Hostname: page-component-8448b6f56d-sxzjt Total loading time: 0 Render date: 2024-04-19T19:22:46.668Z Has data issue: false hasContentIssue false

The potential for dietary factors to prevent or treat osteoarthritis

Published online by Cambridge University Press:  26 February 2014

Jonathan A. Green
Affiliation:
School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ,UK
Kimberley L. Hirst-Jones
Affiliation:
Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
Rose K. Davidson
Affiliation:
School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ,UK
Orla Jupp
Affiliation:
School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ,UK
Yongping Bao
Affiliation:
Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
Alexander J. MacGregor
Affiliation:
Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
Simon T. Donell
Affiliation:
Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
Aedín Cassidy
Affiliation:
Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
Ian M. Clark*
Affiliation:
School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ,UK
*
*Corresponding author: I. M. Clark, fax 01603-592250, email i.clark@uea.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Osteoarthritis (OA) is a degenerative joint disease for which there are no disease-modifying drugs. It is a leading cause of disability in the UK. Increasing age and obesity are both major risk factors for OA and the health and economic burden of this disease will increase in the future. Focusing on compounds from the habitual diet that may prevent the onset or slow the progression of OA is a strategy that has been under-investigated to date. An approach that relies on dietary modification is clearly attractive in terms of risk/benefit and more likely to be implementable at the population level. However, before undertaking a full clinical trial to examine potential efficacy, detailed molecular studies are required in order to optimise the design. This review focuses on potential dietary factors that may reduce the risk or progression of OA, including micronutrients, fatty acids, flavonoids and other phytochemicals. It therefore ignores data coming from classical inflammatory arthritides and nutraceuticals such as glucosamine and chondroitin. In conclusion, diet offers a route by which the health of the joint can be protected and OA incidence or progression decreased. In a chronic disease, with risk factors increasing in the population and with no pharmaceutical cure, an understanding of this will be crucial.

Type
Conference on ‘Nutrition and healthy ageing’
Copyright
Copyright © The Authors 2014 

Abbreviations:
ADAMTS

a disintegrin and metalloproteinase domain with thrombospondin motifs

ASU

avocado-soyabean unsaponifiables

COX

cyclooxygenase

Gla

γ-carboxyglutamic acid

MMP

matrix metalloproteinase

OA

osteoarthritis

Osteoarthritis (OA) is a degenerative joint disease characterised by degradation of articular cartilage, thickening of subchondral bone and osteophyte formation. Incidence and prevalence of OA has been difficult to assess, in part because of heterogeneity in definitions of the disease. A recent meta-analysis suggested that overall prevalence of OA at different anatomical sites was 23·9 % (knee), 10·9 % (hip) and 43·3 % (hand), although only the prevalence of knee OA showed a gender difference between women and men (27·3 and 21 %, respectively)( Reference Pereira, Peleteiro and Araujo 1 ).

Osteoarthritis is a leading cause of disability in the UK. A recent survey( 2 ) found 8·5 million people in the UK with OA, with 71 % of these in constant pain. There are no effective disease-modifying drugs to treat OA and drugs that relieve pain are often insufficient. Joint replacement is offered to patients at end-stage disease with 66 436 hip and 77 578 knee replacements due to OA performed in the UK in 2011( 3 ).

Two major risk factors for OA are increasing age (most affected patients are aged >45 years and the greatest morbidity is seen in patients aged >60 years)( Reference Shane Anderson and Loeser 4 ) and increasing obesity( Reference Richmond, Fukuchi and Ezzat 5 ). With changing demographics, OA is an increasing public health and economic burden. The economic costs of OA in the UK are largely unknown, but direct costs have been estimated at approximately £1 billion/year. With inclusion of indirect costs, estimates from the USA range up to £8 billion/year( Reference Chen, Gupte and Akhtar 6 ).

Although the ability to slow or stop the progression of OA would have individual and population-level benefits, few pharmaceutical companies maintain OA as a disease area. This is in part because there is no precedent. Furthermore, OA generally progresses slowly, and there are no current validated biomarkers for cartilage destruction (joint space narrowing, assessed on X-ray, is the only Food and Drug Administration approved end point in a clinical trial)( Reference Kraus 7 ). Issues of toxicity, in a disease that is not life-threatening, can also make drug development problematic. It is possible to overcome at least some of these issues by selection of the patient group (where particular sub-groups are known to progress more rapidly), and by establishing the dose of drug that gives efficacy within the target tissue (i.e. cartilage)( Reference Jordan, Sowers and Messier 8 ).

Focusing on compounds from the habitual diet that may prevent the onset or slow the progression of OA is an alternative strategy. Since in essence, all of the population can be viewed as at risk for the development of OA in old age, an approach that relies on dietary modification is clearly more attractive in terms of risk/benefit and more likely to be implementable. However, detailed molecular studies ahead of a full clinical trial are required in order to design trials optimally that will examine potential efficacy.

There are currently limited data on the inter-relationship between diet and OA. Data come from a variety of studies: in vitro cell and tissue explant models, animal models, epidemiological associations and intervention trials. There is a large variability between studies, e.g. in animal models, a dietary intake approach would be optimal in order to relate to human exposure, but some studies use intra-articular injection and/or concentrations not achievable through the diet. The intervention trials conducted to date have many different designs, number of patients, time length and outcome measures, often with too few patients and of short duration. There is a need for better quality data before dietary advice can be given. However, clinical trials in OA are expensive and it is not clear who will or should fund them.

This brief review focuses predominantly on potential dietary factors than may reduce the risk or progression of the disease. It focuses only on OA, mainly ignoring data coming from more overtly inflammatory arthritides.

Two pertinent ‘nutraceuticals’ will not be discussed, but should be mentioned: glucosamine and chondroitin. Glucosamine is a sugar and precursor for glycosaminoglycan and therefore proteoglycan biosynthesis. Chondroitin is a glycosaminoglycan, a form of which is found in aggrecan, the major proteoglycan in cartilage. Hydrochloride and sulphate salts of both glucosamine and chondroitin have been extensively examined in laboratory models and clinical trials. The efficacy of these compounds remains controversial, but most recent analyses appear to indicate that high-grade preparations of chondroitin sulphate and glucosamine sulphate, may have efficacy in OA( Reference Black, Clar and Henderson 9 Reference Wu, Huang and Gu 13 ).

Micronutrients

Vitamin C

In prospective studies examining micronutrient intakes, the Framingham study identified a protective association between higher intake of vitamin C and the progression of radiographic knee OA( Reference McAlindon, Jacques and Zhang 14 ) and a higher vitamin C intake was also associated with lower risk of knee pain( Reference McAlindon, Jacques and Zhang 14 , Reference McAlindon 15 ). However, a longitudinal study showed no protective effect of vitamin C supplements on the progression of knee OA, although in multivariate analyses vitamin C supplements were beneficial in preventing the development of knee OA( Reference Peregoy and Wilder 16 ). In healthy subjects, vitamin C intake has been associated with reduced risk of bone marrow lesions on MRI( Reference Wang, Hodge and Wluka 17 ). In these publications, vitamin C has been viewed simply as an antioxidant, but it should not be forgotten that vitamin C is a co-factor enabling the proline and lysine hydroxylation essential for correct collagen biosynthesis. It also has effects on regulating the expression and translation of collagen, a major component of many connective tissues including cartilage and bone( Reference Clark, Rohrbaugh and Otterness 18 ). Animal model data (all from the guinea pig) are conflicting. Early studies showed that dietary ascorbate decreased pathology in surgically induced OA( Reference Schwartz, Oh and Leveille 19 ). In a further study, additional ascorbate in the drinking water showed a protective effect on spontaneous cartilage lesions, but no effect on pathology post-surgery( Reference Meacock, Bodmer and Billingham 20 ). Most recently, ascorbate supplementation increased disease severity in spontaneous OA( Reference Kraus, Huebner and Stabler 21 ).

Vitamin E

The Framingham study identified a weak protective association between higher intake of vitamin E and the progression of radiographic knee OA( Reference McAlindon, Jacques and Zhang 14 ). A study examining tocopherol isoforms and radiographic knee OA suggested complex associations( Reference Jordan, De Roos and Renner 22 ) and intervention trials of vitamin E have to date been contradictory( Reference Canter, Wider and Ernst 23 ). In vitro data in chondrocytes are sparse, but a recent study suggests that vitamin E protects against hydrogen peroxide-induced changes in extracellular matrix gene expression( Reference Bhatti, Mehmood and Wajid 24 ).

Vitamin D

Vitamin D has multiple functions in the musculoskeletal system, particularly in bone health and pathologies( Reference Wolff, Jones and Hansen 25 ). Many studies have explored the association between vitamin D levels and OA. Recent systematic review suggests that low serum concentrations of 25-hydroxyvitamin D are associated with increased radiographic progression of OA, but associations are weaker with symptoms of disease( Reference Cao, Winzenberg and Nguo 26 ). A recent longitudinal study demonstrated the converse, that moderate vitamin D deficiency predicts both knee and hip pain, independent of structural change( Reference Laslett, Quinn and Burgess 27 ). However, a recent 2-year intervention trial showed no decrease in knee pain or structural change in patients with knee OA, with knee function significantly worse following vitamin D intervention( Reference McAlindon, LaValley and Schneider 28 ). Further intervention trials are ongoing( Reference Cao, Jones and Cicuttini 29 ). Vitamin D supplementation in a rat post-surgical model of OA showed a protective effect during the early phase of the disease, but not during the later phase( Reference Castillo, Hernandez-Cueto and Vega-Lopez 30 ). However, this was scored using condyle width, an unusual method. Interestingly vitamin D receptor-deficient mice showed aggravated inflammation and cartilage damage when crossed into a TNF transgenic model( Reference Zwerina, Baum and Axmann 31 ).

