiaixiao, C. Effects of tetrahedral framework nucleic acid/wogonin complexes on osteoarthritis. Bone Res. 2020, 8, 6. [Google Scholar] [CrossRef] [Green Version]
  134. Fang, W.; Zhou, X.; Wang, J.; Xu, L.; Zhou, L.; Yu, W.; Tao, Y.; Zhu, J.; Hu, B.; Liang, C.; et al. Wogonin mitigates intervertebral disc degeneration through the Nrf2/ARE and MAPK signaling pathways. Int. Immunopharmacol. 2018, 65, 539–549. [Google Scholar] [CrossRef]
  135. Zhao, P.; Cheng, J.; Geng, J.; Yang, M.; Zhang, Y.; Zhang, Q.; Wang, Y.; Lu, B. Curcumin protects rabbit articular chondrocytes against sodium nitroprusside-induced apoptosis in vitro. Eur. J. Pharmacol. 2018, 828, 146–153. [Google Scholar] [CrossRef]
  136. Yan, D.; He, B.; Guo, J.; Li, S.; Wang, J. Involvement of TLR4 in the protective effect of intra-articular administration of curcumin on rat experimental osteoarthritis. Acta Cir. Bras. 2019, 34, e201900604. [Google Scholar] [CrossRef] [Green Version]
  137. Jiang, C.; Luo, P.; Li, X.; Liu, P.; Li, Y.; Xu, J. Nrf2/ARE is a key pathway for curcumin-mediated protection of TMJ chondrocytes from oxidative stress and inflammation. Cell Stress Chaperones 2020, 25, 395–406. [Google Scholar] [CrossRef] [PubMed]
  138. Park, H.-J.; Lee, C.-K.; Song, J.-H.; Yun, J.-H.; Lee, A.; Park, H.-J. Highly bioavailable curcumin powder suppresses articular cartilage damage in rats with mono-iodoacetate (MIA)-induced osteoarthritis. Food Sci. Biotechnol. 2020, 29, 251–263. [Google Scholar] [CrossRef] [PubMed]
  139. Chen, Z.; Xue, J.; Shen, T.; Ba, G.; Yu, D.; Fu, Q. Curcumin alleviates glucocorticoid-induced osteoporosis by protecting osteoblasts from apoptosis in vivo and in vitro. Clin. Exp. Pharmacol. Physiol. 2016, 43, 268–276. [Google Scholar] [CrossRef]
  140. Chen, Z.; Xue, J.; Shen, T.; Mu, S.; Fu, Q. Curcumin alleviates glucocorticoid-induced osteoporosis through the regulation of the Wnt signaling pathway. Int. J. Mol. Med. 2016, 37, 329–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Dai, P.; Mao, Y.; Sun, X.; Li, X.; Muhammad, I.; Gu, W.; Zhang, D.; Zhou, Y.; Ni, Z.; Ma, J.; et al. Attenuation of Oxidative Stress-Induced Osteoblast Apoptosis by Curcumin is Associated with Preservation of Mitochondrial Functions and Increased Akt-GSK3β Signaling. Cell. Physiol. Biochem. 2017, 41, 661–677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Gupte, P.A.; Giramkar, S.A.; Harke, S.M.; Kulkarni, S.K.; Desmukh, A.P.; Hingorani, L.L.; Mahajan, M.P.; Bhalerao, S.S. Evaluation of the efficacy and safety of Capsule Longvida® Optimized Curcumin (solid lipid curcumin particles) in knee osteoarthritis: A pilot clinical study. J. Inflamm. Res. 2019, 12, 145–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Kang, C.; Jung, E.; Hyeon, H.; Seon, S.; Lee, D. Acid-activatable polymeric curcumin nanoparticles as therapeutic agents for osteoarthritis. Nanomed. Nanotechnol. Biol. Med. 2020, 23, 102104. [Google Scholar] [CrossRef] [PubMed]
  144. Liu, X.; Hunter, D.J.; Eyles, J.; McLachlan, A.J.; Adiwidjaja, J.; Eagles, S.K.; Wang, X.S. Pharmacokinetic assessment of constituents of Boswellia serrata, pine bark extracts, curcumin in combination including methylsulfonylmethane in healthy volunteers. J. Pharm. Pharmacol. 2020, 72, 121–131. [Google Scholar] [CrossRef]
  145. Heidari-Beni, M.; Moravejolahkami, A.R.; Gorgian, P.; Askari, G.; Tarrahi, M.J.; Bahreini-Esfahani, N. Herbal formulation ‘turmeric extract, black pepper, and ginger’ versus Naproxen for chronic knee osteoarthritis: A randomized, double-blind, controlled clinical trial. Phytother. Res. 2020, 34, 2067–2073. [Google Scholar] [CrossRef]
  146. Liu, X.; Robbins, S.; Eyles, J.; Fedorova, T.; Virk, S.; Deveza, L.A.; McLachlan, A.; Hunter, D. Efficacy and safety of a supplement combination for hand osteoarthritis pain: Protocol for an internet-based randomised placebo-controlled trial (The RADIANT study). BMJ Open 2020, 10, e035672. [Google Scholar] [CrossRef] [Green Version]
  147. Mao, Q.-Q.; Xu, X.-Y.; Cao, S.-Y.; Gan, R.-Y.; Corke, H.; Beta, T.; Li, H.-B. Bioactive Compounds and Bioactivities of Ginger (Zingiber officinale Roscoe). Foods 2019, 8, 185. [Google Scholar] [CrossRef] [Green Version]
  148. Araya-Quintanilla, F.; Gutierrez-Espinoza, H.; Munoz-Yanez, M.J.; Sanchez-Montoya, U.; Lopez-Jeldes, J. Effectiveness of Ginger on Pain and Function in Knee Osteoarthritis: A PRISMA Systematic Review and Meta-Analysis. Pain Physician 2020, 23, E151–E161. [Google Scholar]
  149. Hosseinzadeh, A.; Juybari, K.B.; Fatemi, M.J.; Kamarul, T.; Bagheri, A.; Tekiyehmaroof, N.; Sharifi, A.M. Protective Effect of Ginger (Zingiber officinale Roscoe) Extract against Oxidative Stress and Mitochondrial Apoptosis Induced by Interleukin-1β in Cultured Chondrocytes. Cells Tissues Organs 2017, 204, 241–250. [Google Scholar] [CrossRef]
  150. Abusarah, J.; Benabdoune, H.; Shi, Q.; Lussier, B.; Martel-Pelletier, J.; Malo, M.; Fernandes, J.C.; Pereira de Souza, F.; Fahmi, H.; Benderdour, M. Elucidating the Role of Protandim and 6-Gingerol in Protection Against Osteoarthritis. J. Cell. Biochem. 2017, 118, 1003–1013. [Google Scholar] [CrossRef]
  151. Fan, J.Z.; Yang, X.; Bi, Z.G. The effects of 6-gingerol on proliferation, differentiation, and maturation of osteoblast-like MG-63 cells. Braz. J. Med. Biol. Res. 2015, 48, 637–643. [Google Scholar] [CrossRef] [Green Version]
  152. Hwang, Y.-H.; Kim, T.; Kim, R.; Ha, H. The Natural Product 6-Gingerol Inhibits Inflammation-Associated Osteoclast Differentiation via Reduction of Prostaglandin E2 Levels. Int. J. Mol. Sci. 2018, 19, 2068. [Google Scholar] [CrossRef] [Green Version]
  153. Naderi, Z.; Mozaffari-Khosravi, H.; Dehghan, A.; Nadjarzadeh, A.; Huseini, H.F. Effect of ginger powder supplementation on nitric oxide and C-reactive protein in elderly knee osteoarthritis patients: A 12-week double-blind randomized placebo-controlled clinical trial. J. Tradit. Complement. Med. 2016, 6, 199–203. [Google Scholar] [CrossRef] [Green Version]
  154. Mozaffari-Khosravi, H.; Naderi, Z.; Dehghan, A.; Nadjarzadeh, A.; Fallah Huseini, H. Effect of Ginger Supplementation on Proinflammatory Cytokines in Older Patients with Osteoarthritis: Outcomes of a Randomized Controlled Clinical Trial. J. Nutr. Gerontol. Geriatr. 2016, 35, 209–218. [Google Scholar] [CrossRef]
