t .. . A , :00 . Effluumummm... Grain?! r t . s : 43.1? II! A .¢‘§lU-7. .....+ 3255......» a .2: hi... .4. Sufi”: 3...... an.» E... NL‘ . 5»- Ina-v4?" . S. , t 5. . nag-vi n». g .34. . r in r AW 1.51:... .. . . . 5. £ . . IA . .- ..u...x.r...5z. . .:4..Va..;\r.....&t.. 5 _..u....un..u...u. «63::- « .. . Sanitn.» .EEflI‘V fiafibgiat (1.5... mag . I; SKI? . £31190“... (3‘ .. na‘wi... . It. .7213.» aynolk‘t ”fig Human» . ramfr 4m". . . .5 .V i . . ; by”. .. a”... .E... a... . . _ .I .. ....7. . . 1...: .2... .m_.:..an....ul :HéSlS ‘ LIBRARY loo 7 Michigan State University This is to certify that the thesis entitled DIETARY SUPPLEMENTS AND THEIR EFFECT ON STRESSED CARTILAGE EXPLANTS presented by Robert Harlan has been accepted towards fulfillment of the requirements for the MS. degree in Animal Science ////... idea Major Professor’s Signature 44 // ,4 fl“ ‘4 Date . MSU is an Affinnative Action/Equal Opportunity Institution PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 2/05 p:/ClRC/DateDue.indd-p.1 DIETARY SUPPLEMENTS AND THEIR EFFECT ON STRESSED CARTILAGE EXPLANT S By Robert Harlan A THESIS Submitted to Michigan State University in partial fulfillment of the requirements for the degree of MASTER OF SCIENCE Animal Science 2006 ABSTRACT DIETARY SUPPLEMENTS AND THEIR EFFECT ON STRESSED CARTILAGE EXPLANT S By Robert Harlan We performed two experiments: for the first experiment, three culture schemes: 1) Interleukin-1 (IL-1) (50 ng/ml) treatment with and without GLN + CS for 48 h (short- term non-impact), 2) IL-1 plus mechanical trauma (15 mega pascal) with GLN + CS for 48 h (short-term impact), 3) two-week cultures were cultured with GLN + CS and were exposed to IL-1 (50 ng/ml) on day 2 and day 10 (long-term non-impact). For the second experiment, SAMe was tested at concentrations of 1.0, 0.1, and 0.01 ug/ml (high, medium, and low dose) and ASU was tested at concentrations 10.0, 1.0, and 0.1 ug/rnl (high, medium, and low dose). Interleukin-l (IL-1) at 15 ng/ml was used to initiate inflammatory stress. Nitric oxide (NO) and prostaglandin E2 (PGEZ) were measured as indicators of inflammatory response for both experiments. Glycosaminoglycans (GAG) were measured as an indicator of cartilage turnover for both experiments. For the first experiment, GLN + CS did not affect NO, PGEz, or GAG concentrations in the non-impact short-term model. When mechanical impact was combined with IL-1 , the NO concentration in the GLN + CS treatment was lower than the IL-1 treatment and did not differ from control. The GAG release was also lower in the GLN + CS treatment than in the IL-1 treatment. The non-impact long-term cultures demonstrated that GLN + CS treatment did not affect NO or PGEZ concentration or GAG content in the explants at termination. The second experiment showed no significant treatment effects for either SAMe or ASU. Capyrigh‘t by Robert Harlan 2006 I would like to dedicate this thesis to my parents for all their unconditional love and support. iv ACKNOWLEDGEMENTS I would like to thank the members Dr. Orth’s laboratory, past and present, for all of their help and support. I would like to thank Cara O’Conner for all the help with experiments, spread sheets, and generally bringing my research from chaos to organization. Thanks you to Peggy Wolf for her help and training and for all the good lunch conversations that we had. Thanks also to Pooi-See Chan for answering my questions and training me with RT-PCR. Thanks to Kirsten Neil for her help with RT- PCR and giving me primers when I needed them. Thanks to John Wheeler for all his help with assays, cleaning my lab bench, and cracking jokes. Thanks also to Stacy Endres and Jessica Partlow for all their help with assays. I would also like to thank all of my graduate student friends that I have known through the Animal Science Graduate Student Association for their friendship. Thank you to Dr. Orth for all his guidance and for giving me the opportunity to study and perform research in his laboratory and for being available and encouraging me along the way. Thank you also to Dr. Nielsen, Dr. Bursian, and Dr. Caron for serving on my committee. Thank you to Dr. Tempelmen for helping me with all the statistical analysis of the project. I would like to thank my friends from Spartan Christian Fellowship, Grace Immanuel Reformed Baptist Church, and the Grace Orthodox Presbyterian Church for their friendship, prayers and spiritual support. I would also like to thank my parents for their unconditional love and support through the good times and the bad. Finally, I would like to thank God, my heavenly father, for giving me the strength to make it through when my strength was gone. vi TABLE OF CONTENTS LIST OF TABLES ................................................................... ix LIST OF FIGURES ................................................................... x LIST OF ABBREVIATIONS ....................................................... xi INTRODUCTION ..................................................................... 1 CHAPTER 1 .................................................................................................... 4 LITERATURE REVIEW ................................................................................ 4 The importance to the horse industry .................................................... 4 Articular cartilage .................................................................................. 4 Chondrocytes ........................................................................................ 5 Extra cellular matrix ................................................... 6 Osteoarthritis ................................................................... 7 Onset of disease ........................................................ 7 Inflammatory mediators .............................................. 8 Treatment ....................................................................... 9 Conventional treatments .............................................. 9 Glucosamine and chondroitin sulfate .............................. 10 S-adenosylmethionine ................................................ l4 Avocado/soybean unsaponifiables ................................. 15 Rationale for experiments ................................................... 16 References ..................................................................... 18 CHAPTER 2 .......................................................................... 26 The Effect of Biologically Relevant Concentrations of Glucosamine and Chondroitin Sulfate on Stressed Equine Cartilage Explants ................... 26 Summary ....................................................................... 26 Introduction .................................................................... 27 Materials and Methods ...................................................... 28 Results .......................................................................... 31 Discussion ..................................................................... 32 References ..................................................................... 49 CHAPTER 3 .......................................................................... 52 vii The Effect of Avacado and Soy Unsaponifiables and S-Adenosyl-L- Methionine on Inflammatory Mediators in Bovine Cartilage .................. 52 Summary ...................................................................... 52 Introduction ................................................................... 53 Materials and Methods ...................................................... 55 Results ......................................................................... 57 Discussion ..................................................................... 57 References ..................................................................... 69 viii LIST OF TABLES Chapter 2 Table I. Agents added to short-term explant cultures .......................... 38 Table 11. Agents added to long-term explant cultures ........................... 39 Table III. Mean NO concentration (ng/ml) 2!: SE with n=3 for d 4, 12, and cumulative over 14 days ............................................................ 46 Table IV. Mean GAG concentration (ng/ml) i SE with n=3 for d 4, 12, and cumulative over 14 days ............................................................ 47 Chapter 3 Table 1. Agents added to explant cultures-explants #1 ........................ 61 Table 11. Agents added to explant cultures-explants #2 ........................ 62 ix LIST OF FIGURES Chapter 2 Fig 1: Mean (iSE, n=6 for medium dose, n=4 for low dose) NO, GAG, and PGE2 concentrations in media samples at day 0 and 1 (A, C) or at day 1 (B). IL-1 (50 ng/ml) was added to each treatment except for control. The experimental treatments contained 5 ug/ml GLN + 20 ug/ml CS (medium dose) or 1 ug/ml GLN + 5 itng CS (low dose) ................................. 40 Fig 2: Mean (:tSE, n=3) NO, PGEZ, and GAG concentrations in media samples at day 0 and 1 (A, C) or at day l (B). IL—1 (50 ng/ml) + Impact was added to each treatment except for control. The experimental treatment contained 5 ug/rnl GLN + 20 ug/ml CS (medium dose) ....................... 43 Fig 3: Mean (iSE, n=3) final GAG concentration in papain digest of cartilage explants at day 14. ....................................................... 48 Chapter 3 Fig I: Mean (iSE, n=4) NO, PGEZ, and GAG concentrations in media samples at day l (A-C). IL-1 (15 rig/ml) was added to each treatment except for control. The experimental treatments contained 1 ug/ml SAMe (high dose); 0.1 ug/ml SAMe (medium dose); 0.01 ug/ml SAMe (low dose); 0.3% EthOH (EthOH control); 10 ug/ml ASU (high dose); or 1 ug/ml ASU (medium dose) ........................................................................ 63 Fig 2: Mean (iSE, n=4) NO, PGEZ, and GAG concentrations in media samples at day 1 (A-C). IL-1 (15 ng/ml) was added to each treatment except for control. The treatments contained: 0.3% EthOH (EthOH control); 10 ug/ml ASU (high dose); 1 ug/ml ASU (medium dose); 0.01 ug/ml ASU (low dose) ............................................................................. 