Other micronutrients

In a Japanese population (Research on Osteoarthritis Against Disability), low habitual vitamin K intake was the only dietary factor associated with the increased prevalence of radiographic knee OA in a cross-sectional study( Reference Oka, Akune and Muraki 32 ). This supports data from US cohorts where low vitamin K was associated with OA in the hand and knee( Reference Misra, Booth and Tolstykh 33 , Reference Neogi, Booth and Zhang 34 ). However, a further study, using minimum joint space width and osteophytosis as variables showed an association of vitamins K, B1, B2, B6 and C with the former and vitamins E, K, B1, B2, niacin (B3) and B6 with the latter, both in women only( Reference Muraki, Akune and En-Yo 35 ). Vitamin K is an essential co-factor for the formation of γ-carboxyglutamic acid (Gla) residues, and Gla-containing proteins include osteocalcin and matrix Gla protein, both expressed in the skeleton. Vitamin K regulates mineralisation in both bone and cartilage( Reference Krueger, Westenfeld and Schurgers 36 ). Polymorphisms in the matrix Gla protein gene have been associated with hand OA( Reference Misra, Booth and Crosier 37 ), and serum levels of undercarboxylated osteocalcin may be associated with synovitis in knee OA( Reference Naito, Watari and Obayashi 38 ). Niacinamide, a form of vitamin B3, has been examined in a pilot scale clinical study of OA and reported to show improvements at 12 weeks( Reference Jonas, Rapoza and Blair 39 ).

An association between dietary magnesium intake and knee OA was demonstrated in the Johnston County Osteoarthritis Project, but this varied with ethnicity( Reference Qin, Shi and Samai 40 ). This is supported by data from the Twins UK registry where discordant twin pair analysis showed a decrease in magnesium in co-twins with OA( Reference Hunter, Hart and Snieder 41 ). Selenium has been implicated the osteoarthropathy of Kashin–Beck disease; meta-analysis of supplementation studies supports the benefit of supplementation in children, but highlights the low quality of methodology( Reference Zou, Liu and Wu 42 ).

Lipid metabolism

Recent studies have suggested that OA may be part of metabolic syndrome( Reference Zhuo, Yang and Chen 43 ). Alterations in lipid metabolism may be key to this, with population-based studies suggesting that serum cholesterol is a risk factor for OA (reviewed in( Reference Gkretsi, Simopoulou and Tsezou 44 )). Population studies also suggest that statin use is associated with a reduction in OA incidence and/or progression( Reference Clockaerts, Van Osch and Bastiaansen-Jenniskens 45 , Reference Kadam, Blagojevic and Belcher 46 ), but studies of pain and function in patients with OA have shown no association( Reference Riddle, Moxley and Dumenci 47 ). This area therefore remains controversial. It has been reported that high levels of fat and fatty acids are found in osteoarthritic joint tissues and that this is associated with pathology( Reference Lippiello, Walsh and Fienhold 48 , Reference Plumb and Aspden 49 ). n-3 PUFA, but not n-6 PUFA were found to be associated with the specific loss of cartilage in the Multicenter Osteoarthritis Study population of people at risk of OA( Reference Baker, Matthan and Lichtenstein 50 ). In healthy individuals, consumption of SFA or n-6 PUFA (but not n-3 PUFA) was associated with an increased risk of bone marrow lesions( Reference Wang, Davies-Tuck and Wluka 51 , Reference Wang, Wluka and Hodge 52 ). In animal models, a high-fat diet accelerated progression of OA( Reference Mooney, Sampson and Lerea 53 ), while n-3 PUFA reduced disease( Reference Knott, Avery and Hollander 54 ). Studies in isolated chondrocytes showed that n-3 PUFA inhibited IL-1-induced MMP3, MMP13, ADAMTS4, ADAMTS5 and COX2 (matrix metalloproteinase (MMP); a disintegrin and metalloproteinase domain with thrombospondin motifs (ADAMTS); cyclooxygenase (COX)) expression, while n-6 PUFA had no effect( Reference Hurst, Rees and Randerson 55 , Reference Zainal, Longman and Hurst 56 ). A small improvement in OA in dogs was seen with fish oil supplementation( Reference Hielm-Bjorkman, Roine and Elo 57 , Reference Roush, Dodd and Fritsch 58 ). Interestingly, a supplement rich in fish oil, Phytalgic, was shown to improve function and pain in OA patients( Reference Jacquet, Girodet and Pariente 59 ), although the design of this trial has been criticised( Reference Christensen and Bliddal 60 ).

Diet-derived bioactives

Typically, foods contain multiple bioactive compounds and these can impact upon many biological pathways( Reference Ameye and Chee 61 ). Diet-derived bioactives can be classified into several groups, e.g. flavonoids (and related compounds), carotenoids, plant sterols, glucosinolates and others( Reference Denny and Buttriss 62 ).

Flavonoids

Flavonoids are polyphenols and include flavan-3-ols, flavonols, flavones, isoflavones, flavanones and anthocyanins. More than 6000 different flavonoids have been found and they are widely distributed in plants, with several hundred found in edible plants( Reference Falcone Ferreyra, Rius and Casati 63 , Reference Manach, Scalbert and Morand 64 ).

Flavonols

Flavonols are found in many foods and are exemplified by quercetin, myricetin and kaempferol( Reference Manach, Scalbert and Morand 64 ). Quercetin and kaempferol showed no activity against IL-1-induced MMP13 levels in SW1353 chondrosarcoma cells( Reference Lim, Park and Kim 65 ). However, Lay et al. report that quercetin is able to block aggrecan loss from articular cartilage potentially via inhibition of ADAMTS4 and ADAMTS5( Reference Lay, Samiric and Handley 66 ) and Lee et al. show that myricetin can inhibit IL-1 induction of MMP1 from a synovial cell line( Reference Lee and Choi 67 ).

Flavones

In fruit and vegetables, flavones are found in celery and parsley, mainly luteolin and apigenin. In the skin of citrus fruit, polymethoxylated flavones are also found, e.g. tangeretin, nobiletin and sinensetin( Reference Manach, Scalbert and Morand 64 ). Luteolin and nobiletin have been shown to inhibit aggrecanases ADAMTS4 and ADAMTS5, both in vitro ( Reference Imada, Lin and Liu 68 , Reference Moncada-Pazos, Obaya and Viloria 69 ) and in vivo ( Reference Imada, Lin and Liu 68 ). Luteolin appears to be selective as a better ADAMTS than MMP inhibitor( Reference Moncada-Pazos, Obaya and Viloria 69 ), it also has anti-inflammatory activity, which could play a role in chondroprotection( Reference Lopez-Lazaro 70 ). Nobiletin, tangeretin and sinensetin all repress the IL-1 induction of MMP9 in synovial cells, with nobiletin also active in chondrocytes( Reference Ishiwa, Sato and Mimaki 71 ). Apigenin was shown to be a potent inhibitor of IL-1-induced MMP13 expression in SW1353 chondrosarcoma cells, potentially via activator protein 1 and the JAK/STAT (Janus kinase (JAK) and signal transducer and activator of transcription (STAT)) pathway, with no activity against NF-κB( Reference Lim, Park and Kim 65 ). It has also been shown to block IL-1-induced glycosaminoglycan release( Reference Lim, Park and Kim 65 ) and hyaluronan release( Reference Durigova, Roughley and Mort 72 ) from cartilage explants in vitro.

Flavan-3-ols

These exist as both monomer (catechins) and polymer (proanthocyanidins) forms( Reference Manach, Scalbert and Morand 64 ). Green tea polyphenols were shown to be effective in a model of inflammatory arthritis( Reference Haqqi, Anthony and Gupta 73 ). Catechins from green tea (and also present in other foods including dark chocolate) can inhibit cartilage degradation in vitro, particularly those containing a gallate ester( Reference Adcocks, Collin and Buttle 74 ). Epigallocatechin gallate and epicatechin gallate have been shown to be effective (submicromolar) inhibitors of ADAMTS4 and ADAMTS5 aggrecanase activity, indeed significantly more than their ability to inhibit MMP1 and MMP13 collagenase activity( Reference Vankemmelbeke, Jones and Fowles 75 ). Other anti-inflammatory activities have been described (e.g.( Reference Akhtar and Haqqi 76 )) that suggests promise in OA (reviewed in( Reference Ahmed 77 )), but no human clinical trials have been performed to date.

While not a diet-derived bioactive, Flavocoxid, a mixture of baicalin (a flavone) from Scutellaria baicalensis and catechins from Acacia catechu, is marketed as Limbrel, a ‘medical food’ which inhibits COX2 and 5-lipoxygenase( Reference Burnett, Jia and Zhao 78 ). An assessment of the major catechins from A. catechu suggests that they are predominantly those described earlier found in green tea( Reference Shen, Wu and Wang 79 ). Small clinical trials have suggested that Limbrel shows efficacy in OA (e.g.( Reference Levy, Khokhlov and Kopenkin 80 )), but recently severe liver toxicity has been described in some patients( Reference Chalasani, Vuppalanchi and Navarro 81 ).

A grape seed proanthocyanidin extract is protective in the monosodium iodoacetate model of OA in the rat, showing chondroprotection and decreased pain( Reference Woo, Joo and Jung 82 ). Specifically, procyanidin B3 abrogates cartilage destruction and heterotopic cartilage formation in a surgical model of OA in the mouse( Reference Aini, Ochi and Iwata 83 ). It was shown to block IL-1 repression of matrix gene expression in vitro and also decrease inducible nitric oxide synthase in vitro and in vivo ( Reference Aini, Ochi and Iwata 83 ).

Another mixture not derived from the diet, Pycnogenol is a pine bark extract rich in procyanidins( Reference D'Andrea 84 ). It has been reported to inhibit NF-κB activation and the activity of some MMP( Reference Grimm, Chovanova and Muchova 85 , Reference Grimm, Schafer and Hogger 86 ). Three small clinical trials have been performed in OA with positive outcomes reported (e.g.( Reference Belcaro, Cesarone and Errichi 87 , Reference Cisar, Jany and Waczulikova 88 )). However, a Cochrane review of Pycnogenol in chronic diseases (including OA) stated that it was not possible to reach definite conclusions on either efficacy or safety of Pycnogenol( Reference Schoonees, Visser and Musekiwa 89 ).