  155. Amorndoljai, P.; Taneepanichskul, S.; Niempoog, S.; Nimmannit, U. A Comparative of Ginger Extract in Nanostructure Lipid Carrier (NLC) and 1% Diclofenac Gel for Treatment of Knee Osteoarthritis (OA). J. Med. Assoc. Thai. 2017, 100, 447–456. [Google Scholar] [PubMed]
  156. Bolognesi, G.; Belcaro, G.; Feragalli, B.; Cornelli, U.; Cotellese, R.; Hu, S.; Dugall, M. Movardol® (N-acetylglucosamine, Boswellia serrata, ginger) supplementation in the management of knee osteoarthritis: Preliminary results from a 6-month registry study. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 5198–5204. [Google Scholar] [PubMed]
  157. Rondanelli, M.; Riva, A.; Morazzoni, P.; Allegrini, P.; Faliva, M.A.; Naso, M.; Miccono, A.; Peroni, G.; Agosti, I.D.; Perna, S. The effect and safety of highly standardized Ginger (Zingiber officinale) and Echinacea (Echinacea angustifolia) extract supplementation on inflammation and chronic pain in NSAIDs poor responders. A pilot study in subjects with knee arthrosis. Nat. Prod. Res. 2017, 31, 1309–1313. [Google Scholar] [CrossRef] [PubMed]
  158. Gorzynik-Debicka, M.; Przychoden, P.; Cappello, F.; Kuban-Jankowska, A.; Gammazza, A.M.; Knap, N.; Wozniak, M.; Gorska-Ponikowska, M. Potential Health Benefits of Olive Oil and Plant Polyphenols. Int. J. Mol. Sci. 2018, 19, 686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Chin, K.-Y.; Pang, K.-L. Therapeutic Effects of Olive and Its Derivatives on Osteoarthritis: From Bench to Bedside. Nutrients 2017, 9, 1060. [Google Scholar] [CrossRef] [PubMed]
  160. Horcajada, M.-N.; Sanchez, C.; Membrez-Calfo, F.; Drion, P.; Comblain, F.; Taralla, S.; Donneau, A.-F.; Offord, E.A.; Henrotin, Y. Oleuropein or rutin consumption decreases the spontaneous development of osteoarthritis in the Hartley guinea pig. Osteoarthr. Cartil. 2015, 23, 94–102. [Google Scholar] [CrossRef] [Green Version]
  161. Feng, Z.; Li, X.; Lin, J.; Zheng, W.; Hu, Z.; Xuan, J.; Ni, W.; Pan, X. Oleuropein inhibits the IL-1β-induced expression of inflammatory mediators by suppressing the activation of NF-κB and MAPKs in human osteoarthritis chondrocytes. Food Funct. 2017, 8, 3737–3744. [Google Scholar] [CrossRef]
  162. Hagiwara, K.; Goto, T.; Araki, M.; Miyazaki, H.; Hagiwara, H. Olive polyphenol hydroxytyrosol prevents bone loss. Eur. J. Pharmacol. 2011, 662, 78–84. [Google Scholar] [CrossRef] [PubMed]
  163. García-Martínez, O.; De Luna-Bertos, E.; Ramos-Torrecillas, J.; Ruiz, C.; Milia, E.; Lorenzo, M.L.; Jimenez, B.; Sanchez-Ortiz, A.; Rivas, A. Phenolic Compounds in Extra Virgin Olive Oil Stimulate Human Osteoblastic Cell Proliferation. PLoS ONE 2016, 11, e0150045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Rosillo, M.A.; Montserrat-de-la-Paz, S.; Abia, R.; Castejon, M.L.; Millan-Linares, M.C.; Alarcon-de-la-Lastra, C.; Fernadez-Bolanos, J.G.; Muriana, F.J.G. Oleuropein and its peracetylated derivative negatively regulate osteoclastogenesis by controlling the expression of genes involved in osteoclast differentiation and function. Food Funct. 2020, 11, 4038–4048. [Google Scholar] [CrossRef] [PubMed]
  165. Puel, C.; Mathey, J.; Agalias, A.; Kati-Coulibaly, S.; Mardon, J.; Obled, C.; Davicco, M.-J.; Lebecque, P.; Horcajada, M.-N.; Skaltsounis, A.L.; et al. Dose-response study of effect of oleuropein, an olive oil polyphenol, in an ovariectomy/inflammation experimental model of bone loss in the rat. Clin. Nutr. 2006, 25, 859–868. [Google Scholar] [CrossRef]
  166. Filip, R.; Possemiers, S.; Heyerick, A.; Pinheiro, I.; Raszewski, G.; Davicco, M.-J.; Coxam, V. Twelve-month consumption of a polyphenol extract from olive (Olea europaea) in a double blind, randomized trial increases serum total osteocalcin levels and improves serum lipid profiles in postmenopausal women with osteopenia. J. Nutr. Health Aging 2015, 19, 77–86. [Google Scholar] [CrossRef]
  167. Nguyen, C.; Savouret, J.-F.; Widerak, M.; Corvol, M.-T.; Rannou, F. Resveratrol, Potential Therapeutic Interest in Joint Disorders: A Critical Narrative Review. Nutrients 2017, 9, 45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Meng, X.; Zhou, J.; Zhao, C.-N.; Gan, R.-Y.; Li, H.-B. Health Benefits and Molecular Mechanisms of Resveratrol: A Narrative Review. Foods 2020, 9, 340. [Google Scholar] [CrossRef] [Green Version]
  169. Im, H.-J.; Li, X.; Chen, D.; Yan, D.; Kim, J.; Ellman, M.B.; Stein, G.S.; Cole, B.; Ranjan, K.C.; Cs-Szabo, G.; et al. Biological effects of the plant-derived polyphenol resveratrol in human articular cartilage and chondrosarcoma cells. J. Cell. Physiol. 2012, 227, 3488–3497. [Google Scholar] [CrossRef] [Green Version]
  170. Wang, J.; Gao, J.-S.; Chen, J.-W.; Li, F.; Tian, J. Effect of resveratrol on cartilage protection and apoptosis inhibition in experimental osteoarthritis of rabbit. Rheumatol. Int. 2012, 32, 1541–1548. [Google Scholar] [CrossRef]
  171. Lei, M.; Wang, J.-G.; Xiao, D.-M.; Fan, M.; Wang, D.-P.; Xiong, J.-Y.; Chen, Y.; Dong, Y.; Liu, S.-L. Resveratrol inhibits interleukin 1β-mediated inducible nitric oxide synthase expression in articular chondrocytes by activating SIRT1 and thereby suppressing nuclear factor-κB activity. Eur. J. Pharmacol. 2012, 674, 73–79. [Google Scholar] [CrossRef]
  172. Kang, D.-G.; Lee, H.J.; Lee, C.J.; Park, J.S. Inhibition of the Expression of Matrix Metalloproteinases in Articular Chondrocytes by Resveratrol through Affecting Nuclear Factor-Kappa B Signaling Pathway. Biomol. Ther. 2018, 26, 560–567. [Google Scholar] [CrossRef] [PubMed]
  173. Li, W.; Cai, L.; Zhang, Y.; Cui, L.; Shen, G. Intra-articular resveratrol injection prevents osteoarthritis progression in a mouse model by activating SIRT1 and thereby silencing HIF-2α: Intrapoap-Articular Resveratrol Injection Prevents Osteoarthritis Progression. J. Orthop. Res. 2015, 33, 1061–1070. [Google Scholar] [CrossRef]
  174. Liu, L.; Gu, H.; Liu, H.; Jiao, Y.; Li, K.; Zhao, Y.; An, L.; Yang, J. Protective Effect of Resveratrol against IL-1β-Induced Inflammatory Response on Human Osteoarthritic Chondrocytes Partly via the TLR4/MyD88/NF-κB Signaling Pathway: An “in Vitro Study”. Int. J. Mol. Sci. 2014, 15, 6925–6940. [Google Scholar] [CrossRef] [Green Version]
