Abstract
Chondroitin sulphate (CS) is a natural glycosaminoglycan present in the extracellular matrix and is formed by the 1–3 linkage of D-glucuronic acid to N-acetylgalactosamine. In chondrocytes, CS diminishes interleukin-1 p (IL-1p)-induced increases in p38 mitogen-activated protein kinase (p38MAPK) and signal-regulated kinase 1/2 (Erk1/2) phosphorylation, and decreases nuclear factor-KB (NF-kB) nuclear translocation and as a consequence, reduces the formation of pro-inflammatory cytokines, IL-1 p and TNF-a, and pro-inflammatory enzymes, such as phospholipase A2 (PLA2), cyclooxygenase 2 (COX-2) and nitric oxide synthase-2 (NOS-2). The mechanism of action of CS explains its beneficial effect on the cartilage, synovial membrane and subchondral bone. On the other hand, in vivo, CS given orally prevents hepatic NF-κB nuclear translocation, suggesting that systemic CS may elicit an anti-inflammatory effect in many tissues besides the articulation. There is preliminary evidence showing that in human beings, CS may be of benefit in other diseases where inflammation is an essential marker, such as psoriasis and atherosclerosis. The review of the literature suggest that CS might also be of interest for the treatment of other diseases with an inflammatory and/or autoimmune character, such as inflammatory bowel disease, degenerative diseases of the central nervous system and stroke, multiple sclerosis and other autoimmune diseases.
Keywords: chondroitin sulphate, inflammation, autoimmune diseases, psoriasis, atherosclerosis, inflammatory bowel disease, Alzheimer's disease, Parkinson disease, multiple sclerosis
Biochemistry of chondrotin sulphate
-
Mechanism of action of chondroitin sulphate
- Effect of chondroitin sulphate on the chondrocyte
- Effect of chondroitin sulphate on the synovial membrane
- Effect of chondroitin sulphate on subchondral bone
-
Human use of chondroitin sulphate
- Chondrotin sulphate in osteoarthritis
- Chondrotin sulphate in psoriasis
- Chondrotin sulphate in atherosclerosis
- Chondroitin sulphate in IBD
- Chondroitin sulphate in degenerative diseases of the central nervous system (CNS)
- Other autoimmune diseases that may benefit from chondroitin sulphate
Conclusions
Biochemistry of chondroitin sulphate
Chondroitin sulphate (CS) is a natural glycosaminoglycan (GAG) present in the extracellular matrix surrounding cells, especially in the cartilage, skin, blood vessels, ligaments and tendons, where it forms an essential component of proteoglycans (PG) [1, 2]. CS is the main disaccharide unit of cartilage GAG, formed by the 1–3 linkage of D-glucuronic acid to N-acetylgalactosamine. The disaccharide units are attached by β 1–4 galactosamine links. The galactosamine residues are sulphated either in position 4 (Δdi-4S), 6 (Δdi-6S) or 4 and 6 (Δdi-4,6S). The sulphate groups along with the carboxyl groups of glucuronic acid are ionized, conferring a negative charge. In the extracellular matrix of the cartilage, about 100 chains of CS, each containing 50 to 60 disaccharide units, are covalently attached to a long polypeptide backbone composed of more than 2000 amino acids. The polypeptide with the CS chains is O-linked to hyaluronic acid forming the PG. The domain of the core protein located between the link protein and the CS region is occupied by keratan sulphate chains. The keratan sulphate disaccharide unit is formed by galactosido-1,4-N-acetylglucosamine-6-sulphate. Because of their high degree of hydration, CS-containing PG of articular cartilage are responsible for the viscoelastic proper ties of the tissue [3, 4].
Mechanism of action of chondroitin sulphate
Because CS elicits a key role in the articulation, many research groups have focused on the role of CS on the chondrocytes, the synovial membrane and the subchondral bone.
Effect of chondroitin sulphate on the chondrocyte
Repeated trauma on an articulation increases the release of cytokines such as interleukin-1β (IL-1β) and tumour necrosis factor-α (TNF-α), cytokines that play a key role in the development of osteoarthritis (OA). In chondrocytes, IL-1β activates extracellular signal-regulated kinase 1/2 (Erk1/2) and p38 mitogen-activated protein kinase (p38MAPK), and therefore induces the nuclear translocation of the nuclear factor-κB (NF-κB) and the activator protein-1 (AP-1). These transcription factors bind to consensus sequences of numerous pro-inflammatory genes, and initiate as well as maintain the inflammatory reaction in chondrocytes [5–7]. As a result, IL-1p increases the expression of matrix metallopro-tease-3 (MMP-3) [8], MMP-9 [9], MMP-13 [8, 10, 11], phospholipase A2 (PLA2) and cyclooxygenase 2 (COX-2) [11, 12], nitric oxide synthase-2 (NOS2), IL-1β and TNF-α[13].
Using chondrocytes stimulated by IL-1β as experimental model, it was demonstrated that CS diminishes IL-1β-induced NF-κB nuclear translocation and sodium nitroprusside-induced chondrocyte apoptosis. The effects of CS are mediated by inhibition of p38MAPK and Erk1/2 phosphorylation. These data suggest that the anti-inflammatory activity of CS is associated with the reduction of Erk1/2 and p38MAPK phosphorylation and nuclear transactivation of NF-κB[14].
Effect of chondroitin sulphate on the synovial membrane
Synovial tissue from patients with early osteoarthritis shows activated fibroblast-like synoviocyte (FLS), macrophages, T lymphocytes and mast cells infiltration [15]. Synovial FLS release IL-1β, IL-6, IL-8, MMP-1, MMP-2, MMP-3, MMP-13, MMP-14, MMP-16, tissue inhibitor of metalloproteinases-1 (TIMP-1), receptor activator of nuclear factor-kappa B ligand (RANKL), transforming growth factor-β (TGF-p), vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) [16]. Activation of NF-κB increases FLS proliferation, and changes the phenotype of these cells into highly invasive FLS with great motility and ability to secrete cytokines and MMP-13 [17].
Inhibition of the IκB kinase (IKK) complex impedes the phosphorylation of the inhibitor of κB (κBα) and as a consequence, prevents NF-κB activation. In synovial macrophages, inhibition of IKK diminishes IL-1 β-induced production of IL-6; moreover, in rats with adjuvant-induced arthritis, intra-articular injection of a specific IKK-β inhibitor reduces arthritis activity and bone destruction; synovial inflammation is also decreased as documented by the reduction in synovial cellularity, TNF-α, IL-1β concentrations and reduction of the volume of the paw [18]. The crucial role of NF-κB in the initiation of synovitis is further supported by the fact that the injection of a dominant-negative form of IKK-β in the articulation of the rat with adjuvant-induced arthritis reduces synovial cellularity by 50%, and diminishes synovial concentrations of IL-1β, TNF-α and MMP-3 [19]. These results provide evidence that activation and nuclear translocation of NF-κB is an important step in the development of synovitis [20].
In patients with knee OA, CS diminishes the number of patients with signs of synovitis from 90 of 307 at baseline to 38 (P= 0.01) at the end of 24 weeks of treatment [21]. This confirms, in human beings, observations reported in DBA/1J mice with a type II collagen-induced arthritis and treated for 9 weeks with various dosages of CS; infiltration of inflammatory cells, granulated tissue formation and proliferation of synovial lining cells were partially prevented by treatment with 1000 mg/kg/day of CS for 63 days [22].
The mechanism of action underlying the reduction of synovitis signs by CS remains poorly known. There is evidence that the inflammatory response in OA is closely associated with the activation and nuclear translocation of NF-κB, a phenomenon dependent upon the activation of p38MAPK, Erk1/2 and c-Jun N-terminal kinase (JNK) [23, 24]. Moreover, proliferation, motility, release of cytokines and matrix-degrading activity of FLS are associated with the activation and nuclear translocation of NF-κB[15, 17, 25].
Because in chondrocytes, CS diminishes Erk1/2 and p38MAPK and reduces IL-1 β-induced NF-κB nuclear translocation [14], we might speculate that CS reduces NF-κB nuclear translocation in synovial cells, and so diminishes the synovial inflammatory reaction. Supporting such hypothesis, in human synoviocytes stimulated by IL-1β, the CS disaccharide (Δdi-6S) reduces the nuclear translocation of NF-κB by 65%[26]. This observation is in agreement with the effect of Δdi-4S and κdi-6S disaccharides in chondrocytes, for example they reduce NF-κB nuclear translocation [20]. Oral CS increases plasma concentrations of Δdi-4S and κdi-6S [27], it is therefore conceivable that in human beings the decline of synovitis signs produced by CS may be explained, at least in part, by the reduction in NF-κB nuclear translocation in synoviocytes and macrophages produced by CS disaccharides.
Effect of chondroitin sulphate on subchondral bone
Recent evidence shows that altered osteoblast metabolism plays an important role in subchondral bone alterations, which in turn have been implicated in the progression and/or initiation of OA [28]. In human subchondral bone osteoblasts, CS up-regulates osteoprotegerin (OPG) expression and decreases RANKL expression [29]; as a consequence, CS increases the ratio of OPG/RANKL. Because the expression of RANKL is increased in abnormal osteoblasts with bone destruction [30], CS could exert a positive effect that may result in the reduction of the resorptive activity in subchondral bone. The mechanism of action underlying the effect of CS might be associated with the fact that induction of RANKL expression requires the activation of Erk1/2 and PI-3K/AKT pathways [31].
Human use of chondroitin sulphate
The rationale for using CS as a treatment for OA was the decrease of CS in ageing patients with OA. In the late 1960s, anecdotal reports [32] suggested that CS disaccharides were reduced in synovial fluid of patients with OA, observations that were confirmed three decades later [33].
Osteoarthritis affects a majority of individuals over 60 years of age, and is characterized by focal areas of loss of articular cartilage, with varying degrees of osteophyte formation, subchondral bone change and synovitis, with local inflammation, pain and functional disability [34]. The physiopathology of OA remains controversial. It has been proposed that the use of the joint implies multiple micro-trauma to the articular cartilage with the formation of extracellular matrix fragments (EMFs) and fibronectin (FN-f) (Fig. 1). These fragments bind to the integrin family of cell-surface receptors of the chondrocyte and promote the expression of MMPs, primarily MMP-13, aimed to cleave the EMFs [35–37]. The increase in expression of MMPs is accompanied by an enhanced synthesis of pro-inflammatory cytokines, essentially IL-1β and TNF-α, which will sustain the activation of chondrocytes and, moreover, will promote the formation of MMPs, aggrecanase, reactive oxygen intermediates, nitric oxide and lipid-derivative inflammatory mediators such as prostaglandins and leukotrienes. These mediators will enhance the catabolic activity of the chondrocyte, causing further destruction of the cartilage matrix. On the other hand, EMFs, IL-1β and TNF-α released into the synovial fluid activate macrophages and mastocytes in the synovial membrane originating the synovitis. Activation of synovial cells will result in a further release of IL-1β, TNF-α and MMPs that will contribute to the destruction of the cartilage matrix [16, 34, 38–40].