66 LIST OF ABBREVIATIONS A disintegrin and metalloproteinase with thrombospondin motifs .................. ADAMTS Avocado and Soy Unsaponifiables ............................................................. ASU Chondroitin Sulfate ................................................................................. CS Cyclooxygenase ................................................................................. COX Cyclooxygenase-l ............................................................................. COX-l Cyclooxygenase-2 ............................................................................. COX-2 Dirnethylene blue ............................................................................... DMB Enzyme-Linked Irnmunosorbent Assay ..................................................... ELISA Fetal bovine serum ................................................................................ FBS Glucosamine ...................................................................................... GLN Glycosaminoglycan .............................................................................. GAG Inducible nitric oxide synthase ................................................................ iNOS Insulin transferrin selenite ........................................................................ ITS Interleukin-l ....................................................................................... IL-l Microsomal prostaglandin E synthase-1 .................................................. mPGEsl Mega pascal ....................................................................................... MPa Matrix metalloproteinase ....................................................................... MMP Nitric Oxide ......................................................................................... NO Nonsteroidal anti-inflammatory drugs .................................................... NSAIDs Osteoarthritis ........................................................................................ OA Prostaglandin E2 ................................................................................. PGE2 xi S-Adenosyl-L-Methionine ..................................................................... SAMe Tumor necrosis factor-alpha ...................................................................... TNFa xii INTRODUCTION Osteoarthritis (0A) is a degenerative joint disease marked by progressive cartilage tissue loss, pain, inflammation, and eventual loss of joint function (F elson et a1. 1998; Hashimoto er al. 1998). The significance of 0A to the horse racing industry is that it results in lameness. The onset of OA can be initiated by normal exercise of the animal (Stashak 2002). Since no known way to cure or prevent OA exists, better therapies are needed. In recent years, dietary supplements (now known as nutraceuticals) have been used to treat 0A in both humans and animals. Glucosamine (GLN) and chondroitin sulfate (CS), S-adenosyl-L-methionine (SAMe), and avocado soy unsaponifiables (ASU) have been among some of the dietary supplements taken for joint pain. This project consisted of two objectives: 1) our first objective was to test the anti- inflarnmatory effects of biologically relevant concentrations of GLN plus CS in three different equine cartilage cultures that included conditions for inducing stress; 2) The second objective was to test concentrations of ASU and SAMe in bovine cartilage explant cultures for their ability to mitigate cytokine-induced stress. Explant tissue culture provides an excellent system for testing chondroprotective agents because of the convenience and accuracy of the data collection. Mechanical loading is known to be a risk factor for CA in horses and the combination of GLN and CS at biologically relevant concentrations have neverbeen tested in a mechanical loading tissue culture system. In the first series of experiments, we provide evidence that biologically relevant concentrations of GLN + CS may be effective against harmful inflammatory responses to trauma in equine cartilage tissue. We exposed equine cartilage explant cultures to three different types of conditions: 1) a short-term non-impact culture system utilizing (interleukin-1) IL-1 (50 ng/ml) as the inducer of inflammatory stress with and without GLN + C8, 2) a short-term impact utilizing IL-l plus mechanical trauma to induce inflammatory stress with and without GLN + CS 3) a two-week culture (long term) with continuous exposure to GLN + CS and introduction of IL-1 (50 ng/ml) on day 2 and day 10. In the second series of experiments, we used the short-term culture system mentioned above to test varying concentrations of SAMe and ASU in bovine explants. Both experiments measure tissue inflammatory and breakdown products as the response variables. Similar types of culture systems have been used in our lab previously (Chan et al. 2005a; Chan et al. 2005b; Chan et al. 2005c), and in this study we use an in vitro cartilage explant approach to test GLN and CS, SAMe, and ASU and their ability to mitigate inflammatory responses. REFERENCES Chan, P.S., Caron, J .P. and Orth, M.W. (2005a) Effect of glucosamine and chondroitin sulfate on regulation of gene expression of proteolytic enzymes and their inhibitors in interleukin-l-challenged bovine articular cartilage explants. Am J Vet Res 66, 1870-1876. ' Chan, P.S., Caron, J .P., Rosa, G.J. and Orth, M.W. (2005b) Glucosamine and chondroitin sulfate regulate gene expression and synthesis of nitric oxide and prostaglandin E(2) in articular cartilage explants. Osteoarthritis Cart 13, 387-394. Chan, P.S., Schlueter, A.E., Coussens, P.M., Rosa, G.J., Haut, RC. and Orth, M.W. (2005c) Gene expression profile of mechanically impacted bovine articular cartilage explants. J Orthop Res 23, 1 146-1151. Felson, D.T. and Zhang, Y. (1998) An update on the epidemiology of knee and hip osteoarthritis with a view to prevention. Arthritis Rheum 41, 1343-1355. Hashirnoto, S., Ochs, R.L., Rosen, F., Quach, J ., McCabe, G., Solan, J., Seegmiller, J .E., Terkeltaub, R. and Lotz, M. (1998) Chondrocyte-derived apoptotic bodies and calcification of articular cartilage. Proc Natl Acad Sci U S A 95, 3094-3099. Stashak, TS (2002) Examination for lameness. In: Adams' Lameness in Horses, 5 edn., Ed: D. Troy, Williams and Wilkins, Philadelphia. pp 113-183. Chapter 1 Literature Review Importance of dietary supplements to the horse industry The American Horse Council reported that the equine industry provides 1.4 million full-time jobs in the US. and that there are 9.2 million horses nation-wide (2005). The same report stated that the direct contribution of the horse industry to the US. economy is $39.9 billion annually. Also, they found that 4.6 million people are involved in the equine industry in a professional or volunteer capacity. The significance of osteoarthritis (CA) to the racing industry is that OA results in lameness, reduced performance, and wastage (Murray et a1. 2005); and one of the main factors that determines the value and usefulness of a race horse is soundness of the legs (Jeffcott et al. 1982; Rossdale et al. 1985; Fubini et al. 1999). Lameness is the most significant limiting factor for race horses; the greatest numbers of days lost to training are caused by lameness (Rossdale et al. 1985), and failure to race is most commonly due to lameness (Jeffcott et al. 1982). The presence of 0A is pervasive in the American horse population. A survey of veterinary schools found that at veterinary schools, 33% of equine patients had OA lesions (Rose 1977). In another survey of 140 2-year-old Thoroughbred horses, only 23% were sound (Jeffcott et a1. 1982). The onset of OA can be initiated by injuries sustained during normal exercise normal exercise of the animal and has been linked to activities such as barrel racing, pole bending, dressage, and other types of competions (Stashak 2002). Since no known way to cure or prevent OA exists, better therapies are needed. Articular Cartilage Chondrocytes Articular cartilage tissue at the ends of long bones provides a frictionless surface that allows for joint mobility. Cartilage also provides a shock-absorbing cushion that protects the ends of bones. The tissue is composed of chondrocytes, which are surrounded by an extracellular matrix. These cells only constitute 1% of the tissue volume (Buckwalter and Lane 1997). In healthy cartilage, chondrocytes are capable of breaking down and synthesizing extracellular matrix to maintain a stable equilibrium of catabolism and anabolism (Bullough 1992a). Chondrocytes are also capable of responding to mechanical stimuli, which cause these cells to modulate their metabolism and matrix production to meet the demands of external forces (Buschrnann and Grodzinsky 1995). Extracellular matrix mediates cell signaling and maintenance of phenotype (Bullough 1992a; Blanco et al. 1995). This is substantiated when chondrocytes are removed from the extracellular matrix and cultured in a monolayer, the cells begin to lose their spherical morphology and chondrocyte-specific markers (Domm et al. 2004; Marlovits et al. 2004). Adult cartilage tissue is avascular and therefore chondrocytes absorb nutrients from the synovial fluid, which flows through the matrix as a result of mechanical forces on the joint (Mankin 1984). Chondrocytes absorb oxygen solely from the synovial fluid and carry on a combination of aerobic and glycolytic metabolism (Clegg et al. 2006). Chondrocytes are organized into four distinct zones within the cartilage: superficial, transitional, radial, and calcified zones. Within the superficial zone, chondrocytes have a flattened morphology and are arranged randomly within the matrix (Buckwalter and Mankin 1997). In the transitional zone, they are spherical and are also arranged in a random pattern in the matrix (Buckwalter and Mankin 1997). Chondrocytes in the radial zone are arranged in columns and also demonstrate a spherical morphology (Buckwalter and Mankin 1997). Chondrocytes in the calcified zone have a spherical shape and are arranged in a random pattern in the matrix (Bullough 19923). Extracellular matrix The extracellular matrix of cartilage consists largely of collagen surrounded by proteoglycans. Collagen gives cartilage its tensile strength and provides a rigid framework, which prevents the tissue from swelling to its fullest extent (Maroudas 1976). Collagen fibrils have characteristics appropriate for the each zone of cartilage tissue (Bullough 1992b). Collagen fibrils are arranged parallel to the surface in the superficial zone while in the transitional zone the fibrils appear to be randomly arranged (Bullough 1992b). In the radial zone, the collagen fibrils are arranged perpendicular to the articular surface (Bullough 1992b). The collagen fibrils in the calcified zone however, are calcified by the surrounding chondrocytes (Bullough 1992b). Most of the collagen in the matrix is type II, but type, VI, IX, and XI are also present (Bruckner 1994). Proteoglycans