, . . ua 3. i . 3......aLT.“ 0». L . 32$... .m» . 3 a... .. '4‘. . .317 35?...” . . 2 :7... {4.1. 1 1 ”Ti .33.. 1 tAzltll ”.37. 2...... m... an... 5.3. 3.: .\1 7.1 J. A u... u. .31!" hr nmuwmfifihnfl: .Kphumm4 w. 515.15 a. 13% . [1a . .1.» \ L3... . , s... '19.}; :3: N! O 15?.kamuma s...i.u....u.nu .5 Q11 0". Law». 3395.3. 1. 3.1.5. .1. t... «have! \ .2... “Hanan v.3... avail. 1.3.3.9....“ xfimfv. at}: ... .. 2.... 7.. <2}. 53.2..th 231... . .5 x15 . x... D. h. s. Vx \.L’c:. O! .12. ...in....Har.l, 5?, .3: “(.551 3.... .u. (t! a... . .n. h....9....fi.....; $443.. . . z _‘ Raga . Hm? mils£§ér 5. . 1...”? L... A 3’33: ’3 LIBRARIES V \ UNIVERSITY r—x ~ MICHIGAN STATE 5‘. “13'? EAST LANSING, MICH 48824 1048 (95704296 This is to certify that the dissertation entitled GLUCOSAMINE AND CHONDROITIN SULFATE REGULATE INTERLEUKlN-1 INDUCED MEDIATORS OF OSTEOARTHRITIS IN ARTICULAR CARTILAGE EXPLANTS presented by POOl-SEE CHAN has been accepted towards fulfillment of the requirements for the Ph.D. degree in Animal Science Wade/42¢ Major Professor’ 3 Signature 5/Qfiz>z>s Date MSU is an Affirmative 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 c:ElRC/Da!eDuo.lndd-p.15 GLUCOSAMINE AND CHONDROITIN SULFATE REGULATE INTERLEUKIN-l INDUCED MEDIATORS OF OSTEOARTHRITIS 1N ARTICULAR CARTILAGE EXPLANTS By Pooi-See Chan A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Animal Science 2005 ABSTRACT GLUCOSAMIN E AND CHONDROITIN SULFATE REGULATE INTERLEUKIN-l INDUCED MEDIATORS OF OSTEOARTHRITIS IN ARTICULAR CARTILAGE EXPLANTS By Pooi-See Chan Osteoarthritis (0A) is a significant problem for both humans and animals. Nutraceuticals such as glucosamine and chondroitin sulfate (CS) are widely used to alleviate symptoms of 0A. However, the mechanism(s) of action of these nutraceuticals remains unclear. Bovine articular cartilage explants added with 50 ng/ml interleukin-1 (IL-1) to stimulate cartilage catabolism were used to determine the molecular events of the nutraceuticals. Glucosamine (5 ug/ml) and CS (20 ug/ml), at concentrations attainable in viva, were supplemented individually or in combination. Glucosamine and CS regulated transiently nitric oxide (NO) synthesis and the expression of genes encoding inducible nitric oxide synthase (iNOS) and microsomal prostaglandin E synthase-l (mPGEsl). There was concomitant repression of IL-1 induced expression of cyclooxgenase—Z (COX-2) transcript with prostaglandin E2 (PGEz) production. Glucosamine and CS reduced cytokine up-regulated expressions of matrix metalloproteinase (MMP)-3, MMP-13, aggrecanase (Agg)-l and Agg-Z. The nutraceutical combination was more effective in antagonizing some gene expression than if glucosamine and CS were used individtrally. There were no treatment effects on gene expression of tissue inhibitor of metalloproteinase (TIMP)-l, TIMP-Z, type II collagen and aggrecan although the glucosamine and CS combination tended to elevate TIMP-3 transcript. In a separate study, bovine articular cartilage explants were induced with a subsaturating dose of IL-1 (15 ng/ml) and treated with glucosamine (10 pg/ml) and CS (20 ug/ml). The regulation of molecular events was also characterized at 8, 16 and 24 hours after culture. The combination abrogated IL-l induced gene expression of iNOS, COX-2, mPGEsl and nuclear factor kappa beta p65 subunit at all time points. This was accompanied by decreases in NO and PGE2 synthesis. Stimulated MMP-l3, Agg-l and Agg-2 mRNA abundance were down-regulated while TIMP-3 was increased by the nutraceutical combination at all time points. TIMP-3 protein abundance was elevated by the combination. Proteoglycan release, an indicator of proteoglycan loss was suppressed by glucosamine and CS combination. Thus, glucosamine and CS in combination repress the synthesis and expression of genes encoding inflammatory mediators and matrix catabolic enzymes while up-regulating TIMP-3. This provides a plausible mechanism for their mild anti-inflammatory and chondroprotective properties. This dissertation is dedicated to my dearest husband, Boon-Sam for his unconditional love. iv ACKNOWLEDGEMENTS I would like to thank Peggy Wolf, Angie Schlueter, John Wheeler, Tonya Skuse, Dene Elliot, and Tonia Peters, past and present members from Dr. Orth’s laboratory for all their help. Many thanks also go to Chris Colvin, Xiaoning Ren and Sue Sipkovsky from the Center For Animal Functional Genomics laboratory for technical assistance. I would also like to thank Ling-Chu Chang, Shih—Kai Chiang, Dr. Qinglei Li and Dr. Patty Weber for their aid and advice concerning molecular techniques. Sincere appreciation also goes to Jackie Christie and Barb Sweeney for help pertaining to paper work and administrative issues. My deepest appreciation goes to Dr. Mike Orth for giving me the opportunity to learn from him, for his guidance and encouragement throughout my stay at MSU. I am also indebted to Dr. John Caron for his continuous guidance, support and invaluable