Nuclear Factor Kappa B Inhibition as a Therapeutic Target of Nutraceuticals in Arthritis, Osteoarthritis, and Related Inflammation

Nuclear Factor Kappa B Inhibition as a Therapeutic Target of Nutraceuticals in Arthritis, Osteoarthritis, and Related Inflammation

CHAPTER 25 Nuclear Factor Kappa B Inhibition as a Therapeutic Target of Nutraceuticals in Arthritis, Osteoarthritis, and Related Inflammation Hazem M...

511KB Sizes 0 Downloads 32 Views

CHAPTER 25

Nuclear Factor Kappa B Inhibition as a Therapeutic Target of Nutraceuticals in Arthritis, Osteoarthritis, and Related Inflammation Hazem M. Shaheen*, Abdelwahab A. Alsenosy† *

Department of Pharmacology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt



1. INTRODUCTION Rheumatoid arthritis (RA) is a disease of chronic autoimmunity with an extensive inflammation of the synovial membrane, resulting in erosions of articular cartilage and marginal bone and causing inflammatory joint destruction. Arthritis is chiefly a disease of the joints with a proinflammatory disorder, thus a full understanding of the inflammatory character of arthritis is essential for the success of therapeutic trials.1 Bone tissue primarily contains osteocytes, osteoblasts and matrix osteoid proteins with deposition of salts mainly inorganic mineral. Osteoprotegerin (OPG) and receptor activator of nuclear factor kappa B (NF-κB) ligand (RANKL) played a key role in osteoclast differentiation via its receptor RANK located on the osteoclast membrane. So, diseases such as osteoporosis and arthritis could be regulated by osteoclast differentiation.2 The present molecular biological technologies have extended useful approaches for RA research. Nowadays, gene expression is a prime tool used to investigate pathogenesis in RA, and it provides useful information for understanding RA.3 RA susceptibility loci were discovered through candidate gene, linkage, and genome-wide association studies. The gene signatures in the synovial tissues of rheumatoid arthritis and osteoarthritis have been found using microarray approaches.4 Lately, it is accepted that NF-κB provides a central inflammatory mediator that responds to large varieties of immune receptor signals. Because deregulated NF-κB activation is involved in various inflammatory diseases, targeting the NF-κB signaling pathway is a prominent application for antiinflammatory therapies. Several kinds of inhibitors have been developed to block the different steps of NF-κB signaling.5

Bioactive Food as Dietary Interventions for Arthritis and Related Inflammatory Diseases https://doi.org/10.1016/B978-0-12-813820-5.00025-8

© 2019 Elsevier Inc. All rights reserved.

437

438

Bioactive Food as Dietary Interventions for Arthritis and Related Inflammatory Diseases

2. RHEUMATOID ARTHRITIS Arthritis is an endothelial dysfunction that could be termed a systemic disorder, indicating an inflammation in rheumatoid arthritis with chronic stages.6 The role of chemokines and enzymes of inflammation such as cyclooxygenase-2, 5-lipoxygenase, matrix metalloproteinase, interleukin-1β, IL-6, cytokines, TNF-α, and adhesion molecules in arthritis has been documented. Most of the inflammatory mediators involved in arthritis have been approved to be regulated by NF-κB.7 The multifarious relations between genetic vulnerability and deregulated genes generate rheumatoid arthritis. The clinical signs in arthritic patients are the formation of pannus with atypical proliferation of synovial tissue. Early diagnosis could heighten the prognosis and the quality of life of RA patients. However, the pathogenetic mechanism of RA is indefinite without reasonable biomarkers for early diagnosis in clinical practice.8 The course of action of RA involves a variety of cells containing innate immune cells such as monocytes/macrophages, T and B cells, and synovial fibroblasts. NF-κB mediates the induction of proinflammatory cytokines such as TNF-α, IL-1, and IL-6 in monocytes/macrophages. The canonical and noncanonical NF-κB pathways also mediate RANK ligand-induced differentiation of monocytes/ macrophages into the bone-resorbing osteoclasts, whose deregulation adds to an inflammatory bone loss followed by RA.9 In between the lymphocyte subsets of T cells, Th17 cells are predominantly important for the advancement of RA. NF-κB promotes Th17 segregation indirectly through induction of inflammatory cytokines, IL-1, IL-6, and IL-23, of cellular immune and directly regulates transcription factors of Th17 lineage in T cells.10 Deregulated activation of NF-κB also contributes to atypical survival of self-reactive B cells and autoantibody stimulation that donates to RA pathogenicity.11 Particularly, RA patients often display higher levels of B-cell activating factor in serum belonging to the TNF family associated with unsynchronized stimulation of the NF-κB noncanonical type.12 Therefore, NF-κB mediates the pathogenesis of RA by functioning in various cell types, as shown in Figure 25.1.

3. NF-κB MEDIATOR IN RHEUMATOID ARTHRITIS NF-κB is an initiator in RA pathogenesis and is essential for the production of inflammatory mediators in the synovium. However, while much is recognized about the pathways that result in NF-κB stimulation in transformed cells and in lab animals, these events often differ in the RA linked cells, such as human myeloid cells and cells of the synovium. These events are only being fully explored now, using new approaches such as adenoviral infection. Doubtlessly, cutting-edge technologies such as small inhibiting RNA will also give great insight into the functional roles of these proteins in prospect studies. This will be important to help identify new therapeutic targets of RA and validate those therapies already under development. The exquisitely specific NF-κB response induced by

Nuclear Factor Kappa B Inhibition as a Therapeutic Target

Figure 25.1 Osteoarthritis pathophysiology with attachment of the synovium. Products of cartilage breakdown released into the synovial fluid are phagocytized by synovial cells, intensifying synovial inflammation. In turn, the productions of proinflammatory mediators initiate a surplus production of the proteolytic enzymes, leading to cartilage breakdown. Additionally, the inflamed synovium facilitate the osteophyte assembly via BMPs.

different stimuli in different cells gives hope that treatments can be developed to specifically target NF-κB in the inflamed synovium with no harmful effects on the innate immunity.13 In the synovial cells with RA, activation of the NF-κB pathway could transactivate a responsive gene of the inflammatory phenotype, containing TNF-α from macrophages, matrix metalloproteinases from synovial fibroblasts, and chemokines that hire immune cells to the inflamed pannus. Despite these differences at the molecular level, the significance of NF-κB in inflammation is approved and inhibition of the pathway is widely believed to have great potential as a treatment target in RA. Commercial effort has been exerted into developing inhibitors of NF-κB activation. However, inactivation of the NF-κB leads to an exacerbation of inflammation if TNF-α production by macrophages is not controlled.14 The data gained from arthritic patients suggest that NF-κ B activation is thoroughly involved in the chronic inflammation of the RA synovium. Accordingly, inhibitors of NF-κB are a potential approach as therapeutics for RA. The successful trials for treatments that directly target the products of NF-κB focused genes, such as TNF-α, IL-6, and IL-1, are a major advance in the treatment of RA patients that do not respond to the pattern treatment.15 The useful intensifying evidence is that NF-κB is a major transcription factor controlling inflammation. The activation of the NF-κB pathway is concerned with the progress of chronic inflammatory disorders, including rheumatoid arthritis and inflammatory disease of the bowel.16 An inhibitor of the NF-κB kinase complex formed by the NF-κ kinase inhibitor and IKKb and subunit IKKg that is a regulatory regulates NF-κB proteins. This inhibitor kinase phosphorylates IkB is then ubiquitinated and degraded, leading to the NF-κB activation.17 Moreover, a family of nucleotide-binding domain and leucine-rich repeat containing including NOD1, NOD2, NLR pyrin domain containing (NLRP) 3, and NLRP2 regulate the