consist of a protein core with glycosaminoglycan (GAG) side chains in a radial arrangement giving the molecule a “bottle brush” type structure (10220 1998). The specific proteoglycan that is produced by chondrocytes is called aggrecan, which contains chondroitin sulfate (CS) as the predominant GAG side chain. This GAG is composed of repeating disaccharide units of N-acetyl galactosarnine and glucuronic acid residues. The negatively charged sulfur groups of this disaccharide allow hydrogen bonding with water molecules. The resulting hydration endows aggrecan with the cushioning properties that are typical of articular cartilage (Prydz and Dalen 2000). As mechanical forces are applied to the cartilage tissue, fluid is expelled from between the aggrecan side chains and from the tissue. As the force is removed, fluid returns to the matrix and re-hydrates the negatively charged aggrecan side chains, thus yielding the compressive elasticity and shock absorbing properties of the cartilage matrix. Osteoarthritis Onset of disease Osteoarthritis is a degenerative disease marked by progressive cartilage tissue loss, pain, inflammation, and eventual loss of joint function (Felson et al. 1998; Hashirnoto et al. 1998). When injury or disruption to a joint occurs, enzymes are synthesized that break down the proteoglycan component of the matrix (Spiers et al. 1994a; Spiers et al. 1994b). Chondrocytes respond to this stress by proliferating and increasing proteoglycan synthesis; but this response is sometimes insufficient to overcome the degradation. As a result, the equilibrium is shified toward a gradual loss of total proteoglycan, leading to cartilage destruction (Morales and Roberts 1988; Hedbom and Hauselmann 2002). Onset of OA may be initiated by trauma or chronic mechanical stress (Quinn et al. 2001; Patwari et al. 2003). 0A in horses is believed to usually be the result of trauma resulting from over use of the joint (Mackay-Smith 1962). Factors that can induce OA include intense exercise and fatigue (Hodgson and Rose 1989). The pressure on a joint that a race horse can generate during a race can be millions of foot-pounds per mile Wackay-Smith 1962). Another risk factor in horses is subchondral bone sclerosis, which results from excessive bone remodeling in response to mechanical forces on the bone (Kawcak 2000). Cartilage that is covering areas of the bone affected by sclerosis is vulnerable to the development of OA because the subchondral bone changes shape and increases in density (Kawcak et al. 2001). Acute trauma to a joint can also lead to OA. Also injury to the anterior cruciate ligament can cause OA (Roos et al. 1995). For example, damage to intra-articular ligaments can lead to 0A. This has been demonstrated in dogs, rabbits, and in horses by transection of the cranial cruciate ligament and lateral collateral and lateral collateral sesamoidean ligaments in horses, which leads to destabilization of the joint and cartilage wear (Pond and Nuki 1973; Troyer 1982; Simmons et al. 1999; Clegg et al. 2006). Injury to the meniscus is also a common cause for joint destabilization leading to 0A in humans (Roos et al. 1998; Clegg et al. 2006). Increased concentrations of proteoglycan fragments accumulate in the synovial fluid after injury to the anterior cruciate ligament or meniscus (Lohmander et al. 1989). Stromelysin, an enzyme that breaks down cartilage, can increase up to 40-fold over time in an injured joint in (Lohmander et al. 1993). Inflammatory mediators Joint injury is often accompanied by an inflammatory response that includes the production of pro-inflammatory cytokines such as interleukin-1 beta (ILal). This cytokine is secreted by macrophages, monocytes, and synoviocytes (Kirker-Head 2000; Femandes et al. 2002) and is implicated in the inflammatory symptoms of OA. Another example of an inflammatory cytokine that is implicated in 0A is tumor necrosis factor- alpha (TNFu) (Hardingham et al. 1992; Fernandes et al. 2002; Lopez-Armada et al. 2006), which is produced by macrophages and synoviocytes. The inflammatory activity of IL-1 is mediated by signaling molecules such as nitric oxide (NO) and prostaglandin E2 (PGEZ) (Smalley et al. 1995; Grabowski et al. 1997). IL-l inhibits proteoglycan synthesis and increases proteoglycan breakdown. When inflammation in the joint is prolonged, it leads to a chronic imbalance in tissue turnover, which results in the eventual destruction of the cartilage tissue (Hardingham et al. 1992; Goldring et al. 1994). The imbalance is mediated by enzymes in the joint called matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinase with a thrombospondin type 1 motif (ADAMTS), which are responsible for breakdown of matrix components such as aggrecan and collagen (Struglics et al. 2006). Some examples of enzymes of these types that have been implicated in OA include, MMP-3, MMP-13, ADAMTS-4, and ADAMTS-5 (Patwari et al. 2003; Neil et al. 2005; Struglics et al. 2006). Treatment Conventional treatments Conventional treatments for CA include nonsteroidal anti-inflammatory drugs (NSAIDs) and intra-articular corticosteroids (Hochberg et al. 1995b, 1995a). These treatments are effective for decreasing joint pain, but they do not affect the progression of the disease. They also have significant side effects (Hochberg et al. 1995a). Nonsteroidal anti-inflammatory drugs, amoung other things, inhibit two enzymes known as cyclooxygenase-l and -2 (COX-1 and COX-2) (Harkins et al. 1993). Both of these enzymes produce PGEz, which mediates pain and inflammation in the joint. However, COX-1 is also an important enzyme for maintaining the gastrointestinal lining. As a result of COX-1 inhibition, NSAIDs eventually cause gastrointestinal ulceration if they are taken long-term, which results in thousands of deaths each year (Hochberg et al. 1995a; Griffin 1998). A common NSAID used in horses, called phenylbutazone, is associated with gastric ulcers, weight loss, and diarrhea (T raub et al. 1983). The development of a COX-2-specific inhibitor seemed to be a significant advance promising to become a NSAIDs treatment without significant side effects. Inhibiting COX-2 exclusively, however, resulted in cardiovascular side effects in patients. Clinical trials of some of these drugs demonstrated that adverse cardiovascular events were '3 times more frequent after coronary bypass surgery when COX-2 inhibitors were administered (Ott et al. 2003; Nussmeier et al. 2005). Reports also detail increased risk of cardiovascular events and serious skin reactions (Sibbald 2005; Talhari et al. 2005). This cardiovascular effect may be because COX-2 produces prostaglandin 12 in vascular epithelial tissue, which inhibits platelet aggregation, neutrophil adhesion, and dilatation of bronchial and vascular smooth muscles (Lin 2005). Modulation of such physiological processes could cause thromboembolic events, which is a risk especially to patients with a history of cardiovascular disease (Fitzgerald 2004; Warner et al. 2004). Synthetic corticosteroids can be injected intra-articularly for treatment of OA and have shown efficacy in dramatically decreasing inflammation in horses and in humans (Harkins et al. 1993; Bellamy et al. 2005). These treatments also decrease the production of prostaglandins by COX-2 enzymes (Moses et al. 2001; Frean et al. 2002; Tung et al. 2002). The side effects are eventually damaging to the cartilage, which may include the 10 formation of calcium deposits on the surface of the cartilage, fissuring of the cartilage, and decreased cartilage elasticity (Harkins et al. 1993). Glucosamine and chondroitin sulfate In recent years, dietary supplements (now known as nutraccuticals) have been used to treat OA in both humans and animals. Glucosamine (GLN) and CS have been among the most popular supplements. GLN is a precursor molecule of various components of the cartilage matrix (Hamennan 1989; Muller-Fassbender et al. 1994). Chondroitin sulfate is a component of the cartilage proteoglycan called aggrecan (Conte et al. 1991). Some evidence suggests that glucosarnine is as effective as ibuprofen at relieving pain (Muller-Fassbender et al. 1994) and that the combination of GLN and CS is effective at relieving moderate to severe joint pain in OA patients (Clegg et al. 2006). GLN has an excellent safety profile, when taken orally, based on studies done in humans, dogs, and rats (Pujalte et al. 1980; Tapadinhas et al. 1982; Muller-Fassbender et al. 1994). CS also has an excellent safety profile in humans, dogs, and rats (Conte et al. 1995; Bucsi and Poor 1998; Ronca et al. 1998; Volpi 2002). Dosing in humans is usually 1500 mg of GLN and 1200 mg of CS daily (Clegg et al. 2006) and in horses the dosage is 9 g of GLN and 3 g of CS (Du et al. 2004). Work has been done in animal models to show the effectiveness of these agents. For instance, work done in cultures systems with IL-1 stimulated cartilage explants from Holstein steers show that GLN and CS will decrease cartilage NO and PGE2 synthesis at concentrations found in the blood (Chan et al. 2005b). Another study used fibronectin fragments to initiate cartilage degradation in bovine cartilage explants and showed that 11 mixture of GLN and CS at biologically relevant concentrations was effective at decreasing expression of catabolic mediators MMP-3 and MMP-13 and promoting repair (Homandberg et al. 2006). Studies in equine cartilage explant cultures have shown that GLN and CS are effective at decreasing NO, PGEz , MMP-9, and MMP-13, though the concentrations used here were higher than what is normally found in viva (Orth et al. 2002). Studies in equine chondrocyte cell culture (pellet culture) have shown that relevant concentrations of GLN decrease expression of catabolic mediators such as MMP-13, aggrecanase-1, inducible nitric oxide synthase (iNOS), and COX-2 (Neil et al. 2005) The mechanism of action of these two substances has received significant attention lately. Based on the biochemistry of GLN and CS and GAG synthesis is examined, it appears feasible that these molecules could increase the synthesis of GAG (Mch et al. 2000). McCarty et al. proposed that GLN supplementation provides the rate-limiting substrate for GAG (McCarty et al. 2000). Evidence for this theory was demonstrated by a study in which over-expression of glutamine fructose-6-phosphate aminotransferase, an important rate limiting enzyme in the synthesis of GLN, caused resistance to IL-1 inhibition of GAG synthesis (Gouze et al. 2001). In a study involving dogs with a transected cranial cruciate ligament, a combination of GLN and CS increased