inputs. My gratitude extends to the rest of my guidance committee members, Dr. Jeanne Burton, Dr. Paul Coussens and Dr. George Smith for their brilliant ideas on experimental designs, for their advice, for allowing me to use their facilities and for their constructive criticisms. I would also like to thank Dr. Guilherme Rosa for all his help on statistical analysis. Special thanks goes out to all my brothers and sisters-in-Christ and Auntie Khoo for their friendship, concern and prayers. Appreciation is expressed to my mom, Sau- Lung for her continuous love and support. Love and gratitude also goes to my husband, Boon-Sam for loving and supporting me, and for believing in me. Lastly, I would like to thank the Lord for all the blessings that He has showered upon me. TABLE OF CONTENTS LIST OF TABLES ,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,, viii LIST OF FIGURES ________________________________________________________________________________________________________________ 1X LIST OF ABBREVIATIONS ________________________________________________________________________________________________ xi INTRODUCTION ..................................................................................................................... 1 CHAPTER 1 LITERATURE REVIEW .......................................................................................................... 6 Articular Cartilage _________________________________________________________________________________________________________ 6 Osteoarthritis .................................................................................................................. 8 Mediators of OA ___________________________________________________________________________________________________________ 9 Cytokines ___________________________________________________________________________________________________________ 9 Inflammatory Mediators ________________________________________________________________________________ 14 Nitric Oxide _______________________________________________________________________________________ l4 Prostaglandin E2 _________________________________________________________________________________ 16 Matrix Enzymes _____________________________________________________________________________________________ 17 Matrix Metalloproteinases ________________________________________________________________ 17 Aggrecanases ______________________________________________________________________________________ 19 Tissue Inhibitor of Metalloproteinases _____________________________________________ 20 Glucosamine and Chondroitin Sulfate as Therapeutic Agents for CA ___________________ 21 Structure and Composition ________________________________________________________________ 21 Pharmacokinetics _______________________________________________________________________________ 23 Dosage and Administration _______________________________________________________________ 24 Safety Profile ______________________________________________________________________________________ 25 Clinical Trials _____________________________________________________________________________________ 26 Mechanism(s) of Action ____________________________________________________________________ 28 Rationale for experiments __________________________________________________________________________________________ 31 References ................................................................................................................... 36 CHAPTER 2 GLUCOSAMINE AND CHONDROITIN SULFATE REGULATE GENE EXPRESSION AND SYNTHESIS OF NITRIC OXIDE AND PROSTAGLANDIN E2 IN ARTICULAR CARTILAGE EXPLANT S Summary ______________________________________________________________________________________________________________________ 65 Introduction ,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,, 66 Materials and Methods ,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,, 68 Results __________________________________________________________________________________________________________________________ 72 Discussion ,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,, 75 Acknowledgements _____________________________________________________________________________________________________ 80 References 88 ................................................................................................................... vi CHAPTER 3 GLUCOSAMINE AND CHONDROITIN SULFATE REGULATE GENE EXPRESSION OF PROTEOLYTIC ENZYMES AND THEIR INHIBITORS IN INTERLEUKIN-l CHALLENGED BOVINE ARTICULAR CARTILAGE EXPLANTS Abstract ________________________________________________________________________________________________________________________ 96 Introduction ,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,, 97 Materials and Methods ________________________________________________________________________________________________ 98 Results ________________________________________________________________________________________________________________________ 101 Discussion ,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,, 103 Footnotes ____________________________________________________________________________________________________________________ 108 References _________________________________________________________________________________________________________________ 1 15 CHAPTER 4 SHORT TERM GENE EXPRESSION CHANGES IN INTERLEUKIN-IB STIMULATED CARTILAGE EXPLANT S WITH GLUCOSAMINE AND CHONDROITIN SULFATE Abstract ______________________________________________________________________________________________________________________ 122 Introduction ,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,, 123 Materials and Methods _____________________________________________________________________________________________ 125 Results ________________________________________________________________________________________________________________________ 131 Discussion __________________________________________________________________________________________________________________ 13 3 References _________________________________________________________________________________________________________________ 149 CHAPTER 5 CONCLUSION l55 ..................................................................................................................... vii LIST OF TABLES Chapter I Table 1. Summary of in vitro mechanistic studies with glucosamine and Chondroitin sulfate using articular cartilage explants or articular chondrocytes 33 Chapter 2 Table 1. Description of components found in treatment media of explant cultures ............ 81 Table II. Forward and reverse primers (5'—>3') used for quantitative real-time polymerase chain reaction 32 Table III. Relative gene expression of explants stimulated with rhIL-l B and physiologically relevant concentrations of glucosamine and Chondroitin sulfate ,,,,,,,,,,,,,,, 83 Chapter 3 Table I. Description of components found in treatment media of explant cultures .......... 109 Table 11. Forward and reverse primer sequences (5'—->3') of genes of interest used for quantitative real-time polymerase chain reaction __________________ 110 Chapter 4 Table I. Forward and reverse primer sequences (5'—-)3') of genes of interest used for quantitative real-time polymerase chain reaction 139 Table II, Nitrite and PGE2 release from explants at 8, l6 and 24 hours post-stimulation 140 viii LIST OF FIGURES Chapter 2 Figure 1 (next page). Mean (:hSE) nitrite release into conditioned media 24 hours (Panel A) and 48 hours (Panel B) afier treatments are added. Ctrl= fetal bovine serum control; IL-1= 50 ng/ml rhIL-lB (human recombinant interleukin-1 beta); IL-l + GLN= 50 ng/ml rhIL-IB and 5 ug/ml glucosamine; IL-l + CS= 50 ng/ml rhIL-IB and 20 ug/ml chondroitin sulfate; IL—l + GLN + CS= 50 ng/ml rhlL-lB, 5 ug/ml glucosamine and 20 ug/ml Chondroitin sulfate. Different superscripts indicate significant differences at P<0.05 84 .......................................................................................... Figure 2 (next page). Mean (iSE) prostaglandin E2 (PGE2) release into conditioned media 24 hours (Panel A) and 48 hours (Panel B) after the addition of treatments. Ctrl= fetal bovine serum control; IL-l= 50 ng/ml rhlL-IB (human recombinant interleukin-1 beta); IL-l + GLN= 50 ng/ml rhIL-IB and 5 ug/ml glucosamine; IL-l + CS= 50 ng/ml rhIL-IB and 20 ug/ml Chondroitin sulfate; IL-l + GLN + CS= 50 ng/ml rhIL-lB, 5 ug/ml glucosamine and 20 ug/ml Chondroitin sulfate. Different superscripts indicate significant differences at P<0.05. * Tended to be different than the IL-1 treatment (Pom Nm2 a $3583 coma=oo : 095. am assess on: as»: 2532 35:: 8N Essa... 0.: 2&1 82 8 uses: man. 2858? 0,2. 15w: 2:; m0 58:2 emit”... D... .r , v.8 2:285 5233.8 .8 flea—axe owe—Esq 5:33.:— mE»: 8.5.: £39655 2:. Sings—.3 5...» 8:53 03352—8:— E»? a.» we FEES—5 Q ofiah 33 2m 005 8:2» Nmam» 52.5 $80» <58 $80» :5»... 33 8220 m0 585m 3N ease»... 0<0e as»: San... 240 0525 oom £88m 508mm: (.728 500%me 88:8 0.256 33:8 v.22» £88m TEA, as»: 2-8... mo 58:: com 0328 0.3. £228 .22» £03 02» ER... 3-36 730 ofiscm cam 33:8 222» N”.20.: oz» .aaamdANO Eu... 2 :5»... 2-23 sea... Two 240 cfiscm 2m $22» <72... 5:25 _amame 730 and :m 03:» £88m Nico». 5,88 380» case 823 <73: mozm» oz» _Eaasa cab/~20 583m 9.283 a 033—. 34 53m 8: 055928 332305 .82 5:888:00... H caummqaémmoaoaoflwufihaxo—O ”gubmsuoancm m; @228 2-222» .002» 02» in... RS Ea... 2-3 2.20 2.35 3m 0322 Om» oz» in... 2-2 756 cE>om Cm 2-222» 222» 7.22» N00.2» 02» En: 82 7:0 Quasi vom mmO— goomwafi Ema e. 73 240 oE>om mom <22... 2-222» Ema $-36 20 052cm £5.50 fl oEaH 35 References 1. Kolettas E, Muir HI, Barrett JC, Hardingham TE. Chondrocyte phenotype and cell survival are regulated by culture conditions and by specific cytokines through the expression of Sox-9 transcription factor. Rheumatology 2001; 40:1146-56. 