Fig. 1.

Multiple microtrauma to the articulation induce the production of pro-inflammatory cytokines (IL-1β(5, TNF-α, IL-17), extracellular matrix fragments (EMFs) and fibronectin fragments (FN-f) that bind to membrane receptors of the chondrocyte and activate NF-κB, and therefore promote the expression of matrix metalloproteases (MMPs), which further generate EMFs. Nuclear translocation of NF-κB will also increase the expression of phospholipase A2 (PLA2) and cyclooxygenase-2 (COX-2), enzymes that will release arachidonic acid and form prostaglandin E2 (PGE2) responsible for the inflammation and pain. NF-κB increases the expression of nitric oxide synthase-2 (NOS-2) and the production of nitric oxide (NO) that will contribute to the inflammatory reaction, pain and apoptosis.
Cytokines, FN-f and EMFs by binding to membrane receptors activate signal transduction pathways, such as Erk1/2 and p38MAPK, which induce the nuclear translocation of NF-κB[39]. In the nucleus, NF-κB binds to the promoter of numerous genes and increases the transcription and expression of IL-1β, TNF-α, COX-2, NOS-2, PLA2 and MMP-1, -3, -9 and -13. Released MMPs further contribute to destroy the extracellular matrix and form additional FN-f and EMFs that, together with the cytokines, will perpetrate the inflammatory reaction of the chondrocytes and the synovial membrane. On the other hand, PLA2 will release arachidonic acid (AA) to generate prostaglandin E2 (PGE2) by COX-2, cause of inflammation and pain. The excessive production of nitric oxide contributes to increase the inflammatory reaction and pain [34, 40].
As outlined, cytokines have a crucial role in the onset and progression of OA [41]. For instance, IL-1β and TNF-α are implicated in the early development of arthritis, and IL-1β contributes to sustain the inflammatory reaction in later stages; IL-17 and IL-18 are also pro-inflammatory cytokines in the joint. Other cytokines released in the osteoarthritic joint have a regulatory role of the inflammation (IL-6, IL-8), or an inhibitory or anti-inflammatory function (IL-4, IL-10, IL-11, IL-13, IFN-γ and IL-1 receptor anta gonist), or even an anabolic role, such as insulin-like growth factor 1 (IGF-1), transforming growth factor β (TGF-β), fibroblast growth factor (FGF) and bone morphogenetic protein (BMP) [42, 43].
Chondroitin sulphate in osteoarthritis
Randomized placebo-controlled clinical trials have demonstrated that CS reduces pain and improves articular function [44–46], reduces joint swelling and effusion [21] and prevents joint space narrowing of the knee [44, 46] and fingers [47, 48] more effectively than placebo. Accordingly, CS has been classified as a symptomatic slow-acting drug in osteoarthritis (SYSADOA) and a structure/dis ease-modifying anti-osteoarthritis drug (S/DMOAD) [44, 47].
The beneficial effects of CS in patients with OA result from different effects of CS on articular tissues, primarily the result of the immunomodulatory effect of CS, for example reduction of NF-κB nuclear translocation, decrease in the production of pro-inflammatory cytokines IL-1β and TNF-α and reduction in the expression and activity of NOS-2 and COX-2 (Fig. 2). Other effects of CS may contribute to its beneficial effect, such as the increase in the synthesis of articular cartilage PG, the reduction in the apoptosis of chondrocytes and the reduction of the synthesis and/or activity of MMPs [28, 49, 50].
Fig. 2.

Chondroitin sulphate (CS) by inhibiting reactive oxygen species (H2O2 or O2•) and/or the extracellular signal-regulated kinase 1/2 (Erk1/2) and p38 mitogen-activated protein kinase (p38MAPK), reduces the nuclear translocation of the nuclear factor-κB (NF-κB or p50/p65 heterodimer), and therefore CS diminishes the synthesis of proteolytic enzymes, such as the matrix metalloproteases (MMP-3, -9, -13) and catepsine B, of pro-inflammatory enzymes, for example fosfolipase A2 (PLA2), cyclooxygenase-2 (COX-2) and nitric oxide synthase-2 (NOS-2), and of pro-inflammatory cytokines, for example TNF-α and IL-1β(J.
Following its nuclear translocation, NF-κB enhances the transcription activity of a variety of genes encoding chemokines, cytokines, adhesion molecules, inflammatory-associated enzymes and inhibitors to apoptosis [51]. Moreover, NF-κB signalling has been shown to be involved in lymphopoiesis and in the differentiation and activation of macrophages, osteoclasts, dendritic cells and granulocytes [52]. The NF-kB signalling pathway is closely linked to the regulation of inflammatory responses and survival of immune cells and accordingly, it has been suggested that NF-κB deficiency or its inhibition in vivo should reduce inflammatory responses.
Numerous studies have investigated the molecular mechanisms by which alterations in NF-κB signalling in diverse key cellular processes, including cell proliferation, cell survival, cellular stress response, innate immunity and inflammation are likely to contribute to disease pathology [53]. In this respect, type I diabetes, atherosclerosis, cancer, inflammatory bowel disease (IBD), gastritis, rheumatoid arthritis, systemic lupus erythematosus, asthma, acute respiratory distress syndrome, sepsis and systemic inflammatory response syndrome, surgical major trauma, Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease and multiple sclerosis are human diseases in which experi mental data support a causative role of activation of the NF-κB pathway [52]. The key role of NF-κB in inflammatory responses and immune homeostasis is the basis for the search of compounds targeting any step leading to the nuclear translocation of NF-κB to treat diseases with an inflammatory component.
The beneficial effects of CS for the treatment of OA raise the hypothesis that CS might be effective in other chronic inflammatory processes or diseases because of an autoimmune response. However, in order to accept the hypothesis that CS has a beneficial effect in diseases other than OA, the following question must be answered: can CS inhibition of NF-κB nuclear translocation occur in tissues other than the articulation?
There is preliminary in vivo evidence supporting an effect of CS on hepatic NF-κB nuclear translocation and nitric oxide concentrations; administration of CS (20 mg/kg/day) for 20 and 30 days did not affect NF-κB nuclear translocation in healthy rabbits. However, CS prevented the increase of NF-kB nuclear translocation and in nitric oxide hepatic concentrations triggered by a turpentine-induced inflammatory [54]. These results confirm in vivo that CS prevents NF-κB activation induced by an inflammatory reaction in tissues other than the articulation. The fact that CS modulates NF-κB in several tissues, such as chondrocytes, synovial membrane and liver, supports the hypothesis that CS could be of benefit in the treatment of other diseases with an inflammatory or autoimmune component.
Chondroitin sulphate in psoriasis
Deregulation of NF-κB appears to play an important role in skin pathology, such as psoriasis, inflammatory processes like incontinentia pigmenti, Lyme disease, allergic contact dermatitis and autoimmune diseases as well as in skin carcinogenesis [55]. Total expression of the proteins forming NF-κB, the heterodimer p50 and p65, may not be increased in psoriatic lesions [56], but the active phosphorylated form or nuclear expression of NF-κB is detected in 66% of psoriatic lesions and overexpressed in psoriasis compared with normal skin [57]. In addition, NF-κB function appears deregulated, in the sense that NF-κB DNA binding to the p53 kBsite is decreased, whereas NF-κB binding to the pro-inflammatory interleukin-8 (IL-8) κB site is increased in lesional psoriatic skin compared with non-lesional psoriatic skin [58]. Moreover, the NF-κB-dependent pro-inflammatory cytokines, IL-1β and TNF-a, have a crucial role in the appearance and progression of psoriasis and psoriatic arthritis [59, 60].
The relevancy of NF-κB in psoriasis is supported by a hospital-based, case-control study, including 519 patients with psoriasis vulgarisand 541 matched controls who were genotyped for NFKB1 (encodes for p50 protein) polymorphisms. An association between NFKB1 wild-type genotype and an increased risk for psoriasis vulgaris was found, for example mutations of the gene NFKB1 reduce NF-κB activity and incidence of psoriasis. The association was more evident in the subgroups of onset age ≤40 years, Psoriasis Area and Severity Index (PASI) score >20 and male patients [61].
Further supporting the role of NF-κB in psoriasis are the reports showing that effective treatment of psoriasis diminishes NF-κB nuclear translocation. For instance, one study showed that compared with normal epidermis, active phosphorylated NF-κB/RelA in the epidermis from psoriatic plaques was significantly up-regulated, and etanercept, a recombinant human TNF receptor fusion protein, produced a significant down-regulation of phosphorylated NF-κB/RelA, reduction that correlated with decreases in epidermal thickness, restoration of normal markers of keratinocyte differentiation and clinical outcomes [62].
The iminobenzoxathiolone compound 6-hydroxy-1,3-ben-zoxathiol-2-one is effective for the treatment of psoriasis [63], probably because iminobenzoxathiolones inhibit NF-κB activation and nuclear translocation [64]. Furthermore, the anti-psoriatic effect of avarol-3’-thiosalicylate [65], dimethyl fumarate [66], cur-cumin [67] and tacrolimus [68] depend upon the down-regulation of NF-κB activity.
In an open non-controlled trial, 11 patients with knee OA and long-standing, moderate-to-severe psoriasis resistant to conventional therapy were treated with 800 mg/day of CS for 2 months. Skin biopsies were obtained before and after treatment. All patients but one improved the condition of the skin, with reduction of swelling, redness, flaking and itching, increase in the hydration and softening of the skin and amelioration of scaling. Histopathologically, CS decreased epidermal thickness, the thickness between the stratum basale and the granulosum, reduced the number of keratinocytes and diminished the severity of psoriasis activity [69, 70]. These results strongly suggest that CS might be a helpful tool to treat moderate-to-severe psoriasis; however, controlled double-blinded prospective studies have to be conducted to confirm this report.
Chondroitin sulphate in atherosclerosis
Several reasons prompted researchers to explore whether GAGs are effective in preventing atherosclerosis. Because CS and heparin are GAGs, it was assumed that CS had antithrombotic properties, and as early as in 1955, Kurita reported that intravenous injections of 5 mg/kg chondroitin sulphate C (CSC—mainly composed of Δdi-6S) inhibited the progression of atherosclerosis in rabbits fed with a diet rich in cholesterol [71]. In the early 1960s, Murata confirmed that high doses of sulphated poly-saccharides had antithrombotic properties in animal models and affected the progression of atherosclerosis [72, 73]. With ageing and with advanced atherosclerosis, in the arterial wall there is a decrease in chondroitin sulphate A (CSA – mainly composed of Δdi-4S) and/or CSC, with a concomitant increase in chondroitin sulphate B (CSB – mainly composed of sulphate O-linked in positions 2 of the glucuronic acid and 4 of the N-acetyl-D-galac-tosamine), hence it was hypothesized that administration of CSA might prevent the progression of atherosclerosis [74].