GAG synthesis (Johnson et al. 2001). One of the mechanisms for the preservation of cartilage tissue by GLN maybe the inhibition of aggrecanase (Patwari et al. 2003; Chan et al. 2005a). GLN alone decreases IL-l -induced production of NO and PGE2 and IL-1- induced proteoglycan breakdown (F enton et al. 2000; Gouze et al. 2001; F enton et al. 2002; Chan et al. 2005b). The mechanism of decreasing proteoglycan breakdown is in 12 part due to the inhibition of MMPs, aggrecanases, and collagenase (Fenton et al. 2000; Piperno et al. 2000; Fenton et al. 2002; Dodge and Jimenez 2003; Chan et al. 2005a). GLN can also decrease the gene expression of MMP-1, MMP-3, and MMP-13 (Byron et al. 2003; Dodge and Jimenez 2003; Chan et al. 2005a). CS May stimulate GAG production by binding to membrane proteins of synovial cells (Mch et al. 2000). CS increase proteoglycan synthesis and decreases proteoglycan breakdown (Bassleer et al. 1992; Nerucci et al. 2000). CS reduces proteoglycan loss from articular cartilage in humans (Uebelhart et al. 1998) and rats (Omata et al. 1999) when taken orally. CS may reduce proteoglycan loss in part by decreasing aggrecanase-1 synthesis (Chan et al. 2005a). CS can also decrease IL-l-induced PGE2 and NO production (Bassleer et al. 1992; Chan et al. 2005b). The mechanism for this appears to be that CS decreases the expression of iNOS and microsomal prostaglandin E synthase-l (Chan et al. 2005b). If GLN and CS are taken together, they appear to have a synergistic effect in mitigating the clinical signs of OA. An in vivo study that used a rabbit instability model of osteoarthrosis showed that animals given GLN and CS did not develop OA lesions as severe as rabbits fed GLN or CS alone (Lippiello et al. 2000). In the in vitro portion of the study, the combination of glucosarnine hydrochloride and chondroitin sulfate acted synergistically in stimulating GAG synthesis (Lippiello et al. 2000). GLN and CS also both increase chondrocyte response to mechanical stress and prevent the loss of GAG (Lippiello 2003). The fibronectin fiagrnents study also showed a synergistic relationship between GLN and CS for reversing cartilage damage and promoting repair (Homandberg et al. 2006). GLN and CS have the ability to modify expression of genes that regulate inflammatory mediators such as iNOS, COX-2, and mPGEsl (Chan et al. 2005b; Neil et 13 al. 2005). The concentrations used in these studies (Chan "et al. 2005b; Neil et al. 2005) were within range of those found in the blood of horses after oral administration (Du et al. 2004), and within GLN levels achieved in the synovial fluid of horses when administed by intravenous infusion (Laverty et al. 2005). S-adenosylmethionine S-Adenosyl-L-methionine (SAMe) is synthesized by the body from the essential the amino acid L-methionine and the nucleoside adenosine triphosphate. SAMe acts as a methyl donor for reactions important in the synthesis of proteins, phospholipids, and hormones. It has a variety of roles and is involved in membrane fimction, gene regulation, and brain function (Chiang et al. 1996). SAMe was first sold over the counter in 1999 for treatment of joint pain. The common human dosage is usually between 400 and 1600 mg daily. SAMe has a good safety record, with few side effects and an absence of generation of toxic metabolites (Kagan et al. 1990; Goren et al. 2004). The concentration attained in the synovial fluid in OA patients given SAMe at 400 mg daily for 7 days is 30 to 80 ng/ml (Stramentinoli 1987). Clinical trials show that SAMe supplementation decreases pain and inflammation (Polli et al. 1975; Caruso and Pietrogrande 1987; Soeken et al. 2002). A randomized, double-blind, cross-over study, comparing SAMe with celecoxib (Celebrex) showed that SAMe, over the long term, is just as effective at relieving pain as Celecoxib (Najm et al. 2004). The exact mechanism of action is not known. SAMe also upregulates the proteoglycan synthesis of chondrocytes, and some believe that it may function as a signal of sulfur availability (McCarty and Russell 1999). A study used human chondrocytes in 14 a thick-layer culture model and showed that SAMe increases proteoglycan synthesis (Harmand et al. 1987). SAMe has also demonstrated anti-inflammatory effects by suppressing the effects of TNFa and transcriptional activation of iNOS and COX-2 (Hevia et al. 2004). IL-l suppresses the production of SAMe in the joint. Therefore, since IL-1 production in the joint is upregulated in the disease state of 0A, a supplementation of SAMe may compensate for this deficit (McCarty and Russell 1999). SAMe also decreases with aging in human and rats (di Padova 1987; Stramentinoli 1987). Since aging is a risk factor for 0A, the two may be related and supplementation with age may be a benefit. Avocado/soybean unsaponifiables Avocado soybean unsaponifiables (ASU) are a mixture of vegetable extracts obtained from avocado and soy bean oils that are normally taken orally. Clinical trials with ASU show that it has a good safety record (Lequesne et al. 2002; Little and Parsons 2002), though more study is needed to confirm this. These extracts are obtained when the oil is saponified (heated and broken down by alkaline hydrolysis). The small fraction that is not broken down is known as the “unsaponifiable” fraction (Bassleer et al. 1992). It relieves pain when taken orally, and is usually taken at 300 mg daily (Reginster et al. 2000; Walker—Bone 2003; Soeken 2004). This decrease in pain is attributed to the ability of ASU to decrease inflammatory mediators such as IL-1, NO, and PGE2 as seen in human chondrocyte studies (Bassleer et al. 1992). A study using human chondrocytes in an alginate bead tissue culture system showed that ASU increased the accumulation of GAG in the alginate beads in a dose 15 dependent manner (Bassleer et al. 1992). Avocado soy unsaponifiables also prevent collagen breakdown and thus potentially save cartilage from degradation (Mauviel et al. 1989). This is partially mediated by the ability of ASU to inhibit MMP production, such as MMP-3, as it did in a study of human chondrocytes in culture (Bassleer et al. 1992). There has been no pharrnacokinetic studies done on this supplement and the exact mechanism of action is unknown. Rationale for experiments One objective is to test biologically relevant concentrations of GLN and CS in various equine cultures: 1) a short-term (2 d) in vitro culture model designed for cartilage explants in a 24-wellplate format, 2) a similar short-term culture model including mechanical loading, 3) a long-term culture system (without loading) designed to test the effects of GLN and CS over a 2-week period. Explant tissue culture provides an excellent system for testing chondroprotective agents because of the convenience and accuracy of the data collection. Mechanical loading is known to be a risk factor for CA in horses and the combination of GLN and CS at biologically relevant concentrations have never been tested in a mechanical loading tissue culture system. IL-l will be used to model inflammatory stress in each model and a hydraulic stressor with IL-1 will be used to model mechanical loading in the short-term impact culture. The response variables will consist of concentrations of inflammatory mediators NO and PGE2 in the media. Glycosaminoglycan concentration in the media and within cartilage explants will be measured as an indicator of cartilage breakdown. The concentrations of GLN and CS 16 that will be used in these cultures will be in the range of what is measured in the blood of dogs and horses after oral administration (Adebowale et al. 2002; Du et al. 2004). Another objective is to test varying concentrations of SAMe and ASU in bovine cartilage explant cultures. The system will be a short term 2 (1 culture with IL-1 used to model inflammatory stress. The response variables will consist of NO, PGEz, and GAG concentrations in the media and concentration of GAG in cartilage explants at termination of the experiment. Bovine tissue culture is used here because cartilage tissue from bovine species is readily available. Bovine tissue culture is often used as a model for testing agents that have relevance to human treatment. 17 REFERENCES (2005) Horse Industry Statistics. Retrieved April 30, 2005, from http://www.horsecouncil.org/statistics.htrn. Adebowale, A., Du, J ., Liang, Z., Leslie, J .L. and Eddington, ND. (2002) The bioavailability and pharmacokinetics of glucosarnine hydrochloride and low molecular weight chondroitin sulfate after single and multiple doses to beagle dogs. Biopharm Drug Dispos 23, 217-225. Bassleer, C.T., Combal, J .P., Bougaret, S. and Malaise, M. (1998) Effects of chondroitin sulfate and interleukin-1 beta on human articular chondrocytes cultivated in clusters. Osteoarthritis Cart 6, 196-204. Bellamy, N., Campbell, J ., Robinson, V., Gee, T., Boume, R. and Wells, G. (2005) Intraarticular corticosteroid for treatment of osteoarthritis of the knee. Cochrane Database Syst Rev, CD005328. Blanco, F.J., Geng, Y. and Lotz, M. (1995) Differentiation-dependent effects of IL-1 and TGF-beta on human articular chondrocyte proliferation are related to inducible nitric oxide synthase expression. J Immunol 154, 4018-4026. Bruckner, P., van der Rest, M. (1994) Structure and function of cartilage collagens. Microsc Res Tech 28, 378-384. Buckwalter, J .A. and Lane, NE. (1997) Athletics and osteoarthritis. Am J Sports Med 25, 873-881. Buckwalter, J .A. and Mankin, H. (1997) Articular Cartilage. Part 1. Tissue design and chondrocyte-matrix interactions. J Bone Jt Surg 79A, 600-61 1. Bucsi, L. and Poor, G. (1998) Efficacy and tolerability of oral chondroitin sulfate as a symptomatic slow-acting drug for osteoarthritis (SYSADOA) in the treatment of knee osteoarthritis. Osteoarthritis Cart 6 Suppl A, 31-36. Bullough, P. (1992a) The Pathology of Osteoarthritis. In: Osteoarthritis, Ed: R. Moskowitz, Howell, D., Goldberg, V., Mankin, H., WB Saunders, Philadelphia. pp 39-69. Bullough, P. (1992b) The Pathology of Osteoarthritis, Ed: R. Moskowitz, Howell, D., Goldberg, V., Mankin, H. 18 Buschmann, MD. and Grodzinsky, A.J. (1995) A molecular model of proteoglycan- associated electrostatic forces in cartilage mechanics. J Biomech Eng 117, 179- 192. Byron, C.R., Orth, M.W., Venta, P.J., Lloyd, J.W. and Caron, J .P. (2003) Influence of glucosamine on matrix metalloproteinase expression and activity in lipopolysaccharide-stimulated equine chondrocytes. Am J Vet Res 64, 666-671. Caruso, I. and Pietrogrande, V. (1987) Italian double-blind multicenter study comparing S-adenosylmethionine, naproxen, and