  175. Jiang, M.; Li, X.; Yu, X.; Liu, X.; Xu, X.; He, J.; Gu, H.; Liu, L. Oral Administration of Resveratrol Alleviates Osteoarthritis Pathology in C57BL/6J Mice Model Induced by a High-Fat Diet. Mediat. Inflamm. 2017, 1–11. [Google Scholar] [CrossRef] [PubMed]
  176. Moon, M.-H.; Jeong, J.-K.; Lee, Y.-J.; Seol, J.-W.; Jackson, C.J.; Park, S.-Y. SIRT1, a class III histone deacetylase, regulates TNF-α-induced inflammation in human chondrocytes. Osteoarthr. Cartil. 2013, 21, 470–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Liu, S.; Yang, H.; Hu, B.; Zhang, M. Sirt1 regulates apoptosis and extracellular matrix degradation in resveratrol-treated osteoarthritis chondrocytes via the Wnt/β-catenin signaling pathways. Exp. Ther. Med. 2017, 14, 5057–5062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Qin, N.; Wei, L.; Li, W.; Yang, W.; Cai, L.; Qian, Z.; Wu, S. Local intra-articular injection of resveratrol delays cartilage degeneration in C57BL/6 mice by inducing autophagy via AMPK/mTOR pathway. J. Pharmacol. Sci. 2017, 134, 166–174. [Google Scholar] [CrossRef] [PubMed]
  179. Wei, Y.; Jia, J.; Jin, X.; Tong, W.; Tian, H. Resveratrol ameliorates inflammatory damage and protects against osteoarthritis in a rat model of osteoarthritis. Mol. Med. Rep. 2017, 17, 1493–1498. [Google Scholar] [CrossRef]
  180. Zhang, G.; Zhang, H.; You, W.; Tang, X.; Li, X.; Gong, Z. Therapeutic effect of Resveratrol in the treatment of osteoarthritis via the MALAT1/miR-9/NF-κB signaling pathway. Exp. Ther. Med. 2020, 19, 2343–2352. [Google Scholar] [CrossRef] [PubMed]
  181. Shakibaei, M.; Shayan, P.; Busch, F.; Aldinger, C.; Buhrmann, C.; Lueders, C.; Mobasheri, A. Resveratrol Mediated Modulation of Sirt-1/Runx2 Promotes Osteogenic Differentiation of Mesenchymal Stem Cells: Potential Role of Runx2 Deacetylation. PLoS ONE 2012, 7, e35712. [Google Scholar] [CrossRef] [Green Version]
  182. Yang, X.; Jiang, T.; Wang, Y.; Guo, L. The Role and Mechanism of SIRT1 in Resveratrol-regulated Osteoblast Autophagy in Osteoporosis Rats. Sci. Rep. 2019, 9, 18424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Jiang, Y.; Luo, W.; Wang, B.; Wang, X.; Gong, P.; Xiong, Y. Resveratrol promotes osteogenesis via activating SIRT1/FoxO1 pathway in osteoporosis mice. Life Sci. 2020, 246, 117422. [Google Scholar] [CrossRef]
  184. Wang, W.; Zhang, L.-M.; Guo, C.; Han, J.-F. Resveratrol promotes osteoblastic differentiation in a rat model of postmenopausal osteoporosis by regulating autophagy. Nutr. Metab. 2020, 17, 29. [Google Scholar] [CrossRef] [Green Version]
  185. Huang, Y.; Hui, J.; Liu, F.Q.; Liu, J.; Zhang, X.J.; Guo, C.H.; Song, L.H. Resveratrol Promotes in vitro Differentiation of Osteoblastic MC3T3-E1 Cells via Potentiation of the Calcineurin/NFATc1 Signaling Pathway. Biochemistry 2019, 84, 686–692. [Google Scholar] [CrossRef] [PubMed]
  186. Shakibaei, M.; Buhrmann, C.; Mobasheri, A. Resveratrol-mediated SIRT-1 Interactions with p300 Modulate Receptor Activator of NF-κB Ligand (RANKL) Activation of NF-κB Signaling and Inhibit Osteoclastogenesis in Bone-derived Cells. J. Biol. Chem. 2011, 286, 11492–11505. [Google Scholar] [CrossRef] [Green Version]
  187. Kim, H.-N.; Han, L.; Iyer, S.; De Cabo, R.; Zhao, H.; O’Brien, C.A.; Manolagas, S.C.; Almeida, M. Sirtuin1 Suppresses Osteoclastogenesis by Deacetylating FoxOs. Mol. Endocrinol. 2015, 29, 1498–1509. [Google Scholar] [CrossRef] [PubMed]
  188. Yan, S.; Miao, L.; Lu, Y.; Wang, L. Sirtuin 1 inhibits TNF-α-mediated osteoclastogenesis of bone marrow-derived macrophages through both ROS generation and TRPV1 activation. Mol. Cell. Biochem. 2019, 455, 135–145. [Google Scholar] [CrossRef]
  189. Wong, R.H.X.; Evans, H.M.; Howe, P.R.C. Resveratrol supplementation reduces pain experience by postmenopausal women. Menopause 2017, 24, 916–922. [Google Scholar] [CrossRef]
  190. Hussain, S.; Marouf, B.; Ali, Z.; Ahmmad, R. Efficacy and safety of co-administration of resveratrol with meloxicam in patients with knee osteoarthritis: A pilot interventional study. Clin. Interv. Aging 2018, 13, 1621–1630. [Google Scholar] [CrossRef] [Green Version]
  191. Cao, W.; Guo, X.-W.; Zheng, H.-Z.; Li, D.-P.; Jia, G.-B.; Wang, J. Current progress of research on pharmacologic actions of salvianolic acid B. Chin. J. Integr. Med. 2012, 18, 316–320. [Google Scholar] [CrossRef]
  192. Yang, X.; Liu, S.; Li, S.; Wang, P.; Zhu, W.; Liang, P.; Tan, J.; Cui, S. Salvianolic acid B regulates gene expression and promotes cell viability in chondrocytes. J. Cell. Mol. Med. 2017, 21, 1835–1847. [Google Scholar] [CrossRef] [Green Version]
  193. Lou, Y.; Wang, C.; Zheng, W.; Tang, Q.; Chen, Y.; Zhang, X.; Guo, X.; Wang, J. Salvianolic acid B inhibits IL-1β-induced inflammatory cytokine production in human osteoarthritis chondrocytes and has a protective effect in a mouse osteoarthritis model. Int. Immunopharmacol. 2017, 46, 31–37. [Google Scholar] [CrossRef]
  194. Cui, L.; Li, T.; Liu, Y.; Zhou, L.; Li, P.; Xu, B.; Huang, L.; Chen, Y.; Liu, Y.; Tian, X.; et al. Salvianolic Acid B Prevents Bone Loss in Prednisone-Treated Rats through Stimulation of Osteogenesis and Bone Marrow Angiogenesis. PLoS ONE 2012, 7, e34647. [Google Scholar] [CrossRef] [Green Version]
  195. Zhang, X.; Zou, L.; Li, J.; Xu, B.; Wu, T.; Fan, H.; Xu, W.; Yao, W.; Yang, Y.; Liu, Y.; et al. Salvianolic acid B and danshensu induce osteogenic differentiation of rat bone marrow stromal stem cells by upregulating the nitric oxide pathway. Exp. Ther. Med. 2017, 14, 2779–2788. [Google Scholar] [CrossRef] [Green Version]
  196. He, X.; Shen, Q. Salvianolic acid B promotes bone formation by increasing activity of alkaline phosphatase in a rat tibia fracture model: A pilot study. BMC Complement. Altern. Med. 2014, 14, 493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Wang, N.; Li, Y.; Li, Z.; Liu, C.; Xue, P. Sal B targets TAZ to facilitate osteogenesis and reduce adipogenesis through MEK-ERK pathway. J. Cell. Mol. Med. 2019, 23, 3683–3695. [Google Scholar] [CrossRef] [PubMed]