439

440

Bioactive Food as Dietary Interventions for Arthritis and Related Inflammatory Diseases

Figure 25.2 Genes targeted by NF-κB in inflammation series and development. NF-κB is an inducible transcription factor. Once activated, it can trigger transcription of a series of genes regulating inflammation. In addition, NF-κB targets inflammation by increasing the induction of inflammatory cytokines, chemokines, and adhesion molecules. It also regulates the cell survival and proliferation, apoptosis, and angiogenesis.

activation process of NF-κB.18 A common signaling event is initiation of the canonical NF-κB pathway, which regulates transcriptional induction of proinflammatory cytokines, chemokines, and additional inflammatory mediators in different types of innate immune cells (Figure 25.2).

4. NFKB IN MODELS OF RA Many studies in rheumatoid arthritis propose that NF-κB is essential for the expression of inflammatory mediators such as cytokines and tissue-degrading enzymes. Consistently, NF-κB activation is approved in synovial tissue from arthritic patients, and this emerges as a relation to clinical manifestation. Uncontrolled regulation of proteins that direct the NF-κB pathway contributes to the susceptibility or severity of inflammation during diseases.19 The joints of rheumatic patients’ synovia are infiltrated by immune cells, leading

Nuclear Factor Kappa B Inhibition as a Therapeutic Target

to chronic inflammation, pannus formation, and consequent joint and cartilage damage. The RA synovium is known to possess basically macrophages (30%–40%), T cells (30%), and synovial fibroblast alongside B cells, dendritic cells, other immune cells, and the endothelium.20, 21 RA synovial fluid has been analyzed, and it contains a broad variety of effector molecules including proinflammatory cytokines (such as IL-1β, IL-6, TNFα, and IL-18), chemokines (such as IL-8, IP-10, MCP-1, MIP-1, and RANTES), matrix metalloproteinases (MMPs, such as MMP-1, -3, -9, and -13), and metabolic proteins (such as COX-1, COX-2, and iNOS).22 These correlate in a complex manner that is assumed to cause a malicious cycle of proinflammatory signals, resulting in frequent and chronic inflammation. TNFα is obviously the key inflammatory mediator and also induces apoptosis. Importantly, the genes encoding TNFα and many other listed factors are well known to be controlled by NF-κB transcription factors, suggesting that NF-κB could be a central key of inflammatory cytokine production in RA. Indeed, the activated NF-κB transcription factors have been verified in cultured fibroblasts of the synovium.16 The joints of arthritic humans and animals with experimentally induced RA. Immunohistochemistry has demonstrated the presence of both p50 and p65 in the nuclei cells lining the synovial tissue and macrophages.23 Furthermore, cellular extracts have verified a facility to attach the NF-κB consensus sequence. A practice such as in vivo imaging has been used to demonstrate the bustle of NF-κB in a mouse model of inflammation. By placing the luciferase gene controlled by NF-κB, a marked increase in luminescence was observed in the mice joints. These findings are investigated in mice that carried an NF-κB family member’s c-Rel knockouts genes.24

5. HERBALS WITH POTENTIAL ANTIARTHRITIC ACTIVITIES VIA NF-κB 5.1 Capsicum Genus Natives have traditionally used Capsicum annum fruit for many centuries. It contains chemicals that cause highly selective anesthesia by the breakdown of the capsaicin-sensitive nociceptive nerve endings. It is known to be potent in the stimulation of the receptor potential for vanilloid-1. This is believed to be the main receptor for nociception. It is also suggested to have ability in the inhibition of NF-κB activation for generating the antiinflammatory effect.25 The herb is often mixed with other antiarthritic herbals. It is also used for peripheral neurone disorders and chronic musculoskeletal pain.26

5.2 Curcuma Genus Several members of the Curcuma genus are used in traditional medicine, with the most important being Curcuma longa (CL), or turmeric. Its rhizome has a centuries-long use as a dietary spice as well as a herb for its antiinflammatory properties, hence its utility in arthritic conditions, including RA.27 In an animal arthritis model, a preparation from Curcuma lacking essential oil strongly suppressed joint inflammation and periarticular

441

442

Bioactive Food as Dietary Interventions for Arthritis and Related Inflammatory Diseases

damage with decreased activation of NF-κB and of the resultant cascade of events (involving mediators of inflammation such as chemokines, cyclooxygenase 2, and the receptor activator of NF-κB ligand).28 The ability to prevent the negative changes in joints and bone appears to be comparable to that of betamethasone.29 Liposomal encapsulation may help overcome the poor bioavailability problem generated by the low water solubility. The osteoclast-osteoblast balance is tilted for bone building while halting the osteoarthritis progression.30 In a ratinduced arthritis model, applying ginger and turmeric rhizomes was better than indomethacin (a potent NSAID) regarding the facility to improve both joint histopathological changes and the extraarticular manifestations, including systemic inflammation, malnutrition, and iron deficiency anemia, with no intolerance to kidney function and lessen the cardiovascular risk factors.31 In human clinical studies, a combination of Curcuma longa has been shown to be more efficient than a standard dose of celecoxib in the treatment of osteoarthritis, improving the condition of the patients with no toxicity detectable by laboratory tests.32 Curcuma domestica extracts are useful in knee osteoarthritis, reducing the pain with an efficiency equivalent to ibuprofen, but with low side effects.33 A metaanalysis found related scientific confirmation for the efficacy of turmeric as a therapeutic option in arthritis, but more studies are necessary to definitively fasten it.34 The active ingredient phenolic commonly known as curcumin, which has a complex beneficial action in various fields of pathology due to its ability to favorably influence a variety of signaling pathways and mediators.35 In a rat model of arthritis, Curcuma improved joint inflammation in the few hours after the arthritis-inducing event.36

5.3 Garlic It is well established that IL-1β, once bound to its type 1 receptor, activates NF-κB dimers by triggering phosphorylation and subsequent reduction of the inhibitory IκB proteins. Activation of NF-κB was a necessity for IL-1β-induced MMP-13 secretion in OA chondrocytes.37 Also, Imamura et al. proved that IL-1β and TNF-α inhibited chondrogenesis via the pathway of NF-κB in human mesenchyme stem cells.38 Various garlic products have been studied in osteoarthritis where garlic has demonstrated that it suppressed arthritis through inhibition of NF-κB DNA-binding activity and expression of iNOS and COX-2.39 However, there are few studies on the garlic effect on osteoarthritis. In addition, a previous study revealed that DATS suppressed MMP2–9 expression, which was dependent on NF-κB and ERK-MAPK signaling pathways.40 Mechanistically, garlic was found to be related to the increased levels of IκBα induced by IL-1β, which subsequently mitigated p65 nuclear translocation and the transcriptional activity of NF-κB. Furthermore, a result indicated that IL-1β treatment increased the expression of TNF-α at both the transcription and translation. The combination of these findings suggests that garlic can potentially be applied in OA treatment.