placebo in the treatment of degenerative joint disease. Am J Med 83, 66-71. Chan, P.S., Caron, J .P. and Orth, M.W. (2005a) Effect of glucosamine and chondroitin sulfate on regulation of gene expression of proteolytic enzymes and their inhibitors in interleukin-l-challenged bovine articular cartilage explants. Am J Vet Res 66, 1870-1876. Chan, P.S., Caron, J .P., Rosa, G.J. and Orth, M.W. (2005b) Glucosamine and chondroitin sulfate regulate gene expression and synthesis of nitric oxide and prostaglandin E(2) in articular cartilage explants. Osteoarthritis Cart 13, 387-394. Chiang, P.K., Gordon, R.K., Tal, J ., Zeng, G.C., Doctor, B.P., Pardhasaradhi, K. and McCann, RP. (1996) S-Adenosylmethionine and methylation. F aseb J 10, 471- 480. Clegg, D.O., Reda, D.J., Harris, C.L., Klein, M.A., O'Dell, J .R., Hooper, M.M., Bradley, J .D., Bingharn, C.O., 3rd, Weisman, M.H., Jackson, C.G., Lane, N.E., Cush, J.J., Moreland, L.W., Schumacher, H.R., Jr., Oddis, C.V., Wolfe, F ., Molitor, J .A., Yocum, D.E., Schnitzer, T.J., Furst, D.E., Sawitzke, A.D., Shi, H., Brandt, K.D., Moskowitz, R.W. and Williams, H.J. (2006) Glucosamine, chondroitin sulfate, and the two in combination for painful knee osteoarthritis. N Engl J Med 354, 795-808. Conte, A., Palmieri, L., Segnini, D. and Ronca, G. (1991) Metabolic fate of partially depolymerized chondroitin sulfate administered to the rat. Drugs Exp Clin Res 17, 27-33. Conte, A., Volpi, N., Palmieri, L., Bahous, I. and Ronca, G. (1995) Biochemical and pharrnacokinetic aspects of oral treatment with chondroitin sulfate. Arzneimittelforschung 45, 918-925. di Padova, C. (1987) S-adenosylmethionine in the treatment of osteoarthritis. Review of the clinical studies. The American journal of medicine 83, 60-65. 19 Dodge, GR. and Jimenez, SA. (2003) Glucosamine sulfate modulates the levels of aggrecan and matrix metalloproteinase-3 synthesized by cultured human osteoarthritis articular chondrocytes. Osteoarthritis Cart 1 1, 424-432. Domm, C., Schunke, M., Steinhagen, J ., Freitag, S. and Kurz, B. (2004) Influence of various a1 ginate brands on the redifferentiation of dedifferentiated bovine articular chondrocytes in alginate bead culture under high and low oxygen tension. Tissue Eng 10, 1796-1805. Du, J ., White, N. and Eddington, ND. (2004) The bioavailability and pharmacokinetics of glucosamine hydrochloride and chondroitin sulfate after oral and intravenous single dose administration in the horse. Biopharm Drug Dispos 25, 109-116. Felson, D.T., Anderson, J.J., Lange, M.L., Wells, G. and LaValley, MP. (1998) Should improvement in rheumatoid arthritis clinical trials be defined as fifty percent or seventy percent improvement in core set measures, rather than twenty percent? Arthritis Rheum 41, 1564-1570. Fenton, J .I., Chlebek-Brown, K.A., Caron, J .P. and Orth, M.W. (2002) Effect of glucosamine on interleukin-1 -conditioned articular cartilage. Equine Vet J Suppl, 219-223. Fenton, J .I., Chlebek-Brown, K.A., Peters, T.L., Caron, J .P. and Orth, M.W. (2000) Glucosamine HCl reduces equine articular cartilage degradation in explant culture. Osteoarthritis Cart 8, 258-265. F emandes, J .C., Martel-Pelletier, J. and Pelletier, J .P. (2002) The role of cytokines in osteoarthritis pathophysiology. Biorheology 39, 237-246. Fitzgerald, GA. (2004) Coxibs and cardiovascular disease. N Engl J Med 351, 1709- 1711. Frean, S.P., Cambridge, H. and Lees, P. (2002) Effects of anti-arthritic drugs on proteoglycan synthesis by equine cartilage. J Vet Pharmacol Ther 25, 289-298. Fubini, S.L., Erb, H.N., Freeman, K.P. and Todhunter, R.J. (1999) Prognostic factors affecting survival of 507 horses with joint disease: (1983 to 1990). Can J Vet Res 63, 253-260. Goldring, M.B., Birkhead, J .R., Suen, L.F., Yarnin, R., Mizuno, S., Glowacki, J ., Arbiser, J .L. and Apperley, J .F. (1994) Interleukin-1 beta-modulated gene expression in irnmortalized human chondrocytes. J Clin Invest 94, 2307-2316. Gdren, J .L., Stoll, A.L., Damico, K.E., Sarmiento, LA. and Cohen, BM. (2004) Bioavailability and lack of toxicity of S-adenosyl-L-methionine (SAMe) in humans. Pharmacotherapy 24, 1501 -1 507. 20 Gouze, J .N., Bordji, K., Gulberti, S., Terlain, B., Netter, P., Magdalou, J ., Foumel- Gigleux, S. and Ouzzine, M. (2001) Interleukin-lbeta down-regulates the expression of glucuronosyltransferase I, a key enzyme priming glycosarninoglycan biosynthesis: influence of glucosamine on interleukin-lbcta- mediated effects in rat chondrocytes. Arthritis Rheum 44, 351-360. Gouze, J.N., Gouze, E., Palmer, G.D., Kaneto, H., Ghivizzani, S.C., Grodzinsky, A.J. and Evans, CH. (2004) Adenovirus-mediated gene transfer of glutarnine: fructose-6- phosphate amidotransferase antagonizes the effects of interleukin-1beta on rat chondrocytes. Osteoarthritis Cart 12, 217-224. Grabowski, P.S., Wright, P.K., Van 't Hof, R.J., Helfrich, M.H., Ohshima, H. and Ralston, SH. (1997) Irnmunolocalization of inducible nitric oxide synthase in synovium and cartilage in rheumatoid arthritis and osteoarthritis. Br J Rheumatol 36, 651-655. Griffin, MR. (1998) Epidemiology of nonsteroidal anti-inflammatory drug-associated gastrointestinal injury. Am J Med 104, 23S-29S; discussion 41 S-42S. Hamerman, D. (1989) The biology of osteoarthritis. N Engl J Med 320, 1322-1330. Hardingham, T.E., Bayliss, M.T., Rayan, V. and Noble, DP. (1992) Effects of growth factors and cytokines on proteoglycan turnover in articular cartilage. Br J Rheumatol 31 Suppl 1, 1-6. Harkins, J .D., Carney, J .M. and Tobin, T. (1993) Clinical use and characteristics of the corticosteroids. Vet Clin North Am Equine Pract 9, 543-562. Harmand, M.F., Vilamitjana, J ., Maloche, E., Duphil, R. and Ducassou, D. (1987) Effects of S-adenosyhnethionine on human articular chondrocyte differentiation. An in vitro study. Am J Med 83, 48-54. Hashirnoto, S., Ochs, R.L., Rosen, F ., Quach, J., McCabe, G., Solan, J ., Seegrniller, J .E., Terkeltaub, R. and Lotz, M. (1998) Chondrocyte-derived apoptotic bodies and calcification of articular cartilage. Proc Natl Acad Sci U S A 95, 3094-3099. Hedbom, E. and Hauselmann, H.J. (2002) Molecular aspects of pathogenesis in osteoarthritis: the role of inflammation. Cell Mol Life Sci 59, 45-53. Henrotin, Y.E., Labasse, A.H., Jaspar, J .M., De Groote, D.D., Zheng, S.X., Guillou, GB. and Reginster, J .Y. (1998) Effects of three avocado/soybean unsaponifiable mixtures on metalloproteinases, cytokines and prostaglandin E2 production by human articular chondrocytes. Clin Rheumatol 17, 31-39. 21 Henrotin, Y.E., Sanchez, C., Deberg, M.A., Piccardi, N., Guillou, G.B., Msika, P. and Reginster, J .Y. (2003) Avocado/soybean unsaponifiables increase aggrecan synthesis and reduce catabolic and proinflammatory mediator production by human osteoarthritic chondrocytes. J Rheumatol 30, 1825-1834. Hevia, H., Varela-Rey, M., Corrales, P.J., Berasain, C., Martinez-Chantar, M.L., Latasa, M.U., Lu, S.C., Mato, J .M., Garcia-Trevijano, ER. and Avila, MA. (2004) 5'- methylthioadenosine modulates the inflammatory response to endotoxin in mice and in rat hepatocytes. Hepatology 39, 1088-1098. Hochberg, M.C., Altman, R.D., Brandt, K.D., Clark, B.M., Dieppe, P.A., Griffin, M.R., Moskowitz, R.W. and Schnitzer, T.J. (1995a) Guidelines for the medical management of osteoarthritis. Part I. Osteoarthritis of the hip.American College of Rheumatology. Arthritis Rheum 38, 1535-1540. Hochberg, M.C., Altman, R.D., Brandt, K.D., Clark, B.M., Dieppe, P.A., Griffin, M.R., Moskowitz, R.W. and Schnitzer, T.J. (1995b) Guidelines for the medical management of osteoarthritis. Part II. Osteoarthritis of the knee.American College of Rheumatology. Arthritis Rheum 38, 1541-1546. Hodgson, DR. and Rose, R.J. ( 1989) Principles and practice of equine sports medicine: The athletic horse, W.B. Saunders Company, Philadelphia. Homandberg, G.A., Guo, D., Ray, L.M. and Ding, L. (2006) Mixtures of glucosamine and chondroitin sulfate reverse fibronectin fragment mediated damage to cartilage more effectively than either agent alone. Osteoarthritis Cart. Epub ahead of print. Iozzo, R.V. (1998) Matrix proteoglycans: from molecular design to cellular function. Annu Rev Biochem 67, 609-652. Jeffcott, L.B., Rossdale, P.D., Freestone, J ., Frank, C]. and Towers-Clark, RF. (1982) An assessment of wastage in thoroughbred racing from conception to 4 years of age. Equine VetJ14, 185-198. Johnson, K.A., Hulse, D.A., Hart, R.C., Kochevar, D. and Chu, Q. (2001) Effects of an orally administered mixture of chondroitin sulfate, glucosamine hydrochloride and manganese ascorbate on synovial fluid chondroitin sulfate 333 and 7D4 epitope in a canine cruciate ligament transection model of osteoarthritis. Osteoarthritis Cart 9, 14-21. Kagan, B.L., Sultzer, D.L., Rosenlicht, N. and Gemer, RH. (1990) Oral S- adenosylmethionine in depression: a randomized, double-blind, placebo- controlled trial. Am J Psychiatry 147, 591-595. Kawcak, CE. (2000) Pathophysiology of equine joint disease. Suppl compend Contin Educ Pract Vet 22, 4-9. 22 Kawcak, C.E., McIlwraith, C.W., Norrdin, R.W., Park, RD. and James, SP. (2001) The role of subchondral bone in joint disease: a review. Equine Vet J 33, 120-126. Lane, J .M., Brighton, QT. and Menkowitz, B.J. (1977) Anaerobic and aerobic metabolism in articular cartilage. J Rheumatol 4, 334-342. Laverty, S., Sandy, J .D., Celeste, C., Vachon, P., Marier, J .F . and Plaas, AH. (2005) Synovial fluid levels and serum pharmacokinetics in a large animal model following treatment with oral glucosamine at clinically relevant doses. Arthritis Rheum 52,181-191. Lequesne, M., Maheu, E., Cadet, C. and Dreiser, R.L. (2002) Structural effect of avocado/soybean unsaponifiables on joint space loss in osteoarthritis of the hip. Arthritis Rheum 47, 50-58. Lin, S. (2005) The Fall of Titans: The Need to Reassess COX-2 Inhibitors. WM 2, 138- 141. Lippiello, L. (2003) Glucosamine and chondroitin sulfate: biological response modifiers of chondrocytes under simulated conditions of joint stress. Osteoarthritis Cart 11, 335-342. Lippiello, L., Woodward, J ., Karpman, R. and Hammad, TA. (2000) In vivo chondroprotection and metabolic synergy of glucosamine and chondroitin sulfate. Clin Orthop Relat Res, 229-240. Little, CV. and Parsons, T. (2002) Herbal therapy for treating osteoarthritis, The Cochrane Library. Lohmander, L.S., Dahlberg, L., Ryd, L. and Heinegard, D. (1989) Increased levels of proteoglycan fragments in knee joint fluid after injury. Arthritis Rheum 32, 1434- 1442. Lohmander, L.S., Hoerrner, L.A., Dahlberg, L., Roos, H., Bjomsson, S. and Lark, M.W. (1993) Stromelysin, tissue inhibitor of metalloproteinases and proteoglycan fragments in human knee joint fluid after injury. J Rheumatol 20, 1362-1368. Lopez-Armada, M.J., Carames, B., Lites-Dean, M., Cillero-Pastor, B., Ruiz-Romero, C., Galdo, F. and Blanco, El (2006) Cytokines, tumor necrosis factor-alpha and interleukin-lbeta, differentially regulate apoptosis in osteoarthritis cultured human chondrocytes. Osteoarthritis Cart. Mackay-Smith, MP. (1962) Pathogenesis and pathology of equine osteoarthritis. J Vet Med Sci 141, 1246-1252. 