A clinical trial compared the evolution of 60 patients with coronary heart disease given initially 10 g daily of CSA for 3 months, followed by doses of 1.5 to 3 g daily for 6 to 30 months with the evolution of 60 patients with coronary heart disease who were not taking CSA. At the end of the trial, three patients of the CSA-treated group presented a coronary event, compared with 21 patients in the control group; however, cardiovascular mortality did not differ [75]. The same patients were followed for 6 years with a dose of CSA reduced by half, for example 0.75 to 1.5 g; after 6 years of treatment with CSA, 6 patients (10%) presented an acute cardiac event, of which four died, compared with 42 patients (70%) of which 14 died in the control group [76]. No adverse effects or abnormal laboratory were found in patients receiving CSA for 6 years. These long-term studies in human beings warrant further double-blind randomized placebo-controlled trials.
There is strong evidence that the incidence and progression of atherosclerosis and subsequent cardiovascular diseases, for example myocardial infarction and stroke, are closely associated to inflammation. Pro-inflammatory cytokines and cellular adhesion molecules involved in the attachment of monocytes to the endothelial wall are critical in early atherogenesis and atherosclerotic lesions are infiltrated with cellular components associated with inflammation; furthermore, in response to acute ischaemia, there is an influx of neutrophils into the walls of the epicardial vessels, and the sites of acute plaque rupture are preferentially associated with inflammatory components [77]. The Cholesterol and Recurrent Events (CARE) trial demonstrated that the presence of inflammation after a myocardial infarction is associated with increased risk of recurrent coronary events [78]. There is growing evidence supporting that pro-inflammatory cytokines TNF-α and IL-1β play a crucial role in the disruption of macrovascular and microvascular circulation both in vivo and in vitro[79]. Moreover, markers of inflammation are highly predictive of cardiovascular events [80].
Keeping in mind the role of inflammation on vascular dysfunction, how CS might exert a beneficial effect on the progression of atherosclerosis and cardiovascular pathology? In first place, there is strong evidence that NF-κB is a pivotal transcription factor in the heart and cardiovascular system [81, 82]. The beneficial effect of acetylsalicylic acid (ASA) on cardiovascular events has been in part explained by a reduction in NF-κB nuclear translocation [83]. RANKL and its soluble decoy receptor OPG modulate vascular calcification, as demonstrated by several reports: (i) mice genetically deficient in OPG show a vascular calcification phenotype, (ii) in calcified arteries, the expression of RANKL is increased and that of OPG is diminished and (iii) there is a clinical association between coronary disease and serum OPG and RANKL levels [84]. The mechanism underlying the role of RANKL/OPG in atherosclerosis appears to be associated with the fact that binding of RANKL to its cognate receptor RANK activates NF-κB nuclear transcription, which stimulates osteoclastic differentiation in preosteoclasts and induces BMP-2 expression in chondrocytes; on the other hand, OPG reduces the effect of RANKL [85].
In summary, there is evidence that in human beings the formation of the arterial atherosclerotic plaque and the progression of atherosclerosis is closely modulated by local and systemic inflammatory reactions, where NF-κB and pro-inflammatory cytokines play a pivotal role. We postulate that CS might limit the progression of atherosclerosis [71–76] by diminishing NF-κB nuclear translocation [14] and by increasing the ratio of OPG/RANKL [29].
A recent study [86] examined the effect of CS on vascular injury and on markers of systemic inflammation in a rabbit model of atherosclerosis aggravated by systemic inflammation provoked by chronic antigen-induced arthritis [87]. Administration of CS prophylactically reduced serum concentrations of C-reactive protein and IL-6. Likewise, CS inhibited the expression of chemokine ligand 2 (CCL2)/monocyte chemoattractant protein-1 (MCP-1) and COX-2, and reduced the nuclear translocation of NF-kB in peripheral blood mononuclear cells. In femoral lesions, CS diminished the expression of CCL2 and COX-2, as well as the ratio of the intima/media thickness. Moreover, CS reduced the percentage of rabbits that developed vascular lesions in the aorta. These results support that CS may prevent/limit the progression of atherosclerosis, probably by diminishing local and systemic inflammatory reactions.
Recently, the possibility was raised that therapies targeting chronic low-grade inflammation may provide novel future strategies for cardiovascular disease prevention [88]. The data presented here support that CS might be a candidate to be used in the prevention of atherosclerotic cardiovascular events. Indeed, randomized, double-blind placebo-controlled trials have to be conducted to demonstrate such hypothesis.
Chondroitin sulphate in IBD
IBD, ulcerative colitis (UC) and Crohn's disease (CD), are chronic, relapsing gastro-intestinal disorders (GI) disorders of unknown aetiology. IBD is caused by the hyper-activation of effector immune cells through toll-like receptors (TLRs), primarily in macrophages, which produce high levels of pro-inflammatory cytokines TNF-α, IL-1α, IL-6 and IFN, resulting in colonic tissue damage [89]. NF-κB activation is markedly induced in IBD patients and through its ability to promote the expression of various pro-inflammatory genes, NF-κB strongly influences the course of mucosal inflammation [90]. Moreover, the NF-κB-dependent cytokines TNF-α and IL-1α are essential to the inflammatory reaction in UC and CD [91, 92].
Keeping in mind that IBD results from the activation of TLRs and NF-κB, and excessive production of TNF-α and IL-1β, treatment of IBD at each one of these levels has been proposed. To date, there is no effective tool to block directly TLRs [93], but there is preliminary evidence that the activation of the peroxisome proli -ferator-activated receptor-γ (PPARγ), a natural TLRs suppressor and antagonist, by thiazolidenedione ligands, for example troglitazone, pioglitazone and rosiglitazone, reduces colonic inflammation [94]. The murine dextran sulphate sodium-induced colitis (DSS-IC) is significantly attenuated by pioglitazone and netoglitazone, both prophylactically and therapeutically [95]. A recent multi-centre, randomized, double-blind, placebo-controlled clinical trial compared the efficacy of rosiglitazone with placebo for 12 weeks in 105 patients with mild to moderately active UC; after 12 weeks of therapy, 23 patients (44%) treated with rosiglitazone and 12 patients (23%) treated with placebo achieved clinical response; remission was achieved in nine patients (17%) treated with rosiglitazone and one patient (2%) treated with placebo, and moreover, quality of life was improved in patients receiving rosiglitazone. The authors concluded that rosiglitazone was effective in the treatment of mild to moderately active UC [96].
There is evidence that in intestinal biopsies from patients with CD, the phosphorylation of NF-κB is considerably increased [97], as is the concentration of TNF-α and IL-1β[98]. Oxymatrine (OMT) and matrine (MT) are quinolizidine alkaloids with anti-inflammatory properties that improve the DSS-IC by reducing serum levels of TNF-α and the expression of NF-κB in colonic mucosa [99]. Phytosteryl ferulates are hydroxycinnamic acid derivatives that partially prevent the DSS-IC very probably because they inhibit NF-κB activation [100]. Based on the evidence brought by studies in animals using genetic approaches to inhibit NF-κB activity, it was proposed that blocking NF-κB might offer particular promise to treat IBD [101]. A recent Cochrane review concluded that TNF-α blocking agents, infliximab, CDP571, adalimumab and certolizumab, were effective for the maintenance of remission in patients with CD who have responded to induction therapy [102]. Moreover, current data suggest that infliximab is an effective treatment for patients with moderate-to-severe UC with an inadequate response to conventional glucocorticoid treatment [103].
Is it reasonable to speculate that CS by inhibiting NF-κB phosphorylation and TNF-α and IL-1β release could be beneficial in patients with IBD? There is preliminary evidence suggesting that this may be the case. Using the model of DSS-IC, it was shown that CS improved the symptoms of bloody stools, erosion and increased white blood cells more effectively than 5-aminosalicylic acid [104]. However, more recently, Ota et al. showed a mild but non-significant effect of CS on DSS-IC, and in contrast a salmon-derived PG elicited a significant beneficial effect [105]. Interestingly, with the same DSS-IC model, it was shown that another naturally occurring monosaccharide, glucosamine, improved the clinical symptoms and suppressed colonic inflammation and tissue injury; moreover, glucosamine inhibited the activation of intestinal epithelial cells, as demonstrated by the reduced activation of NF-κB in the intestinal mucosa [106]. The experimental evidence reviewed support the hypothesis that CS might reduce the incidence and severity of relapse of IBD in human beings.
Chondroitin sulphate in degenerative diseases of the central nervous system (CNS)
There is strong evidence that uncontrolled neuroinflammatory processes, where activated astrocytes and microglia and their cytotoxic agents have a crucial role, contribute to the cascade of events leading to neuronal cell death, as occurs in neurodegenerative disorders such as Parkinson's disease and Alzheimer's disease [107].
The neuropathological hallmarks of Alzheimer's disease include deposits of amyloid-β peptide (Aβ), formation of Aβ plaques, accumulation of abnormal tau protein filaments in neu-rofibrillary tangles, extensive neurodegeneration and signs of chronic inflammation. The Aβ peptides activate NF-κB, resulting in the up-regulation of pro-inflammatory cytokines TNF-α and IL-1β, NOS-2, and COX-2 [108, 109]. However, there is debate on whether the inflammation contributes to neurotoxic effects or represents a secondary reaction to Aβ deposition. Several, but not all non-steroideal anti-inflammatory drugs (NSAIDs) reduce Aβ burden in transgenic mouse models of Alzheimer's disease, effect that may be mediated by either inhibition of COX or by a direct effect on γ-secretase, thereby reducing Aβ generation independently of COX inhibition [110].
Most epidemiological studies suggest that the risk of suffering Alzheimer's disease is reduced in patients treated with NSAIDs [111]. However, clinical trials on anti-inflammatory drugs inclu ding prednisone, hydroxychloroquine and the selective COX-2 inhibitors celecoxib and rofecoxib, showed no effects on cognition. The first large-scale clinical trial on both non-selective and COX-2 selective NSAIDs was also disappointing [112]. A possible explanation is that these drugs might be protective only at the initial stages of the disease, or even before disease initiation, but may not reverse the degenerative process in patients with esta blished pathology.