Nuclear Factor Kappa B Inhibition as a Therapeutic Target

5.4 Ginger Zingiber officinalis (ZO), also known as ginger, is a common spice used in Asian cuisine and a traditional remedy for joint diseases in ethnomedicine.27 Nevertheless, the ginger extract inconsistently increased the synthesis of proinflammatory cytokines (TNF-α, IL-6, and monocyte chemotactic protein-1) in cell culture. The exogenous administration of the ginger extract had a twofold effect on TNF-α synthesis in mice in peritoneal cells: ZO extract primarily improved it, but with repeated administrations reduced it.41 In addition, it augmented the serum corticosterone level and this may contribute to the antiinflammatory effect of ZO. A recent human clinical study found that ZO powder supplementation can reduce the serum level of nitric oxide and high-sensitivity reactive protein hs-CRP in OA patients. The inflammatory markers started to decrease after 3 weeks of treatment.42 Several studies showed a clinical improvement in OA patients with ZO extract as evaluated by the pain score with VA and a reduction in intake of free medication, having mostly mild gastrointestinally adverse events. Sharp-tasting constituents of ZO were thought to contribute to the antiinflammatory activity of this medicinal plant. For instance, ginger inhibited kB kinase activity required for NF-κB activation and suppressed NF-κB-regulated expression of inflammatory genes in the lipopolysaccharide S-activated macrophage.43 6-Dehydrogingerdione attenuated iNOS, COX-2, IL-1β, IL-6, and TNF-α gene expression in vitro in RAW 264.7 macrophages.44

5.5 Licorice About 300 polyphenols have been isolated from licorice, including phenolic acids, flavonoids, flavans, chalcones, isoflavan, and isoflavonoids. Thus far, the main antiinflammatory active polyphenols in licorice are chalcones, isoflavan, and isoflavonoids. The mechanisms for the antiinflammatory activities of chalcones have been fully investigated. LCA, LCB, ISL, and EC all inhibited the production of NO, IL-6, and PGE2 while LCA, LCB, and LCD all exhibited potent inhibition of lipid peroxidation.45, 46 Furthermore, LCC decreased the expression of iNOS and modulated the antioxidant system activity of SOD, catalase, and glutathione peroxidase.47 LCE efficiently inhibited PKC/JNK and ERK1/2; reduced the expression of iNOS, COX-2, IL-6, IL-1β, IL-12p40, TNF-α, AKT, and p38 mitogen-activated protein kinase (MAPK); and lessened IκBα degradation and NF-κB activities as well as the transcriptional activity of activator protein AP-1.48 Besides chalcones, other flavonoids in licorice, including DGC, DGD, ISOA, GLD, LID, and LIA, also revealed tremendous antiinflammatory activities. DGC, DGD, and ISOA all showed well-built ferric reducing activities and effectively scavenged DPPH, ABTS+, and singlet oxygen radicals.49 Furthermore, DGC increased the expression of haemeoxygenase-1 and MAPK phosphatase-1, suppressed the inflammationmediated neurodegeneration, production of TNF-α, NO, ROS, NF-κB and phosphorylation of p38 MAPKs, ERK1/2, IκB-α, and p65.50 GLD significantly inhibited NO

443

444

Bioactive Food as Dietary Interventions for Arthritis and Related Inflammatory Diseases

and IL-1β release51 and decreased the iNOS mRNA expression under high-glucose levels, which indicated that GLD could be applied to diabetes-induced vascular dysfunction.52 LID and LIA inhibited the secretion of IL-6, chemokine (C-C motif) ligand 5, and MMP-7, -8, and -9. The suppression of cytokine and MMP secretion by LID and LIA was associated with the inhibition of NF-κB p65 in periodontitis therapeutic trials.53

5.6 Pomegranate In vivo studies have defined a clear role for NF-κB in the modulation of inflammation by pomegranate extracts, findings that have been confirmed in vitro. Pomegranate juice, POMxTM extract,54 and their bioactive compounds punicalagin55 or delphinidin56 all suppressed NF-κB activation in different cells. It was found that pomegranate inhibited the expression of NF-κB target genes, including IL-6 and interleukin 8 (IL-8), upon exposure to proinflammatory stimuli in intestinal cells.57 Also, EA and POMxTM58 declined NF-κB activation in various subsets of immune cells, and anthocyanin delphinidin reduced inflammation in rheumatoid arthritis cells. Taken together, these results suggest that pomegranate and other bioactive compounds present in its juice show antiinflammatory effects in vitro, and that the mechanisms involved appear to be related to inactivation of NF-κB signaling. Administration of pomegranate-derived products has been shown to reduce inflammation in a respiratory inflamed model of mice59 and in the joints of RA model mice.60 There also exists data support suggesting that pomegranate extract exerts antiinflammatory effects that may alleviate the symptoms of IBD and inflammation were all recovered by pomegranate fruit supplementation in rodent models of IBD.61 The mechanisms involved appear to be related to the inhibition of NF-κB,62 c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase, and signal transducer and activator of transcription 3 phosphorylation.63

5.7 Resveratrol Resveratrol has a variety of concentrations in different plants and the higher concentrations are believed to be derived from knotweed or Polygonum cuspidatum and red wine grapes. In plants, it is located in the skin, which serves as a phytoalexin in the protection of the plant against infection and UV radiation as well as a broad plant defense system. It is known to acquire antimutation, antioxidant, antiinflammatory, and DNA protection entities when consumed by animals and humans. Numerous studies have been established in neuro and cardio protection. This is used in the management of arthritic joint pain. Intraarticular injections of resveratrol showed protective effects on the cartilage through the reduction of the inflammatory reactions caused by osteoarthritis in the knees. This has also been justified with the reduction of the edema in experimental animal models supposedly related to the inhibition of production of the prostaglandins.64 It is also a powerful inhibitor of the TNF-α and IL-1β-induced NF-κB activation. Similarly, the

Nuclear Factor Kappa B Inhibition as a Therapeutic Target

antiinflammatory activities may be suppressing the COX-2 pathways through blocking NF-κB activation in the joints. Resveratrol is extracted from many sources. However, when administered, it is converted to transresveratrol, which is the active form, with no significant side effects and no safety issues in numerous studies of resveratrol.65