23 Mankin, H.J.aB., K. D. (1984) Biochemistry and metabolism of cartilage in osteoarthritis. In: Osteoarthritis, Ed: R.W. Moskowitz, Howell, D. S., Goldberg, V. M. and Mankin, H. J ., WB Saunders, Philadelphia. pp 43-79. Marlovits, S., Hombauer, M., Tamandl, D., Vecsei, V. and Schlegel, W. (2004) Quantitative analysis of gene expression in human articular chondrocytes in monolayer culture. Int J Mol Med 13, 281-287. Maroudas, AL (1976) Balance between swelling pressure and collagen tension in normal and degenerate cartilage. Nature 260, 808-809. Mauviel, A., Daireaux, M., Hartrnann, D.J., Galera, P., Loyau, G. and Pujol, J .P. (1989) [Effects of unsaponifiable extracts of avocado/soy beans (PIAS) on the production of collagen by cultures of synoviocytes, articular chondrocytes and skin fibroblasts]. Rev Rhum Mal Osteoartic 56, 207-211. McCarty, MP. and Russell, AL. (1999) Niacinarnide therapy for osteoarthritis-does it inhibit nitric oxide synthase induction by interleukin 1 in chondrocytes? Med Hypotheses 53, 350-360. McCarty, M.F., Russell, AL. and Seed, MP. (2000) Sulfated glycosaminoglycans and glucosamine may synergize in promoting synovial hyaluronic acid synthesis. Med Hypotheses 54, 798-802. Morales, T.I. and Roberts, AB. (1988) Transforming growth factor beta regulates the metabolism of proteoglycans in bovine cartilage organ cultures. J Biol Chem 263, 12828-12831. Morris, E.A., McDonald, B.S., Webb, AC. and Rosenwasser, L.J. (1990) Identification of interleukin-1 in equine osteoarthritic joint effusions. Am J Vet Res 51, 59-64. Moses, V.S., Hardy, J ., Bertone, AL. and Weisbrode, SE. (2001) Effects of anti- inflammatory drugs on lipopolysaccharide-challenged and -unchallenged equine synovial explants. Am J Vet Res 62, 54-60. Moskowitz, R.W., Davis, W., Sammarco, J ., Martens, M., Baker, J., Mayor, M., Burstein, AH. and Frankel, V.H. (1973) Experimentally induced degenerative joint lesions following partial meniscectomy in the rabbit. Arthritis Rheum 16, 397-405. Muller-Fassbender, H., Bach, G.L., Haase, W., Rovati, LC. and Setnikar, I. (1994) Glucosamine sulfate compared to ibuprofen in osteoarthritis of the knee. Osteoarthritis Cart 2, 61-69. Murray, R.C., Dyson, S.J., Weekes, J .S., Short, C. and Branch, M.V. (2005) Scintigraphic evaluation of the distal tarsal region in horses with distal tarsal pain. Vet Radiol Ultrasound 46, 171-178. 24 Najm, W.I., Reinsch, S., Hoehler, F ., Tobis, J .S. and Harvey, P.W. (2004) S-adenosyl methionine (SAMe) versus celecoxib for the treatment of osteoarthritis symptoms: a double-blind cross-over trial. [ISRCTN3623 3495]. BMC Musculoskelet Disord 5, 6. Neil, K.M., Orth, M.W., Coussens, P.M., Chan, RS. and Caron, J .P. (2005) Effects of glucosamine and chondroitin sulfate on mediators of osteoarthritis in cultured equine chondrocytes stimulated by use of recombinant equine interleukin-1 beta. Am J Vet Res 66, 1861 -1 869. Nerucci, F ., Fioravanti, A., Cicero, M.R., Collodel, G. and Marcolongo, R. (2000) Effects of chondroitin sulfate and interleukin-1 beta on human chondrocyte cultures exposed to pressurization: a biochemical and morphological study. Osteoarthritis Cart 8, 279-287. Nussmeier, N.A., Whelton, A.A., Brown, M.T., Langford, R.M., Hoefi, A., Parlow, J .L., Boyce, SW. and Verburg, KM. (2005) Complications of the COX-2 inhibitors parecoxib and valdecoxib after cardiac surgery. N Engl J Med 352, 1081-1091. Omata, T., Segawa, Y., Itokazu, Y., Inoue, N. and Tanaka, Y. (1999) Effects of chondroitin sulfate-C on bradykinin-induced proteoglycan depletion in rats. Arzneimittel-Forschung 49, 577-581. Orth, M.W., Peters, T.L. and Hawkins, J.N. (2002) Inhibition of articular cartilage degradation by glucosamine-HCI and chondroitin sulphate. Equine Vet J Suppl, 224-229. Ott, B., Nussmeier, N.A., Duke, P.C., Feneck, R.O., Alston, R.P., Snabes, M.C., Hubbard, R.C., Hsu, P.H., Saidman, L.J. and Mangano, D.T. (2003) Efficacy and safety of the cyclooxygenase 2 inhibitors parecoxib and valdecoxib in patients undergoing coronary artery bypass surgery. J Thorac Cardiovasc Surg 125, 1481- 1492. Patwari, P., Cook, M.N., DiMicco, M.A., Blake, S.M., James, I.E., Kumar, S., Cole, A.A., Lark, M.W. and Grodzinsky, A.J. (2003) Proteoglycan degradation afier injurious compression of bovine and human articular cartilage in vitro: interaction with exogenous cytokines. Arthritis Rheum 48, 1292-1301. Patwari, F., Kurz, B., Sandy, J .D. and Grodzinsky, A.J. (2000) Mannosamine inhibits aggrecanase-mediated changes in the physical properties and biochemical composition of articular cartilage. Arch Biochem Biophys 374, 79-85. Pipemo, M., Reboul, F., Hellio Le Graverand, M.F., Peschard, M.J., Annefeld, M., Richard, M. and Vignon, E. (2000) Glucosamine sulfate modulates dysregulated 25 activities of human osteoarthritic chondrocytes in vitro. Osteoarthritis Cart 8, 207-212. Polli, B., Cortellaro, M., Parrini, L., Tessari, L. and Cherie Ligniere, G. (1975) [Pharmacological and clinical aspects of S-adenosylmethionine (SAMe) in primary degenerative arthropathy (osteoarthrosis)]. Minerva Med 66, 4443-4459. Pond, MJ. and Nuki, G. (1973) Experimentally-induced osteoarthritis in the dog. Ann Rheum Dis 32, 387-388. Prydz, K. and Dalen, K.T. (2000) Synthesis and sorting of proteoglycans. J Cell Sci 113 Pt 2, 193-205. Pujalte, J .M., Llavore, ER and Ylescupidez, FR. (1980) Double-blind clinical evaluation of oral glucosamine sulphate in the basic treatment of osteoarthrosis. Curr Med Res Opin 7, 110-114. Quinn, T.M., Allen, R.G., Schalet, B.J., Perumbuli, P. and Hunziker, EB. (2001) Matrix and cell injury due to sub-impact loading of adult bovine articular cartilage explants: effects of strain rate and peak stress. J Orthop Res 19, 242-249. Reginster, J .Y., Gillot, V., Bruyere, O. and Henrotin, Y. (2000) Evidence of nutriceutical effectiveness in the treatment of osteoarthritis. Curr Rheumatol Rep 2, 472-477. Ronca, F ., Palmieri, L., Panicucci, P. and Ronca, G. (1998) Anti-inflammatory activity of chondroitin sulfate. Osteoarthritis Cart 6 Suppl A, 14-21. Roos, H., Adalberth, T., Dahlberg, L. and Lohmander, LS. (1995) Osteoarthritis of the knee after injury to the anterior cruciate ligament or meniscus: the influence of time and age. Osteoarthritis Cart 3, 261-267. Roos, H., Lauren, M., Adalberth, T., Roos, B.M., Jonsson, K. and Lohmander, LS. (1998) Knee osteoarthritis after meniscectomy: prevalence of radiographic changes after twenty-one years, compared with matched controls. Arthritis Rheum 41, 687-693. Rose, R. (1977) An analysis of conditions causing lameness in the horse. In: 54th Annual Meeting of the Australian Veterinary Association. pp 99-102. Rossdale, P.D., Hopes, R., Digby, NJ. and offord, K. (1985) Epidemiological study of wastage among racehorses 1982 and 1983. Vet Rec 116, 66-69. Setnikar, I., Giacchetti, C. and Zanolo, G. (1986) Phannacokinetics of glucosamine in the dog and in man. Arzneimittel-Forschung 36, 729-735. 26 Setnikar, 1., Pacini, MA. and Revel, L. (1991) Antiarthritic effects of glucosamine sulfate studied in animal models. Arzneimittelforschung 41, 542-545. Setnikar, 1., Palumbo, R., Canali, S. and Zanolo, G. (1993) Pharrnacokinetics of glucosamine in man. Arzneimittelforschung 43, 1109-1113. Setnikar, I. and Rovati, LC. (2001) Absorption, distribution, metabolism and excretion of glucosamine sulfate. A review. Arzneimittelforschung 51, 699-725. Sibbald, B. (2005) Pfizer to withdraw valdecoxib (Bextra) at Health Canada's request. CMAJ 10, e1298. Simmons, B.J., Bertone, AL. and Weisbrode, SE. (1999) Instability-induced osteoarthritis in the metacarpophalangeal joint of horses. Am J Vet Res 60, 7-13. Smalley, W.B., Ray, W.A., Daugherty, J .R. and Griffin, MR. (1995) Nonsteroidal anti- inflammatory drugs and the incidence of hospitalizations for peptic ulcer disease in elderly persons. Am J Epidemiol 141, 539-545. Soeken, KL. (2004) Selected CAM therapies for arthritis-related pain: the evidence from systematic reviews. Clin J Pain 20, 13-18. Soeken, K.L., Lee, W.L., Bausell, R.B., Agelli, M. and Berman, BM. (2002) Safety and efficacy of S-adenosylmethionine (SAMe) for osteoarthritis. J F am Pract 51, 425- 430. Spiers, S., May, S.A., Bennett, D. and Edwards, G.B. (1994a) Cellular sources of proteolytic enzymes in equine joints. Equine Vet J 26, 43-47. Spiers, S., May, S.A., Harrison, L.J., Bennett, D. and Edwards, GB. (1994b) Proteolytic enzymes in equine joints with infectious arthritis. Equine Vet J26, 48-50. Stashak, TS. (2002) Examination for lameness. In: Adams' Lameness in horses, 5 edn., Ed: D. Troy, Williams and Wilkins, Philadelphia. pp 113-183. Stramentinoli, G. (1987) Pharrnacologic aspects of S-adenosylmethionine. Pharmacokinetics and pharmacodynamics. Am J Med 83, 35-42. Struglics, A., Larsson, S., Pratta, M.A., Kumar, S., Lark, M.W. and Lohmander, LS. (2006) Human osteoarthritis synovial fluid and joint cartilage contain both aggrecanase- and matrix metalloproteinase-generated aggrecan fragments. Osteoarthritis Cart 14, 101-113. Talhari, C., Lauceviciute, 1., Enderlein, E., Ruzicka, T. and Homey, B. (2005) COX-2- selective inhibitor valdecoxib induces severe allergic skin reactions. J Allergy Clin Immunol 115, 1089-1090. 27 Tapadinhas, M.J., Rivera, LC. and Bignamini, AA. (1982) Oral glucosamine sulphate in the management of arthrosis: report on a multi-centre open investigation in Portugal. Pharmatherapeutica 3, 157-168. Traub, J.L., Gallina, A.M., Grant, B.D., Reed, S.M., Gavin, RR. and Paulsen, L.M. (1983) Phenylbutazone toxicosis in the foal. Am J Vet Res 44, 1410-1418. Troyer, H. (1982) Experimental models of osteoarthritis: a review. Semin Arthritis Rheum 11, 362-374. Tung, J .T., Venta, P.J., Eberhart, S.W., Yuzbasiyan-Gurkan, V., Alexander, L. and Caron, J .P. (2002) Effects of anti-arthritis preparations on gene expression and enzyme activity of cyclooxygenase-2 in cultured equine chondrocytes. Am J Vet Res 63, 1134-1139. Uebelhart, D., Thonar, B.J., Delrnas, P.D., Chantraine, A. and Vignon, E. (1998) Effects of oral chondroitin sulfate on the progression of knee osteoarthritis: a pilot study. Osteoarthritis and Cart / OARS, Osteoarthritis Research Society 6 Suppl A, 39- 46. Volpi, N. (2002) Oral bioavailability of chondroitin sulfate (Condrosulf) and its constituents in healthy male volunteers. Osteoarthritis Cart 10, 768-777. Walker-Bone, K. (2003) 'Natural remedies' in the treatment of osteoarthritis. Drugs Aging 20, 517-526. Warner, T.D., Vojnovic, I., Giuliano, F., Jimenez, R., Bishop-Bailey, D. and Mitchell, J .A. (2004) Cyclooxygenases 1, 2, and 3 and the production of prostaglandin 12: investigating the activities of acetaminophen and cyclooxygenase-2-selective inhibitors in rat tissues. J Pharmacol Exp Ther 310, 642-647. 