  198. Ma, D.; Yu, T.; Peng, L.; Wang, L.; Liao, Z.; Xu, W. PIM1, CYP1B1, and HSPA2 Targeted by Quercetin Play Important Roles in Osteoarthritis Treatment by Achyranthes bidentata. Evid. Based Complement. Alternat. Med. 2019, 1–10. [Google Scholar] [CrossRef] [Green Version]
  199. Weng, X.; Lin, P.; Liu, F.; Chen, J.; Li, H.; Huang, L.; Zhen, C.; Xu, H.; Liu, X.; Ye, H.; et al. Achyranthes bidentata polysaccharides activate the Wnt/β-catenin signaling pathway to promote chondrocyte proliferation. Int. J. Mol. Med. 2014, 34, 1045–1050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Ma, D.; Li, Y.; Xiao, W.; Peng, L.; Wang, L.; Liao, Z.; Hu, L. Achyranthes bidentata extract protects chondrocytes functions through suppressing glycolysis and apoptosis via MAPK/AKT signaling axis. Am. J. Transl. Res. 2020, 12, 142–152. [Google Scholar] [PubMed]
  201. Zhang, D.; Wang, C.; Hou, X.; Yan, C. Structural characterization and osteoprotective effects of a polysaccharide purified from Achyranthes bidentata. Int. J. Biol. Macromol. 2019, 139, 1063–1073. [Google Scholar] [CrossRef] [PubMed]
  202. He, G.; Guo, W.; Lou, Z.; Zhang, H. Achyranthes bidentata saponins promote osteogenic differentiation of bone marrow stromal cells through the ERK MAPK signaling pathway. Cell Biochem. Biophys. 2014, 70, 467–473. [Google Scholar] [CrossRef]
  203. Song, D.; Cao, Z.; Huang, S.; Tickner, J.; Li, N.; Qiu, H.; Chen, X.; Wang, C.; Chen, K.; Sun, Y.; et al. Achyranthes bidentata polysaccharide suppresses osteoclastogenesis and bone resorption via inhibiting RANKL signaling. J. Cell. Biochem. 2018, 119, 4826–4835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Yan, C.; Zhang, S.; Wang, C.; Zhang, Q. A fructooligosaccharide from Achyranthes bidentata inhibits osteoporosis by stimulating bone formation. Carbohydr. Polym. 2019, 210, 110–118. [Google Scholar] [CrossRef] [PubMed]
  205. Mussard, E.; Cesaro, A.; Lespessailles, E.; Legrain, B.; Berteina-Raboin, S.; Toumi, H. Andrographolide, a Natural Antioxidant: An Update. Antioxidants 2019, 8, 571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Mussard, E.; Jousselin, S.; Cesaro, A.; Legrain, B.; Lespessailles, E.; Esteve, E.; Berteina-Raboin, S.; Toumi, H. Andrographis paniculata and its Bioactive Diterpenoids Against Inflammation and Oxidative Stress in Keratinocytes. Antioxidants 2020, 9, 530. [Google Scholar] [CrossRef] [PubMed]
  207. Mussard, E.; Jousselin, S.; Cesaro, A.; Legrain, B.; Lespessailles, E.; Esteve, E.; Berteina-Raboin, S.; Toumi, H. Andrographis paniculata and its Bioactive Diterpenoids Protect Dermal Fibroblasts against Inflammation and Oxidative Stress. Antioxidants 2020, 9, 432. [Google Scholar] [CrossRef] [PubMed]
  208. Villedieu-Percheron, E.; Ferreira, V.; Campos, J.F.; Destandau, E.; Pichon, C.; Berteina-Raboin, S. Quantitative determination of Andrographolide and related compounds in Andrographis paniculata extracts and biological evaluation of their Anti-Inflammatory Activity. Foods 2019, 8, 683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Hancke, J.L.; Srivastav, S.; Cáceres, D.D.; Burgos, R.A. A double-blind, randomized, placebo-controlled study to assess the efficacy of Andrographis paniculata standardized extract (ParActin®) on pain reduction in subjects with knee osteoarthritis. Phytother. Res. 2019, 33, 1469–1479. [Google Scholar] [CrossRef]
  210. Ding, Q.; Ji, X.; Cheng, Y.; Yu, Y.; Qi, Y.; Wang, X. Inhibition of matrix metalloproteinases and inducible nitric oxide synthase by andrographolide in human osteoarthritic chondrocytes. Mod. Rheumatol. 2013, 23, 1124–1132. [Google Scholar] [CrossRef]
  211. Li, B.; Jiang, T.; Liu, H.; Miao, Z.; Fang, D.; Zheng, L.; Zhao, J. Andrographolide protects chondrocytes from oxidative stress injury by activation of the Keap1-Nrf2-Are signaling pathway. J. Cell. Physiol. 2018, 234, 561–571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Chen, S.; Luo, Z.; Chen, X. Andrographolide mitigates cartilage damage via miR-27-3p-modulated matrix metalloproteinase13 repression. J. Gene Med. 2020, 22, e3187. [Google Scholar] [CrossRef] [PubMed]
  213. Jiang, T.; Zhou, B.; Huang, L.; Wu, H.; Huang, J.; Liang, T.; Liu, H.; Zheng, L.; Zhao, J. Andrographolide Exerts Pro-Osteogenic Effect by Activation of Wnt/β-Catenin Signaling Pathway in Vitro. Cell. Physiol. Biochem. 2015, 36, 2327–2339. [Google Scholar] [CrossRef]
  214. Li, B.; Hu, R.-Y.; Sun, L.; Luo, R.; Lu, K.-H.; Tian, X.-B. Potential role of andrographolide in the proliferation of osteoblasts mediated by the ERK signaling pathway. Biomed. Pharmacother. 2016, 83, 1335–1344. [Google Scholar] [CrossRef] [PubMed]
  215. Shah, M.M.R.; Liang, Y.; Cheng, J.J.; Daroch, M. Astaxanthin-Producing Green Microalga Haematococcus pluvialis: From Single Cell to High Value Commercial Products. Front. Plant Sci. 2016, 7, 531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Fakhri, S.; Aneva, I.Y.; Farzaei, M.H.; Sobarzo-Sánchez, E. The Neuroprotective Effects of Astaxanthin: Therapeutic Targets and Clinical Perspective. Molecules 2019, 24, 2640. [Google Scholar] [CrossRef] [Green Version]
  217. Mashhadi, N.S.; Zakerkish, M.; Mohammadiasl, J.; Zarei, M.; Mohammadshahi, M.; Haghighizadeh, M.H. Astaxanthin improves glucose metabolism and reduces blood pressure in patients with type 2 diabetes mellitus. Asia Pac. J. Clin. Nutr. 2018, 27, 341–346. [Google Scholar] [CrossRef]
  218. Han, J.H.; Ju, J.H.; Lee, Y.S.; Park, J.H.; Yeo, I.J.; Park, M.H.; Roh, Y.S.; Han, S.B.; Hong, J.T. Astaxanthin alleviated ethanol-induced liver injury by inhibition of oxidative stress and inflammatory responses via blocking of STAT3 activity. Sci. Rep. 2018, 8, 14090. [Google Scholar] [CrossRef]
  219. Kumar, A.; Dhaliwal, N.; Dhaliwal, J.; Dharavath, R.N.; Chopra, K. Astaxanthin attenuates oxidative stress and inflammatory responses in complete Freund-adjuvant-induced arthritis in rats. Pharmacol. Rep. 2020, 72, 104–114. [Google Scholar] [CrossRef]