5.8 Sesamum Oil Sesame oil (SO) extracted from Sesamum indicum has been used in various Asian traditional medicines to lighten pain in inflammatory conditions of the various tissues such as joints, teeth, and skin.66 In rat-induced arthritis, SO was tested and was capable of reducing the biochemical consequences of oxidative stress: lower plasma thiobarbituric acid reactive substance levels and reduced gamma-glutamyltransferase activity in the joints.67 In a rat model of destructive arthritis, SO strongly delayed the inflammatory reaction and lowered the levels of inflammatory mediators, hindering the NF-κB activity within the mast cells and the activation of complement systems.66 In another arthritic model, SO alleviated the inflammatory pain by inhibiting oxidative aggression via falling lipid peroxidation and production of superoxide anion and peroxynitrite in the muscles.68 SO is active in experimentally induced arthritis through its minor constituents which without it is inactive, decreasing the clinically visible joint inflammation, in addition to its serum markers including oxidative stress related molecules, RA markers, inflammatory eicosanoids and cytokines and the activity of hydrolytic enzymes; additionally, bone loss was also diminished.69 In a study on knee OA patients, orally administrated sesame produced better outcomes in terms of objective and subjective manifestations in comparison to standard therapy alone.70 In OA patients, a placebo trial on sesame seed management was associated with a noticed drop in serum levels of malondialdehyde and of high-sensitivity C-reactive protein (hs-CRP); it also significantly lowered levels of IL6 after treatment.71 The ability of SI to protect from the forward consequences of inflammation and oxidative stress is due to the presence of lignans. It contains sesamin and its hydroxylated counterpart, sesamolin. Similar biological activity has a phenolic compound, sesamol (3,4-methylene-dioxy-phenol), which results from the degradation of sesamolin.72 Sesamol has been approved to alleviate joint inflammation and cartilage degradation in an adjuvant-induced arthritis animal model. This action was paralleled by a drop in the level of proinflammatory cytokines and in the activity of tissue-destructive enzymes.73 In addition, restorations of the oxidant homeostasis reflected in decreased oxidative stress markers and a boost in the activity of protective enzymes were noticed, which leads to the inhibition of this inflammation-promoting enzyme.72

5.9 Whitania somnifera Withania somnifera (WS) also called ashwagandha, is a powerful antiosteoarthritic and antiinflammatory plant.74 WS extract was studied in vitro, and it was observed to inhibit

445

446

Bioactive Food as Dietary Interventions for Arthritis and Related Inflammatory Diseases

liposaccharide S-induced production of proinflammatory mediators (TNF-α, IL-1β, and IL-12) in mononuclear cells from rheumatoid arthritis in humans.75 The WS extract also showed inhibitory effects on collagenase activity in the Achilles tendon that may be useful in joint disease treatment.76 In an experimental rat-induced arthritis model, WS root powder had shielding effect on bone collagen.77 The aqueous extract of WS produced a significant reduction of scores for pain, stiffness, and disability in patients with knee joint inflammation in a randomized, double-blind, placebo study.78 Withaferin A suppresses NF-κB activation by targeting a crucial cysteine 179 in IKKβ, a kinase, and by inhibition of the NF-κB essential modulator complex formation, according to molecular studies.79, 80

6. NUTRACEUTICALS TARGET THE NF-κB PATHWAY IN ARTHRITIS 6.1 Camel Milk Camel milk revealed various antiinflammatory effects via downregulation of TNF-α, COX-2, iNOS, and its upstream effector NF-κB, together with enhancing the IL-10 antiinflammatory pathway. These antiinflammatory mechanisms are described additionally in further studies, which reported that camel milk possesses marked antiinflammatory actions in experimental models.81, 82 The observed downregulation of activated NF-κB p65 in the pouch lining confirms the effectual antiinflammatory performance of camel milk.83 Meanwhile, the observed downregulation of iNOS, TNF-α, and COX-2 in arthritic rats is prone to the downregulation of their upstream NF-κB transcription factor.84, 85

6.2 Glucosamine Glucosamine is a building block of polysaccharide chain glycosaminoglycans correlated to a protein in proteoglycan molecules called aggrecans, forming a component of the cartilage matrix. Glucosamine administration exerts specific outcomes on chondrocytes and cartilage in osteoarthritis.86 Glucosamine affects the molecular expression of arthritic cartilage, and its therapeutic effects are linked to its anticatabolic activities.87 Glucosamine is given in vitro to reduce PGE2 production and inactivation of the NF-κB pathway, thus inhibiting the cytokine intracellular signaling cascade in chondrocytes and synovial cells. In osteoarthritis, glucosamine induces a setback of the inflammation and jointdegenerating drawbacks of IL-1.86 Interleukin-1β is a persuasive proinflammatory cytokine produced in the arthritic joint, where it triggers the upregulation of inflammatory mediators such as COX-2, iNOS, IL-6, and TNF-α. IL-1β also induces cells to produce more IL-1β and matrix degradation factors, such as metalloproteinases (MMPs) and a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member TSs (ADAMTSs). Most of these genes are under the transcriptional control of NF-κB.

Nuclear Factor Kappa B Inhibition as a Therapeutic Target

Glucosamine at appropriate concentrations lessens the gene expression of COX-2, iNOS, and microsomal prostaglandin Esynthase-1 (mPGEs1) and PGE2 synthesis after IL-1β induction, signifying that glucosamine can regulate the triggered cascade by inflammatory stimuli.88 Transcription of IL-6, IL-8, IL-24, and TNF-α genes is controlled by multiple transcription factors, such as NF-κB.89 Attractively, the functions of NF-κB are controlled by OGlcNAc modification.90 Furthermore, GlcN inhibits the TNF-α-induced chemokine expression by rat smooth muscle cells via the O-GlcNAc modification of NF-κB p65.91 Moreover, it was previously revealed that GlcN inhibits the expression of chemokine and the adhesion molecule by endothelial cells via O-GlcNAc modification of NF-κB p65.89 GlcN had been confirmed to enhance the O-GlcNAc modification of NF-κB p65 but suppresses the expression of cytokines in MH7A cells. These observations likely suggest that the expression of proinflammatory cytokine genes may be regulated by the mechanism involving the O-GlcNAc modification of NF-κB.