28 Chapter 2 The Effect of Biologically Relevant Concentrations of Glucosamine and Chondroitin Sulfate on Stressed Equine Cartilage Explants Summary Reasons for performing study: In the past decade, dietary supplements such as glucosamine (GLN) and chondroitin sulfate (CS) have gained popularity in treating joint pain and inflammation due to osteoarthritis (0A) in humans and animals. Although GLN and CS have established anti-inflammatory properties, to our knowledge, no study has tested biologically relevant concentrations of these molecules in equine cartilage explants. Hypothesis: Glucosamine and CS have anti-inflammatory properties at biologically relevant concentrations when tested in equine cartilage explant cultures. Methods: Three culture schemes were used: 1) IL-1 (50 ng/ml) treatment with and without GLN + CS (short-term non-impact) 2) IL-1 plus mechanical trauma with and without GLN + CS (short-term impact) 3) two-week cultures were cultured with and without GLN + CS and were exposed to IL-1 (50 ng/ml) on day 2 and day 10 (long- term non-impact). Nitric oxide (N O) and prostaglandin E2 (PGE2) were measured as indicators of inflammatory response. Glycosarninoglycans (GAG) were measured as an indicator of cartilage turnover. Results: Glucosamine + CS did not affect NO, PGEZ, or GAG concentrations in the non- impact short-term model. When mechanical impact was combined with IL-1 the NO production in the GLN + CS treatment was lower than the IL-1 treatment and did not 29 differ from control. The GAG release was also lower in the GLN + CS treatment than the IL-1 treatment, although the PGE2 concentration was not affected by GLN + CS. The non-impact long-term cultures demonstrated that GLN + CS treatment decreased GAG release and did not affect NO or PGE2 concentration or GAG content in the explants at termination. Conclusions: This study suggests that biologically relevant concentrations of GLN + CS may have protective effects against some indicators of mechanically-induced trauma in explants. Potential relevance: These in vitro data provide support that GLN and CS may be beneficial for equine joint health. Introduction In recent years, dietary supplements (now known as nutraceuticals) have been used to treat osteoarthritis (0A) in both humans and animals. Glucosamine (GLN) and chondroitin sulfate (CS) have been among the most popular dietary supplements taken for joint pain. Glucosamine is a precursor of N-acetyl galactosarnine, which is a component of glycosaminoglycans (GAG). Glycosaminoglycans, such as CS, are components of a proteoglycan called aggrecan, which gives cartilage its distinctive properties. Both of these molecules increase synthesis (Bassleer et al. 1992; Lippiello et al. 2000) and decrease breakdown (Orth et al. 2002; Lippiello 2003) of GAG in cartilage tissue, and they may be an effective treatment for decreasing joint pain in clinical trials (Clegg et al. 2006; Qui et al. 2005). 30 We use three different culture systems with equine” cartilage explants: short-term non-impact used previously in our lab (Chan et al. 2005b), short-term impact system (Ewers et al. 2001), and a long-term non-impact system. Our objective was to test the anti-inflammatory effects of biologically relevant concentrations of GLN and CS in these three different equine cartilage cultures that. We provide evidence that biologically relevant concentrations of GLN and CS may be effective against harmful inflammatory responses to trauma in equine cartilage tissue. Materials and methods Explant Cultures Articular cartilage was isolated from antebrachial-carpal and middle carpal joints of 2- to 9- year-old horses, of several breeds (Thoroughbred, Appaloosa, Tennessee Walking Horse, Arabian, and Paint). Horses were euthanized at The Diagnostic Center for Population and Animal Health at Michigan State University for reasons other than lameness. Cartilage discs (6 mm in diameter) were isolated and randomly distributed to be cultured in 24-well Falcon platesb with two discs per well. Each well contained 1 m1 of media. The medium (Ham media: 1:] Dulbecco’s Modified Eagles Medium: nutrient mixture F-12)a was supplemented as previously described (Fenton et al. 2000). Medium was also supplemented with all 20 amino acids6 at 25% the concentration as previously described (Rosselot et al. 1992). Human recombinant interleukin-1 betae (IL-1) at a concentration of 50 ng/ml was used to induce inflammatory stress. Glucosamine Hle (1, 5, or 10 rig/ml) and low 31 molecular weight (16.9 kilodaltons) chondroitin sulfatef (5, 20, or 50 ug/ml) were all within the range of concentrations known to occur in equine blood after oral administration (Adebowale et al. 2002; Du et al. 2004). Short- Term Non-Impact Model Explants for short-term experiments were harvested from six different horses and equilibrated for 2 d in media without fetal bovine serum21 (FBS). Media was removed and replaced with fresh media 1 d after harvesting. Cultures were then incubated in media supplemented with 10% FBS3 and treatments 1-4 in Table 1 for 2 d. There were 6 to 12 wells per treatment depending on the availability of cartilage from a given horse. Treatments were non-impact treatments were assigned as listed in Table 1. Media were replaced daily until termination of the experiment at 4 d after harvest. Short-term Impact Model Explants from three horses were mechanically impacted as a single acute load of 15 mega pascal (MPa) for 50 msec using a Servo-Hydraulic Testing Machine (Model 1331).g The unconfined explants were compressed between highly polished stainless steel plates and peak load was recorded electronically. There were 6 wells per treatment and the impact treatments were assigned as listed in Table 1. Long- Term Model 32 Explants in long-term cultures were equilibrated as above for 2 d and the media was changed at 24 h after harvesting. Cultures were then incubated in serum-free media supplemented with 1 ul/ml insulin transferring seleniteh (ITS) to termination. There were 9 wells per treatment assigned as listed in Table 11. Media were replaced every 2 (1 until termination of the experiment at 14 d. The cartilage explants were then stored at -20° C for later papain digestion and GAG analysis. All conditioned media samples were stored at 4° C for later assay. All cultures were maintained at 37° C in a humidified incubator with 7% C02. For the short-term non-impact system, there were six horses used (11 = 6) for all treatments except the low dose, which only had four horses (n = 4). For both the short-term impact and the long- terrn non-impact there were three horses (n = 3). Biochemical Analyses: Nitrite levels in the media were assayed as previously described (Blanco et al. 1995; Chan et al. 2005b). Absorbance was detected at 540 nm by a Spectromax 300 plate reader.l Results are expressed as nmol NO/ml. GAG release into the media was assayed by dimethylene blueJ (DMB) assay as previously described (Chandrasekhar 1987). The total amount of GAG in the explant was obtained from papainc-digested cartilage. Results of explant DMB assays are expressed as pg GAG/mg wet weight of cartilage. Prostaglandin E2 was assayed using a commercially available kite according to the manufacturer’s instructions concentrations are reported in pg/ml. Indomethacinc (10 33 ug/ml) was added to the media at collection and samples were stored at -20° C until analysis. To bring the samples within range of the standard curve, samples were diluted 1:10 or 1:20 depending on the PGE2 level of the sample, and PGE2 was detected at 405 nm with a wavelength correction set at 590 nm with a spectrometer. Statistical Analysis Data were analyzed using Fisher’s Least Significant Difference in the PROC MIXED procedure of SAS software.k The data from all wells were pooled according to treatment, with individual horses as replicates. The random effects included horse, treatrnent*horse, and horse*day. The long-term data had the same random effects and were analyzed for each day using Fisher’s Least Significant Difference in the PROC MDIED procedure by using daily average and the repeated measures option. We also analyzed the long-term cumulative total for GAG and NO for all days of the long term using Fisher’s Least Significant Difference, except that the horse*day effect and the repeated measure options were omitted since only one measurement was being analyzed for a total time period. GAG content in explants were also measured with Fisher Least Significant Difference with no horse*day effect and no repeated measures option. A p value of <0.05 was considered significant and p values <0.1 were considered a trend. Results Short- Term Non-Impact Model 34 The explants in the IL-l-only treatment released significantly more NO, PGE2, and GAG compared with the control. The NO concentration in the media in the 5 ug/ml GLN + 20 ug/ml CS (medium dose), and 1 rig/ml GLN + 5 ug/ml CS (low dose) did not differ from the IL-l-only treatment (Fig 1A). The low dose did not differ from the control (Fig 1A). The nutraceutical treatments had no effect on PGEZ or GAG release into the media. (Fig 18, C). Short-term Impact Model The IL-l + impact explants released more NO (p = 0.03) and GAG (p = 0.02) than the control (Fig 2A-C). The impact model included only the medium dose of GLN + CS because of lack of available tissue. The NO concentration in the IL-1 + impact treatment containing GLN + CS was lower than in the IL-1 + impact treatment (p = 0.013) and did not differ from the control (Fig 2A). Neither the IL-1 nor the GLN + CS treatment had a significant effect on PGE2 release into the media (Fig 2B). GAG release in the GLN + CS treatment was lower than in the IL-1 + impact treatment (p = 0.023) (Fig 2C). Long- Term Model The IL-1 treatment resulted in increased NO release in all treatments with IL-1 on d 4, d 12, and total for the two week accumulation (Table 3). The GLN + CS treatments did not affect the IL-l-induced NO release (Table 3). The