Anthocyanins

Anthocyanins are responsible for the red/blue pigmentation in fruits and vegetables( Reference Manach, Scalbert and Morand 64 ). To date most studies have been performed using fruit juices or extracts that are rich in anthocyanins. A recent clinical trial examined tart cherry juice in patients with knee OA( Reference Schumacher, Pullman-Mooar and Gupta 90 ). No difference in disease scores compared with placebo was uncovered, but high-sensitivity C-reactive protein was significantly lowered and this was associated with decreased score( Reference Schumacher, Pullman-Mooar and Gupta 90 ). Pomegranate juice or extracts, which have been reported to contain anthocyanins and many other flavonoids including flavanols, have been shown to inhibit IL-1-induced MMP expression in chondrocytes via inhibition of MAP kinases and NF-κB( Reference Ahmed, Wang and Hafeez 91 Reference Jean-Gilles, Li and Vaidyanathan 93 ). Such extracts also show efficacy in the monosodium iodoacetate model of OA in mice( Reference Hadipour-Jahromy and Mozaffari-Kermani 94 ). Raspberry extract( Reference Jean-Gilles, Li and Ma 95 ) and red orange extract( Reference Frasca, Panico and Bonina 96 ) have also been reported to have some efficacy in vitro and in vivo.

Isoflavones

Isoflavones are diphenolic compounds with structural similarity to oestrogens, and are consequently referred to as phytoestrogens. They are found mainly in legumes and soya is a major source of isoflavones in the diet( Reference Manach, Scalbert and Morand 64 ). Data in chondrocytes show that one isoflavone, genistein, reduces the production of inflammatory molecules such as COX2 and nitric oxide( Reference Hooshmand, Soung do and Lucas 97 ). Extracellular matrix synthesis in cartilage may increase or decrease, potentially with increasing dose( Reference Claassen, Briese and Manapov 98 , Reference Yu, Xing and Dong 99 ). In the rat inflammatory collagen-induced arthritis model, soya protein appears to be protective( Reference Mohammad Shahi, Rashidi and Mahboob 100 ), however, no significant effect of soya intake was measurable on OA severity in Cynomolgus monkeys( Reference Ham, Loeser and Lindgren 101 ). One human study suggested beneficial effects of soya protein supplementation on function, symptoms and biochemical markers of OA, particularly in men( Reference Arjmandi, Khalil and Lucas 102 ).

Flavanones

Flavanones are present in the diet at high concentrations only in citrus fruits including naringenin from grapefruit, hesperetin from oranges and eriodictyol from lemons( Reference Manach, Scalbert and Morand 64 ). No effect was seen for naringenin on IL-1-induced MMP13 production in SW1353 chondrosarcoma cells( Reference Lim, Park and Kim 65 ). However, hesperetin, its glycoside hesperidin or its derivatives, show efficacy in inflammatory models of arthritis( Reference Choi and Lee 103 Reference Umar, Kumar and Sajad 105 ). Red orange juice extract showed repression of inflammatory molecules in chondrocytes as mentioned earlier( Reference Frasca, Panico and Bonina 96 ).

Carotenoids

β-Carotene is the most widely known carotenoid and is a precursor to vitamin A( Reference Maiani, Caston and Catasta 106 ). Vitamin A and its derivatives, retinoids, are known to have profound effects on cartilage and the skeleton and may contribute to OA( Reference Davies, Ribeiro and Downey-Jones 107 ). The Framingham study identified a weak protective association between intake of β-carotene and the progression of radiographic knee OA( Reference McAlindon, Jacques and Zhang 14 ). A case–control study in the Johnston Couny Osteoarthritis Project examined the association between serum levels of several carotenoids (lutein, zeaxanthin, β-cryptoxanthin, lycopene, α-carotene and β-carotene) and OA( Reference De Roos, Arab and Renner 108 ). People with high levels of lutein or β-cryptoxanthin were less likely to have knee OA, while those with high levels of trans-β-carotene or zeaxanthin were more likely to have knee OA. Similarly, a cross-sectional study in a Japanese population with radiographic knee OA examined the association between serum levels of several carotenoids (lutein, zeaxanthin, cantaxanthin, cryptoxanthin, lycopene, α-carotene and β-carotene) and OA, but found nothing significant( Reference Seki, Hasegawa and Yamaguchi 109 ). It is worth noting that there is evidence that β-cryptoxanthin is associated with a decreased risk of inflammatory arthritis (e.g.( Reference Pattison, Symmons and Lunt 110 )). In healthy, middle-aged people, lutein and zeaxanthin intake were associated with decreased risk of cartilage defects on MRI and β-cryptoxanthin intake was inversely associated with tibial plateau bone area( Reference Wang, Hodge and Wluka 17 ).

Plant sterols

As discussed earlier, there is a positive association between serum cholesterol and OA, with statin use appearing to show efficacy in disease incidence and/or progression. Intake of plant phytosterols/stanols significantly reduce LDL cholesterol and total cholesterol in intervention trials( Reference Kamal-Eldin and Moazzami 111 , Reference Wu, Fu and Yang 112 ) and of the three phytosterols tested, (stigmasterol, sitosterol and campesterol), stigmasterol bound best to chondrocyte membranes( Reference Gabay, Sanchez and Salvat 113 ). It inhibited IL-1-induced MMP and ADAMTS4 expression, although had no effect on ADAMTS5, potentially via its ability to inhibit NF-κB activation( Reference Gabay, Sanchez and Salvat 113 ). Intra-articular injection of stigmasterol was shown to suppress MMP expression and reduce cartilage degradation in a rabbit anterior cruciate ligament transection model of OA( Reference Chen, Yu and Hu 114 ).

Glucosinolates

Glucosinolates are found in cruciferous vegetables and are the precursors of isothiocyanates. Broccoli is rich in glucoraphanin, and when the vegetable is chopped or chewed, it is exposed to the action of an enzyme myrosinase to yield sulforaphane, the isothiocyanate. In chondrocytes, sulforaphane was initially shown to decrease shear stress-induced apoptosis( Reference Healy, Lee and Gao 115 ). More recently, it has been shown to exhibit pro-survival and anti-apoptotic activities when cell death is induced by a variety of stimuli( Reference Facchini, Stanic and Cetrullo 116 ). Sulforaphane has been shown to block IL-1 and TNFα induction of MMP1 and MMP13 expression, as well as PGE2 and nitric oxide in chondrocytes( Reference Kim, Yeo and Kim 117 ) and inhibit cartilage degradation in vitro ( Reference Kim, Yeo and Jung 118 ). Later work showed that it was effective in inhibiting expression of ADAMTS4 and ADAMTS5, and abrogating cartilage destruction in the ‘destabilisation of the medial meniscus’ model of OA in the mouse, acting as a direct inhibitor of NF-κB( Reference Davidson, Jupp and de Ferrars 119 ).

Resveratrol

Resveratrol is a plant-derived phenol of the stilbenoid class, found at high concentrations in the skin of red grapes and in red wine. It has come to the fore as an activator of the histone deacetylase Sirt1, which has important roles in cell survival and as a mimic of caloric restriction that extends lifespan in many models( Reference Lam, Peterson and Ravussin 120 ). Sirt1 is intimately involved in OA with deletion of Sirt1 in mice causing more rapid development of OA in a post-surgical model( Reference Matsuzaki, Matsushita and Takayama 121 ). Resveratrol decreases OA score when directly injected intraarticularly in the rabbit anterior cruciate ligament transection model of OA( Reference Elmali, Esenkaya and Harma 122 , Reference Wang, Gao and Chen 123 ). It is an NF-κB inhibitor in chondrocytes and blocks inflammation and apoptosis( Reference Csaki, Keshishzadeh and Fischer 124 Reference Shakibaei, Csaki and Nebrich 126 ). It has also been shown to decrease proteolysis (e.g. MMP and ADAMTS) and enhance extracellular matrix synthesis( Reference Im, Li and Chen 127 ).

Interestingly, resveratrol has been shown to display synergistic effects on chondrocyte phenotype and apoptosis with curcumin (see later)( Reference Csaki, Mobasheri and Shakibaei 128 , Reference Shakibaei, Mobasheri and Buhrmann 129 ). These compounds both inhibit NF-κB, but are known to act via different mechanisms.

Curcumin

Curcumin is the major curcuminoid found in the spice, turmeric. It has been shown to be an NF-κB inhibitor( Reference Singh and Aggarwal 130 ), and used in chondrocytes as an inhibitor of oncostatin M-, IL-1- and TNFα-induced signalling( Reference Li, Dehnade and Zafarullah 131 Reference Liacini, Sylvester and Li 133 ). Here it was shown to inhibit c-Jun N-terminal kinase, activator protein 1, STAT and mitogen-activated protein kinase signalling, to inhibit expression of key MMP in cartilage and proposed to have potential clinical utility. Innes et al. used a turmeric extract in a clinical trial of OA in the dog, with clinical assessments showing significant improvement( Reference Innes, Fuller and Grover 134 ). The anti-catabolic effects of curcumin in human articular chondrocytes were confirmed( Reference Schulze-Tanzil, Mobasheri and Sendzik 135 ) and its impact extended to include anti-apoptotic activity( Reference Shakibaei, Schulze-Tanzil and John 136 ), pro-anabolic effects on matrix expression( Reference Lay, Samiric and Handley 66 , Reference Shakibaei, Schulze-Tanzil and John 136 ), inhibition of COX2 expression and other inflammatory mediators( Reference Mathy-Hartert, Jacquemond-Collet and Priem 137 , Reference Shakibaei, John and Schulze-Tanzil 138 ). Efficacy was also shown in cartilage explants( Reference Lay, Samiric and Handley 66 , Reference Clutterbuck, Mobasheri and Shakibaei 139 ) and murine models of inflammatory arthritis( Reference Mun, Kim and Kim 140 ), although not yet OA. Curcumin itself has poor solubility and bioavailability( Reference Henrotin, Clutterbuck and Allaway 141 ), but a curcumin–phophatidylcholine complex (Meriva), designed to overcome this, has shown some efficacy in small-scale clinical trials( Reference Belcaro, Cesarone and Dugall 142 , Reference Belcaro, Cesarone and Dugall 143 ). As discussed earlier, a thorough understanding of mechanism of action has led to experiments showing synergy between curcumin and resveratrol( Reference Csaki, Mobasheri and Shakibaei 128 , Reference Shakibaei, Mobasheri and Buhrmann 129 ).