2. Bruckner P, van der Rest M. Structure and function of cartilage collagens. Microsc Res Tech 1994; 28:378-84. 3. Maroudas AI. Balance between swelling pressure and collagen tension in normal and degenerate cartilage. Nature 1976; 260:808-9. 4. Carney SL, Muir H. The structure and function of cartilage proteoglycans. Physiol Rev 1988; 68:858-910. 5. Hardingham TE, Muir H. Binding of oligosaccharides of hyaluronic acid to proteoglycans. Biochem J 1973; 135:905-8. 6. Hascall VC, Riolo RL. Characteristics of the protein-keratan sulfate core and of keratan sulfate prepared from bovine nasal cartilage proteoglycan. J Biol Chem 1972; 247:4529-38. 7. Maroudas A, Bannon C. Measurement of swelling pressure in cartilage and comparison with the osmotic pressure of constituent proteoglycans. Biorheology 1981; 18:619-32. 8. Mankin HJ, Lippiello L. The glycosaminoglycans of normal and arthritic cartilage. J Clin Invest 1971; 50: 1712-9. 9. Mollenhauer JA, Erdmann S. Introduction: molecular and biomechanical basis of osteoarthritis. Cell Mol Life Sci 2002; 59:3-4. 10. Vingard E, Sandmark H, Alfredsson L. Musculoskeletal disorders in former athletes. A cohort study in 114 track and field champions. Acta Orthop Scand 1995; 66:289-91. 11. Felson DT, Naimark A, Anderson J, Kazis L, Castelli W, Meenan RF. The prevalence of knee osteoarthritis in the elderly. The Framingham Osteoarthritis Study. Arthritis Rheum 1987; 30:914-8. 12. Spector TD, Harris PA, Hart DJ, Cicuttini FM, Nandra D, Etherington J, et al. Risk of osteoarthritis associated with long-term weight-bearing sports: a radiologic survey of the hips and knees in female ex-athletes and population controls. Arthritis Rheum 1996; 39:988-95. 13. Dunlop DD, Manheim LM, Yelin EH, Song J, Chang RW. The costs of arthritis. Arthritis Rheum 2003; 49:101-13. 36 14. Ayral X, Dougados M, Listrat V, Bonvarlet JP, Simonnet J, Amor B. Arthroscopic evaluation of chondropathy in osteoarthritis of the knee. J Rheumatol 1996; 23:698-706. 15. Buckland-Wright JC, Macfarlane DG, Williams SA, Ward RJ. Accuracy and precision of joint space width measurements in standard and macroradiographs of osteoarthritic knees. Ann Rheum Dis 1995; 54:872-80. 16. Fubini SL, Erb HN, Freeman KP, Todhunter RJ. Prognostic factors affecting survival of 507 horses with joint disease: (1983 to 1990). Can J Vet Res 1999; 63:253-60. 17. Malone ED, Les CM, Turner TA. Severe carpometacarpal osteoarthritis in older Arabian horses. Vet Surg 2003; 32: 191-5. 18. Moore GE, Burkrnan KD, Carter MN, Peterson MR. Causes of death or reasons for euthanasia in military working dogs: 927 cases (1993-1996). J Am Vet Med Assoc 2001; 219:209-14. 19. Johnston SA. Osteoarthritis. Joint anatomy, physiology, and pathobiology. Vet Clin 1997; 27:699-723. 20. Schwarz T, Weller R, Dickie AM, Konar M, Sullivan M. Imaging of the canine and feline temporomandibular joint: a review. Vet Radiol Ultrasound 2002; 43:85-97. 21. Jergensen B. Osteochondrosis/osteoarthrosis and claw disorders in sows, associated with leg weakness. Acta Vet Scand 2000; 41 :123-38. 22. Scott PR. Osteoarthritis of the elbow joint in adult sheep. Vet Rec 2001; 149:652-4. 23. Mankin I-IJ, Dorfrnan H, Lippiello L, Zarins A. Biochemical and metabolic abnormalities in articular cartilage from osteo-arthritic human hips. II. Correlation of morphology with biochemical and metabolic data. J Bone Joint Surg Am 1971; 53:523- 37. 24. Donohue JM, Buss D, Oegema TR, Jr., Thompson RC, Jr. The effects of indirect blunt trauma on adult canine articular cartilage. J Bone Joint Surg Am 1983; 65:948-57. 25. Yoshimura N, Nishioka S, Kinoshita H, Hori N, Nishioka T, Ryujin M, et al. Risk factors for knee osteoarthritis in Japanese women: heavy weight, previous joint injuries, and occupational activities. J Rheumatol 2004; 3 1 : 157-62. 26. Englund M, Roos EM, Lohmander LS. Impact of type of meniscal tear on radiographic and symptomatic knee osteoarthritis: a sixteen-year followup of meniscectomy with matched controls. Arthritis Rheum 2003; 48:2178-87. 27. Felson DT, Anderson JJ, Naimark A, Walker AM, Meenan RF. Obesity and knee osteoarthritis. The Framingham Study. Ann Intern Med 1988; 109:18-24. 37 28. Verzijl N, Bank RA, TeKoppele JM, DeGroot J. AGEing and osteoarthritis: a different perspective. Curr Opin Rheumtol 2003; 15:616-22. 29. Knowlton RG, Katzenstein PL, Moskowitz RW, Weaver EJ, Malemud CJ, Pathria MN, et al. Genetic linkage of a polymorphism in the type II procollagen gene (COL2A1) to primary osteoarthritis associated with mild chondrodysplasia. New Engl J Med 1990; 322:526-30. 30. Smith GK, Mayhew PD, Kapatkin AS, McKelvie PJ, Shofer F S, Gregor TP. Evaluation of risk factors for degenerative joint disease associated with hip dysplasia in German Shepherd Dogs, Golden Retrievers, Labrador Retrievers, and Rottweilers. J Am Vet Med Assoc 2001; 219: 1719-24. 31. Dean DD, Martel-Pelletier J, Pelletier JP, Howell DS, Woessner JF, Jr. Evidence for metalloproteinase and metalloproteinase inhibitor imbalance in human osteoarthritic cartilage. J Clin Invest 1989; 84:678-85. 