The PPARs are a family of nuclear hormone receptors that regu late immune and inflammatory responses, with an anti-inflammatory effect. Thiadiazolidinone derivatives, PPARγ agonists, are potent neuroprotector compounds. Thiadiazolidinones inhibit the inflammatory activation of cultured brain astrocytes and microglia by diminishing lipopolysaccharide (LPS)-induced IL-6, TNF-α, NOS-2 and COX-2 expression. Neuroprotective effects of thiadiazolidinones are completely inhibited by PPARs antagonists [113]. Clinical trials showed that rosiglitazone improves cognitive functions in apolipoprotein E4 (ApoE4)-negative patients [114]. Actually, there is evidence that reducing local inflammation with PPAR agonists improves the evolution of diseases such as Alzheimer's and Parkinson [115, 116], confirming that inflammation is a target for treatment.
In the CNS, synthesis of matrix CS by macrophages and oligo-dendrocyte progenitors is strongly up-regulated after a trauma [117]; CS inhibits post-injury axonal regeneration [118]. The CS growth inhibitory effect is considered to be a major obstacle for regeneration, and explain in part the lack of effective CNS recovery [119]. Interestingly, inhibition of CS synthesis immediately after injury impairs functional motor recovery and increases tissue loss, however, allowing CS synthesis during 48 hrs following injury, with subsequent inhibition, improves recovery by directly activating microglia/macrophages viathe CD44 receptor [120].
The Δdi-6S disaccharide modulates neuronal and microglial activities; in vitro, in neurons, Δdi-6S promotes neurite outgrowth and protects against neuronal toxicity and axonal collapse via protein kinase Cα (PKCα) and proline-rich tyrosine kinase 2 (PYK2) intracellular signalling pathways; in microglia, Δdi-6S transforms the phenotype of microglia in neuroprotective by means of the activation of Erk1/2 and PYK2. It is noteworthy that in vivo, systemically or locally injected Δdi-6S protects neurons in mice subjected to glutamate or aggregated β–amyloid intoxication [121, 122]. Recent evidence shows that by regulating multiple genes, Δdi-6S confers to microglia behaviour a phagocytic and anti-inflammatory profile [123]. The Δdi-6S may be a promising candidate for pharmacological development as a neuroprotective therapy for acute and chronic neurodegenerative disorders [124].
Glycosaminoglycans bind Aβ and can promote its aggregation [125]. The synthetic glycosaminoglycan 3-amino-1-propanesul-fonic acid (tramiprosate) was designed to interfere with the binding of glycosaminoglycans and Aβ. This interference should prevent conformational transitions that lead to the assembly of oligomers, protofibrils and fibrils, which ultimately results in plaque deposition. In vitro, tramiprosate has demonstrated anti-amyloid activity including inhibition of Aβ-induced neurotoxicity in neuronal cell cultures. In a phase II clinical trial in patients with mild-to-moderate Alzheimer's disease, tramiprosate decreased the CSF Aβ42 levels, reflecting a decrease of Aβ in the brain. However, there were no differences in the cognitive and clinical assessments between patients treated with tamiprosate or placebo, probably because 3 months follow-up is too short to record differences [126].
In human neuroblastoma SH-SY5Y cells subjected to oxida-tive stress, CS reduces the formation of reactive oxygen species (ROS) induced by both H2O2 (extracellular ROS) and Rot/oligo (intracellular ROS), effects associated with an increased AKT/PI3K phosphorylation and heme oxygenase-1 expression [127]. These studies are of particular interest, because it is known that antioxidants elicit a small beneficial effect on the evolution of Alzheimer's and Parkinson diseases [67, 128, 129].
It has been proposed that glaucoma may be considered as a neurodegenerative disease and treated as other neurodegenerative diseases are treated [130]. In vivo, the Δdi-6S disaccharide protects retinal ganglion cells from death caused by elevated intra ocular pressure in part through its control of microglial activity, for example Δdi-6S disaccharide activated the microglia through the activation of Erk1/2 and PYK2 but without increasing TNF-α secretion [131].
Following the intravenous administration of 131I labelled CS to rats, CS and disaccharides are found in the brain at similar concentrations, for example 0.2 and 0.3 μg/g, respectively, compared with blood concentrations of 1.9 and 0.4 μg/ml respec-tively; on the other hand, following oral administration of 131I labelled CS, brain concentrations of CS and disaccharides were 0.2 and 2.3 μg/g, respectively, compared with 2.1 and 6.3 μg/ml in blood [132]. In agreement with these results, systemic administration of the Δdi-6S disaccharide to mice elicits directly or indirectly an effect in the CNS [121, 122, 132]. These studies demonstrate that CS and its disaccharides penetrate the brain when given systemically.
Taken together, because of the anti-inflammatory and antioxi-dant effects of CS, further animal and human studies are warranted to determine whether glycosaminoglycans could become a new therapeutic strategy for neurodegenerative diseases.
Other autoimmune diseases that may benefit from chondroitin sulphate
The physiopathology of multiple sclerosis (MS) is complex and still incompletely characterized, but there is growing evidence that several factors contribute to the autoimmune response in MS lesions. MS is an autoimmune demyelinating disease of the CNS primarily mediated by Th1, Th2 and/or Th17 cells, which cross the blood-brain barrier [133, 134]. There is compelling evidence that the increase of Th17 by IL-6 plays a pivotal role in the appearance of autoimmunity, closely associated with the release of IL-17 [134, 135]. IL-17 induces the release of IL-1β and TNF-α from macrophages [136], by activating Erk1/2, p38MAPK and NF-κB[137].
In actively demyelinating plaques, the RelA, c-Rel and p50 sub-units of NF-κB are all present in macrophage nuclei in both parenchymal and perivascular areas; RelA is also found in the nuclei of a subset of hypertrophic astrocytes, suggesting that activation of NF-κB has a role in the evolution of MS [138]. In active MS lesions, NF-κB and JNK are up-regulated in oligodendrocytes located at the edge of active lesions and in microglia/macrophages throughout plaques [139]. There is evidence that NF-κB plays a central role in triggering molecular events in T cells responsible for acute relapse of MS [140].
The most abundant gene transcript present in early active MS lesions is αB-crystallin (CRYAB), whereas it is absent in normal brain tissue [141]. CRYAB has anti-apoptotic and neuroprotective functions [142], and it is the major target of CD4+ T-cell immunity to the myelin sheath from MS brain [143]. Astrocytes null for CRYAB display an activation of Erk1/2 and p38MAPK and up-regulated expression of NF-κB active subunits p65 and p105/p50, whereas the negative regulator κBa is down-regulated; in addition, mice Cryab−/- show significantly higher proliferation and secretion of the Th1 cytokines, for example IL-2, IFN-c, TNF-α and IL-12 and Th17 cytokines, for example IL-17 [144].
Taken together, the information available suggest that Th17, IL-17, the phosphorylation of Erk1/2 and p38MAPK and the activation of NF-κB nuclear translocation with further production and release of pro-inflammatory cytokines are important in the appearance and progression of MS. The sequence of events leading to MS allows raising the hypothesis that CS could block or diminish these events. Several arguments support such hypothesis. Firstly, there is evidence that blocking Th2 and mast cell activation improves the experimental autoimmune encephalomyelitis (EAE), an animal model of human MS [133], and CS is capable to down-regulate Th2 response [145, 146]. Secondly, it has been reported that inhibition of PLA2 and COX-2 delays the onset, prevents the development and reduces the severity of EAE and greatly reduces antigen-induced production of Th1 and Th17-type cytokines associated with autoimmune response [147]; it is well documented that CS down-regulates PLA2 [132] and COX-2 [148, 149]. Finally, glucosamine, a natural glucose derivative and an essential component of glycoproteins and PG, suppresses acute EAE, diminishing CNS inflammation and demyelination, effect probably associated with the blockade of Th1 response [150]. This review supports the hypothesis that CS might be an agent beneficial for the treatment of MS, and prompts further studies.
Conclusions
Despite the limitations of the in vitro and in vivo animal models because of differences in CS dosages, routes of administration and duration of exposure, this review supports that by inhibiting nuclear translocation of NF-κB and subsequent production of pro-inflammatory cytokines, and COX-2 and PLA2 expression and activity (Fig. 3), CS might potentially be of interest for the treatment of many inflammatory and autoimmune diseases, besides OA [151, 152]. Being CS a SYSADOA, we predict that an effect of CS could only be detected on long-term treatments, for example months. Because of the nature, the evolution and the difficulty to treat the inflammatory or autoimmune diseases listed in Figure 3, animal and human studies are warranted to test whether CS provides any beneficial effect.
Fig. 3.

Chondroitin sulphate (CS) by inhibiting the nuclear translation of the nuclear factor-KB (NF-κB), and consequently, the synthesis of pro-inflammatory cytokines, for example TNF-α and IL-1β, and of pro-inflammatory enzymes, for example cyclooxygenase-2 (COX-2) and phospholipase A2 (PLA2), may be of benefit for multiple autoimmune diseases.
Acknowledgments
This work was supported by “Cátedra UAM/Bioibérica de Inflamación Crónica y citoprotección (CABICYC), UAM, Madrid, Spain. We thank Marta Pulido, M.D., for editorial assistance.