Avocado-soyabean unsaponifiables

While not truly dietary-derived, avocado-soyabean unsaponifiables (ASU), Piascledine, has been developed by Laboratoire Expanscience and is the unsaponifiable fraction of one-third avocado oil and two-thirds soyabean oil. It is a mixture of tocopherols, plant sterols and other molecules( Reference Msika, Baudouin and Saunois 144 ). A recent moderate-sized trial of Piascledine in hip OA (the ERADIAS study) over 3 years showed that while there was no significant difference in mean joint space width loss between treatment and placebo, there were significantly less progressors in the treatment group. There was no difference in clinical outcomes including pain or analgesic/non-steroidal anti-inflammatory drug use( Reference Maheu, Cadet and Marty 145 ). This was somewhat similar to an earlier smaller study examining structural modification( Reference Lequesne, Maheu and Cadet 146 ), but very different from other earlier trials, where ASU demonstrated reductions in pain, functional disability or non-steroidal anti-inflammatory drug use in patients with hip or knee OA over 3–6 months( Reference Appelboom, Schuermans and Verbruggen 147 Reference Maheu, Mazieres and Valat 149 ). In a dog anterior cruciate ligament transection model of OA, ASU reduced disease severity and decreased MMP13 production( Reference Boileau, Martel-Pelletier and Caron 150 ), although in an ovine model of post-meniscectomy OA, ASU was described to have a ‘subtle, but statistically significant’ effect on cartilage( Reference Cake, Read and Guillou 151 ). In vitro data showed that ASU exhibit anti-catabolic (MMP expression), anti-inflammatory (PGE2, nitric oxide, COX2) and pro-anabolic (type II collagen and aggrecan synthesis) in chondrocytes. It has also been shown to inhibit NF-κB activity( Reference Gabay, Gosset and Levy 152 Reference Henrotin, Sanchez and Deberg 154 ). It should also be pointed out that other formulations of ASU exist and one from Nutramax has been shown to have similar in vitro activity in chondrocytes( Reference Au, Al-Talib and Au 155 ). Data from equine chondrocytes suggest that this ASU can act synergistically with epigallocatechin gallate( Reference Heinecke, Grzanna and Au 156 ). The relative merits of each preparation have been the subject of debate( Reference Msika, Baudouin and Saunois 144 , Reference Frondoza 157 , Reference Henrotin 158 ).

Ginger

There have been several small clinical trials exploring the efficacy of ginger extract in the treatment of OA. Trials using Zingiber officinale extract showed variable outcome and a review found that evidence for its efficacy in OA was weak( Reference Leach and Kumar 159 ). A mixture of extracts from Z. officinale and Alpinia galangal used in a short (6-week) study showed a significant effect in reducing clinical symptoms( Reference Altman and Marcussen 160 ). In vitro research suggests that ginger extract can decrease production of inflammatory mediators from chondrocytes( Reference Shen, Hong and Kim 161 ) and synoviocytes( Reference Ribel-Madsen, Bartels and Stockmarr 162 ).

Sulphur-containing compounds

A cross-sectional study in twins demonstrated that consumption of both allium vegetables and also non-citrus fruits showed a protective association with hip OA( Reference Williams, Skinner and Spector 163 ). Furthermore, diallyl disulphide, a compound from garlic, was shown to inhibit IL-1-induced MMP1, MMP3 and MMP13 expression( Reference Williams, Skinner and Spector 163 ). Diallyl sulphide has also been shown to block expression of these enzymes and ameliorate cartilage destruction when administered intraarticularly in the rabbit anterior cruciate ligament transection model of OA( Reference Chen, Tang and Bao 164 ).

Others

Interestingly, data on the progression of knee OA, coming from the Osteoarthritis Initiative showed that frequent soft drink consumption is associated with increased disease progression in men, independent of obesity( Reference Lu, Ahmad and Zhang 165 ). This obviously requires replication. An extract of edible bird's nest (which is made from swiftlet saliva), has been shown to have anti-catabolic, anti-inflammatory and pro-anabolic activity on human osteoarthritic chondrocytes( Reference Chua, Lee and Nagandran 166 ). Sesamin, a lignan from sesame seeds has been reported to be chondroprotective in an explant assay, decreasing MMP expression and activation( Reference Phitak, Pothacharoen and Settakorn 167 ). An extract of a variety of mint which overexpressed rosmarinic acid inhibits lipopolysaccharide-induced glycosaminoglycan release and inflammatory mediators from porcine cartilage explants( Reference Pearson, Fletcher and Kott 168 ).

Conclusions

There are many compounds present in the habitual diet, which have been shown to have activity in both laboratory models of OA and/or human disease. Where examined, many of these compounds appear to be inhibitors of the NF-κB pathway. This signalling pathway has been shown to play a role in the development and progression of OA( Reference Marcu, Otero and Olivotto 169 ). Two studies suggest that using a combination of compounds, which inhibit the NF-κB pathway via different mechanisms gives a synergistic response( Reference Csaki, Mobasheri and Shakibaei 128 , Reference Shakibaei, Mobasheri and Buhrmann 129 ). It would thus be important to understand the mode of NF-κB inhibition for all compounds with this activity. In order to achieve synergy, it will also be important to discover compounds which do not act via this mechanism. Since habitual dietary intakes vary widely, an understanding of food combinations, which protect the joint, may be key and this may also be a means to develop specific food products or offer targeted advice to reduce risk.

Basic science provides information on the mechanisms of cartilage protection in healthy tissue and the prevention of cartilage destruction in disease. The design of randomised clinical trials in the longer term needs to include ‘at risk’ populations (in which incidence of OA can be used as an outcome measure), as well as patients with existing OA. This is in line with the current European Food Standards Agency recommendations that the design of human trials must demonstrate a preventative effect on the healthy joint, separately from an impact on established OA per se to establish claims in both areas.

In summary, diet offers a route by which the health of the joint can be protected and OA incidence or progression decreased. In a chronic disease, with risk factors increasing in the population and with no pharmaceutical cure, an understanding of this will be crucial.

Acknowledgements

We would like to thank all members of the Clark laboratory present and past and our collaborators in research related to this review.

Financial Support

Research in this area in Clark laboratory is funded by the BBSRC Diet and Health Research Industry Club grant BB/I006060/1 and PhD studentships BB/J500112/1, Arthritis Research UK grant 19371, Orthopaedic Research UK grant 487 and previously Dunhill Medical Trust grant R73/0208. These funders had no role in the design, analysis or writing of this article.

Conflicts of Interest

None.

Authorship

All authors have contributed to writing and/or critically reviewing and editing the manuscript.

Footnotes

These authors contributed equally to this review.