GAG release was higher in all treatments with IL-1 than those without IL-1 on d 4, but the IL-1 did not effect the GAG release on d 12 (Table 4). The high dose 35 GLN and CS treatment had lower GAG release than the IL-1 treatment on d 4 (p < 0.001) (Table 4). For the cumulative totals, all treatments containing IL-1 were lower than the IL-l-only, except for the control, which tended to be lower (p = 0.093) (Table 4). The high dose GLN and CS treatment had lower GAG release over the two-week period (p = 0.001) and the low dose tended to be lower (p = 0.055). The assay for GAG content in explants did not show any difference between the control and the IL-l-only treatment (Fig 3). Furthermore, the IL-l-only treatment did not differ significantly from the control or any other treatment. Discussion We used equine cartilage explant cultures including short-term, long-term, and mechanical trauma (impact) culture systems. The short-term model has been used befor in out laboratory to test these agents (Chan et al. 20053-c). To our knowledge, this is the first study to test physiologically relevant concentrations of GLN + CS in equine articular cartilage using trauma to stress the explants. This is a relevant way to induce stress is to apply mechanical impact, since OA often results from acute trauma to the joint and subsequent inflammation (Quinn et al. 2001; Patwari et al. 2003). We modeled acute trauma to the cartilage using a hydraulic impact concomitant with IL-1 treatment as previously reported (Quinn et al. 2001; Patwari et al. 2003). A previous study (Dvoracek-Driksna 2001) has demonstrated that impact treatment at 15 MPa pressure causes fissuring and cell death in the superficial tangential zone of bovine cartilage. We used this treatment concomitantly with IL-1, to model trauma and inflammatory stress found in vivo. The long-term study was used to see if there were long-term effects of 36 GLN and CS on cartilage explants. The advantage of the long-term system is that total release of NO and GAG can be calculated at the end of an experiment as well as the average concentration of GAG within the explant for each treatment. This cumulative effect of the treatments may reveal effects that may not be seen over the short-term, especially since these agents are known to be slow acting when compared with conventional treatments (Muller-Fassbender et al. 1994). NO is an inorganic free radical released into the media by the cartilage tissue. Studies using rabbit and human cartilage have shown that NO can increase GAG loss and decrease GAG synthesis (Taskiran et al. 1994; Hardy et al. 2002). Nitric oxide promotes inflammation and cartilage breakdown by increasing cytokine production, suppressing matrix synthesis, and increasing matrix metalloproteinase (MMP) synthesis (T askiran et al. 1994; Evens 1995; Murrell et al. 1995). We demonstrated that the NO release was lower in cartilage explants treated with GLN + CS and impact + IL-l than in cartilage explants treated with impact + IL-l alone, though GLN + CS were not effective without impact as they were in a previous study (Chan et al. 2005b). Prostaglandin E2 was also measured because it is implicated in the pain, inflammation, and cartilage breakdown observed in OA (Hardy et al. 2002; Kirker-Head et al. 2000; Schueter and Orth 2004). Why the GLN and CS treatments were ineffective at decreasing the PGE2 release or why concentration of PGE2 was so much higher in the GLN and CS low dose treatment than even in the IL- l—only treatment in the short-term non-impact model is not known (Fig 1B). In contrast, a previous study has demonstrated success in effecting a change in PGEz release with GLN + CS at these concentrations in explants from Holstein steers (Chan et al. 2005b). Other studies that used equine 37 cartilage demonstrated positive affects of GLN (alone) on PGEz, but at much higher (up to 100 times) concentrations of GLN used in the present study (Orth et al. 2002; Schlueter and Orth 2004). IL-l-induced MMP synthesis causes cartilage matrix breakdown, which we quantified by measuring GAG release (Chan et al. 2005a). GLN alone decreases IL-l- induced proteoglycan breakdown (Fenton et al. 2000; Gouze et al. 2001; Fenton et al. 2002). CS also may reduce proteoglycan loss in part by decreasing MMP-13 (Chan et al. 2005a). Another study showed a synergistic relationship between GLN and CS for reversing cartilage damage and promoting repair (Homandberg et al. 2006). In the current study, the high dose was most effective at protecting cartilage against the loss of GAG (Table 4), and there was also an effective protection using the medium dose in the impact system Gig 2 C). There was no treatment effect on the GAG content in the explants at termination (Fig 4). Chan et al. (2005b) have previously demonstrated that genes responsible for NO and PGE2 synthesis are down-regulated when bovine explant cultures are exposed to physiologically relevant concentrations of GLN + CS. Metalloprotienases responsible for breakdown of the cartilage matrix are also down regulated (Chan et al. 20053). The present study tested the same physiologically relevant concentrations of GLN and CS as those used by Chan et al. (2005a-b). There were instances in this study where the GLN + CS treatments were less effective than reported in earlier studies. It is unknown why the PGE2 concentration was unaffected by the GLN + CS treatment in either the short-term impact experiment or the short-term non-impact experiments. It is also difficult to explain the lack of responsiveness of NO and GAG level to GLN + CS treatment in the short- 38 term non-impact experiment. Significant differences may not have been found because the GLN + CS concentrations may have been too low to effect a change. Also, the variation between horses, which differed with respect to breed and age, may have been too high to detect a statistically significant difference between treatments. The Chan et al. (2005a-b) studies used bovine species of a single breed and common age, which may have decreased the variation in these studies and made finding differences more statistically possible. Conclusion Our hypothesis was that GLN + CS would decrease inflammatory markers and cartilage breakdown at physiologically relevant concentrations. Our results indicate that GLN + C8 are effective in mitigating harmful inflammatory responses due to mechanical impact + IL-1 in equine cartilage tissue. The advantage of this impact model is the combination of impact and IL-1. The reason for this is that 0A is often initiated by trauma to the joint, which is then followed by IL-l-mediated inflammation (Quinn et al. 2001; Patwari et al. 2003). This makes the conditions in this system somewhat similar to those found in vivo. We demonstrated that the NO release was lower in cartilage explants treated with GLN + CS and impact + IL-l than in cartilage explants treated with impact + IL-l alone. We have also shown that the GAG release was lower in cartilage explants treated with GLN + CS and impact + IL-l than in cartilage explants treated with impact + IL-l alone. We also demonstrated that, over a two-week period, the GAG release was lower in cartilage explants treated with GLN + CS and IL-1 than in cartilage explants treated with IL-1 alone. This study provide some support these supplements are 39 beneficial for horses, in maintaining equine joint health, although in vivo research should be done to confirm the findings. Manufacturer's address aGibco, Grand Island, New York, USA. bFisher Scientific, Pittsburgh, Pennsylvania, USA. cSigma Chemical, St Louis, Missouri, USA. dJ.T. Baker, Phillipsburg, New Jersey, USA. eR&D Systems, Minneapolis, Minnesota, USA. {NutIamax Laboratories, Edgewood, Maryland, USA. gInstron Corrperation, Canto, Massachusetts, USA. hRoche Diagnostics Corporation, Indianapolis, Indiana, USA. iMolecular Devices, Sunnyvale, California, USA. jPolyscience, Inc., Warrington, Pennsylvania, USA. kSAS Institute, Inc., Cary, North Carolina, USA. 40 no 3 - 2 38 82.8.2 awe. + 7... a «.0 o? - 2 moon :9: 39¢. + T... A» m.o - - 2 .295 + T... a - - :3 2 $8 33 22% + 7... a - - to 9 38 528.2 22% + 7... a. .. - F 2 $8 :2: 225 + 3. 8 - - - 2 28-3. 3 - - - - 3. oz “.928 : Ac\ov 2533 9833 9535 locum Dm< 652d 7.: E953; 255.8: so as shoe 2 8%.... emu so: “a EeEtquoAmcgao 39on 2 tonne £53. . 2an 41 no to m. 9.8 26.. 2w< + 7... 6 no 3 B 88 53.8.2 3m< + r... a no 3: 2 88 :9... 2w... + 7... a. no - 8 .295 + w... a - - m? 25.7.: N - - - w... oz .228 2 as 9553 seas roam. :9... 7... Eoeao: 22258.. E 5 £85 o. n88 mm“. so: Na LeoEteqxfmmcSEo 33qu 9 peace 3:63. = 2an 42 mammmmmmmo. .1. Fig I: Mean (iSE, n=6 for medium dose, n# for low dose) NO, GAG, and PGE2 concentrations in media samples at d 1 (A, C) or at d 1 (B). IL-1 (50 ng/ml) was added to each treatment except for control. The experimental treatments contained 5 11ng GLN + 20 11me CS (medium dose) or 1 ug/ml GLN + 5 ug/ml CS (low dose). 45 Fig 2: Mean (iSE, n=3) NO, PGEz, and GAG concentrations in media samples at d O and 1 (A, C) or at d 1 (B). IL-1 (50 ng/ml) + Impact was added to each treatment except for control. The experimental treatment contained 5 ug/ml GLN + 20 ug/ml CS (medium dose). GLN + CS treatment differed from IL-l-only treatment in NO (p = 0.0013) and GAG (p = 0.023) concentrations. 48 .2... 3 8.8.2. .85 A a .r. 2.. cases 55 v a .. 2.. 38...... 3o Vao 2:? a ”550% 8a Am .8 «52.53.: 7.: 05 Sea moocoaobmu .eosmwfim .Aomou :wEv m0 ER: on + 750 web: 3 .8 .686 8:625 me .529. 8 + 2.6 2%... m .38.. so: me .529. m + 7:0 :5»... 2 853:8 358.8: 3885...... 2F .3-.. 5.3 mm a totem 08: e 3 .295 «:68 E cote—22838 02 mo 33 952383 Ea moron S e was v u .36 no 22323588 CZ 532 am. EEK 2.2 a 32 E H 8.8 S a to... moon 5...: me + 25 + 2-... a «.3 u." 32 no a 8.8 as a as 38 528.2 me + 2.9 + 3. C E. a 32 a; s New we .2 So 88 25.. me + 2.6 + T... 6 92 a 39 no o 38 car a «.3 7... 3 Eco H was 33 o 2. 2 i? u 4.2 $8 :9: me + 50 .4 im: a m. x: is a 2.: i: a 5.. 88 53.8.2 me + 2.6 8 $2.9 .2 Es imam a 5.2 i? a 4.3 88 25.. me + 250 Am ENS « on: ram .2 t. 3 i? o 0.2 585. oz. 7... oz ”.9280 9 9:52:30 ~_. .80 330 2953.... name I .620 9522:2230 new N“ .v ED mu: 5.2. mm o is 8.28.2828 oz :83. _: aim... 49 .222... .3 323:2: Sod A a .2. .3 350%.: Ed v a .2. .3 @2865 36 v he 62? a “2305 2.1m av Eugen: 7.: 05 Sea meanest? 88:3 .68.. 8.5 8 ES... S + 2.5 ES: 2 8 .988 8288 mo in: om + 2.6 new: A .98.. so; we :5»... m + 750 BE: 2 358.80 858382 3:68:88 2:. .w 8 m 35.58: 8 Beta 33 GEE: any 7.: Am": 53> mm HV totem 2:: v 3 BE «62: E 8:228:08 02 mo :32 028—253 28 cocoa E u use v w .98 no 8:888:00 O