32. Lohmander LS. Articular cartilage and osteoarthrosis. The role of molecular markers to monitor breakdown, repair and disease. J Anat 1994; 184:477-92. 33. Hamerman D. Aging and osteoarthritis: basic mechanisms. J Am Geriatr Soc 1993; 41 :760-70. 34. Tsai CL, Liu TK, Chen TJ. Estrogen and osteoarthritis: a study of synovial estradiol and estradiol receptor binding in human osteoarthritic knees. Biochem Biophys Res Commun 1992; 183:1287-91. 35. Dinarello CA. Interleukin-1. Ann N Y Acad Sci 1988; 546:122-32. 36. Caron JP, Femandes JC, Martel-Pelletier J, Tardif G, Mineau F, Geng C, et al. Chondroprotective effect of intraarticular injections of interleukin-1 receptor antagonist in experimental osteoarthritis. Suppression of collagenase-1 expression. Arthritis Rheum 1996; 39:1535-44. 37. Westacott CI, Sharif M. Cytokines in osteoarthritis: mediators or markers of joint destruction? Semin Arthritis Rheum 1996; 25:254-72. 38. Dinarello CA, Savage N. Interleukin-1 and its receptor. Crit Rev Immunol 1989; 9:1- 20. 39. Pelletier JP, DiBattista JA, Roughley P, McCollum R, Martel-Pelletier J. Cytokines and inflammation in cartilage degradation. Rheum Dis Clin North Am 1993; 19:545-68. 40. Towle CA, Trice ME, Ollivierre F, Awbrey BJ, Treadwell BV. Regulation of cartilage remodeling by IL- 1: evidence for autocrine synthesis of IL-1 by chondrocytes. J Rheumatol 1987; 14 Spec No:11-3. 38 41. Wood DD, Ihrie EJ, Dinarello CA, Cohen PL. Isolation of an interleukin-l-like factor from human joint effusions. Arthritis Rheum 1983; 26:975-83. 42. Nouri AM, Panayi GS, Goodman SM. Cytokines and the chronic inflammation of rheumatic disease. I. The presence of interleukin-1 in synovial fluids. Clin Exp Immunol 1984; 55:295-302. 43. March CJ, Mosley B, Larsen A, Cerretti DP, Braedt G, Price V, et a1. Cloning, sequence and expression of two distinct human interleukin-l complementary DNAs. Nature 1985; 315:641-7. 44. Martel-Pelletier J, McCollum R, DiBattista J, Faure MP, Chin JA, Fournier S, et al. The interleukin-1 receptor in normal and osteoarthritic human articular chondrocytes. Identification as the type 1 receptor and analysis of binding kinetics and biologic function. Arthritis Rheum 1992; 35:530-40. 45. Pujol JP, Loyau G. Interleukin-1 and osteoarthritis. Life Sci 1987; 41:1187-98. 46. Kronheim SR, Mumma A, Greenstreet T, Glackin PJ, Van Ness K, March CJ, et a1. Purification of interleukin-1 beta converting enzyme, the protease that cleaves the interleukin-1 beta precursor. Arch Biochem Biophys 1992; 296:698-703. 47. Boileau C, Martel-Pelletier J, Moldovan F, Jouzeau JY, Netter P, Manning PT, et al. The in situ up-regulation of chondrocyte interleukin- l-converting enzyme and interleukin-18 levels in experimental osteoarthritis is mediated by nitric oxide. Arthritis Rheum 2002; 46:2637-47. 48. Bankers-Fulbright JL, Kalli KR, McKean DJ. Interleukin-l signal transduction. Life Sci 1996; 59:61-83. 49. Geng Y, Valbracht J, Lotz M. Selective activation of the mitogen-activated protein kinase subgroups c-Jun NH2 terminal kinase and p38 by IL-1 and TNF in human articular chondrocytes. J Clin Invest 1996; 98:2425-30. 50. Mengshol JA, Vincenti MP, Coon CI, Barchowsky A, Brinckerhoff CE. Interleukin-l induction of collagenase 3 (matrix metalloproteinase 13) gene expression in chondrocytes requires p3 8, c-Jun N-terrninal kinase, and nuclear factor kappaB: differential regulation of collagenase 1 and collagenase 3. Arthritis Rheum 2000; 43:801-11. 51. Hui A, Min WX, Tang J, Cruz TF. Inhibition of activator protein 1 activity by paclitaxel suppresses interleukin-l-induced collagenase and stromelysin expression by bovine chondrocytes. Arthritis Rheum 1998; 41 :869-76. 52. Liacini A, Sylvester J, Li WQ, Zafarullah M. Inhibition of interleukin-l-stimulated MAP kinases, activating protein-l (AP-1) and nuclear factor kappa B (NF -kappa B) 39 transcription factors down-regulates matrix metalloproteinase gene expression in articular chondrocytes. Matrix Biol 2002; 21 :25 1-62. 53. Malemud CJ, Islam N, Haqqi TM. Pathophysiological mechanisms in osteoarthritis lead to novel therapeutic strategies. Cells Tissues Organs 2003; 174:34-48. 54. Nolan JC, Gathright CE, Wagner LE. The effect of calcium channel blockers and calmodulin inhibitors on the macrophage factor-stimulated synthesis of collagenase by rabbit chondrocytes. Agents Actions 1988; 25:71-6. 55. Richard M, Broquet P, Vignon E, Peschard MJ, Carret JP, Louisot P. Calmodulin- dependent collagenase and proteoglycanase activities in chondrocytes from human osteoarthritic cartilage. Biochem Biophys Res Commun 1991; 174: 1204-7. 56. Chadjichristos C, Ghayor C, Kypriotou M, Martin G, Renard E, Ala-Kokko L, et al. Spl and Sp3 transcription factors mediate interleukin-1 beta down-regulation of human type II collagen gene expression in articular chondrocytes. J Biol Chem 2003; 278:39762-72. 