References
- 1.Martel-Pelletier J, Boileau C, Pelletier JP, et al. Cartilage in normal and osteoarthritis conditions. Best Pract Res Clin Rheumatol. 2008;22:351–84. doi: 10.1016/j.berh.2008.02.001. [DOI] [PubMed] [Google Scholar]
- 2.Hardingham T. Extracellular matrix and pathogenic mechanisms in osteoarthritis. Curr Rheumatol Rep. 2008;10:30–6. doi: 10.1007/s11926-008-0006-9. [DOI] [PubMed] [Google Scholar]
- 3.Bali JP, Cousse H, Neuzil E. Biochemical basis of the pharmacologic action of chon-droitin sulfates on the osteoarticular system. Semin Arthritis Rheum. 2001;31:58–68. doi: 10.1053/sarh.2000.24874. [DOI] [PubMed] [Google Scholar]
- 4.Volpi N. Analytical aspects of pharmaceutical grade chondroitin sulfates. J Pharm Sci. 2007;96:3168–80. doi: 10.1002/jps.20997. [DOI] [PubMed] [Google Scholar]
- 5.Domagala F, Martin G, Bogdanowicz P, et al. Inhibition of interleukin-1beta-induced activation of MEK/ERK pathway and DNA binding of NF-kappaB and AP-1: potential mechanism for Diacerein effects in osteoarthritis. Biorheology. 2006;43:577–87. [PubMed] [Google Scholar]
- 6.Roman-Blas JA, Jimenez SA. NF-kappaB as a potential therapeutic target in osteoarthritis and rheumatoid arthritis. Osteoarthritis Cartilage. 2006;14:839–48. doi: 10.1016/j.joca.2006.04.008. [DOI] [PubMed] [Google Scholar]
- 7.Agarwal S, Deschner J, Long P, et al. Role of NF-kappaB transcription factors in antiinflammatory and proinflammatory actions of mechanical signals. Arthritis Rheum. 2004;50:3541–8. doi: 10.1002/art.20601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Liacini A, Sylvester J, Li WQ, et al. 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. 2002;21:251–62. doi: 10.1016/s0945-053x(02)00007-0. [DOI] [PubMed] [Google Scholar]
- 9.Lianxu C, Hongti J, Changlong Y. NF-kappaBp65-specific siRNA inhibits expression of genes of COX-2, NOS-2 and MMP-9 in rat IL-1beta-induced and TNF-alpha-induced chondrocytes. Osteoarthritis Cartilage. 2006;14:467–76. doi: 10.1016/j.joca.2005.10.009. [DOI] [PubMed] [Google Scholar]
- 10.Mengshol JA, Vincenti MP, Coon CI, et al. Interleukin-1 induction of collage-nase 3 (matrix metalloproteinase 13) gene expression in chondrocytes requires p38, c-Jun N-terminal kinase, and nuclear factor kappaB: differential regulation of colla-genase 1 and collagenase 3. Arthritis Rheum. 2000;43:801–11. doi: 10.1002/1529-0131(200004)43:4<801::AID-ANR10>3.0.CO;2-4. [DOI] [PubMed] [Google Scholar]
- 11.Wada Y, Shimada K, Sugimoto K, et al. Novel p38 mitogen-activated protein kinase inhibitor R-130823 protects cartilage by down-regulating matrix metallo-proteinase-1,-13 and prostaglandin E2 production in human chondrocytes. Int Immunopharmacol. 2006;6:144–55. doi: 10.1016/j.intimp.2005.07.009. [DOI] [PubMed] [Google Scholar]
- 12.Berenbaum F, Humbert L, Bereziat G, et al. Concomitant recruitment of ERK1/2 and p38 MAPK signalling pathway is required for activation of cytoplasmic phos-pholipase A2 via ATP in articular chondro-cytes. J Biol Chem. 2003;278:13680–7. doi: 10.1074/jbc.M211570200. [DOI] [PubMed] [Google Scholar]
- 13.Wen D, Nong Y, Morgan JG, et al. A Selective small molecule IkappaB inhibitor blocks nuclear factor kappaB-mediated inflammatory responses in human fibrob-last-like synoviocytes, chondrocytes and mast cells. J Pharmacol Exp Ther. 2006;317:989–1001. doi: 10.1124/jpet.105.097584. [DOI] [PubMed] [Google Scholar]
- 14.Jomphe C, Gabriac M, Hale TM, et al. Chondroitin sulfate inhibits the nuclear translocation of nuclear factor-kappaB in interleukin-1beta-stimulated chondro-cytes. Basic Clin Pharmacol Toxicol. 2008;102:59–65. doi: 10.1111/j.1742-7843.2007.00158.x. [DOI] [PubMed] [Google Scholar]
- 15.Benito MJ, Veale DJ, FitzGerald O, et al. Synovial tissue inflammation in early and late osteoarthritis. Ann Rheum Dis. 2005;64:1263–7. doi: 10.1136/ard.2004.025270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Firestein GS. Evolving concepts of rheumatoid arthritis. Nature. 2003;423:356–61. doi: 10.1038/nature01661. [DOI] [PubMed] [Google Scholar]
- 17.Li X, Makarov SS. An essential role of NF-kappaB in the “tumor-like” phenotype of arthritic synoviocytes. Proc Natl Acad Sci USA. 2006;103:17432–7. doi: 10.1073/pnas.0607939103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Tas SW, Vervoordeldonk MJ, Hajji N, et al. Local treatment with the selective IkappaB kinase beta inhibitor NEMO-binding domain peptide ameliorates synovial inflammation. Arthritis Res Ther. 2006;8:R86. doi: 10.1186/ar1958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Tas SW, Hajji N, Stenvers DJ, et al. Reduction of proinflammatory cytokine expression in the synovium by targeting IKKbeta in vivo in a rat model. Arthritis Rheum. 2006;54:3716–8. doi: 10.1002/art.22188. [DOI] [PubMed] [Google Scholar]
- 20.Iovu M, Dumais G, du Souich P. Anti-inflammatory activity of chondroitin sul-fate. Osteoarthritis Cartilage. 2008;16:S14–8. doi: 10.1016/j.joca.2008.06.008. [DOI] [PubMed] [Google Scholar]
- 21.Clegg DO, Reda DJ, Harris CL, et al. Glucosamine, chondroitin sulfate, and the two in combination for painful knee osteoarthritis. N Engl J Med. 2006;354:795–808. doi: 10.1056/NEJMoa052771. [DOI] [PubMed] [Google Scholar]
- 22.Omata T, Itokazu Y, Inoue N, et al. Effects of chondroitin sulfate-C on articular cartilage destruction in murine collagen-induced arthritis. Arzneimittelforschung. 2000;50:148–53. doi: 10.1055/s-0031-1300180. [DOI] [PubMed] [Google Scholar]
- 23.Lauder SN, Carty SM, Carpenter CE, et al. Interleukin-1beta induced activation of nuclear factor-kappab can be inhibited by novel pharmacological agents in osteoarthritis. Rheumatology. 2007;46:752–8. doi: 10.1093/rheumatology/kel419. [DOI] [PubMed] [Google Scholar]
- 24.Saklatvala J. Inflammatory signaling in cartilage: MAPK and NF-kappaB pathways in chondrocytes and the use of inhibitors for research into pathogenesis and therapy of osteoarthritis. Curr Drug Targets. 2007;8:305–13. doi: 10.2174/138945007779940115. [DOI] [PubMed] [Google Scholar]
- 25.Xu H, He Y, Yang X, et al. Anti-malarial agent artesunate inhibits TNF-alpha-induced production of proinflammatory cytokines via inhibition of NF-kappaB and PI3 kinase/Akt signal pathway in human rheumatoid arthritis fibroblast-like synoviocytes. Rheumatology. 2007;46:920–6. doi: 10.1093/rheumatology/kem014. [DOI] [PubMed] [Google Scholar]
- 26.Alvarez-Soria MA, Largo R, Santillana J, et al. Differential anticatabolic profile of glucosamine sulfate versus other antios-teoarthritic drugs on human osteoarthritic chondrocytes and synovial fibroblast in culture. Osteoarthritis Cartilage. 2005;13:S153.27. [Google Scholar]
- 27.Volpi N. Oral bioavailability of chondroitin sulfate (Condrosulf) and its constituents in healthy male volunteers. Osteoarthritis Cartilage. 2002;10:768–77. doi: 10.1053/joca.2002.0824. [DOI] [PubMed] [Google Scholar]
- 28.Monfort J, Pelletier JP, Garcia-Giralt N, et al. Biochemical basis of the effect of chondroitin sulphate on osteoarthritis articular tissues. Ann Rheum Dis. 2008;67:735–40. doi: 10.1136/ard.2006.068882. [DOI] [PubMed] [Google Scholar]
- 29.Tat SK, Pelletier JP, Verges J, et al. Chondroitin and glucosamine sulfate in combination decrease the pro-resorptive properties of human osteoarthritis sub-chondral bone osteoblasts: a basic science study. Arthritis Res Ther. 2007;9:R117. doi: 10.1186/ar2325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kwan Tat S, Pelletier JP, Lajeunesse D, et al. The differential expression of osteoprotegerin (OPG) and receptor activator of nuclear factor kappaB ligand (RANKL) in human osteoarthritic subchondral bone osteoblasts is an indicator of the metabolic state of these disease cells. Clin Exp Rheumatol. 2008;26:295–304. [PMC free article] [PubMed] [Google Scholar]
- 31.Tsubaki M, Kato C, Manno M, et al. Macrophage inflammatory protein-1alpha (MIP-1alpha) enhances a receptor activator of nuclear factor kappaB ligand (RANKL) expression in mouse bone marrow stromal cells and osteoblasts through MAPK and PI3K/Akt pathways. Mol Cell Biochem. 2007;304:53–60. doi: 10.1007/s11010-007-9485-7. [DOI] [PubMed] [Google Scholar]
- 32.Barker SA, Hawkins CF, Hewins M. Mucopolysaccharides in synovial fluid detection of chondroitin sulphate. Ann Rheum Dis. 1966;25:209–13. doi: 10.1136/ard.25.3.209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Lewis S, Crossman M, Flannelly J, et al. Chondroitin sulphation patterns in synovial fluid in osteoarthritis subsets. Ann Rheum Dis. 1999;58:441–5. doi: 10.1136/ard.58.7.441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Aigner T, Sachse A, Gebhard PM, et al. Osteoarthritis: pathobiology-targets and ways for therapeutic intervention. Adv Drug Deliv Rev. 2006;58:128–49. doi: 10.1016/j.addr.2006.01.020. [DOI] [PubMed] [Google Scholar]
- 35.Forsyth CB, Pulai J, Loeser RF. Fibronectin fragments and blocking antibodies to alpha2beta1 and alpha5beta1 integrins stimulate mitogen-activated protein kinase signaling and increase collage-nase 3 (matrix metalloproteinase 13) production by human articular chondrocytes. Arthritis Rheum. 2002;46:2368–76. doi: 10.1002/art.10502. [DOI] [PubMed] [Google Scholar]