References

1. Pereira, D, Peleteiro, B, Araujo, J et al. (2011) The effect of osteoarthritis definition on prevalence and incidence estimates: a systematic review. Osteoarthritis Cartilage 19, 12701285.CrossRefGoogle ScholarPubMed
4. Shane Anderson, A & Loeser, RF (2010) Why is osteoarthritis an age-related disease? Best Pract Res Clin Rheumatol 24, 1526.CrossRefGoogle ScholarPubMed
5. Richmond, SA, Fukuchi, RK, Ezzat, A et al. (2013) Are joint injury, sport activity, physical activity, obesity, or occupational activities predictors for osteoarthritis? A systematic review. J Orthop Sports Phys Ther 43, 515524.CrossRefGoogle ScholarPubMed
6. Chen, A, Gupte, C, Akhtar, K et al. (2012) The global economic cost of osteoarthritis: how the UK compares. Arthritis 2012, 698709.CrossRefGoogle ScholarPubMed
7. Kraus, VB (2012) Patient evaluation and OA study design: OARSI/Biomarker qualification. HSS J 8, 6465.CrossRefGoogle ScholarPubMed
8. Jordan, JM, Sowers, MF, Messier, SP et al. (2011) Methodologic issues in clinical trials for prevention or risk reduction in osteoarthritis. Osteoarthritis Cartilage 19, 500508.CrossRefGoogle ScholarPubMed
9. Black, C, Clar, C, Henderson, R et al. (2009) The clinical effectiveness of glucosamine and chondroitin supplements in slowing or arresting progression of osteoarthritis of the knee: a systematic review and economic evaluation. Health Technol Assess 13, 1148.CrossRefGoogle ScholarPubMed
10. Hochberg, M, Chevalier, X, Henrotin, Y et al. (2013) Symptom and structure modification in osteoarthritis with pharmaceutical-grade chondroitin sulfate: what's the evidence? Curr Med Res Opin 29, 259267.CrossRefGoogle ScholarPubMed
11. Reginster, JY, Neuprez, A, Lecart, MP et al. (2012) Role of glucosamine in the treatment for osteoarthritis. Rheumatol Int 32, 29592967.CrossRefGoogle ScholarPubMed
12. Wildi, LM, Martel-Pelletier, J, Abram, F et al. (2013) Assessment of cartilage changes over time in knee osteoarthritis disease-modifying osteoarthritis drug trials using semiquantitative and quantitative methods: pros and cons. Arthritis Care Res (Hoboken) 65, 686694.CrossRefGoogle ScholarPubMed
13. Wu, D, Huang, Y, Gu, Y et al. (2013) Efficacies of different preparations of glucosamine for the treatment of osteoarthritis: a meta-analysis of randomised, double-blind, placebo-controlled trials. Int J Clin Pract 67, 585594.CrossRefGoogle ScholarPubMed
14. McAlindon, TE, Jacques, P, Zhang, Y et al. (1996) Do antioxidant micronutrients protect against the development and progression of knee osteoarthritis? Arthritis Rheum 39, 648656.CrossRefGoogle ScholarPubMed
15. McAlindon, TE (2006) Nutraceuticals: do they work and when should we use them? Best Pract Res Clin Rheumatol 20, 99115.CrossRefGoogle Scholar
16. Peregoy, J & Wilder, FV (2011) The effects of vitamin C supplementation on incident and progressive knee osteoarthritis: a longitudinal study. Public Health Nutr 14, 709715.CrossRefGoogle ScholarPubMed
17. Wang, Y, Hodge, AM, Wluka, AE et al. (2007) Effect of antioxidants on knee cartilage and bone in healthy, middle-aged subjects: a cross-sectional study. Arthritis Res Ther 9, R66.CrossRefGoogle ScholarPubMed
18. Clark, AG, Rohrbaugh, AL, Otterness, I et al. (2002) The effects of ascorbic acid on cartilage metabolism in guinea pig articular cartilage explants. Matrix Biol 21, 175184.CrossRefGoogle ScholarPubMed
19. Schwartz, ER, Oh, WH & Leveille, CR (1981) Experimentally induced osteoarthritis in guinea pigs: metabolic responses in articular cartilage to developing pathology. Arthritis Rheum 24, 13451355.CrossRefGoogle ScholarPubMed
20. Meacock, SC, Bodmer, JL & Billingham, ME (1990) Experimental osteoarthritis in guinea-pigs. J Exp Pathol (Oxford) 71, 279293.Google ScholarPubMed
21. Kraus, VB, Huebner, JL, Stabler, T et al. (2004) Ascorbic acid increases the severity of spontaneous knee osteoarthritis in a guinea pig model. Arthritis Rheum 50, 18221831.CrossRefGoogle Scholar
22. Jordan, JM, De Roos, AJ, Renner, JB et al. (2004) A case-control study of serum tocopherol levels and the alpha- to gamma-tocopherol ratio in radiographic knee osteoarthritis: the Johnston County Osteoarthritis Project. Am J Epidemiol 159, 968977.CrossRefGoogle ScholarPubMed
23. Canter, PH, Wider, B & Ernst, E (2007) The antioxidant vitamins A, C, E and selenium in the treatment of arthritis: a systematic review of randomized clinical trials. Rheumatology (Oxford) 46, 12231233.CrossRefGoogle Scholar
24. Bhatti, FU, Mehmood, A, Wajid, N et al. (2013) Vitamin E protects chondrocytes against hydrogen peroxide-induced oxidative stress in vitro . Inflamm Res 62, 781789.CrossRefGoogle ScholarPubMed
25. Wolff, AE, Jones, AN & Hansen, KE (2008) Vitamin D and musculoskeletal health. Nat Clin Pract Rheumatol 4, 580588.CrossRefGoogle ScholarPubMed
26. Cao, Y, Winzenberg, T, Nguo, K et al. (2013) Association between serum levels of 25-hydroxyvitamin D and osteoarthritis: a systematic review. Rheumatology (Oxford) 52, 13231334.CrossRefGoogle ScholarPubMed
27. Laslett, LL, Quinn, S, Burgess, JR et al. (2013) Moderate vitamin D deficiency is associated with changes in knee and hip pain in older adults: a 5-year longitudinal study. Ann Rheum Dis (In the Press).Google ScholarPubMed
28. McAlindon, T, LaValley, M, Schneider, E et al. (2013) Effect of vitamin D supplementation on progression of knee pain and cartilage volume loss in patients with symptomatic osteoarthritis: a randomized controlled trial. JAMA 309, 155162.CrossRefGoogle ScholarPubMed
29. Cao, Y, Jones, G, Cicuttini, F et al. (2012) Vitamin D supplementation in the management of knee osteoarthritis: study protocol for a randomized controlled trial. Trials 13, 131.CrossRefGoogle ScholarPubMed
30. Castillo, EC, Hernandez-Cueto, MA, Vega-Lopez, MA et al. (2012) Effects of vitamin D supplementation during the induction and progression of osteoarthritis in a rat model. Evid Based Complement Alternat Med 2012, 156563.CrossRefGoogle ScholarPubMed
31. Zwerina, K, Baum, W, Axmann, R et al. (2011) Vitamin D receptor regulates TNF-mediated arthritis. Ann Rheum Dis 70, 11221129.CrossRefGoogle ScholarPubMed
32. Oka, H, Akune, T, Muraki, S et al. (2009) Association of low dietary vitamin K intake with radiographic knee osteoarthritis in the Japanese elderly population: dietary survey in a population-based cohort of the ROAD study. J Orthop Sci 14, 687692.CrossRefGoogle Scholar
33. Misra, D, Booth, SL, Tolstykh, I et al. (2013) Vitamin K deficiency is associated with incident knee osteoarthritis. Am J Med 126, 243248.CrossRefGoogle ScholarPubMed
34. Neogi, T, Booth, SL, Zhang, YQ et al. (2006) Low vitamin K status is associated with osteoarthritis in the hand and knee. Arthritis Rheum 54, 12551261.CrossRefGoogle ScholarPubMed
35. Muraki, S, Akune, T, En-Yo, Y et al. (2013) Association of dietary intake with joint space narrowing and osteophytosis at the knee in Japanese men and women: the ROAD study. Mod Rheumatol (In the Press).CrossRefGoogle ScholarPubMed
36. Krueger, T, Westenfeld, R, Schurgers, L et al. (2009) Coagulation meets calcification: the vitamin K system. Int J Artif Organs 32, 6774.CrossRefGoogle ScholarPubMed
37. Misra, D, Booth, SL, Crosier, MD et al. (2011) Matrix Gla protein polymorphism, but not concentrations, is associated with radiographic hand osteoarthritis. J Rheumatol 38, 19601965.CrossRefGoogle Scholar
38. Naito, K, Watari, T, Obayashi, O et al. (2012) Relationship between serum undercarboxylated osteocalcin and hyaluronan levels in patients with bilateral knee osteoarthritis. Int J Mol Med 29, 756760.Google ScholarPubMed
39. Jonas, WB, Rapoza, CP & Blair, WF (1996) The effect of niacinamide on osteoarthritis: a pilot study. Inflamm Res 45, 330334.CrossRefGoogle ScholarPubMed
40. Qin, B, Shi, X, Samai, PS et al. (2012) Association of dietary magnesium intake with radiographic knee osteoarthritis: results from a population-based study. Arthritis Care Res (Hoboken) 64, 13061311.CrossRefGoogle ScholarPubMed
41. Hunter, DJ, Hart, D, Snieder, H et al. (2003) Evidence of altered bone turnover, vitamin D and calcium regulation with knee osteoarthritis in female twins. Rheumatology (Oxford) 42, 13111316.CrossRefGoogle ScholarPubMed
42. Zou, K, Liu, G, Wu, T et al. (2009) Selenium for preventing Kashin-Beck osteoarthropathy in children: a meta-analysis. Osteoarthritis Cartilage 17, 144151.CrossRefGoogle ScholarPubMed
43. Zhuo, Q, Yang, W, Chen, J et al. (2012) Metabolic syndrome meets osteoarthritis. Nat Rev Rheumatol 8, 729737.CrossRefGoogle ScholarPubMed
44. Gkretsi, V, Simopoulou, T & Tsezou, A (2011) Lipid metabolism and osteoarthritis: lessons from atherosclerosis. Prog Lipid Res 50, 133140.CrossRefGoogle ScholarPubMed
45. Clockaerts, S, Van Osch, GJ, Bastiaansen-Jenniskens, YM et al. (2012) Statin use is associated with reduced incidence and progression of knee osteoarthritis in the Rotterdam study. Ann Rheum Dis 71, 642647.CrossRefGoogle ScholarPubMed
46. Kadam, UT, Blagojevic, M & Belcher, J (2013) Statin use and clinical osteoarthritis in the general population: a longitudinal study. J Gen Intern Med 28, 943949.CrossRefGoogle ScholarPubMed
47. Riddle, DL, Moxley, G & Dumenci, L (2013) Associations between statin use and changes in pain, function and structural progression: a longitudinal study of persons with knee osteoarthritis. Ann Rheum Dis 72, 196203.CrossRefGoogle ScholarPubMed