57. Kubota E, Kubota T, Matsumoto J, Shibata T, Murakami KI. Synovial fluid cytokines and proteinases as markers of temporomandibular joint disease. J Oral Maxillofac Surg 1998; 56:192-8. 58. Lefebvre V, Peeters-Joris C, Vaes G. Modulation by interleukin 1 and tumor necrosis factor alpha of production of collagenase, tissue inhibitor of metalloproteinases and collagen types in differentiated and dedifferentiated articular chondrocytes. Biochim Biophys Acta 1990; 1052:366-78. 59. Ohta S, Imai K, Yamashita K, Matsumoto T, Azumano 1, Okada Y. Expression of matrix metalloproteinase 7 (matrilysin) in human osteoarthritic cartilage. Lab Invest 1998; 78:79-87. 60. Yamada H, Kikuchi T, Nemoto O, Obata K, Sato H, Seiki M, et al. Effects of indomethacin on the production of matrix metalloproteinase-3 and tissue inhibitor of metalloproteinases-1 by human articular chondrocytes. J Rheumatol 1996; 23:1739-43. 61. Shingu M, Miyauchi S, Nagai Y, Yasutake C, Horie K. The role of IL-4 and IL-6 in IL-l-dependent cartilage matrix degradation. British J Rheumatol 1995; 34: 101-6. 62. Bunning RA, Richardson HJ, Crawford A, Skjodt H, Hughes D, Evans DB, et al. The effect of interleukin-1 on connective tissue metabolism and its relevance to arthritis. Agents Actions 1986; 18:131-52. 63. Campbell 1K, Roughley PJ, Mort J S. The action of human articular-cartilage metalloproteinase on proteoglycan and link protein. Similarities between products of degradation in situ and in vitro. Biochem J 1986; 237:117-22. 40 64. Tetlow LC, Adlam DJ, Woolley DE. Matrix metalloproteinase and proinflammatory cytokine production by chondrocytes of human osteoarthritic cartilage: associations with degenerative changes. Arthritis Rheum 2001; 44:585-94. 65. Reboul P, Pelletier JP, Tardif G, Cloutier JM, Martel-Pelletier J. The new collagenase, collagenase-3, is expressed and synthesized by human chondrocytes but not by synoviocytes. A role in osteoarthritis. J Clin Invest 1996; 97:2011-9. 66. Borden P, Solymar D, Sucharczuk A, Lindman B, Cannon P, Heller RA. Cytokine control of interstitial collagenase and collagenase-3 gene expression in human chondrocytes. J Biol Chem 1996; 271 :23577-81. 67. Shlopov BV, Lie WR, Mainardi CL, Cole AA, Chubinskaya S, Hasty KA. Osteoarthritic lesions: involvement of three different collagenases. Arthritis Rheum 1997; 40:2065-74. 68. Amer EC, Hughes CE, Decicco CP, Caterson B, Tortorella MD. Cytokine-induced cartilage proteoglycan degradation is mediated by aggrecanase. Osteoarthritis Cart 1998; 62214-28. 69. McCachren SS, Greer PK, Niedel J. Regulation of human synovial fibroblast collagenase messenger RNA by interleukin-1. Arthritis Rheum 1989; 32:1539—45. 70. Vincenti MP, Coon CI, Lee 0, Brinckerhoff C. Regulation of collagenase gene expression by IL-1 beta requires transcriptional and post-transcriptional mechanisms. Nucleic Acids Res 1994; 22:4818-27. 71. Femandes JC, Martel-Pelletier J, Pelletier JP. The role of cytokines in osteoarthritis pathophysiology. Biorheology 2002; 39:237-46. 72. Shingu M, Nagai Y, Isayama T, Naono T, Nobunaga M. The effects of cytokines on metalloproteinase inhibitors (TIMP) and collagenase production by human chondrocytes and TIMP production by synovial cells and endothelial cells. Clin Exp Immunol 1993; 94: 145-9. 73. Stefanovic-Racic M, Morales TI, Taskiran D, McIntyre LA, Evans CH. The role of nitric oxide in proteoglycan turnover by bovine articular cartilage organ cultures. J Immunol 1996; 156:1213-20. 74. Pettipher ER, Higgs GA, Henderson B. Interleukin 1 induces leukocyte infiltration and cartilage proteoglycan degradation in the synovial joint. Proc Natl Acad Sci U S A 1986; 83:8749-53. 75. Saklatvala J, Pilsworth LM, Sarsfield SJ, Gavrilovic J, Heath JK. Pig catabolin is a form of interleukin 1. Cartilage and bone resorb, fibroblasts make prostaglandin and 41 collagenase, and thymocyte proliferation is augmented in response to one protein. Biochem J 1984; 224:461-6. 76. Leizer T, Clarris BJ, Ash PE, van Damme J, Saklatvala J, Hamilton JA. Interleukin-l beta and interleukin-1 alpha stimulate the plasminogen activator activity and prostaglandin E2 levels of human synovial cells. Arthritis Rheum 1987; 30:562-6. 77. Hauselmann HJ, Oppliger L, Michel BA, Stefanovic-Racic M, Evans CH. Nitric oxide and proteoglycan biosynthesis by human articular chondrocytes in alginate culture. FEBS Lett 1994; 352:361-4. 78. Benton HP, Tyler JA. Inhibition of cartilage proteoglycan synthesis by interleukin 1. Biochem Biophys Res Commun 1988; 154:421-8. 79. van de Loo FA, Joosten LA, van Lent PL, Amtz OJ, van den Berg WB. Role of interleukin-1, tumor necrosis factor alpha, and interleukin-6 in cartilage proteoglycan metabolism and destruction. Effect of in situ blocking in murine antigen- and zyrnosan- induced arthritis. Arthritis Rheum 1995; 38: 164-72. 80. Goldring MB, Birkhead J, Sandell LJ, Kimura T, Krane SM. Interleukin 1 suppresses expression of cartilage-specific types 11 and IX collagens and increases types I and HI collagens in human chondrocytes. J Clin Invest 1988; 82:2026-37. 