- 36.Homandberg GA, Costa V, Wen C. Fibronectin fragments active in chondrocytic chondrolysis can be chemically cross-linked to the alpha5 integrin receptor subunit. Osteoarthritis Cartilage. 2002;10:938–49. doi: 10.1053/joca.2002.0854. [DOI] [PubMed] [Google Scholar]
- 37.Reyes CD, Petrie TA, Garcia AJ. Mixed extracellular matrix ligands synergistically modulate integrin adhesion and signaling. J Cell Physiol. 2008;217:450–8. doi: 10.1002/jcp.21512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Loeser RF. Molecular mechanisms of cartilage destruction: mechanics, inflammatory mediators, and aging collide. Arthritis Rheum. 2006;54:1357–60. doi: 10.1002/art.21813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Li Y, Xu L, Olsen BR. Lessons from genetic forms of osteoarthritis for the pathogenesis of the disease. Osteoarthritis Cartilage. 2007;15:1101–5. doi: 10.1016/j.joca.2007.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Goldring MB. Update on the biology of the chondrocyte and new approaches to treating cartilage diseases. Best Pract Res Clin Rheumatol. 2006;20:1003–25. doi: 10.1016/j.berh.2006.06.003. [DOI] [PubMed] [Google Scholar]
- 41.Luyten FP, Lories RJ, Verschueren P, et al. Contemporary concepts of inflammation, damage and repair in rheumatic diseases. Best Pract Res Clin Rheumatol. 2006;20:829–48. doi: 10.1016/j.berh.2006.06.009. [DOI] [PubMed] [Google Scholar]
- 42.Ge Z, Hu Y, Heng BC, et al. Osteoarthritis and therapy. Arthritis Rheum. 2006;55:493–500. doi: 10.1002/art.21994. [DOI] [PubMed] [Google Scholar]
- 43.Goldring MB. The role of cytokines as inflammatory mediators in osteoarthritis: lessons from animal models. Connect Tissue Res. 1999;40:1–11. doi: 10.3109/03008209909005273. [DOI] [PubMed] [Google Scholar]
- 44.Uebelhart D, Malaise M, Marcolongo R, et al. Intermittent treatment of knee osteoarthritis with oral chondroitin sulfate: a one-year, randomized, double-blind, multi-center study versus placebo. Osteoarthritis Cartilage. 2004;12:269–76. doi: 10.1016/j.joca.2004.01.004. [DOI] [PubMed] [Google Scholar]
- 45.Mazieres B, Combe B, Phan Van A, et al. Chondroitin sulfate in osteoarthritis of the knee: a prospective, double blind, placebo controlled multicenter clinical study. J Rheumatol. 2001;28:173–81. [PubMed] [Google Scholar]
- 46.Uebelhart D, Thonar EJ, Delmas PD, et al. Effects of oral chondroitin sulfate on the progression of knee osteoarthritis: a pilot study. Osteoarthritis Cartilage. 1998;6:39–46. doi: 10.1016/s1063-4584(98)80011-3. [DOI] [PubMed] [Google Scholar]
- 47.Verbruggen G, Goemaere S, Veys EM. Systems to assess the progression of finger joint osteoarthritis and the effects of disease modifying osteoarthritis drugs. Clin Rheumatol. 2002;21:231–43. doi: 10.1007/s10067-002-8290-7. [DOI] [PubMed] [Google Scholar]
- 48.Rovetta G, Monteforte P, Molfetta G, et al. Chondroitin sulfate in erosive osteoarthritis of the hands. Int J Tissue React. 2002;24:29–32. [PubMed] [Google Scholar]
- 49.Volpi N. The pathobiology of osteoarthritis and the rationale for using the chondroitin sulfate for its treatment. Curr Drug Targets Immune Endocr Metabol Disord. 2004;4:119–27. doi: 10.2174/1568008043339929. [DOI] [PubMed] [Google Scholar]
- 50.Legendre F, Bauge C, Roche R, et al. Chondroitin sulfate modulation of matrix and inflammatory gene expression in IL-1beta-stimulated chondrocytes–study in hypoxic alginate bead cultures. Osteoarthritis Cartilage. 2008;16:105–14. doi: 10.1016/j.joca.2007.05.020. [DOI] [PubMed] [Google Scholar]
- 51.Pahl HL. Activators and target genes of Rel/NF-kappaB transcription factors. Oncogene. 1999;18:6853–66. doi: 10.1038/sj.onc.1203239. [DOI] [PubMed] [Google Scholar]
- 52.Uwe S. Anti-inflammatory interventions of NF-kappaB signaling: potential applications and risks. Biochem Pharmacol. 2008;75:1567–79. doi: 10.1016/j.bcp.2007.10.027. [DOI] [PubMed] [Google Scholar]
- 53.Courtois G, Gilmore TD. Mutations in the NF-kappaB signaling pathway: implications for human disease. Oncogene. 2006;25:6831–43. doi: 10.1038/sj.onc.1209939. [DOI] [PubMed] [Google Scholar]
- 54.Montell E, Iovu M, Héroux L, et al. Chronic administrationof chondroitin sulfate does not affect cytochrome P450 nor NADPH P450 reductase. Osteoarthritis Cartilage. 2008;16:S226–7. [Google Scholar]
- 55.Bell S, Degitz K, Quirling M, et al. Involvement of NF-kappaB signalling in skin physiology and disease. Cell Signal. 2003;15:1–7. doi: 10.1016/s0898-6568(02)00080-3. [DOI] [PubMed] [Google Scholar]
- 56.Westergaard M, Henningsen J, Johansen C, et al. Expression and localization of peroxisome proliferator-activated receptors and nuclear factor kappaB in normal and lesional psoriatic skin. J Invest Dermatol. 2003;121:1104–17. doi: 10.1046/j.1523-1747.2003.12536.x. [DOI] [PubMed] [Google Scholar]
- 57.Abdou AG, Hanout HM. Evaluation of sur-vivin and NF-kappaB in psoriasis, an immunohistochemical study. J Cutan Pathol. 2008;35:445–51. doi: 10.1111/j.1600-0560.2007.00841.x. [DOI] [PubMed] [Google Scholar]
- 58.Johansen C, Flindt E, Kragballe K, et al. Inverse regulation of the nuclear factor-kappaB binding to the p53 and interleukin-8 kappaB response elements in lesional pso-riatic skin. J Invest Dermatol. 2005;124:1284–92. doi: 10.1111/j.0022-202X.2005.23749.x. [DOI] [PubMed] [Google Scholar]
- 59.Victor FC, Gottlieb AB, Menter A. Changing paradigms in dermatology: tumor necrosis factor alpha (TNF-alpha) blockade in psoriasis and psoriatic arthritis. Clin Dermatol. 2003;21:392–7. doi: 10.1016/j.clindermatol.2003.08.015. [DOI] [PubMed] [Google Scholar]
- 60.Yano S, Banno T, Walsh R, et al. Transcriptional responses of human epidermal keratinocytes to cytokine inter-leukin-1. J Cell Physiol. 2008;214:1–13. doi: 10.1002/jcp.21300. [DOI] [PubMed] [Google Scholar]
- 61.Li H, Gao L, Shen Z, et al. Association study of NFKB1 and SUMO4 polymorphisms in Chinese patients with psoriasis vulgaris. Arch Dermatol Res. 2008;300:425–33. doi: 10.1007/s00403-008-0843-4. [DOI] [PubMed] [Google Scholar]
- 62.Lizzul PF, Aphale A, Malaviya R, et al. Differential expression of phosphorylated NF-kappaB/RelA in normal and psoriatic epidermis and downregulation of NF-kappaB in response to treatment with etanercept. J Invest Dermatol. 2005;124:1275–83. doi: 10.1111/j.0022-202X.2005.23735.x. [DOI] [PubMed] [Google Scholar]
- 63.Wildfeuer A. [6-hydroxy-1,3-ben-zoxathiol- 2-one, an antipsoriatic with antibacterial and antimycotic properties] Arzneimittelforschung. 1970;20:824–31. [PubMed] [Google Scholar]
- 64.Kim MH, Lee HY, Roh E, et al. Novel imi-nobenzoxathiolone compound inhibits nuclear factor-kappaB activation targeting inhibitory kappaB kinase beta and down-regulating interleukin-1beta expression in lipopolysaccharide-activated macrophages. Biochem Pharmacol. 2008;76:373–81. doi: 10.1016/j.bcp.2008.05.013. [DOI] [PubMed] [Google Scholar]
- 65.Amigo M, Paya M, De Rosa S, et al. Antipsoriatic effects of avarol-3’-thiosalicy-late are mediated by inhibition of TNF-alpha generation and NF-kappaB activation in mouse skin. Br J Pharmacol. 2007;152:353–65. doi: 10.1038/sj.bjp.0707394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Meili-Butz S, Niermann T, Fasler-Kan E, et al. Dimethyl fumarate, a small molecule drug for psoriasis, inhibits nuclear factor-kappaB and reduces myocardial infarct size in rats. Eur J Pharmacol. 2008;586:251–8. doi: 10.1016/j.ejphar.2008.02.038. [DOI] [PubMed] [Google Scholar]
- 67.Hatcher H, Planalp R, Cho J, et al. Curcumin: from ancient medicine to current clinical trials. Cell Mol Life Sci. 2008;65:1631–52. doi: 10.1007/s00018-008-7452-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Lan CC, Yu HS, Wu CS, et al. FK506 inhibits tumour necrosis factor-alpha secretion in human keratinocytes via regulation of nuclear factor-kappaB. Br J Dermatol. 2005;153:725–32. doi: 10.1111/j.1365-2133.2005.06779.x. [DOI] [PubMed] [Google Scholar]
- 69.Vergés J, Montell E, Herrero M, et al. Clinical and histopathological improvement of psoriasis in patients with osteoarthritis treated with chondroitin sul-fate: report of 3 cases. Med Clin. 2004;123:739–42. doi: 10.1016/s0025-7753(04)74654-0. [DOI] [PubMed] [Google Scholar]
- 70.Verges J, Montell E, Herrero M, et al. Clinical and histopathological improvement of psoriasis with oral chondroitin sulfate: a serendipitous finding. Dermatol Online J. 2005;11:31. [PubMed] [Google Scholar]
- 71.Kurita H. Über den einfluss des chon-droitin sulfates auf die experimentelle ath-erosklerose in kaninchen. Med J Shinshu University. 1955;1:23–7. [Google Scholar]
- 72.Murata K. Infra-red spectroscopic evidence of chondroitin polysulphate and its relation to anticoagulant activity. Nature. 1962;193:578–9. doi: 10.1038/193578a0. [DOI] [PubMed] [Google Scholar]
- 73.Murata K. The effects of sulfated polysac-charides obtained from seaweeds on experimental atherosclerosis. J Gerontol. 1962;17:30–6. doi: 10.1093/geronj/17.1.30. [DOI] [PubMed] [Google Scholar]
- 74.Nakamura M, Ishihara Y, Sata T, et al. Acid mucopolysaccharides and lipids of Japanese arteries, with special reference to the absence of hyaluronic acid in Japanese cerebral artery. J Atheroscler Res. 1966;6:132–50. doi: 10.1016/s0368-1319(66)80018-2. [DOI] [PubMed] [Google Scholar]