6.3 Chondroitin and Glucosamine Combination The chondroitin and glucosamine combination restrained the IL-1-induced gene upregulation of iNOS, COX- 2, m PGEs, and NF-κB in inflamed cartilage. This led to decreased NO and PGE2 production, which is the mediator for chondrocyte cell death and inflammatory reactions.92 Both in a mixture could diminish production of the COX2 enzyme. One way is the reduction of the IL-1β-induced NF-κB pathway by glucosamine results in reduced synthesis of the COX-2 enzyme.93 A further way in which glucosamine inhibits COX-2 is the downregulation of COX-2 N-glycosylation and the facilitation of COX-2 protein yield.94 Chondroitin alone reduces the nuclear translocation of NF-κB, which lessens the production of proinflammatory cytokines IL-1β and TNF-α and enzymes such as COX-2 and NOS-2.95 The antiinflammatory capability of CS was a reduction of the proinflammatory molecules C-reactive protein and IL-6 and the expression of MCP-1 and COX-2 in the mononuclear cells. It also inhibited NF-κB that initiates induction of inflammatory processes.96 Furthermore, the metalloproteinase-3 (TIMP-3), a potent inhibitor of ADAMTS, was upregulated. Glucosamine alone inhibited the activation process of MMP-2 and MMP-9 expression via inhibition of the NF-κB pathway.97 Inflammatory mediators are keys for narrowed biosynthesis of cartilage material. In rats, chondrocytes models have shown that IL-1β inhibits the key enzyme in the biosynthesis of cartilage GAG chains and a dosedependent glucosamine could reduce this inhibition.98

6.4 Omega 3 Fatty Acids Omega-3 polyunsaturated fatty acids such as linolenic acid and eicosapentaenoic acid are found in plant and fish oils.51 Their antiinflammatory action has been approved in several

447

448

Bioactive Food as Dietary Interventions for Arthritis and Related Inflammatory Diseases

studies. It has been successfully used in clinical trials, mainly to treat rheumatoid arthritis. In vitro studies on omega-3 fatty acids revealed that they can augment collagen synthesis and diminish the inflammatory mediator PGE2.99 Omega-3 fatty acids decrease IL-1induced aggrecanase and collagenase activity and reduce mRNA transcription of ADAMTS-4, COX-2, IL-1α, and TNF-α. Furthermore, they decline the levels of numerous MMP proteins.100 Inflammatory diseases are regularly related with activation of NF-κB transcription factor and release of inflammatory cytokines and therefore, this is an important target for n-3 PUFAs. Associations between dietary fats and DNA methylation in the NF-κB pathway, measured using the Infinium 450 k array, were observed in the Greek preadolescent cross-sectional cohort. Therefore, this may provide valuable targets within intervention studies.101

7. CONCLUSION Because dysregulated NF-κB activation is implicated in various inflammatory diseases, targeting the NF-κB signaling pathway represents a striking application for antiinflammatory trials in arthritis. It’s noticed that the use of nutrients in rheumatology is atypical and eventually, the enhanced interest in arthritic patients in the nutrient approach will be a reality. Therefore, rheumatologists should advise their patients with scientific tools and the finest data available. In this sense, the antiarthritic ability of nutrients sets its position, which supports precise scientific evaluation on all approaches that improve the therapy of rheumatic diseases. At present, the nutrient approaches are possibly advantageous for RA patients, although there is still a long way ahead in terms of research to draw firm conclusions. Until now, no long-term studies, nor studies to assess series of joint damage; conversely some complementary therapies may signify a prospect to broaden the worth of our patient’s life, may be along with integrated management of arthritic patients in the future.

REFERENCES 1. Smolen JS, Breedveld FC, Burmester GR, et al. Treating rheumatoid arthritis to target: 2014 update of the recommendations of an international task force. Ann Rheum Dis. 2016;75(1):3–15. 2. Sauer S, Plauth A. Health-beneficial nutraceuticals-myth or reality? Appl Microbiol Biotechnol. 2017;101 (3):951–961.28070666. 3. Burska AN, Roget K, Blits M, et al. Gene expression analysis in RA: towards personalized medicine. Pharmacogenomics J. 2014;14:93. 4. van der Pouw Kraan TCTM, van Gaalen FA, Kasperkovitz PV, et al. Rheumatoid arthritis is a heterogeneous disease: evidence for differences in the activation of the STAT-1 pathway between rheumatoid tissues. Arthritis Rheum. 2003;48(8):2132–2145. 5. Liu T, Zhang L, Joo D, Sun S-C. NF-κB signaling in inflammation. Signal Transduct Target Ther. 2017;2:17023. 6. Chimenti MS, Triggianese P, Conigliaro P, Candi E, Melino G, Perricone R. The interplay between inflammation and metabolism in rheumatoid arthritis. Cell Death Dis. 2015;6:e1887.

Nuclear Factor Kappa B Inhibition as a Therapeutic Target

7. Kumar A, Takada Y, Boriek AM, Aggarwal BB. Nuclear factor-κB: its role in health and disease. J Mol Med. 2004;82(7):434–448. 8. Hao R, Du H, Guo L, et al. Identification of dysregulated genes in rheumatoid arthritis based on bioinformatics analysis. PeerJ. 2017;5:e3078. 9. Baum R, Gravallese EM. Bone as a target organ in rheumatic disease: impact on osteoclasts and osteoblasts. Clin Rev Allergy Immunol. 2016;51(1):1–15. 10. Teng MWL, Bowman EP, McElwee JJ, et al. IL-12 and IL-23 cytokines: from discovery to targeted therapies for immune-mediated inflammatory diseases. Nat Med. 2015;21:719. 11. Wei F, Chang Y, Wei W. The role of BAFF in the progression of rheumatoid arthritis. Cytokine. 2015;76(2):537–544. 12. Sellam J, Berenbaum F. The role of synovitis in pathophysiology and clinical symptoms of osteoarthritis. Nat Rev Rheumatol. 2010;6:625. 13. Foxwell B, Browne K, Bondeson J, et al. Efficient adenoviral infection with IκBα reveals that macrophage tumor necrosis factor α production in rheumatoid arthritis is NF-κB dependent. Proc Natl Acad Sci. 1998;95(14):8211–8215. 14. Perkins ND. Integrating cell-signalling pathways with NF-κB and IKK function. Nat Rev Mol Cell Biol. 2007;8:49. 15. Moran EM, Mullan R, McCormick J, et al. Human rheumatoid arthritis tissue production of IL-17A drives matrix and cartilage degradation: synergy with tumour necrosis factor-α, Oncostatin M and response to biologic therapies. Arthritis Res Ther. 2009;11(4):R113. 16. Hayden MS, Ghosh S. NF-kappaB, the first quarter-century: remarkable progress and outstanding questions. Genes Dev. 2012;26(3):203–234. 22302935 [Epub 2012/02/04. eng]. 17. Oeckinghaus A, Hayden MS, Ghosh S. Crosstalk in NF-κB signaling pathways. Nat Immunol. 2011;12:695. 18. Fontalba A, Gutierrez O, Fernandez-Luna JL. NLRP2, an inhibitor of the NF-κB pathway, is transcriptionally activated by NF-κB and exhibits a nonfunctional allelic variant. J Immunol. 2007;179 (12):8519–8524. 19. Lawrence T. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb Perspect Biol. 2009;1(6):a001651. 20457564 [Epub 2010/05/12. eng]. 20. Choy E. Understanding the dynamics: pathways involved in the pathogenesis of rheumatoid arthritis. Rheumatology. 2012;51(suppl 5):v3–v11. 21. Gibofsky A. Overview of epidemiology, pathophysiology, and diagnosis of rheumatoid arthritis. Am J Manag Care. 2012;18(13 suppl):S295–S302.23327517. 22. Herkenham M, Rathore P, Brown P, Listwak SJ. Cautionary notes on the use of NF-κB p65 and p50 antibodies for CNS studies. J Neuroinflamm. 2011;8(1):141. 23. Giopanou I, Bravou V, Papanastasopoulos P, et al. Metadherin, p50, and p65 expression in epithelial ovarian neoplasms: an immunohistochemical study. Biomed Res Int. 2014;2014:8. 24. Mann DA. The NFκB luciferase mouse: a new tool for real time measurement of NFκB activation in the whole animal. Gut. 2002;51(6):769–770. 25. Caterina MJ, Julius D. The vanilloid receptor: a molecular gateway to the pain pathway. Annu Rev Neurosci. 2001;24(1):487–517.11283319. 26. Surh Y-J, Chun K-S, Cha H-H, et al. Molecular mechanisms underlying chemopreventive activities of anti-inflammatory phytochemicals: down-regulation of COX-2 and iNOS through suppression of NF-κB activation. Mutat Res. 2001;480–481:243–268. 27. Tetali P, Waghchaure C, Daswani PG, Antia NH, Birdi TJ. Ethnobotanical survey of antidiarrhoeal plants of Parinche valley, Pune district, Maharashtra, India. J Ethnopharmacol. 2009;123(2):229–236. 28. Funk JL, Frye JB, Oyarzo JN, et al. Efficacy and mechanism of action of turmeric supplements in the treatment of experimental arthritis. Arthritis Rheum. 2006;54(11):3452–3464. 29. Kamarudin TA, Othman F, Mohd Ramli ES, Md Isa N, Das S. Protective effect of curcumin on experimentally induced arthritic rats: detailed histopathological study of the joints and white blood cell count. EXCLI J. 2012;11:226–236. 30. Yeh C-C, Su Y-H, Lin Y-J, et al. Evaluation of the protective effects of curcuminoid (curcumin and bisdemethoxycurcumin)-loaded liposomes against bone turnover in a cell-based model of osteoarthritis. Drug Des Dev Ther. 2015;9:2285–2300.