48. Lippiello, L, Walsh, T & Fienhold, M (1991) The association of lipid abnormalities with tissue pathology in human osteoarthritic articular cartilage. Metabolism 40, 571576.CrossRefGoogle ScholarPubMed
49. Plumb, MS & Aspden, RM (2004) High levels of fat and (n-6) fatty acids in cancellous bone in osteoarthritis. Lipids Health Dis 3, 12.CrossRefGoogle ScholarPubMed
50. Baker, KR, Matthan, NR, Lichtenstein, AH et al. (2012) Association of plasma n-6 and n-3 polyunsaturated fatty acids with synovitis in the knee: the MOST study. Osteoarthritis Cartilage 20, 382387.CrossRefGoogle ScholarPubMed
51. Wang, Y, Davies-Tuck, ML, Wluka, AE et al. (2009) Dietary fatty acid intake affects the risk of developing bone marrow lesions in healthy middle-aged adults without clinical knee osteoarthritis: a prospective cohort study. Arthritis Res Ther 11, R63.CrossRefGoogle ScholarPubMed
52. Wang, Y, Wluka, AE, Hodge, AM et al. (2008) Effect of fatty acids on bone marrow lesions and knee cartilage in healthy, middle-aged subjects without clinical knee osteoarthritis. Osteoarthritis Cartilage 16, 579583.CrossRefGoogle Scholar
53. Mooney, RA, Sampson, ER, Lerea, J et al. (2011) High-fat diet accelerates progression of osteoarthritis after meniscal/ligamentous injury. Arthritis Res Ther 13, R198.CrossRefGoogle ScholarPubMed
54. Knott, L, Avery, NC, Hollander, AP et al. (2011) Regulation of osteoarthritis by omega-3 (n-3) polyunsaturated fatty acids in a naturally occurring model of disease. Osteoarthritis Cartilage 19, 11501157.CrossRefGoogle Scholar
55. Hurst, S, Rees, SG, Randerson, PF et al. (2009) Contrasting effects of n-3 and n-6 fatty acids on cyclooxygenase-2 in model systems for arthritis. Lipids 44, 889896.CrossRefGoogle ScholarPubMed
56. Zainal, Z, Longman, AJ, Hurst, S et al. (2009) Relative efficacies of omega-3 polyunsaturated fatty acids in reducing expression of key proteins in a model system for studying osteoarthritis. Osteoarthritis Cartilage 17, 896905.CrossRefGoogle Scholar
57. Hielm-Bjorkman, A, Roine, J, Elo, K et al. (2012) An un-commissioned randomized, placebo-controlled double-blind study to test the effect of deep sea fish oil as a pain reliever for dogs suffering from canine OA. BMC Vet Res 8, 157.CrossRefGoogle ScholarPubMed
58. Roush, JK, Dodd, CE, Fritsch, DA et al. (2010) Multicenter veterinary practice assessment of the effects of omega-3 fatty acids on osteoarthritis in dogs. J Am Vet Med Assoc 236, 5966.CrossRefGoogle ScholarPubMed
59. Jacquet, A, Girodet, PO, Pariente, A et al. (2009) Phytalgic, a food supplement, vs placebo in patients with osteoarthritis of the knee or hip: a randomised double-blind placebo-controlled clinical trial. Arthritis Res Ther 11, R192.CrossRefGoogle ScholarPubMed
60. Christensen, R & Bliddal, H (2010) Is Phytalgic(R) a goldmine for osteoarthritis patients or is there something fishy about this nutraceutical? A summary of findings and risk-of-bias assessment. Arthritis Res Ther 12, 105.CrossRefGoogle ScholarPubMed
61. Ameye, LG & Chee, WS (2006) Osteoarthritis and nutrition. From nutraceuticals to functional foods: a systematic review of the scientific evidence. Arthritis Res Ther 8, R127.CrossRefGoogle ScholarPubMed
62. Denny, A & Buttriss, J (2007) Plant food and health: focus on plant bioactives. Synthesis Report No 4. http://www.ipfn.ie/download/pdf/eurofir_report_plant_bioactives.pdf Google Scholar
63. Falcone Ferreyra, ML, Rius, SP, Casati, P et al. (2012) Flavonoids: biosynthesis, biological functions, and biotechnological applications. Front Plant Sci 3, 222.CrossRefGoogle ScholarPubMed
64. Manach, C, Scalbert, A, Morand, C et al. (2004) Polyphenols: food sources and bioavailability. Am J Clin Nutr 79, 727747.CrossRefGoogle ScholarPubMed
65. Lim, H, Park, H & Kim, HP (2011) Effects of flavonoids on matrix metalloproteinase-13 expression of interleukin-1beta-treated articular chondrocytes and their cellular mechanisms: inhibition of c-Fos/AP-1 and JAK/STAT signaling pathways. J Pharmacol Sci 116, 221231.CrossRefGoogle ScholarPubMed
66. Lay, E, Samiric, T, Handley, CJ et al. (2012) Short- and long-term exposure of articular cartilage to curcumin or quercetin inhibits aggrecan loss. J Nutr Biochem 23, 106112.CrossRefGoogle ScholarPubMed
67. Lee, YS & Choi, EM (2010) Myricetin inhibits IL-1beta-induced inflammatory mediators in SW982 human synovial sarcoma cells. Int Immunopharmacol 10, 812814.CrossRefGoogle ScholarPubMed
68. Imada, K, Lin, N, Liu, C et al. (2008) Nobiletin, a citrus polymethoxy flavonoid, suppresses gene expression and production of aggrecanases-1 and -2 in collagen-induced arthritic mice. Biochem Biophys Res Commun 373, 181185.CrossRefGoogle ScholarPubMed
69. Moncada-Pazos, A, Obaya, AJ, Viloria, CG et al. (2011) The nutraceutical flavonoid luteolin inhibits ADAMTS-4 and ADAMTS-5 aggrecanase activities. J Mol Med (Berl) 89, 611619.CrossRefGoogle ScholarPubMed
70. Lopez-Lazaro, M (2009) Distribution and biological activities of the flavonoid luteolin. Mini Rev Med Chem 9, 3159.CrossRefGoogle ScholarPubMed
71. Ishiwa, J, Sato, T, Mimaki, Y et al. (2000) A citrus flavonoid, nobiletin, suppresses production and gene expression of matrix metalloproteinase 9/gelatinase B in rabbit synovial fibroblasts. J Rheumatol 27, 2025.Google ScholarPubMed
72. Durigova, M, Roughley, PJ & Mort, JS (2008) Mechanism of proteoglycan aggregate degradation in cartilage stimulated with oncostatin M. Osteoarthritis Cartilage 16, 98104.CrossRefGoogle ScholarPubMed
73. Haqqi, TM, Anthony, DD, Gupta, S et al. (1999) Prevention of collagen-induced arthritis in mice by a polyphenolic fraction from green tea. Proc Natl Acad Sci USA 96, 45244529.CrossRefGoogle ScholarPubMed
74. Adcocks, C, Collin, P & Buttle, DJ (2002) Catechins from green tea (Camellia sinensis) inhibit bovine and human cartilage proteoglycan and type II collagen degradation in vitro . J Nutr 132, 341346.CrossRefGoogle ScholarPubMed
75. Vankemmelbeke, MN, Jones, GC, Fowles, C et al. (2003) Selective inhibition of ADAMTS-1, -4 and -5 by catechin gallate esters. Eur J Biochem 270, 23942403.CrossRefGoogle ScholarPubMed
76. Akhtar, N & Haqqi, TM (2011) Epigallocatechin-3-gallate suppresses the global interleukin-1beta-induced inflammatory response in human chondrocytes. Arthritis Res Ther 13, R93.CrossRefGoogle ScholarPubMed
77. Ahmed, S (2010) Green tea polyphenol epigallocatechin 3-gallate in arthritis: progress and promise. Arthritis Res Ther 12, 208.CrossRefGoogle ScholarPubMed
78. Burnett, BP, Jia, Q, Zhao, Y et al. (2007) A medicinal extract of Scutellaria baicalensis and Acacia catechu acts as a dual inhibitor of cyclooxygenase and 5-lipoxygenase to reduce inflammation. J Med Food 10, 442451.CrossRefGoogle ScholarPubMed
79. Shen, D, Wu, Q, Wang, M et al. (2006) Determination of the predominant catechins in Acacia catechu by liquid chromatography/electrospray ionization-mass spectrometry. J Agric Food Chem 54, 32193224.CrossRefGoogle ScholarPubMed
80. Levy, RM, Khokhlov, A, Kopenkin, S et al. (2010) Efficacy and safety of flavocoxid, a novel therapeutic, compared with naproxen: a randomized multicenter controlled trial in subjects with osteoarthritis of the knee. Adv Ther 27, 731742.CrossRefGoogle ScholarPubMed
81. Chalasani, N, Vuppalanchi, R, Navarro, V et al. (2012) Acute liver injury due to flavocoxid (Limbrel), a medical food for osteoarthritis: a case series. Ann Intern Med 156, 857860, W297–W300.CrossRefGoogle ScholarPubMed
82. Woo, YJ, Joo, YB, Jung, YO et al. (2011) Grape seed proanthocyanidin extract ameliorates monosodium iodoacetate-induced osteoarthritis. Exp Mol Med 43, 561570.CrossRefGoogle ScholarPubMed
83. Aini, H, Ochi, H, Iwata, M et al. (2012) Procyanidin B3 prevents articular cartilage degeneration and heterotopic cartilage formation in a mouse surgical osteoarthritis model. PLoS ONE 7, e37728.CrossRefGoogle Scholar
84. D'Andrea, G (2010) Pycnogenol: a blend of procyanidins with multifaceted therapeutic applications? Fitoterapia 81, 724736.CrossRefGoogle ScholarPubMed
85. Grimm, T, Chovanova, Z, Muchova, J et al. (2006) Inhibition of NF-kappaB activation and MMP-9 secretion by plasma of human volunteers after ingestion of maritime pine bark extract (Pycnogenol). J Inflamm (Lond) 3, 1.CrossRefGoogle ScholarPubMed
86. Grimm, T, Schafer, A & Hogger, P (2004) Antioxidant activity and inhibition of matrix metalloproteinases by metabolites of maritime pine bark extract (pycnogenol). Free Radic Biol Med 36, 811822.CrossRefGoogle ScholarPubMed
87. Belcaro, G, Cesarone, MR, Errichi, S et al. (2008) Treatment of osteoarthritis with Pycnogenol. The SVOS (San Valentino Osteo-arthrosis Study). Evaluation of signs, symptoms, physical performance and vascular aspects. Phytother Res 22, 518523.CrossRefGoogle ScholarPubMed
88. Cisar, P, Jany, R, Waczulikova, I et al. (2008) Effect of pine bark extract (Pycnogenol) on symptoms of knee osteoarthritis. Phytother Res 22, 10871092.CrossRefGoogle ScholarPubMed
89. Schoonees, A, Visser, J, Musekiwa, A et al. (2012) Pycnogenol(R) for the treatment of chronic disorders. Cochrane Database Syst Rev 2, CD008294.Google Scholar
90. Schumacher, HR, Pullman-Mooar, S, Gupta, SR et al. (2013) Randomized double-blind crossover study of the efficacy of a tart cherry juice blend in treatment of osteoarthritis (OA) of the knee. Osteoarthritis Cartilage 21, 10351041.CrossRefGoogle ScholarPubMed