81. MacDonald MH, Stover SM, Willits NH, Benton HP. Regulation of matrix metabolism in equine cartilage explant cultures by interleukin 1. Am J Vet Res 1992; 53:2278-85. 82. Tyler JA, Benton HP. Synthesis of type II collagen is decreased in cartilage cultured with interleukin 1 while the rate of intracellular degradation remains unchanged. Coll Relat Res 1988; 8:393-405. 83. Gouze JN, Bordji K, Gulberti S, Terlain B, Netter P, Magdalou J, et al. Interleukin- 1beta down-regulates the expression of glucuronosyltransferase I, a key enzyme priming glycosaminoglycan biosynthesis: influence of glucosamine on interleukin-1beta-mediated effects in rat chondrocytes. Arthritis Rheum 2001; 44:351-60. 84. Mobasheri A, Vannucci SJ, Bondy CA, Carter SD, Innes JF, Arteaga MF, et al. Glucose transport and metabolism in chondrocytes: a key to understanding chondrogenesis, skeletal development and cartilage degradation in osteoarthritis. Histopathol 2002; 17: 1239-67. 85. Windhaber RA, Wilkins RJ, Meredith D. Functional characterisation of glucose transport in bovine articular chondrocytes. Pflugers Arch 2003; 446:572-7. 42 86. Dingle JT, Page Thomas DP, Hazleman B. The role of cytokines in arthritic diseases: in vitro and in vivo measurements of cartilage degradation. Int J Tissue React 1987; 9:349-54. 87. Stadler J, Stefanovic-Racic M, Billiar TR, Curran RD, McIntyre LA, Georgescu H1, et a1. Articular chondrocytes synthesize nitric oxide in response to cytokines and lipopolysaccharide. J Immunol 1991; 147:3915-20. 88. Campbell 1K, Piccoli DS, Hamilton JA. Stimulation of human chondrocyte prostaglandin E2 production by recombinant human interleukin-1 and tumour necrosis factor. Biochim Biophys Acta 1990; 1051:310-8. 89. Smith RJ, Chin JE, Sam LM, Justen JM. Biologic effects of an interleukin-1 receptor antagonist protein on interleukin-l-stimulated cartilage erosion and chondrocyte responsiveness. Arthritis Rheum 1991; 34:78-83. 90. Goldring MB. The role of cytokines as inflammatory mediators in osteoarthritis: lessons from animal models. Connect Tissue Res 1999; 40: 1-1 1. 91. van den Berg WB. Lessons for joint destruction from animal models. Curr Opin Rheumatol 1997; 9:221-8. 92. van den Berg WB, van de Loo F, Joosten LA, Amtz OJ. Animal models of arthritis in NOSZ-deficient mice. Osteoarthritis Cart 1999; 7:413-5. 93. Saklatvala J. Tumour necrosis factor alpha stimulates resorption and inhibits synthesis of proteoglycan in cartilage. Nature 1986; 322:547-9. 94. Barakat AF, Elson CJ, Westacott C1. Susceptibility to physiological concentrations of IL-lbeta varies in cartilage at different anatomical locations on human osteoarthritic knee joints. Osteoarthritis Cart 2002; 10:264-9. 95. Henrotin YE, De Groote DD, Labasse AH, Gaspar SE, Zheng SX, Geenen VG, et al. Effects of exogenous IL-1 beta, TNF alpha, IL-6, IL-8 and LIF on cytokine production by human articular chondrocytes. Osteoarthritis Cart 1996; 4: 163-73. 96. Nietfeld JJ, Wilbrink B, Helle M, van Roy JL, den Otter W, Swaak AJ, et al. Interleukin- 1 -induced interleukin-6 is required for the inhibition of proteoglycan synthesis by interleukin-1 in human articular cartilage. Arthritis Rheum 1990; 33: 1695- 701. 97. Gueme PA, Carson DA, Lotz M. IL-6 production by human articular chondrocytes. Modulation of its synthesis by cytokines, growth factors, and hormones in vitro. J Immunol 1990; 144:499-505. 43 98. Gueme PA, Zuraw BL, Vaughan JH, Carson DA, Lotz M. Synovium as a source of interleukin 6 in vitro. Contribution to local and systemic manifestations of arthritis. J Clin Invest 1989; 83:585-92. 99. Waage A, Kaufrnann C, Espevik T, Husby G. Interleukin-6 in synovial fluid from patients with arthritis. Clin Immunol Immunopathol 1989; 50:394-8. 100. Tilg H, Trehu E, Atkins MB, Dinarello CA, Mier JW. Interleukin-6 (IL-6) as an anti-inflammatory cytokine: induction of circulating IL-1 receptor antagonist and soluble tumor necrosis factor receptor p55. Blood 1994; 83:1 13-8. 101. Ito A, Itoh Y, Sasaguri Y, Morimatsu M, Mori Y. Effects of interleukin-6 on the metabolism of connective tissue components in rheumatoid synovial fibroblasts. Arthritis Rheum 1992; 35:1197-201. 102. Sato T, Ito A, Mori Y. Interleukin 6 enhances the production of tissue inhibitor of metalloproteinases (TIMP) but not that of matrix metalloproteinases by human fibroblasts. Biochem Biophys Res Commun 1990; 170:824-9. 103. Aderka D, Le JM, Vilcek J. IL-6 inhibits lipopolysaccharide-induced tumor necrosis factor production in cultured human monocytes, U937 cells, and in mice. J Immunol 1989; 143:3517-23. 104. Malfait AM, Verbruggen G, Veys EM, Lambert J, De Ridder L, Comelissen M. Comparative and combined effects of interleukin 6, interleukin 1 beta, and tumor necrosis factor alpha on proteoglycan metabolism of human articular chondrocytes cultured in agarose. J Rheumatol 1994; 21:314-20. 