449

450

Bioactive Food as Dietary Interventions for Arthritis and Related Inflammatory Diseases

31. Ramadan G, El-Menshawy O. Protective effects of ginger-turmeric rhizomes mixture on joint inflammation, atherogenesis, kidney dysfunction and other complications in a rat model of human rheumatoid arthritis. Int J Rheum Dis. 2013;16(2):219–229. 32. Prabhavathi K, Chandra USJ, Soanker R, Rani PU. A randomized, double blind, placebo controlled, cross over study to evaluate the analgesic activity of Boswellia serrata in healthy volunteers using mechanical pain model. Indian J Pharmacol. 2014;46(5):475–479. 33. Kuptniratsaikul V, Dajpratham P, Taechaarpornkul W, et al. Efficacy and safety of Curcuma domestica extracts compared with ibuprofen in patients with knee osteoarthritis: a multicenter study. Clin Interv Aging. 2014;9:451–458. 34. Daily JW, Yang M, Park S. Efficacy of turmeric extracts and curcumin for alleviating the symptoms of joint arthritis: a systematic review and meta-analysis of randomized clinical trials. J Med Food. 2016;19 (8):717–729. 35. Ghosh S, Banerjee S, Sil PC. The beneficial role of curcumin on inflammation, diabetes and neurodegenerative disease: a recent update. Food Chem Toxicol. 2015;83:111–124. 36. Nonose N, Pereira JA, Machado PRM, Rodrigues MR, Sato DT, Martinez CAR. Oral administration of curcumin (Curcuma longa) can attenuate the neutrophil inflammatory response in zymosan-induced arthritis in rats. Acta Cir Bras. 2014;29:727–734. 37. Kim SR, Jung YR, An HJ, et al. Anti-wrinkle and anti-inflammatory effects of active garlic components and the inhibition of MMPs via NF-κB signaling. PLoS ONE. 2013;8(9):e73877. 38. Stacie KT, Robert ES. Immune system involvement in specific pain conditions. Mol Pain. 2017;13. https://doi.org/10.1177/1744806917724559. 39. Ban JO, Hwang IG, Kim TM, et al. Inhibition of cell growth and induction of apoptosis via inactivation of NF-kappa B by a sulfur compound isolated from garlic in human colon cancer cells. J Pharmacol Sci. 2007;104. 40. Liu Y, Zhu P, Wang Y, et al. Antimetastatic therapies of the polysulfide diallyl trisulfide against triplenegative breast cancer (TNBC) via suppressing MMP2/9 by blocking NF-κB and ERK/MAPK signaling pathways. PLoS ONE. 2015;10(4)e0123781. 41. Ueda H, Ippoushi K, Takeuchi A. Repeated oral administration of a squeezed ginger (Zingiber officinale) extract augmented the serum corticosterone level and had anti-inflammatory properties. Biosci Biotechnol Biochem. 2010;74(11):2248–2252. 42. Naderi Z, Mozaffari-Khosravi H, Dehghan A, Nadjarzadeh A, Huseini HF. 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(3):199–203. 43. Lee HY, Park SH, Lee M, et al. 1-Dehydro-[10]-gingerdione from ginger inhibits IKKβ activity for NF-κB activation and suppresses NF-κB-regulated expression of inflammatory genes. Br J Pharmacol. 2012;167(1):128–140. 44. Huang S-H, Lee C-H, Wang H-M, et al. 6-Dehydrogingerdione restrains lipopolysaccharide-induced inflammatory responses in RAW 264.7 macrophages. J Agric Food Chem. 2014;62(37):9171–9179. 45. Fu Y, Chen J, Li Y-J, Zheng Y-F, Li P. Antioxidant and anti-inflammatory activities of six flavonoids separated from licorice. Food Chem. 2013;141(2):1063–1071. 46. Honda H, Nagai Y, Matsunaga T, et al. Isoliquiritigenin is a potent inhibitor of NLRP3 inflammasome activation and diet-induced adipose tissue inflammation. J Leukoc Biol. 2014;96(6):1087–1100. 25210146 [Epub 2014/09/12. eng]. 47. Wang Z, Cao Y, Paudel S, Yoon G, Cheon SH. Concise synthesis of licochalcone C and its regioisomer, licochalcone H. Arch Pharm Res. 2013;36(12):1432–1436. 48. Lee H, Cho H, Lim D, Kang Y-H, Lee K, Park J. Mechanisms by which licochalcone E exhibits potent anti-inflammatory properties: studies with phorbol ester-treated mouse skin and lipopolysaccharide-stimulated murine macrophages. Int J Mol Sci. 2013;14(6):10926. https://doi. org/10.3390/ijms140610926. 49. Kim HJ, Seo J-Y, Suh H-J, Lim SS, Kim J-S. Antioxidant activities of licorice-derived prenylflavonoids. Nutr Res Pract. 2012;6(6):491–498. 50. Kim J, Kim J, Shim J, et al. Licorice-derived dehydroglyasperin C increases MKP-1 expression and suppresses inflammation-mediated neurodegeneration. Neurochem Int. 2013;63(8):732–740.