- 75.Morrison LM. Response of ischemic heart disease to chondroitin sulfate-A. J Am Geriatr Soc. 1969;17:913–23. doi: 10.1111/j.1532-5415.1969.tb02328.x. [DOI] [PubMed] [Google Scholar]
- 76.Morrison LM, Enrick N. Coronary heart disease: reduction of death rate by chondroitin sulfate A. Angiology. 1973;24:269–87. doi: 10.1177/000331977302400503. [DOI] [PubMed] [Google Scholar]
- 77.Ridker PM. Inflammation, infection, and cardiovascular risk: how good is the clinical evidence? Circulation. 1998;97:1671–4. doi: 10.1161/01.cir.97.17.1671. [DOI] [PubMed] [Google Scholar]
- 78.Ridker PM, Rifai N, Pfeffer MA, et al. Inflammation, pravastatin, and the risk of coronary events after myocardial infarction in patients with average cholesterol levels. Cholesterol and Recurrent Events (CARE) investigators. Circulation. 1998;98:839–44. doi: 10.1161/01.cir.98.9.839. [DOI] [PubMed] [Google Scholar]
- 79.Zhang H, Park Y, Wu J, et al. Role of TNF-alpha in vascular dysfunction. Clin Sci. 2009;116:219–30. doi: 10.1042/CS20080196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Ridker PM, Hennekens CH, Buring JE, et al. C-reactive protein and other markers of inflammation in the prediction of cardiovascular disease in women. N Engl J Med. 2000;342:836–43. doi: 10.1056/NEJM200003233421202. [DOI] [PubMed] [Google Scholar]
- 81.Gutierrez SH, Kuri MR, del Castillo ER. Cardiac role of the transcription factor NF-kappaB. Cardiovasc Hematol Disord Drug Targets. 2008;8:153–60. doi: 10.2174/187152908784533702. [DOI] [PubMed] [Google Scholar]
- 82.Chung JH, Seo AY, Chung SW, et al. Molecular mechanism of PPAR in the regulation of age-related inflammation. Ageing Res Rev. 2008;7:126–36. doi: 10.1016/j.arr.2008.01.001. [DOI] [PubMed] [Google Scholar]
- 83.Steinhubl SR, Badimon JJ, Bhatt DL, et al. Clinical evidence for anti-inflammatory effects of antiplatelet therapy in patients with atherothrombotic disease. Vasc Med. 2007;12:113–22. doi: 10.1177/1358863X07077462. [DOI] [PubMed] [Google Scholar]
- 84.Tintut Y, Demer L. Role of osteoprotegerin and its ligands and competing receptors in atherosclerotic calcification. J Investig Med. 2006;54:395–401. doi: 10.2310/6650.2006.06019. [DOI] [PubMed] [Google Scholar]
- 85.Montecucco F, Steffens S, Mach F. The immune response is involved in atherosclerotic plaque calcification: could the RANKL/RANK/OPG system be a marker of plaque instability? Clin Dev Immunol. 2007;2007:75805. doi: 10.1155/2007/75805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Herrero-Beaumont G, Marcos ME, Sanchez-Pernaute O, et al. Effect of chondroitin sulphate in a rabbit model of atherosclerosis aggravated by chronic arthritis. Br J Pharmacol. 2008;154:843–51. doi: 10.1038/bjp.2008.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Largo R, Sanchez-Pernaute O, Marcos ME, et al. Chronic arthritis aggravates vascular lesions in rabbits with atherosclerosis: a novel model of atherosclerosis associated with chronic inflammation. Arthritis Rheum. 2008;58:2723–34. doi: 10.1002/art.23765. [DOI] [PubMed] [Google Scholar]
- 88.Ridker PM. Inflammation, atherosclerosis, and cardiovascular risk: an epidemiologic view. Blood Coagul Fibrinolysis. 1999;10:S9–12. [PubMed] [Google Scholar]
- 89.Zhang SZ, Zhao XH, Zhang DC. Cellular and molecular immunopathogenesis of ulcerative colitis. Cell Mol Immunol. 2006;3:35–40. [PubMed] [Google Scholar]
- 90.Atreya I, Atreya R, Neurath MF. NF-kappaB in inflammatory bowel disease. J Intern Med. 2008;263:591–6. doi: 10.1111/j.1365-2796.2008.01953.x. [DOI] [PubMed] [Google Scholar]
- 91.Danese S. Mechanisms of action of infliximab in inflammatory bowel disease: an anti-inflammatory multitasker. Dig Liver Dis. 2008;40:S225–8. doi: 10.1016/S1590-8658(08)60530-7. [DOI] [PubMed] [Google Scholar]
- 92.Ferrero-Miliani L, Nielsen OH, Andersen PS, et al. Chronic inflammation: importance of NOD2 and NALP3 in interleukin-1beta generation. Clin Exp Immunol. 2007;147:227–35. doi: 10.1111/j.1365-2249.2006.03261.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Harris G, KuoLee R, Chen W. Role of Tolllike receptors in health and diseases of gastrointestinal tract. World J Gastroenterol. 2006;12:2149–60. doi: 10.3748/wjg.v12.i14.2149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Saubermann LJ, Nakajima A, Wada K, et al. Peroxisome proliferator-activated receptor gamma agonist ligands stimulate a Th2 cytokine response and prevent acute colitis. Inflamm Bowel Dis. 2002;8:330–9. doi: 10.1097/00054725-200209000-00004. [DOI] [PubMed] [Google Scholar]
- 95.Takaki K, Mitsuyama K, Tsuruta O, et al. Attenuation of experimental colonic injury by thiazolidinedione agents. Inflamm Res. 2006;55:10–5. doi: 10.1007/s00011-005-0002-8. [DOI] [PubMed] [Google Scholar]
- 96.Lewis JD, Lichtenstein GR, Deren JJ, et al. Rosiglitazone for active ulcerative colitis: a randomized placebo-controlled trial. Gastroenterology. 2008;134:688–95. doi: 10.1053/j.gastro.2007.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Schreiber S, Nikolaus S, Hampe J. Activation of nuclear factor kappa B inflammatory bowel disease. Gut. 1998;42:477–84. doi: 10.1136/gut.42.4.477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Schreiber S, Nikolaus S, Hampe J, et al. Tumour necrosis factor alpha and interleukin 1beta in relapse of Crohn's disease. Lancet. 1999;353:459–61. doi: 10.1016/S0140-6736(98)03339-X. [DOI] [PubMed] [Google Scholar]
- 99.Zheng P, Niu FL, Liu WZ, et al. Anti-inflammatory mechanism of oxymatrine in dextran sulfate sodium-induced colitis of rats. World J Gastroenterol. 2005;11:4912–5. doi: 10.3748/wjg.v11.i31.4912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Islam MS, Murata T, Fujisawa M, et al. Anti-inflammatory effects of phytosteryl ferulates in colitis induced by dextran sulphate sodium in mice. Br J Pharmacol. 2008;154:812–24. doi: 10.1038/bjp.2008.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Egan LJ, Toruner M. NF-kappaB signaling: pros and cons of altering NF-kappaB as a therapeutic approach. Ann N Y Acad Sci. 2006;1072:114–22. doi: 10.1196/annals.1326.009. [DOI] [PubMed] [Google Scholar]
- 102.Behm BW, Bickston SJ. Tumor necrosis factor-alpha antibody for maintenance of remission in Crohn's disease. Cochrane Database Syst Rev. 2008:CD006893. doi: 10.1002/14651858.CD006893. [DOI] [PubMed] [Google Scholar]
- 103.Wilhelm SM, McKenney KA, Rivait KN, et al. A review of infliximab use in ulcera-tive colitis. Clin Ther. 2008;30:223–30. doi: 10.1016/j.clinthera.2008.02.014. [DOI] [PubMed] [Google Scholar]
- 104.Hori Y, Hoshino J, Yamazaki C, et al. Effects of chondroitin sulfate on colitis induced by dextran sulfate sodium in rats. Jpn J Pharmacol. 2001;85:155–60. doi: 10.1254/jjp.85.155. [DOI] [PubMed] [Google Scholar]
- 105.Ota S, Yoshihara S, Ishido K, et al. Effects of proteoglycan on dextran sulfate sodium-induced experimental colitis in rats. Dig Dis Sci. 2008;53:3176–83. doi: 10.1007/s10620-008-0304-0. [DOI] [PubMed] [Google Scholar]
- 106.Yomogida S, Kojima Y, Tsutsumi-Ishii Y, et al. Glucosamine, a naturally occurring amino monosaccharide, suppresses dex-tran sulfate sodium-induced colitis in rats. Int J Mol Med. 2008;22:317–23. [PubMed] [Google Scholar]
- 107.Rogers J, Mastroeni D, Leonard B, et al. Neuroinflammation in Alzheimer's disease and Parkinson's disease: are microglia pathogenic in either disorder? Int Rev Neurobiol. 2007;82:235–46. doi: 10.1016/S0074-7742(07)82012-5. [DOI] [PubMed] [Google Scholar]
- 108.Memet S. NF-kappaB functions in the nervous system: from development to disease. Biochem Pharmacol. 2006;72:1180–95. doi: 10.1016/j.bcp.2006.09.003. [DOI] [PubMed] [Google Scholar]
- 109.Sriram K, O’Callaghan JP. Divergent roles for tumor necrosis factor-alpha in the brain. J Neuroimmune Pharmacol. 2007;2:140–53. doi: 10.1007/s11481-007-9070-6. [DOI] [PubMed] [Google Scholar]
- 110.Eriksen JL, Sagi SA, Smith TE, et al. NSAIDs and enantiomers of flurbiprofen target gamma-secretase and lower Abeta 42 in vivo. J Clin Invest. 2003;112:440–9. doi: 10.1172/JCI18162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Aisen PS. The potential of anti-inflammatory drugs for the treatment of Alzheimer's disease. Lancet Neurol. 2002;1:279–84. doi: 10.1016/s1474-4422(02)00133-3. [DOI] [PubMed] [Google Scholar]
- 112.Aisen PS, Schafer KA, Grundman M, et al. Effects of rofecoxib or naproxen vs placebo on Alzheimer disease progression: a randomized controlled trial. JAMA. 2003;289:2819–26. doi: 10.1001/jama.289.21.2819. [DOI] [PubMed] [Google Scholar]
- 113.Luna-Medina R, Cortes-Canteli M, Alonso M, et al. Regulation of inflammatory response in neural cells in vitro by thiadiazolidinones derivatives through peroxisome proliferator-activated receptor gamma activation. J Biol Chem. 2005;280:21453–62. doi: 10.1074/jbc.M414390200. [DOI] [PubMed] [Google Scholar]
- 114.Risner ME, Saunders AM, Altman JF, et al. Efficacy of rosiglitazone in a genetically defined population with mild-to-moderate Alzheimer's disease. Pharmacogenomics J. 2006;6:246–54. doi: 10.1038/sj.tpj.6500369. [DOI] [PubMed] [Google Scholar]
- 115.Bright JJ, Kanakasabai S, Chearwae W, et al. PPAR regulation of inflammatory signaling in CNS diseases. PPAR Res. 2008;2008:658520. doi: 10.1155/2008/658520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Chaturvedi RK, Beal MF. PPAR: a therapeutic target in Parkinson's disease. J Neurochem. 2008;106:506–18. doi: 10.1111/j.1471-4159.2008.05388.x. [DOI] [PubMed] [Google Scholar]