91. Ahmed, S, Wang, N, Hafeez, BB et al. (2005) Punica granatum L. extract inhibits IL-1beta-induced expression of matrix metalloproteinases by inhibiting the activation of MAP kinases and NF-kappaB in human chondrocytes in vitro . J Nutr 135, 20962102.CrossRefGoogle ScholarPubMed
92. Haseeb, A, Chen, D & Haqqi, TM (2013) Delphinidin inhibits IL-1beta-induced activation of NF-kappaB by modulating the phosphorylation of IRAK-1(Ser376) in human articular chondrocytes. Rheumatology (Oxford) 52, 9981008.CrossRefGoogle ScholarPubMed
93. Jean-Gilles, D, Li, L, Vaidyanathan, VG et al. (2013) Inhibitory effects of polyphenol punicalagin on type-II collagen degradation in vitro and inflammation in vivo . Chem Biol Interact 205, 9099.CrossRefGoogle ScholarPubMed
94. Hadipour-Jahromy, M & Mozaffari-Kermani, R (2010) Chondroprotective effects of pomegranate juice on monoiodoacetate-induced osteoarthritis of the knee joint of mice. Phytother Res 24, 182185.CrossRefGoogle ScholarPubMed
95. Jean-Gilles, D, Li, L, Ma, H et al. (2012) Anti-inflammatory effects of polyphenolic-enriched red raspberry extract in an antigen-induced arthritis rat model. J Agric Food Chem 60, 57555762.CrossRefGoogle Scholar
96. Frasca, G, Panico, AM, Bonina, F et al. (2010) Involvement of inducible nitric oxide synthase and cyclooxygenase-2 in the anti-inflammatory effects of a red orange extract in human chondrocytes. Nat Prod Res 24, 14691480.Google ScholarPubMed
97. Hooshmand, S, Soung do, Y, Lucas, EA et al. (2007) Genistein reduces the production of proinflammatory molecules in human chondrocytes. J Nutr Biochem 18, 609614.CrossRefGoogle ScholarPubMed
98. Claassen, H, Briese, V, Manapov, F et al. (2008) The phytoestrogens daidzein and genistein enhance the insulin-stimulated sulfate uptake in articular chondrocytes. Cell Tissue Res 333, 7179.CrossRefGoogle ScholarPubMed
99. Yu, SB, Xing, XH, Dong, GY et al. (2012) Excess genistein suppresses the synthesis of extracellular matrix in female rat mandibular condylar cartilage. Acta Pharmacol Sin 33, 918923.CrossRefGoogle ScholarPubMed
100. Mohammad Shahi, M, Rashidi, MR, Mahboob, S et al. (2012) Protective effect of soy protein on collagen-induced arthritis in rat. Rheumatol Int 32, 24072414.CrossRefGoogle ScholarPubMed
101. Ham, KD, Loeser, RF, Lindgren, BR et al. (2002) Effects of long-term estrogen replacement therapy on osteoarthritis severity in cynomolgus monkeys. Arthritis Rheum 46, 19561964.CrossRefGoogle ScholarPubMed
102. Arjmandi, BH, Khalil, DA, Lucas, EA et al. (2004) Soy protein may alleviate osteoarthritis symptoms. Phytomedicine 11, 567575.CrossRefGoogle ScholarPubMed
103. Choi, EM & Lee, YS (2010) Effects of hesperetin on the production of inflammatory mediators in IL-1beta treated human synovial cells. Cell Immunol 264, 13.CrossRefGoogle ScholarPubMed
104. Li, R, Cai, L, Ren, DY et al. (2012) Therapeutic effect of 7, 3¢-dimethoxy hesperetin on adjuvant arthritis in rats through inhibiting JAK2-STAT3 signal pathway. Int Immunopharmacol 14, 157163.CrossRefGoogle Scholar
105. Umar, S, Kumar, A, Sajad, M et al. (2013) Hesperidin inhibits collagen-induced arthritis possibly through suppression of free radical load and reduction in neutrophil activation and infiltration. Rheumatol Int 33, 657663.CrossRefGoogle ScholarPubMed
106. Maiani, G, Caston, MJ, Catasta, G et al. (2009) Carotenoids: actual knowledge on food sources, intakes, stability and bioavailability and their protective role in humans. Mol Nutr Food Res 53, Suppl. 2, S194S218.CrossRefGoogle ScholarPubMed
107. Davies, MR, Ribeiro, LR, Downey-Jones, M et al. (2009) Ligands for retinoic acid receptors are elevated in osteoarthritis and may contribute to pathologic processes in the osteoarthritic joint. Arthritis Rheum 60, 17221732.CrossRefGoogle ScholarPubMed
108. De Roos, AJ, Arab, L, Renner, JB et al. (2001) Serum carotenoids and radiographic knee osteoarthritis: the Johnston County Osteoarthritis Project. Public Health Nutr 4, 935942.CrossRefGoogle ScholarPubMed
109. Seki, T, Hasegawa, Y, Yamaguchi, J et al. (2010) Association of serum carotenoids, retinol, and tocopherols with radiographic knee osteoarthritis: possible risk factors in rural Japanese inhabitants. J Orthop Sci 15, 477484.CrossRefGoogle ScholarPubMed
110. Pattison, DJ, Symmons, DP, Lunt, M et al. (2005) Dietary beta-cryptoxanthin and inflammatory polyarthritis: results from a population-based prospective study. Am J Clin Nutr 82, 451455.CrossRefGoogle ScholarPubMed
111. Kamal-Eldin, A & Moazzami, A (2009) Plant sterols and stanols as cholesterol-lowering ingredients in functional foods. Recent Pat Food Nutr Agric 1, 114.CrossRefGoogle ScholarPubMed
112. Wu, T, Fu, J, Yang, Y et al. (2009) The effects of phytosterols/stanols on blood lipid profiles: a systematic review with meta-analysis. Asia Pac J Clin Nutr 18, 179186.Google ScholarPubMed
113. Gabay, O, Sanchez, C, Salvat, C et al. (2010) Stigmasterol: a phytosterol with potential anti-osteoarthritic properties. Osteoarthritis Cartilage 18, 106116.CrossRefGoogle ScholarPubMed
114. Chen, WP, Yu, C, Hu, PF et al. (2012) Stigmasterol blocks cartilage degradation in rabbit model of osteoarthritis. Acta Biochim Pol 59, 537541.CrossRefGoogle ScholarPubMed
115. Healy, ZR, Lee, NH, Gao, X et al. (2005) Divergent responses of chondrocytes and endothelial cells to shear stress: cross-talk among COX-2, the phase 2 response, and apoptosis. Proc Natl Acad Sci U S A 102, 1401014015.CrossRefGoogle ScholarPubMed
116. Facchini, A, Stanic, I, Cetrullo, S et al. (2011) Sulforaphane protects human chondrocytes against cell death induced by various stimuli. J Cell Physiol 226, 17711779.CrossRefGoogle ScholarPubMed
117. Kim, HA, Yeo, Y, Kim, WU et al. (2009) Phase 2 enzyme inducer sulphoraphane blocks matrix metalloproteinase production in articular chondrocytes. Rheumatology (Oxford) 48, 932938.CrossRefGoogle ScholarPubMed
118. Kim, HA, Yeo, Y, Jung, HA et al. (2012) Phase 2 enzyme inducer sulphoraphane blocks prostaglandin and nitric oxide synthesis in human articular chondrocytes and inhibits cartilage matrix degradation. Rheumatology (Oxford) 51, 10061016.CrossRefGoogle ScholarPubMed
119. Davidson, RK, Jupp, O, de Ferrars, R et al. (2013) Sulforaphane represses matrix-degrading proteases and protects cartilage from destruction in vitro and in vivo . Arthritis Rheum 65, 31303140.CrossRefGoogle ScholarPubMed
120. Lam, YY, Peterson, CM & Ravussin, E (2013) Resveratrol vs. calorie restriction: data from rodents to humans. Exp Gerontol 48, 10181024.CrossRefGoogle ScholarPubMed
121. Matsuzaki, T, Matsushita, T, Takayama, K et al. (2013) Disruption of Sirt1 in chondrocytes causes accelerated progression of osteoarthritis under mechanical stress and during ageing in mice. Ann Rheum Dis (In the Press).Google ScholarPubMed
122. Elmali, N, Esenkaya, I, Harma, A et al. (2005) Effect of resveratrol in experimental osteoarthritis in rabbits. Inflamm Res 54, 158162.CrossRefGoogle ScholarPubMed
123. Wang, J, Gao, JS, Chen, JW et al. (2012) Effect of resveratrol on cartilage protection and apoptosis inhibition in experimental osteoarthritis of rabbit. Rheumatol Int 32, 15411548.CrossRefGoogle ScholarPubMed
124. Csaki, C, Keshishzadeh, N, Fischer, K et al. (2008) Regulation of inflammation signalling by resveratrol in human chondrocytes in vitro . Biochem Pharmacol 75, 677687.CrossRefGoogle Scholar
125. Lei, M, Wang, JG, Xiao, DM et al. (2012) Resveratrol inhibits interleukin 1beta-mediated inducible nitric oxide synthase expression in articular chondrocytes by activating SIRT1 and thereby suppressing nuclear factor-kappaB activity. Eur J Pharmacol 674, 7379.CrossRefGoogle ScholarPubMed
126. Shakibaei, M, Csaki, C, Nebrich, S et al. (2008) Resveratrol suppresses interleukin-1beta-induced inflammatory signaling and apoptosis in human articular chondrocytes: potential for use as a novel nutraceutical for the treatment of osteoarthritis. Biochem Pharmacol 76, 14261439.CrossRefGoogle ScholarPubMed
127. Im, HJ, Li, X, Chen, D et al. (2012) Biological effects of the plant-derived polyphenol resveratrol in human articular cartilage and chondrosarcoma cells. J Cell Physiol 227, 34883497.CrossRefGoogle ScholarPubMed
128. Csaki, C, Mobasheri, A & Shakibaei, M (2009) Synergistic chondroprotective effects of curcumin and resveratrol in human articular chondrocytes: inhibition of IL-1beta-induced NF-kappaB-mediated inflammation and apoptosis. Arthritis Res Ther 11, R165.CrossRefGoogle ScholarPubMed
129. Shakibaei, M, Mobasheri, A & Buhrmann, C (2011) Curcumin synergizes with resveratrol to stimulate the MAPK signaling pathway in human articular chondrocytes in vitro . Genes Nutr 6, 171179.CrossRefGoogle ScholarPubMed
130. Singh, S & Aggarwal, BB (1995) Activation of transcription factor NF-kappa B is suppressed by curcumin (diferuloylmethane) [corrected]. J Biol Chem 270, 2499525000.CrossRefGoogle ScholarPubMed
131. Li, WQ, Dehnade, F & Zafarullah, M (2001) Oncostatin M-induced matrix metalloproteinase and tissue inhibitor of metalloproteinase-3 genes expression in chondrocytes requires Janus kinase/STAT signaling pathway. J Immunol 166, 34913498.CrossRefGoogle ScholarPubMed