Nuclear Factor Kappa B Inhibition as a Therapeutic Target

51. Thiyagarajan P, Chandrasekaran CV, Deepak HB, Agarwal A. Modulation of lipopolysaccharideinduced pro-inflammatory mediators by an extract of Glycyrrhiza glabra and its phytoconstituents. Inflammopharmacology. 2011;19(4):235–241. 52. Yehuda I, Madar Z, Leikin-Frenkel A, Tamir S. Glabridin, an isoflavan from licorice root, downregulates iNOS expression and activity under high-glucose stress and inflammation. Mol Nutr Food Res. 2015;59(6):1041–1052. 53. La VD, Tanabe S-I, Bergeron C, Gafner S, Grenier D. Modulation of matrix metalloproteinase and cytokine production by licorice isolates licoricidin and licorisoflavan a: potential therapeutic approach for periodontitis. J Periodontol. 2010;82(1):122–128. 54. Danesi F, Ferguson L. Could pomegranate juice help in the control of inflammatory diseases? Nutrients. 2017;9(9):958. https://doi.org/10.3390/nu9090958. 55. Kim YE, Hwang CJ, Lee HP, et al. Inhibitory effect of punicalagin on lipopolysaccharide-induced neuroinflammation, oxidative stress and memory impairment via inhibition of nuclear factor-kappaB. Neuropharmacology. 2017;117:21–32. 56. Seong A-R, Yoo J-Y, Choi K, et al. Delphinidin, a specific inhibitor of histone acetyltransferase, suppresses inflammatory signaling via prevention of NF-κB acetylation in fibroblast-like synoviocyte MH7A cells. Biochem Biophys Res Commun. 2011;410(3):581–586. 57. Hollebeeck S, Winand J, Herent M-F, et al. Anti-inflammatory effects of pomegranate (Punica granatum L.) husk ellagitannins in Caco-2 cells, an in vitro model of human intestine. Food Funct. 2012; 3(8):875–885. 58. Promsong A, Chung WO, Satthakarn S, Nittayananta W. Ellagic acid modulates the expression of oral innate immune mediators: potential role in mucosal protection. J Oral Pathol Med. 2015;44(3):214–221. 59. Husari A, Hashem Y, Bitar H, Dbaibo G, Zaatari G, El Sabban M. Antioxidant activity of pomegranate juice reduces emphysematous changes and injury secondary to cigarette smoke in an animal model and human alveolar cells. Int J Chron Obstruct Pulmon Dis. 2016;11:227–237. 60. Shukla M, Gupta K, Rasheed Z, Khan KA, Haqqi TM. Consumption of hydrolyzable tannins-rich pomegranate extract suppresses inflammation and joint damage in rheumatoid arthritis. Nutrition. 2008;24(7):733–743. 61. Kim H, Banerjee N, Ivanov I, et al. Comparison of anti-inflammatory mechanisms of mango (Mangifera indica L.) and pomegranate (Punica granatum L.) in a preclinical model of colitis. Mol Nutr Food Res. 2016;60(9):1912–1923. 62. Shah TA, Parikh M, Patel KV, Patel KG, Joshi CG, Gandhi TR. Evaluation of the effect of Punica granatum juice and punicalagin on NFκB modulation in inflammatory bowel disease. Mol Cell Biochem. 2016;419(1):65–74. 63. Rosillo MA, Sanchez-Hidalgo M, Ca´rdeno A, Alarco´n de la Lastra C. Protective effect of ellagic acid, a natural polyphenolic compound, in a murine model of Crohn’s disease. Biochem Pharmacol. 2011;82 (7):737–745. 64. Elmali N, Baysal O, Harma A, Esenkaya I, Mizrak B. Effects of resveratrol in inflammatory arthritis. Inflammation. 2007;30(1):1–6. 65. Sumpio BE, Cordova AC, Berke-Schlessel DW, Qin F, Chen QH. Green tea, the “Asian paradox,” and cardiovascular disease. J Am Coll Surg. 2006;202(5):813–825. 66. Hsu D-Z, Chen S-J, Chu P-Y, Liu M-Y. Therapeutic effects of sesame oil on monosodium urate crystal-induced acute inflammatory response in rats. Springerplus. 2013;2(1):659. 67. Bauerova´ K, Ponisˇt S, Mihalova´ D, Dra´fi F, Kuncı´rova´ V. Utilization of adjuvant arthritis model for evaluation of new approaches in rheumatoid arthritis therapy focused on regulation of immune processes and oxidative stress. Interdiscip Toxicol. 2011;33. 68. Hsu D-Z, Chu P-Y, Jou IM. Daily sesame oil supplement attenuates joint pain by inhibiting muscular oxidative stress in osteoarthritis rat model. J Nutr Biochem. 2016;29:36–40. 69. Yadav NV, Sadashivaiah, Ramaiyan B, Acharya P, Belur L, Talahalli RR. Sesame oil and rice bran oil ameliorates adjuvant-induced arthritis in rats: distinguishing the role of minor components and fatty acids. Lipids. 2016;51(12):1385–1395. 70. Eftekhar Sadat B, Khadem Haghighian M, Alipoor B, Malek Mahdavi A, Asghari Jafarabadi M, Moghaddam A. Effects of sesame seed supplementation on clinical signs and symptoms in patients with knee osteoarthritis. Int J Rheum Dis. 2013;16(5):578–582.

451

452

Bioactive Food as Dietary Interventions for Arthritis and Related Inflammatory Diseases