  220. Park, M.H.; Jung, J.C.; Hill, S.; Cartwright, E.; Dohnalek, M.H.; Yu, M.; Jun, H.J.; Han, S.B.; Hong, J.T.; Son, D.J. FlexPro MD®, a Combination of Krill Oil, Astaxanthin and Hyaluronic Acid, Reduces Pain Behavior and Inhibits Inflammatory Response in Monosodium Iodoacetate-Induced Osteoarthritis in Rats. Nutrients 2020, 12, 956. [Google Scholar] [CrossRef] [Green Version]
  221. Sun, K.; Luo, J.; Jing, X.; Guo, J.; Yao, X.; Hao, X.; Ye, Y.; Liang, S.; Lin, J.; Wang, G.; et al. Astaxanthin protects against osteoarthritis via Nrf2: A guardian of cartilage homeostasis. Aging 2019, 11, 10513–10531. [Google Scholar] [CrossRef]
  222. Huang, L.; Chen, W.-P. Astaxanthin ameliorates cartilage damage in experimental osteoarthritis. Mod. Rheumatol. 2015, 25, 768–771. [Google Scholar] [CrossRef] [PubMed]
  223. Hwang, Y.-H.; Kim, K.-J.; Kim, S.-J.; Min, S.-K.; Hong, S.-G.; Son, Y.-J.; Yee, S.-T. Suppression Effect of Astaxanthin on Osteoclast Formation In Vitro and Bone Loss In Vivo. Int. J. Mol. Sci. 2018, 19, 912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Balci Yuce, H.; Lektemur Alpan, A.; Gevrek, F.; Toker, H. Investigation of the effect of astaxanthin on alveolar bone loss in experimental periodontitis. J. Periodontal Res. 2018, 53, 131–138. [Google Scholar] [CrossRef] [PubMed]
  225. Zeng, X.; Guo, F.; Ouyang, D. A review of the pharmacology and toxicology of aucubin. Fitoterapia 2020, 140, 104443. [Google Scholar] [CrossRef]
  226. Wang, S.-N.; Xie, G.-P.; Qin, C.-H.; Chen, Y.-R.; Zhang, K.-R.; Li, X.; Wu, Q.; Dong, W.-Q.; Yang, J.; Yu, B. Aucubin prevents interleukin-1 beta induced inflammation and cartilage matrix degradation via inhibition of NF-κB signaling pathway in rat articular chondrocytes. Int. Immunopharmacol. 2015, 24, 408–415. [Google Scholar] [CrossRef]
  227. Young, I.-C.; Chuang, S.-T.; Hsu, C.-H.; Sun, Y.-J.; Liu, H.-C.; Chen, Y.-S.; Lin, F.-H. Protective effects of aucubin on osteoarthritic chondrocyte model induced by hydrogen peroxide and mechanical stimulus. BMC Complement. Altern. Med. 2017, 17, 91. [Google Scholar] [CrossRef] [Green Version]
  228. Huang, T.-L.; Yang, S.-H.; Chen, Y.-R.; Liao, J.-Y.; Tang, Y.; Yang, K.-C. The therapeutic effect of aucubin-supplemented hyaluronic acid on interleukin-1beta-stimulated human articular chondrocytes. Phytomedicine 2019, 53, 1–8. [Google Scholar] [CrossRef]
  229. Wang, B.-W.; Jiang, Y.; Yao, Z.-I.; Chen, P.; Yu, B.; Wang, S. Aucubin Protects Chondrocytes Against IL-1β-Induced Apoptosis In Vitro And Inhibits Osteoarthritis In Mice Model. Drug Des. Devel. Ther. 2019, 13, 3529–3538. [Google Scholar] [CrossRef] [Green Version]
  230. Abdel-Tawab, M.; Werz, O.; Schubert-Zsilavecz, M. Boswellia serrata: An overall assessment of in vitro, preclinical, pharmacokinetic and clinical data. Clin. Pharmacokinet. 2011, 50, 349–369. [Google Scholar] [CrossRef]
  231. Majeed, M.; Majeed, S.; Narayanan, N.K.; Nagabhushanam, K. A pilot, randomized, double-blind, placebo-controlled trial to assess the safety and efficacy of a novel Boswellia serrata extract in the management of osteoarthritis of the knee. Phytother. Res. 2019, 33, 1457–1468. [Google Scholar] [CrossRef] [Green Version]
  232. Sengupta, K.; Kolla, J.N.; Krishnaraju, A.V.; Yalamanchili, N.; Rao, C.V.; Golakoti, T.; Raychaudhuri, S.; Raychaudhuri, S.P. Cellular and molecular mechanisms of anti-inflammatory effect of Aflapin: A novel Boswellia serrata extract. Mol. Cell. Biochem. 2011, 354, 189–197. [Google Scholar] [CrossRef] [PubMed]
  233. Blain, E.J.; Ali, A.Y.; Duance, V.C. Boswellia frereana (frankincense) suppresses cytokine-induced matrix metalloproteinase expression and production of pro-inflammatory molecules in articular cartilage. Phytother. Res. 2010, 24, 905–912. [Google Scholar] [CrossRef] [PubMed]
  234. Wang, Q.; Pan, X.; Wong, H.H.; Wagner, C.A.; Lahey, L.J.; Robinson, W.H.; Sokolove, J. Oral and topical boswellic acid attenuates mouse osteoarthritis. Osteoarthr. Cartil. 2014, 22, 128–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Takada, Y.; Ichikawa, H.; Badmaev, V.; Aggarwal, B.B. Acetyl-11-Keto-β-Boswellic Acid Potentiates Apoptosis, Inhibits Invasion, and Abolishes Osteoclastogenesis by Suppressing NF-κB and NF-κB-Regulated Gene Expression. J. Immunol. 2006, 176, 3127–3140. [Google Scholar] [CrossRef] [Green Version]
  236. Bai, F.; Chen, X.; Yang, H.; Xu, H.-G. Acetyl-11-Keto-β-Boswellic Acid Promotes Osteoblast Differentiation by Inhibiting Tumor Necrosis Factor-α and Nuclear Factor-κB Activity. J. Craniofac. Surg. 2018, 29, 1996–2002. [Google Scholar] [CrossRef] [PubMed]
  237. Lv, H.; Jiang, L.; Zhu, M.; Li, Y.; Luo, M.; Jiang, P.; Tong, S.; Zhang, H.; Yan, J. The genus Tripterygium: A phytochemistry and pharmacological review. Fitoterapia 2019, 137, 104190. [Google Scholar] [CrossRef]
  238. Ding, Q.-H.; Cheng, Y.; Chen, W.-P.; Zhong, H.-M.; Wang, X.-H. Celastrol, an inhibitor of heat shock protein 90β potently suppresses the expression of matrix metalloproteinases, inducible nitric oxide synthase and cyclooxygenase-2 in primary human osteoarthritic chondrocytes. Eur. J. Pharmacol. 2013, 708, 1–7. [Google Scholar] [CrossRef]
  239. Wang, W.; Ha, C.; Lin, T.; Wang, D.; Wang, Y.; Gong, M. Celastrol attenuates pain and cartilage damage via SDF-1/CXCR4 signalling pathway in osteoarthritis rats. J. Pharm. Pharmacol. 2018, 70, 81–88. [Google Scholar] [CrossRef]
  240. Feng, K.; Chen, H.; Xu, C. Chondro-protective effects of celastrol on osteoarthritis through autophagy activation and NF-κB signaling pathway inhibition. Inflamm. Res. 2020, 69, 385–400. [Google Scholar] [CrossRef]
  241. Cascão, R.; Vidal, B.; Finnila, M.A.J.; Lopes, I.P.; Teixeira, R.L.; Saarakkala, S.; Moita, L.F.; Fonseca, J.E. Effect of celastrol on bone structure and mechanics in arthritic rats. RMD Open 2017, 3, e000438. [Google Scholar] [CrossRef] [Green Version]
  242. Gan, K.; Xu, L.; Feng, X.; Zhang, Q.; Wang, F.; Zhang, M.; Tan, W. Celastrol attenuates bone erosion in collagen-Induced arthritis mice and inhibits osteoclast differentiation and function in RANKL-induced RAW264.7. Int. Immunopharmacol. 2015, 24, 239–246. [Google Scholar] [CrossRef] [PubMed]
  243. Mancuso, C.; Santangelo, R. Panax ginseng and Panax quinquefolius: From pharmacology to toxicology. Food Chem. Toxicol. 2017, 107, 362–372. [Google Scholar] [CrossRef] [PubMed]