132. Liacini, A, Sylvester, J, Li, WQ et al. (2003) Induction of matrix metalloproteinase-13 gene expression by TNF-alpha is mediated by MAP kinases, AP-1, and NF-kappaB transcription factors in articular chondrocytes. Exp Cell Res 288, 208217.CrossRefGoogle ScholarPubMed
133. Liacini, A, Sylvester, J, Li, WQ et al. (2002) Inhibition of interleukin-1-stimulated MAP kinases, activating protein-1 (AP-1) and nuclear factor kappa B (NF-kappa B) transcription factors down-regulates matrix metalloproteinase gene expression in articular chondrocytes. Matrix Biol 21, 251262.CrossRefGoogle ScholarPubMed
134. Innes, JF, Fuller, CJ, Grover, ER et al. (2003) Randomised, double-blind, placebo-controlled parallel group study of P54FP for the treatment of dogs with osteoarthritis. Vet Rec 152, 457460.CrossRefGoogle Scholar
135. Schulze-Tanzil, G, Mobasheri, A, Sendzik, J et al. (2004) Effects of curcumin (diferuloylmethane) on nuclear factor kappaB signaling in interleukin-1beta-stimulated chondrocytes. Ann N Y Acad Sci 1030, 578586.CrossRefGoogle ScholarPubMed
136. Shakibaei, M, Schulze-Tanzil, G, John, T et al. (2005) Curcumin protects human chondrocytes from IL-l1beta-induced inhibition of collagen type II and beta1-integrin expression and activation of caspase-3: an immunomorphological study. Ann Anat 187, 487497.CrossRefGoogle ScholarPubMed
137. Mathy-Hartert, M, Jacquemond-Collet, I, Priem, F et al. (2009) Curcumin inhibits pro-inflammatory mediators and metalloproteinase-3 production by chondrocytes. Inflamm Res 58, 899908.CrossRefGoogle ScholarPubMed
138. Shakibaei, M, John, T, Schulze-Tanzil, G et al. (2007) Suppression of NF-kappaB activation by curcumin leads to inhibition of expression of cyclo-oxygenase-2 and matrix metalloproteinase-9 in human articular chondrocytes: implications for the treatment of osteoarthritis. Biochem Pharmacol 73, 14341445.CrossRefGoogle ScholarPubMed
139. Clutterbuck, AL, Mobasheri, A, Shakibaei, M et al. (2009) Interleukin-1beta-induced extracellular matrix degradation and glycosaminoglycan release is inhibited by curcumin in an explant model of cartilage inflammation. Ann N Y Acad Sci 1171, 428435.CrossRefGoogle Scholar
140. Mun, SH, Kim, HS, Kim, JW et al. (2009) Oral administration of curcumin suppresses production of matrix metalloproteinase (MMP)-1 and MMP-3 to ameliorate collagen-induced arthritis: inhibition of the PKCdelta/JNK/c-Jun pathway. J Pharmacol Sci 111, 1321.CrossRefGoogle ScholarPubMed
141. Henrotin, Y, Clutterbuck, AL, Allaway, D et al. (2010) Biological actions of curcumin on articular chondrocytes. Osteoarthritis Cartilage 18, 141149.CrossRefGoogle ScholarPubMed
142. Belcaro, G, Cesarone, MR, Dugall, M et al. (2010) Efficacy and safety of Meriva(R), a curcumin-phosphatidylcholine complex, during extended administration in osteoarthritis patients. Altern Med Rev 15, 337344.Google Scholar
143. Belcaro, G, Cesarone, MR, Dugall, M et al. (2010) Product-evaluation registry of Meriva(R), a curcumin-phosphatidylcholine complex, for the complementary management of osteoarthritis. Panminerva Med 52, 5562.Google Scholar
144. Msika, P, Baudouin, C, Saunois, A et al. (2008) Avocado/soybean unsaponifiables, ASU EXPANSCIENCE, are strictly different from the nutraceutical products claiming ASU appellation. Osteoarthritis Cartilage 16, 12751276.CrossRefGoogle ScholarPubMed
145. Maheu, E, Cadet, C, Marty, M et al. (2014) Randomised, controlled trial of avocado-soybean unsaponifiable (Piascledine) effect on structure modification in hip osteoarthritis: the ERADIAS study. Ann Rheum Dis 73, 376384.CrossRefGoogle ScholarPubMed
146. Lequesne, M, Maheu, E, Cadet, C et al. (2002) Structural effect of avocado/soybean unsaponifiables on joint space loss in osteoarthritis of the hip. Arthritis Rheum 47, 5058.CrossRefGoogle ScholarPubMed
147. Appelboom, T, Schuermans, J, Verbruggen, G et al. (2001) Symptoms modifying effect of avocado/soybean unsaponifiables (ASU) in knee osteoarthritis. A double blind, prospective, placebo-controlled study. Scand J Rheumatol 30, 242247.Google ScholarPubMed
148. Blotman, F, Maheu, E, Wulwik, A et al. (1997) Efficacy and safety of avocado/soybean unsaponifiables in the treatment of symptomatic osteoarthritis of the knee and hip. A prospective, multicenter, three-month, randomized, double-blind, placebo-controlled trial. Rev Rhum Engl Ed 64, 825834.Google Scholar
149. Maheu, E, Mazieres, B, Valat, JP et al. (1998) Symptomatic efficacy of avocado/soybean unsaponifiables in the treatment of osteoarthritis of the knee and hip: a prospective, randomized, double-blind, placebo-controlled, multicenter clinical trial with a six-month treatment period and a two-month followup demonstrating a persistent effect. Arthritis Rheum 41, 8191.3.0.CO;2-9>CrossRefGoogle Scholar
150. Boileau, C, Martel-Pelletier, J, Caron, J et al. (2009) Protective effects of total fraction of avocado/soybean unsaponifiables on the structural changes in experimental dog osteoarthritis: inhibition of nitric oxide synthase and matrix metalloproteinase-13. Arthritis Res Ther 11, R41.CrossRefGoogle ScholarPubMed
151. Cake, MA, Read, RA, Guillou, B et al. (2000) Modification of articular cartilage and subchondral bone pathology in an ovine meniscectomy model of osteoarthritis by avocado and soya unsaponifiables (ASU). Osteoarthritis Cartilage 8, 404411.CrossRefGoogle Scholar
152. Gabay, O, Gosset, M, Levy, A et al. (2008) Stress-induced signaling pathways in hyalin chondrocytes: inhibition by Avocado-Soybean Unsaponifiables (ASU). Osteoarthritis Cartilage 16, 373384.CrossRefGoogle ScholarPubMed
153. Henrotin, YE, Deberg, MA, Crielaard, JM et al. (2006) Avocado/soybean unsaponifiables prevent the inhibitory effect of osteoarthritic subchondral osteoblasts on aggrecan and type II collagen synthesis by chondrocytes. J Rheumatol 33, 16681678.Google ScholarPubMed
154. Henrotin, YE, Sanchez, C, Deberg, MA et al. (2003) Avocado/soybean unsaponifiables increase aggrecan synthesis and reduce catabolic and proinflammatory mediator production by human osteoarthritic chondrocytes. J Rheumatol 30, 18251834.Google ScholarPubMed
155. Au, RY, Al-Talib, TK, Au, AY et al. (2007) Avocado soybean unsaponifiables (ASU) suppress TNF-alpha, IL-1beta, COX-2, iNOS gene expression, and prostaglandin E2 and nitric oxide production in articular chondrocytes and monocyte/macrophages. Osteoarthritis Cartilage 15, 12491255.CrossRefGoogle ScholarPubMed
156. Heinecke, LF, Grzanna, MW, Au, AY et al. (2010) Inhibition of cyclooxygenase-2 expression and prostaglandin E2 production in chondrocytes by avocado soybean unsaponifiables and epigallocatechin gallate. Osteoarthritis Cartilage 18, 220227.CrossRefGoogle ScholarPubMed
157. Frondoza, CG (2008) Response to letter to editor entitled: “Avocado/soybean unsaponfiables, ASU Expanscience, are strictly different from the nutraceutical products claiming ASU appellation” (4365). Osteoarthritis Cartilage 16, 15901591.CrossRefGoogle ScholarPubMed
158. Henrotin, Y (2008) Avocado/soybean unsaponifiable (ASU) to treat osteoarthritis: a clarification. Osteoarthritis Cartilage 16, 11181119; author reply 1120.CrossRefGoogle ScholarPubMed
159. Leach, MJ & Kumar, S (2008) The clinical effectiveness of Ginger (Zingiber officinale) in adults with osteoarthritis. Int J Evid Based Healthc 6, 311320.Google ScholarPubMed
160. Altman, RD & Marcussen, KC (2001) Effects of a ginger extract on knee pain in patients with osteoarthritis. Arthritis Rheum 44, 25312538.3.0.CO;2-J>CrossRefGoogle ScholarPubMed
161. Shen, CL, Hong, KJ & Kim, SW (2005) Comparative effects of ginger root (Zingiber officinale Rosc.) on the production of inflammatory mediators in normal and osteoarthrotic sow chondrocytes. J Med Food 8, 149153.CrossRefGoogle ScholarPubMed
162. Ribel-Madsen, S, Bartels, EM, Stockmarr, A et al. (2012) A synoviocyte model for osteoarthritis and rheumatoid arthritis: response to Ibuprofen, betamethasone, and ginger extract-a cross-sectional in vitro study. Arthritis 2012, 505842.CrossRefGoogle ScholarPubMed
163. Williams, FM, Skinner, J, Spector, TD et al. (2010) Dietary garlic and hip osteoarthritis: evidence of a protective effect and putative mechanism of action. BMC Musculoskelet Disord 11, 280.CrossRefGoogle ScholarPubMed
164. Chen, WP, Tang, JL, Bao, JP et al. (2011) Effects of diallyl sulphide in chondrocyte and cartilage in experimental osteoarthritis in rabbit. Phytother Res 25, 351356.CrossRefGoogle ScholarPubMed
165. Lu, B, Ahmad, O, Zhang, FF et al. (2013) Soft drink intake and progression of radiographic knee osteoarthritis: data from the osteoarthritis initiative. BMJ Open 3, e002993.CrossRefGoogle ScholarPubMed
166. Chua, KH, Lee, TH, Nagandran, K et al. (2013) Edible bird's nest extract as a chondro-protective agent for human chondrocytes isolated from osteoarthritic knee: in vitro study. BMC Complement Altern Med 13, 19.CrossRefGoogle ScholarPubMed
167. Phitak, T, Pothacharoen, P, Settakorn, J et al. (2012) Chondroprotective and anti-inflammatory effects of sesamin. Phytochemistry 80, 7788.CrossRefGoogle ScholarPubMed
168. Pearson, W, Fletcher, RS, Kott, LS et al. (2010) Protection against LPS-induced cartilage inflammation and degradation provided by a biological extract of Mentha spicata. BMC Complement Altern Med 10, 19.CrossRefGoogle ScholarPubMed
169. Marcu, KB, Otero, M, Olivotto, E et al. (2010) NF-kappaB signaling: multiple angles to target OA. Curr Drug Targets 11, 599613.CrossRefGoogle ScholarPubMed