71. Khadem Haghighian M, Alipoor B, Malek Mahdavi A, Eftekhar Sadat B, Asghari Jafarabadi M, Moghaddam A. Effects of sesame seed supplementation on inflammatory factors and oxidative stress biomarkers in patients with knee osteoarthritis. Acta Med Iran. 2015;7 [Epub 2015-04-15]. 72. Yashaswini PS, Rao AGA, Singh SA. Inhibition of lipoxygenase by sesamol corroborates its potential anti-inflammatory activity. Int J Biol Macromol. 2017;94:781–787. 73. Hemshekhar M, Thushara RM, Jnaneshwari S, Devaraja S, Kemparaju K, Girish KS. Attenuation of adjuvant-induced arthritis by dietary sesamol via modulation of inflammatory mediators, extracellular matrix degrading enzymes and antioxidant status. Eur J Nutr. 2013;52(7):1787–1799. 74. Pratte MA, Nanavati KB, Young V, Morley CP. An alternative treatment for anxiety: a systematic review of human trial results reported for the ayurvedic herb ashwagandha (Withania somnifera). J Altern Complement Med. 2014;20(12):901–908. 75. Singh D, Aggarwal A, Maurya R, Naik S. Withania somnifera inhibits NF-κB and AP-1 transcription factors in human peripheral blood and synovial fluid mononuclear cells. Phytother Res. 2007;21 (10):905–913. 76. Ganesan K, Sehgal PK, Mandal AB, Sayeed S. Protective effect of Withania somnifera and Cardiospermum halicacabum extracts against collagenolytic degradation of collagen. Appl Biochem Biotechnol. 2011;165 (3):1075–1091. 77. Rasool M, Varalakshmi P. Protective effect of Withania somnifera root powder in relation to lipid peroxidation, antioxidant status, glycoproteins and bone collagen on adjuvant-induced arthritis in rats. Fundam Clin Pharmacol. 2007;21(2):157–164. 17391288 [Epub 2007/03/30. eng]. 78. Ramakanth GS, Uday Kumar C, Kishan PV, Usharani P. A randomized, double blind placebo controlled study of efficacy and tolerability of Withaina somnifera extracts in knee joint pain. J Ayurveda Integr Med. 2016;7(3):151–157. 27647541 [Epub 2016/09/21. eng]. 79. Grover A, Shandilya A, Punetha A, Bisaria VS, Sundar D. Inhibition of the NEMO/IKKβ association complex formation, a novel mechanism associated with the NF-κB activation suppression by Withania somnifera’s key metabolite withaferin A. BMC Genomics. 2010;11(4):S25. 80. Heyninck K, Lahtela-Kakkonen M, Van der Veken P, Haegeman G, Vanden Berghe W. Withaferin A inhibits NF-kappaB activation by targeting cysteine 179 in IKKβ. Biochem Pharmacol. 2014;91 (4):501–509. 81. Darwish HA, Abd Raboh NR, Mahdy A. Camel’s milk alleviates alcohol-induced liver injury in rats. Food Chem Toxicol. 2012;50(5):1377–1383. 82. Zhu W-W, Kong G-Q, Ma M-M, et al. Short communication: Camel milk ameliorates inflammatory responses and oxidative stress and downregulates mitogen-activated protein kinase signaling pathways in lipopolysaccharide-induced acute respiratory distress syndrome in rats. J Dairy Sci. 2016;99 (1):53–56. 83. Ebaid H, Abdel-salam B, Hassan I, Al-Tamimi J, Metwalli A, Alhazza I. Camel milk peptide improves wound healing in diabetic rats by orchestrating the redox status and immune response. Lipids Health Dis. 2015;14(1):132. 84. Phillips DC, Dias HK, Kitas GD, Griffiths HR. Aberrant reactive oxygen and nitrogen species generation in rheumatoid arthritis (RA): causes and consequences for immune function, cell survival, and therapeutic intervention. Antioxid Redox Signal. 2010;12(6):743–785. 19686039 [Epub 2009/08/19. eng]. 85. Rosillo MA´, Alcaraz MJ, Sa´nchez-Hidalgo M, Ferna´ndez-Bolan˜os JG, Alarco´n-de-la-Lastra C, Ferra´ndiz ML. Anti-inflammatory and joint protective effects of extra-virgin olive-oil polyphenol extract in experimental arthritis. J Nutr Biochem. 2014;25(12):1275–1281. 86. Lucio CR, Federica G, Stefano P. Crystalline glucosamine sulfate in the management of knee osteoarthritis: efficacy, safety, and pharmacokinetic properties. Ther Adv Musculoskelet Dis. 2012;4 (3):167–180. 87. Reginster J-Y, Neuprez A, Lecart M-P, Sarlet N, Bruyere O. Role of glucosamine in the treatment for osteoarthritis. Rheumatol Int. 2012;32(10):2959–2967. 88. Chan PS, Caron JP, Rosa GJM, Orth MW. Glucosamine and chondroitin sulfate regulate gene expression and synthesis of nitric oxide and prostaglandin E2 in articular cartilage explants. Osteoarthr Cartil. 2005;13(5):387–394.

Nuclear Factor Kappa B Inhibition as a Therapeutic Target

89. Zachara NE. The roles of O-linked β-N-acetylglucosamine in cardiovascular physiology and disease. Am J Phys Heart Circ Phys. 2012;302(10):H1905–H1918.22287582. 90. Ramakrishnan P, Clark PM, Mason DE, Peters EC, Hsieh-Wilson LC, Baltimore D. Activation of the transcriptional function of the NF-kappaB protein c-Rel by O-GlcNAc glycosylation. Sci Signal. 2013; 6(290):ra75. 23982206 [Epub 2013/08/29. eng]. 91. Xing D, Gong K, Feng W, et al. O-GlcNAc modification of NFκB p65 inhibits TNF-α-induced inflammatory mediator expression in rat aortic smooth muscle cells. PLoS ONE. 2011;6(8):e24021. 92. Chan P-S, Caron JP, Orth MW. Short-term gene expression changes in cartilage explants stimulated with interleukin beta plus glucosamine and chondroitin sulfate. J Rheumatol. 2006;33(7):1329–1340. 93. Largo R, Alvarez-Soria MA, Dı´ez Ortego I, et al. Glucosamine inhibits IL-1β-induced NFκB activation in human osteoarthritic chondrocytes. Osteoarthr Cartil. 2003;11(4):290–298. 94. Jang BC, Sung SH, Park JG, et al. Glucosamine hydrochloride specifically inhibits COX-2 by preventing COX-2 N-glycosylation and by increasing COX-2 protein turnover in a proteasome-dependent manner. J Biol Chem. 2007;282(38):27622–27632. 17635918 [Epub 2007/07/20. eng]. 95. du Souich P, Garcı´a AG, Verges J, Montell E. Immunomodulatory and anti-inflammatory effects of chondroitin sulphate. J Cell Mol Med. 2009;13(8a):1451–1463. 96. Herrero-Beaumont G, Marcos ME, Sa´nchez-Pernaute O, et al. Effect of chondroitin sulphate in a rabbit model of atherosclerosis aggravated by chronic arthritis. Br J Pharmacol. 2008;154(4):843–851. 97. Rajapakse N, Mendis E, Kim M-M, Kim S-K. Sulfated glucosamine inhibits MMP-2 and MMP-9 expressions in human fibrosarcoma cells. Bioorg Med Chem. 2007;15(14):4891–4896. 98. Gouze J-N, Bordji K, Gulberti S, et al. Interleukin-1β down-regulates the expression of glucuronosyltransferase I, a key enzyme priming glycosaminoglycan biosynthesis: influence of glucosamine on interleukin-1β–mediated effects in rat chondrocytes. Arthritis Rheum. 2001;44(2):351–360. 99. Hankenson KD, Watkins BA, Schoenlein IA, Allen KGD, Turek JJ. Omega-3 fatty acids enhance ligament fibroblast collagen formation in association with changes in interleukin-6 production. Proc Soc Exp Biol Med. 2000;223(1):88–95. 100. Curtis CL, Rees SG, Little CB, et al. Pathologic indicators of degradation and inflammation in human osteoarthritic cartilage are abrogated by exposure to n-3 fatty acids. Arthritis Rheum. 2002;46 (6):1544–1553. 101. Voisin S, Almen MS, Moschonis G, Chrousos GP, Manios Y, Schi€ oth HB. Dietary fat quality impacts genome-wide DNA methylation patterns in a cross-sectional study of Greek preadolescents. Eur J Hum Genet. 2014;23:654.

453