- 117.Jones LL, Yamaguchi Y, Stallcup WB, et al. NG2 is a major chondroitin sulfate proteoglycan produced after spinal cord injury and is expressed by macrophages and oligodendrocyte progenitors. J Neurosci. 2002;22:2792–803. doi: 10.1523/JNEUROSCI.22-07-02792.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Grimpe B, Silver J. The extracellular matrix in axon regeneration. Prog Brain Res. 2002;137:333–49. doi: 10.1016/s0079-6123(02)37025-0. [DOI] [PubMed] [Google Scholar]
- 119.Rhodes KE, Fawcett JW. Chondroitin sulphate proteoglycans: preventing plasticity or protecting the CNS? J Anat. 2004;204:33–48. doi: 10.1111/j.1469-7580.2004.00261.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Rolls A, Shechter R, London A, et al. Two faces of chondroitin sulfate proteoglycan in spinal cord repair: a role in microglia/macrophage activation. PLoS Med. 2008;5:e171. doi: 10.1371/journal.pmed.0050171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Rolls A, Avidan H, Cahalon L, et al. A dis-accharide derived from chondroitin sulphate proteoglycan promotes central nervous system repair in rats and mice. Eur J Neurosci. 2004;20:1973–83. doi: 10.1111/j.1460-9568.2004.03676.x. [DOI] [PubMed] [Google Scholar]
- 122.Rolls A, Cahalon L, Bakalash S, et al. A sulfated disaccharide derived from chondroitin sulfate proteoglycan protects against inflammation-associated neurode-generation. FASEB J. 2006;20:547–9. doi: 10.1096/fj.05-4540fje. [DOI] [PubMed] [Google Scholar]
- 123.Ebert S, Schoeberl T, Walczak Y, et al. Chondroitin sulfate disaccharide stimulates microglia to adopt a novel regulatory phenotype. J Leukoc Biol. 2008;84:736–40. doi: 10.1189/jlb.0208138. [DOI] [PubMed] [Google Scholar]
- 124.Rolls A, Shechter R, Schwartz M. The bright side of the glial scar in CNS repair. Nat Rev Neurosci. 2009;10:235–41. doi: 10.1038/nrn2591. [DOI] [PubMed] [Google Scholar]
- 125.van Horssen J, Wesseling P, van den Heuvel LP, et al. Heparan sulphate proteo-glycans in Alzheimer's disease and amyloid-related disorders. Lancet Neurol. 2003;2:482–92. doi: 10.1016/s1474-4422(03)00484-8. [DOI] [PubMed] [Google Scholar]
- 126.Aisen PS, Saumier D, Briand R, et al. A phase II study targeting amyloid-beta with 3APS in mild-to-moderate Alzheimer disease. Neurology. 2006;67:1757–63. doi: 10.1212/01.wnl.0000244346.08950.64. [DOI] [PubMed] [Google Scholar]
- 127.Canas N, Valero T, Villarroya M, et al. Chondroitin sulfate protects SH-SY5Y cells from oxidative stress by inducing heme oxygenase-1 via phosphatidylinositol 3-kinase/Akt. J Pharmacol Exp Ther. 2007;323:946–53. doi: 10.1124/jpet.107.123505. [DOI] [PubMed] [Google Scholar]
- 128.Ancelin ML, Christen Y, Ritchie K. Is antioxidant therapy a viable alternative for mild cognitive impairment? Examination of the evidence. Dement Geriatr Cogn Disord. 2007;24:1–19. doi: 10.1159/000102567. [DOI] [PubMed] [Google Scholar]
- 129.Vafeiadou K, Vauzour D, Spencer JP. Neuroinflammation and its modulation by flavonoids. Endocr Metab Immune Disord Drug Targets. 2007;7:211–24. doi: 10.2174/187153007781662521. [DOI] [PubMed] [Google Scholar]
- 130.Schwartz M. Neuroprotection as a treatment for glaucoma: pharmacological and immunological approaches. Eur J Ophthalmol. 2001;11:S7–11. doi: 10.1177/112067210101102s01. [DOI] [PubMed] [Google Scholar]
- 131.Bakalash S, Rolls A, Lider O, et al. Chondroitin sulfate-derived disaccharide protects retinal cells from elevated intraocular pressure in aged and immunocompromised rats. Invest Ophthalmol Vis Sci. 2007;48:1181–90. doi: 10.1167/iovs.05-1213. [DOI] [PubMed] [Google Scholar]
- 132.Ronca F, Palmieri L, Panicucci P, et al. Anti-inflammatory activity of chondroitin sulfate. Osteoarthritis Cartilage. 1998;6:14–21. doi: 10.1016/s1063-4584(98)80006-x. [DOI] [PubMed] [Google Scholar]
- 133.Theoharides TC, Kempuraj D, Kourelis T, et al. Human mast cells stimulate activated T cells: implications for multiple sclerosis. Ann N Y Acad Sci. 2008;1144:74–82. doi: 10.1196/annals.1418.029. [DOI] [PubMed] [Google Scholar]
- 134.Awasthi A, Murugaiyan G, Kuchroo VK. Interplay between effector Th17 and regulatory T cells. J Clin Immunol. 2008;28:660–70. doi: 10.1007/s10875-008-9239-7. [DOI] [PubMed] [Google Scholar]
- 135.Montes M, Zhang X, Berthelot L, et al. Oligoclonal myelin-reactive T-cell infiltrates derived from multiple sclerosis lesions are enriched in Th17 cells. Clin Immunol. 2009;130:133–44. doi: 10.1016/j.clim.2008.08.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Jovanovic DV, Di Battista JA, Martel-Pelletier J, et al. IL-17 stimulates the production and expression of proinflamma-tory cytokines, IL-beta and TNF-alpha, by human macrophages. J Immunol. 1998;160:3513–21. [PubMed] [Google Scholar]
- 137.Martel-Pelletier J, Mineau F, Jovanovic D, et al. Mitogen-activated protein kinase and nuclear factor kappaB together regulate interleukin-17-induced nitric oxide production in human osteoarthritic chondrocytes: possible role of transactivating factor mitogen-activated protein kinase-activated proten kinase (MAPKAPK) Arthritis Rheum. 1999;42:2399–409. doi: 10.1002/1529-0131(199911)42:11<2399::AID-ANR19>3.0.CO;2-Y. [DOI] [PubMed] [Google Scholar]
- 138.Gveric D, Kaltschmidt C, Cuzner ML, et al. Transcription factor NF-kappaB and inhibitor I kappaBalpha are localized in macrophages in active multiple sclerosis lesions. J Neuropathol Exp Neurol. 1998;57:168–78. doi: 10.1097/00005072-199802000-00008. [DOI] [PubMed] [Google Scholar]
- 139.Bonetti B, Stegagno C, Cannella B, et al. Activation of NF-kappaB and c-jun transcription factors in multiple sclerosis lesions. Implications for oligodendrocyte pathology. Am J Pathol. 1999;155:1433–8. doi: 10.1016/s0002-9440(10)65456-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Satoh J, Misawa T, Tabunoki H, et al. Molecular network analysis of T-cell tran-scriptome suggests aberrant regulation of gene expression by NF-kappaB as a bio-marker for relapse of multiple sclerosis. Dis Markers. 2008;25:27–35. doi: 10.1155/2008/824640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Chabas D, Baranzini SE, Mitchell D, et al. The influence of the proinflammatory cytokine, osteopontin, on autoimmune demyelinating disease. Science. 2001;294:1731–5. doi: 10.1126/science.1062960. [DOI] [PubMed] [Google Scholar]
- 142.Masilamoni JG, Jesudason EP, Baben B, et al. Molecular chaperone alpha-crystallin prevents detrimental effects of neuroin-flammation. Biochim Biophys Acta. 2006;1762:284–93. doi: 10.1016/j.bbadis.2005.11.007. [DOI] [PubMed] [Google Scholar]
- 143.van Noort JM, van Sechel AC, Bajramovic JJ, et al. The small heat-shock protein alpha B-crystallin as candidate autoantigen in multiple sclerosis. Nature. 1995;375:798–801. doi: 10.1038/375798a0. [DOI] [PubMed] [Google Scholar]
- 144.Ousman SS, Tomooka BH, van Noort JM, et al. Protective and therapeutic role for alphaB-crystallin in autoimmune demyeli-nation. Nature. 2007;448:474–9. doi: 10.1038/nature05935. [DOI] [PubMed] [Google Scholar]
- 145.Sakai S, Akiyama H, Harikai N, et al. Effect of chondroitin sulfate on murine splenocytes sensitized with ovalbumin. Immunol Lett. 2002;84:211–6. doi: 10.1016/s0165-2478(02)00181-5. [DOI] [PubMed] [Google Scholar]
- 146.Sakai S, Akiyama H, Sato Y, et al. Chondroitin sulfate intake inhibits the IgE-mediated allergic response by down-regulating Th2 responses in mice. J Biol Chem. 2006;281:19872–80. doi: 10.1074/jbc.M509058200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Marusic S, Thakker P, Pelker JW, et al. Blockade of cytosolic phospholipase A2 alpha prevents experimental autoimmune encephalomyelitis and diminishes development of Th1 and Th17 responses. J Neuroimmunol. 2008;204:29–37. doi: 10.1016/j.jneuroim.2008.08.012. [DOI] [PubMed] [Google Scholar]
- 148.Chan PS, Caron JP, Orth MW. Effect of glu-cosamine and chondroitin sulfate on regulation of gene expression of proteolytic enzymes and their inhibitors in interleukin-1-challenged bovine articular cartilage explants. Am J Vet Res. 2005;66:1870–6. doi: 10.2460/ajvr.2005.66.1870. [DOI] [PubMed] [Google Scholar]
- 149.Orth MW, Peters TL, Hawkins JN. Inhibition of articular cartilage degradation by glucosamine-HCl and chondroitin sulphate. Equine Vet J Suppl. 2002:224–9. doi: 10.1111/j.2042-3306.2002.tb05423.x. [DOI] [PubMed] [Google Scholar]
- 150.Zhang GX, Yu S, Gran B, et al. Glucosamine abrogates the acute phase of experimental autoimmune encephalomyelitis by induction of Th2 response. J Immunol. 2005;175:7202–8. doi: 10.4049/jimmunol.175.11.7202. [DOI] [PubMed] [Google Scholar]
- 151.Brown KD, Claudio E, Siebenlist U. The roles of the classical and alternative nuclear factor-kappaB pathways: potential implications for autoimmunity and rheumatoid arthritis. Arthritis Res Ther. 2008;10:212. doi: 10.1186/ar2457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Dejardin E. The alternative NF-kappaB pathway from biochemistry to biology: pitfalls and promises for future drug development. Biochem Pharmacol. 2006;72:1161–79. doi: 10.1016/j.bcp.2006.08.007. [DOI] [PubMed] [Google Scholar]