  244. Lee, J.H.; Lim, H.; Shehzad, O.; Kim, Y.S.; Kim, H.P. Ginsenosides from Korean red ginseng inhibit matrix metalloproteinase-13 expression in articular chondrocytes and prevent cartilage degradation. Eur. J. Pharmacol. 2014, 724, 145–151. [Google Scholar] [CrossRef]
  245. Kim, S.-H.; Na, J.-Y.; Song, K.-B.; Choi, D.-S.; Kim, J.-H.; Kwon, Y.-B.; Kwon, J. Protective Effect of Ginsenoside Rb1 on Hydrogen Peroxide-induced Oxidative Stress in Rat Articular Chondrocytes. J. Ginseng Res. 2012, 36, 161–168. [Google Scholar] [CrossRef] [Green Version]
  246. Na, J.-Y.; Kim, S.; Song, K.; Lim, K.-H.; Shin, G.-W.; Kim, J.-H.; Kim, B.; Kwon, Y.-B.; Kwon, J. Anti-apoptotic Activity of Ginsenoside Rb1 in Hydrogen Peroxide-treated Chondrocytes: Stabilization of Mitochondria and the Inhibition of Caspase-3. J. Ginseng Res. 2012, 36, 242–247. [Google Scholar] [CrossRef] [Green Version]
  247. Huang, Y.; Wu, D.; Fan, W. Protection of ginsenoside Rg1 on chondrocyte from IL-1β-induced mitochondria-activated apoptosis through PI3K/Akt signaling. Mol. Cell. Biochem. 2014, 392, 249–257. [Google Scholar] [CrossRef]
  248. Cheng, W.; Wu, D.; Zuo, Q.; Wang, Z.; Fan, W. Ginsenoside Rb1 prevents interleukin-1 beta induced inflammation and apoptosis in human articular chondrocytes. Int. Orthop. 2013, 37, 2065–2070. [Google Scholar] [CrossRef] [Green Version]
  249. Wang, W.; Zeng, L.; Wang, Z.; Zhang, S.; Rong, X.-F.; Li, R.-H. Ginsenoside Rb1 inhibits matrix metalloproteinase 13 through down-regulating Notch signaling pathway in osteoarthritis. Exp. Biol. Med. 2015, 240, 1614–1621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  250. Chen, Y.; Lin, S.; Sun, Y.; Pan, X.; Xiao, L.; Zou, L.; Ho, K.W.; Li, G. Translational potential of ginsenoside Rb1 in managing progression of osteoarthritis. J. Orthop. Transl. 2016, 6, 27–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  251. Cheng, W.; Jing, J.; Wang, Z.; Wu, D.; Huang, Y. Chondroprotective Effects of Ginsenoside Rg1 in Human Osteoarthritis Chondrocytes and a Rat Model of Anterior Cruciate Ligament Transection. Nutrients 2017, 9, 263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Zhang, P. Ginsenoside-Rg5 treatment inhibits apoptosis of chondrocytes and degradation of cartilage matrix in a rat model of osteoarthritis. Oncol. Rep. 2017, 37, 1497–1502. [Google Scholar] [CrossRef]
  253. Xie, J.-J.; Chen, J.; Guo, S.-K.; Gu, Y.-T.; Yan, Y.-Z.; Guo, W.-J.; Yao, C.-L.; Jin, M.-Y.; Xie, C.-L.; Wang, X.; et al. Panax quinquefolium saponin inhibits endoplasmic reticulum stress-induced apoptosis and the associated inflammatory response in chondrocytes and attenuates the progression of osteoarthritis in rat. Biomed. Pharmacother. 2018, 97, 886–894. [Google Scholar] [CrossRef]
  254. Zhang, Y.; Cai, W.; Han, G.; Zhou, S.; Li, J.; Chen, M.; Li, H. Panax notoginseng saponins prevent senescence and inhibit apoptosis by regulating the PI3K-AKT-mTOR pathway in osteoarthritic chondrocytes. Int. J. Mol. Med. 2020, 45, 1225–1236. [Google Scholar] [CrossRef]
  255. Siddiqi, M.H.; Siddiqi, M.Z.; Ahn, S.; Kang, S.; Kim, Y.-J.; Veerappan, K.; Yang, D.-U.; Yang, D.-C. Stimulative Effect of Ginsenosides Rg5:Rk1 on Murine Osteoblastic MC3T3-E1 Cells. Phytother. Res. 2014, 10, 1447–1455. [Google Scholar] [CrossRef]
  256. Siddiqi, M.H.; Siddiqi, M.Z.; Ahn, S.; Kim, Y.-J.; Yang, D.C. Ginsenoside Rh1 induces mouse osteoblast growth and differentiation through the bone morphogenetic protein 2/runt-related gene 2 signalling pathway: Rh1, osteoblast growth and differentiation. J. Pharm. Pharmacol. 2014, 66, 1763–1773. [Google Scholar] [CrossRef] [PubMed]
  257. Siddiqi, M.Z.; Siddiqi, M.H.; Kim, Y.-J.; Jin, Y.; Huq, M.A.; Yang, D.C. Effect of Fermented Red Ginseng Extract Enriched in Ginsenoside Rg3 on the Differentiation and Mineralization of Preosteoblastic MC3T3-E1 Cells. J. Med. Food 2015, 18, 542–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  258. Bei, J.; Zhang, X.; Wu, J.; Hu, Z.; Xu, B.; Lin, S.; Cui, L.; Wu, T.; Zou, L. Ginsenoside Rb1 does not halt osteoporotic bone loss in ovariectomized rats. PLoS ONE 2018, 13, e0202885. [Google Scholar] [CrossRef] [PubMed]
  259. Cheng, B.; Li, J.; Du, J.; Lv, X.; Weng, L.; Ling, C. Ginsenoside Rb1 inhibits osteoclastogenesis by modulating NF-κB and MAPKs pathways. Food Chem. Toxicol. 2012, 50, 1610–1615. [Google Scholar] [CrossRef] [PubMed]
  260. Zhang, X.; Chen, K.; Wei, B.; Liu, X.; Lei, Z.; Bai, X. Ginsenosides Rg3 attenuates glucocorticoid-induced osteoporosis through regulating BMP-2/BMPR1A/Runx2 signaling pathway. Chem. Biol. Interact. 2016, 256, 188–197. [Google Scholar] [CrossRef] [PubMed]
  261. Chrubasik, S.; Conradt, C.; Roufogalis, B.D. Effectiveness of Harpagophytum extracts and clinical efficacy. Phytother. Res. 2004, 18, 187–189. [Google Scholar] [CrossRef]
  262. Harpagophytum procumbens (devil’s claw). Monograph. Altern. Med. Rev. 2008, 13, 248–252.
  263. Haseeb, A.; Ansari, M.Y.; Haqqi, T.M. Harpagoside suppresses IL-6 expression in primary human osteoarthritis chondrocytes. J. Orthop. Res. 2017, 35, 311–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Chung, H.-J.; Kim, W.K.; Park, H.J.; Cho, L.; Kim, M.-R.; Kim, M.J.; Shin, J.-S.; Lee, J.H.; Ha, I.-H.; Lee, S.K. Anti-osteoporotic activity of harpagide by regulation of bone formation in osteoblast cell culture and ovariectomy-induced bone loss mouse models. J. Ethnopharmacol. 2016, 179, 66–75. [Google Scholar] [CrossRef] [PubMed]
  265. Wegener, T.; Lüpke, N.-P. Treatment of patients with arthrosis of hip or knee with an aqueous extract of devil’s claw (Harpagophytum procumbens DC). Phytother. Res. 2003, 17, 1165–1172. [Google Scholar] [CrossRef] [PubMed]
Table 1. Natural-alkaloid-based pharmacology therapy for osteoarthritis (OA).
Table 1. Natural-alkaloid-based pharmacology therapy for osteoarthritis (OA).
Compound
(Source)
CategoryStructureTherapeutic TargetTreatmentRef.
Berberine
(Berberis vulgaris)
Benzyl
isoquinolin alkaloid
Antioxidants 10 00265 i001Activation of AMPK signaling and inhibition of p38 MAPK/NF-κB pathways in chondrocytes.
Activation of p38 MAPK signaling in osteoblasts.
Anti-inflammatory, anti-apoptotic, and anti-degradation in cartilage;
Induction of bone formation.
[22,23,24,25,26,27,28]
Table 2. Natural-flavonoid-based pharmacology therapy for OA.
Table 2. Natural-flavonoid-based pharmacology therapy for OA.
Compound
(Source)
CategoryStructureTherapeutic TargetTreatmentRef.
Apigenin
(chamomile, thyme, tea, extra virgin oil)
4′,5,7-Trihydroxyflavone Antioxidants 10 00265 i002Inhibition of IL-1β-induced effects and NF-κB, Hif-2α, and TGFβ/Smad2/3 pathways in chondrocytes.Anti-inflammatory effect, prevent cartilage degradation.[32,33]
Increases BMP-2, BMP-7, APL, and Col I in osteoblasts. Induces JNK and p38 MAPK pathways in osteoblasts.Promotes osteoblastic differentiation.[34,35]
Astragalin
(Cuscuta chinensis)
Kaempferol 3-glucoside Antioxidants 10 00265 i003Inhibition of IL-1β/NF-κB and MAPK in chondrocytes.Anti-inflammatory effect, suppresses bone destruction.[38,39]
Baicalein
(Scutellaria baicalensis)
5,6,7-Trihydroxyflavone Antioxidants 10 00265 i004Inhibition of IL-1β-induced effects in chondrocytes. Increases secretion of GAG and Col II.Anti-catabolic and anti-apoptotic effects.[42,43,44]
Increases osteoblast differentiation and inhibits osteoclast differentiation.Attenuated OA in pre-clinical models.
Inhibition of bone loss.
[45,46]
Chrysin
(Passiflora caerulea, Scutellaria baicalensis)
5,7-Dihydroxyflavone Antioxidants 10 00265 i005Inhibition of IL-1β/NF-κB induction. Dowregulates the expression of iNOS, COX-2, MMP-1, MMP-3, MMP-13, ADAMTS-4, ADAMTS-5, and HMGB-1 in chondrocytes. The level of NO, PGE2 decreases.Anti-inflammatory and anti-apoptotic effects.[48,49]
Activation of ERK/MAPK signaling in osteoblasts and upregulation of Runx-2 and Osx.Induction of osteoblast differentiation.[50,51]
Genistein
(Genista tinctoria)
Isoflavone Antioxidants 10 00265 i006Inhibition of IL-1β-induced effects via the activation of Nrf2/HO-1 signaling in chondrocytes.Anti-catabolic effect. Attenuated OA in pre-clinical models.[58,59]
Increases osteoblast differentiation via MAPK activation and inhibits osteoclast differentiation via NF-κB inhibition.Inhibition of bone loss.[60,61,62,63]
Icariin
(Epimedium)
Flavonoid glycoside Antioxidants 10 00265 i007Inhibition of IL-1β/TNF-α/LPS-induced effects via the inhibition of NF-κB and the activation of Nrf2/HO-1 signaling in chondrocytes. Increases the secretion of ACAN and Col II. Decreases the expression of MMP-1, 3, 9, 13, COX-2, and iNOS.Anti-inflammatory and anti-catabolic effects. Increased cartilage repair in pre-clinical OA models.[64,65,66,67,68]
Increases osteoblast differentiation via the activation of ERK, JUNK, and miR-153/Runx2 signaling. Increases the secretion of Col I APL.Inhibition of bone loss. Improved bone remodeling in pre-clinical models.[69,70,71,72]
Kaempferol
(Kaempferia galanga)
3,4′,5,7-Tetrahydroxyflavone Antioxidants 10 00265 i008Attenuation of IL-1β-induced effects by inhibiting p38 MAPK/NF-κB pathways in chondrocytes.Anti-inflammatory effect.[74,75]
Increases osteoblast differentiation via the activation of Wnt/β-catenin and mTOR signaling, increasing BMP-2, Rux-2, Osx, and Col I expression. Inhibits osteoclastogenesis by downregulating MAPK, c-Fos, and NFATc1.Inhibition of bone loss and stimulation of bone formation.[76,77,78,79,80,81]
Luteolin
(Salvia tomentosa, Artemisia asiatica)
3,4′,5,7-Tetrahydroxyflavone Antioxidants 10 00265 i009Attenuation of IL-1β-induced effects by inhibiting NF-κB pathways and the activation of Foxo3a in chondrocytes. Decreases the expression of COX-2, iNOS, MMPs, and ADAMTS-4,5. Attenuates cartilage degradation and increases Col II secretion.Anti-inflammatory and anti-catabolic effects. Attenuation of cartilage degradation.[83,84,85,86,87]
Increases osteoblast differentiation via the regulation of ERK/Lrp-5/GSK-3β signaling, increasing BMP-7, Rux-2, Osx, Osc, APL, TGF-β1, and Col I expression. Inhibition of osteoclast differentiation.Inhibition of bone loss and stimulation of bone formation.[34,88,89,90,91,92,93,94]
Naringin
(Citrus × paradisi)
Flavanone-7-O-glycoside Antioxidants 10 00265 i010Alleviation of IL-1β/TNFα/LPS-induced effects via inhibiting MAPK p38 and NF-κB signaling and the activation of Foxo3a in chondrocytes. Decreases the expression of MMPs and ADAMTS-4,5. Attenuates cartilage degradation.Anti-inflammatory and anti-catabolic effects. Attenuation of cartilage degradation.[95,96,97,98]
Increases osteoblast proliferation and differentiation. Increases the expression of Rux-2, Osx, Osc, BMP-2, OPN, and Col I expression. Inhibits osteoclast differentiation.Inhibition of bone loss and promotes bone formation.[99,100,101,102,103]
Puerarin
(Pueraria lobate)
Isoflavone Antioxidants 10 00265 i011Blocks the anti-catabolic effects in chondrocytes via the action of the AMPK/PGC-1α signaling pathway. Attenuates cartilage degradation.Anti-inflammatory and anti-catabolic effects. Attenuation of cartilage degradation.[105,106]
Promotes bone formation via the activation of p38 MAPK, ERK1/2-Runx2, and Wnt/β-catenin signaling and by inhibiting TRPM3/miR-204 expression. Inhibits osteoclastogenesis by downregulating CREB/PGC1β/c-Fos/NFATc1 signaling.Inhibition of bone loss and promotes bone formation.[107,108,109,110,111,112,113,114]
Silibinin/Silymarin
(Silybum marianum)
Flavone Antioxidants 10 00265 i012Inhibition of IL-1β-induced effects by inhibiting PI3K/Akt and NF-κB signaling. Decreases the expression of iNOS, MMPs, and ADAMTS-4,5. Diminishes the secretion of NO, PEG2, TNF-α, and IL-6. Attenuates cartilage degradation and synovitis in vivo.Anti-inflammatory, anti-oxidant, and anti-catabolic effects. Attenuation of cartilage degradation and synovitis.[118,119,120]
Induces osteoblast differentiation, increasing the expression of Runx-2, BMP-2, ALP, and Col I. Inhibits osteoclastogenesis by disturbing TRAF6-c-Src signaling.Anti-oxidant and anti-apoptotic effects in osteoblasts. Inhibition of bone loss.[121,122,123,124,125,126]
Wogonin
(Scutellaria baicalensis)
O-methylated flavone Antioxidants 10 00265 i013Inhibition of IL-1β-induced effects by inhibiting c-Fos/AP-1 and JAK/STAT signaling and activating ROS/ERK/Nrf2 signaling. Decreases the expression of iNOS, MMPs, and ADAMTS-4,5. Diminishes the secretion of NO, PEG2, TNF-α, and IL-6. Attenuates cartilage degradation and synovitis in vivo.Anti-inflammatory, anti-oxidant, and anti-catabolic effects. Attenuation of cartilage degradation and synovitis.[129,130,131,132,133,134]
Table 3. Natural-phenol-based pharmacology therapy for OA.
Table 3. Natural-phenol-based pharmacology therapy for OA.
CompoundCategoryStructureTherapeutic TargetTreatmentRef.
Curcumine
(Curcuma longa)
Diferuloyl-methane Antioxidants 10 00265 i014Inhibits the expression of IL-6, iNOS, COX-2, MMPs, and ADAMTS4,5 and increases the expression of SOX-9 and Col II by inhibiting 5-LOX/NF-κB signaling and activating Nrf2/ARE signaling.Anti-inflammatory, antioxidant, anti-apoptotic, and anti-catabolic effects.[12,135,136,137,138]
Induces osteoblast differentiation, increasing the expression of Runx-2, Osx, Osc, and Col I by regulating Wnt signaling.Attenuation of cartilage degradation and synovitis.
Bone protection.
[139,140,141,142,143,144,145,146]
Gingerly/ginger
(Zingier officinal)
6-Gingerol Antioxidants 10 00265 i015Inhibits IL-1β-induced effects via the activation of Nrf2 signaling in chondrocytes.Anti-apoptotic, antioxidant, and anti-inflammatory effects.[149,150]
Induces osteoblasts differentiation and inhibits osteoclast differentiation. Inhibition of bone loss.[151,152]
Oleuropein
(Olea europea)
Secoiridoid glycoside Antioxidants 10 00265 i016Inhibits of IL-1β-induced effects by suppressing NF-κB and MAPK signaling. Decreases the expression of COX-2, iNOS, MMP-1, MMP-13, and ADAMTS-5. Anti-inflammatory effects. Decreases synovitis, cartilage degradation, and osteophyte formation.[160,161]
Increases calcium deposits and inhibits osteoclast formation and differentiation. Bone protection.[162,163,164,165,166]
Resveratrol
(red grapes, blueberries, raspberries, mulberries)
3,5,4′-Trihydroxy-trans-stilbene Antioxidants 10 00265 i017Inhibits IL-1β-induced effects by suppressing NF-κB signaling and increasing SIRT-1 expression via the AMPK/mTOR pathway. It decreases the expression of iNOS, MMP1, MMP-3, MMP-13, and ADAMTS-4,5. Anti-inflammatory and anti-apoptotic effects. Prevents cartilage degradation and maintains the homeostasis of cartilage and bone.[171,172,173,174,175,176,177,178,179,180]
Induces osteoblast differentiation by modulating Sirt-1/Runx-2/Fox-1 and PI3K/AKT/mTOR signaling. Inhibits osteoclastogenesis via the activation of SIRT-1 and FoxOs.Bone protection.[181,182,183,184,185,186,187,188]
Salvianolic acid B
(Radix salvia miltiorrhiza)
Polyphenol Antioxidants 10 00265 i018Inhibits IL-1β-induced effects by suppressing NF-κB signaling. Decreases the expression of iNOS, COX-2, MMP-13, and ADAMTS-5.Anti-inflammatory and anti-catabolic effects. Reduces cartilage degradation.[192,193]
Induces osteoblast differentiation through the activation of ERK signaling, upregulating the expression of Runx-2, OPN, and Osx.Bone protection and induces bone formation[194,195,196,197]
Table 4. Natural-polysaccharide-based pharmacology therapy for OA.
Table 4. Natural-polysaccharide-based pharmacology therapy for OA.
Compound
(Source)
CategoryTherapeutic TargetTreatmentRef.
Achyranthes bidentata extractsVarious polysaccharidesInduces chondrocyte proliferation, Wnt/β-catenin pathway activation, and inhibits apoptosis via MAPK/Akt signaling.Anti-apoptotic effect and induces proliferation.[199,200]
Promotes bone formation and inhibits osteoclastogenesis via the inhibition of RANK.Bone formation.[201,202,203]
Table 5. Natural-terpenoid-based pharmacology therapy for OA.
Table 5. Natural-terpenoid-based pharmacology therapy for OA.
Compound
(Source)
CategoryStructureTherapeutic TargetTreatmentRef.
Andrographolide
(Andrographis paniculate)
Diterpenoid Antioxidants 10 00265 i019Reduces oxidative stress and inhibits MMP-13 expression. Attenuates cartilage degradation via miR-27-3p/MMP13 signaling.Anti-oxidant effects. Reduces the degradation of cartilage.[210,211,212,213,214,215,216,217,218,219,220,221,222]
Promotes bone formation by inhibiting NF-κB signaling.Bone formation[213,214]
Astaxanthin
(Haematococcus pluvialis)
Tetraterpenoid Antioxidants 10 00265 i020Anti-catabolic effects via the activation of Nrf2–ARE signaling. Reduces cartilage degradation via MAPK signaling inhibition.Antioxidant and anti-inflammatory effects. Attenuates degradation of cartilage[221,222]
Aucubin
(Aucuba japonica)
Iridoid glycoside Antioxidants 10 00265 i021Inhibits IL-1β-induced effects. Inhibits iNOS expression and NO productionAnti-inflammatory and antioxidant effects. Prevents OA progression.[226,227,228,229]
Boswellia serrata11-Keto-β-boswellic, acetyl-11-keto-β-boswellic acid Antioxidants 10 00265 i022Inhibits IL-1β/oncostatin-M-induced effect, decreasing the expression of MMP-9, MMP-13, and COX-2 and reducing the production of NO and PGE2. Inhibits 5-LOX and TNF-α.Anti-inflammatory and antioxidant effects.[15,232,233,234]
Antioxidants 10 00265 i023Promotes osteoblast differentiation and suppresses osteoclastogenesis by inhibiting TNF-α and NF-κB signaling.Bone protection.[235,236]
Celastrol
(Celastrus regelii, Tripterygium wilfordii)
Triterpenoid Antioxidants 10 00265 i024Diminishes the IL-1β-induced catabolic effect, decreasing the expression of MMP-1, MMP-3, MMP-13, iNOS, and COX-2. Reduces cartilage degradation by inhibiting NF-κB signaling and activating SDF-1/CXCR4 signaling. Anti-inflammatory and anti-catabolic effects.[238,239,240]
Suppresses osteoclastogenesis by inhibiting MAPK and NF-κB signaling.Bone protection.[241,242]
Ginsenoside
(Panax)
Terpenoid glycoside Antioxidants 10 00265 i025Inhibits the IL-1β-induced effect, decreasing the expression of MMP-1, MMP-13, iNOS, and COX-2; the level of PGE2; promoting the expression of ACAN and Col II.Anti-inflammatory, anti-apoptotic, antioxidant, and anti-degradative effects.[244,245,246,247,248,249,250,251,252,253,254]
Promotes osteoblast differentiation and suppresses osteoclastogenesis by inhibiting MAPK and NF-κB signaling.Bone protection[255,256,257,258,259,260]
Harpagophytum procumbensIridoid glycosides Antioxidants 10 00265 i026Inhibits IL-1β-induced anti-inflammatory effects, decreasing the expression of IL-6 and MMP-13 via the suppression of c-Fos/AP-1 activity.Anti-inflammatory effect.[263]
Stimulates osteoblast dif-ferentiation and inhibits osteoclast differentiation.Bone protection.[264]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pérez-Lozano, M.-L.; Cesaro, A.; Mazor, M.; Esteve, E.; Berteina-Raboin, S.; Best, T.M.; Lespessailles, E.; Toumi, H. Emerging Natural-Product-Based Treatments for the Management of Osteoarthritis. Antioxidants 2021, 10, 265. https://doi.org/10.3390/antiox10020265

AMA Style

Pérez-Lozano M-L, Cesaro A, Mazor M, Esteve E, Berteina-Raboin S, Best TM, Lespessailles E, Toumi H. Emerging Natural-Product-Based Treatments for the Management of Osteoarthritis. Antioxidants. 2021; 10(2):265. https://doi.org/10.3390/antiox10020265

Chicago/Turabian Style

Pérez-Lozano, Maria-Luisa, Annabelle Cesaro, Marija Mazor, Eric Esteve, Sabine Berteina-Raboin, Thomas M. Best, Eric Lespessailles, and Hechmi Toumi. 2021. "Emerging Natural-Product-Based Treatments for the Management of Osteoarthritis" Antioxidants 10, no. 2: 265. https://doi.org/10.3390/antiox10020265

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