105. van de Loo FA, Kuiper S, van Enckevort FH, Amtz OJ, van den Berg WB. Interleukin-6 reduces cartilage destruction during experimental arthritis. A study in interleukin-6-deficient mice. Am J Pathol 1997; 151:177-91. 106. Kandel RA, Petelycky M, Dinarello CA, Minden M, Pritzker KP, Cruz TF. Comparison of the effect of interleukin 6 and interleukin 1 on collagenase and proteoglycan production by chondrocytes. J Rheumatol 1990; 17:953-7. 107. Peveri P, Walz A, Dewald B, Baggiolini M. A novel neutrophil-activating factor produced by human mononuclear phagocytes. J Exp Med 1988; 167:1547-59. 108. Elford PR, Cooper PH. Induction of neutrophil-mediated cartilage degradation by interleukin-8. Arthritis Rheum 1991; 34:325-32. 109. Van Damme J, Bunning RA, Conings R, Graham R, Russell G, Opdenakker G. Characterization of granulocyte chemotactic activity from human cytokine-stimulated chondrocytes as interleukin 8. Cytokine 1990; 2: 106-1 1. 110. Sunyer T, Rothe L, J iang X, Osdoby P, Collin-Osdoby P. Proinflammatory agents, IL-8 and IL-10, upregulate inducible nitric oxide synthase expression and nitric oxide production in avian osteoclast-like cells. J Cell Biochem 1996; 60:469-83. 1 11. Muller B. Cytokine imbalance in non-immunological chronic disease. Cytokine 2002; 18:334-9. 112. van den Berg WB. The role of cytokines and growth factors in cartilage destruction in osteoarthritis and rheumatoid arthritis. Z Rheumatol 1999; 58:136-41. 113. Nathan C, Xie QW. Nitric oxide synthases: roles, tolls, and controls. Cell 1994; 78:915-8. 114. Ignarro LJ. Endothelium-derived nitric oxide: actions and properties. F ASEB J 1989; 3:31-6. 115. Melchiorri C, Meliconi R, Frizziero L, Silvestri T, Pulsatelli L, Mazzetti I, et al. Enhanced and coordinated in vivo expression of inflammatory cytokines and nitric oxide synthase by chondrocytes from patients with osteoarthritis. Arthritis Rheum 1998; 41 :2165-74. 116. Vuolteenaho K, Moilanen T, Hamalainen M, Moilanen E. Regulation of nitric oxide production in osteoarthritic and rheumatoid cartilage. Role of endogenous IL-l inhibitors. Scand J Rheumatol 2003; 32:19-24. 117. Tamura T, Nakanishi T, Kimura Y, Hattori T, Sasaki K, Norimatsu H, et al. Nitric oxide mediates interleukin-l-induced matrix degradation and basic fibroblast growth factor release in cultured rabbit articular chondrocytes: a possible mechanism of pathological neovascularization in arthritis. Endocrinology 1996; 137:3729-37. 118. Taskiran D, Stefanovic-Racic M, Georgescu H, Evans C. Nitric oxide mediates suppression of cartilage proteoglycan synthesis by interleukin-1. Biochem Biophys Res Commun 1994; 200:142-8. 119. Murrell GA, Jang D, Williams RJ. Nitric oxide activates metalloprotease enzymes in articular cartilage. Biochem Biophys Res Commun 1995; 206: 1 5-21. 120. Abramson SB, Attur M, Amin AR, Clancy R. Nitric oxide and inflammatory mediators in the perpetuation of osteoarthritis. Curr Rheumatol Rep 2001; 3:535-41. 121. Jang D, Murrell GA. Nitric oxide in arthritis. Free Radic Biol Med 1998; 24:1511-9. 122. Evans CH. Nitric oxide: what role does it play in inflammation and tissue destruction? Agents Actions 1995; 47:107-16. 45 123. Jouzeau JY, Cipolletta C, Presle N, Netter P, Terlain B. Modulation of IL-1 effects on cartilage by NO synthase inhibitors: pharmacological studies in rats. Osteoarthritis Cart 1999; 7:382-5. 124. Craven PA, Studer RK, Felder J, Phillips S, DeRubertis FR Nitric oxide inhibition of transforming growth factor-beta and collagen synthesis in mesangial cells. Diabetes 1997; 46:671-81. 125. Studer R, Jaffurs D, Stefanovic-Racic M, Robbins PD, Evans CH. Nitric oxide in osteoarthritis. Osteoarthritis Cart 1999; 7:3 77-9. 126. Hashimoto S, Takahashi K, Amiel D, Coutts RD, Lotz M. Chondrocyte apoptosis and nitric oxide production during experimentally induced osteoarthritis. Arthritis Rheum 1998; 41 :1266-74. 127. Pelletier JP, Jovanovic D, Femandes JC, Manning P, Connor JR, Currie MG, et al. Reduced progression of experimental osteoarthritis in vivo by selective inhibition of inducible nitric oxide synthase. Arthritis Rheum 1998; 41 : 1275-86. 128. Clancy R, Rediske J, Koehne C, Stoyanovsky D, Amin A, Attur M, et al. Activation of stress-activated protein kinase in osteoarthritic cartilage: evidence for nitric oxide dependence. Osteoarthritis Cart 2001; 9:294-9. 129. Mendes AF, Caramona MM, Carvalho AP, Lopes MC. Role of mitogen-activated protein kinases and tyrosine kinases on IL-l-Induced NF-kappaB activation and iNOS expression in bovine articular chondrocytes. Nitric Oxide 2002; 6:3 5-44. 130. Badger AM, Cook MN, Lark MW, Newman-Tarr TM, Swifi BA, Nelson AH, et a1. SB 203580 inhibits p38 mitogen-activated protein kinase, nitric oxide production, and inducible nitric oxide synthase in bovine cartilage-derived chondrocytes. J Immunol 1998; 161:467-73. 131. Needleman P, Isakson PC. The discovery and function of COX-2. J Rheumatol 1997; 24 Suppl 49:6-8. 132. Crofford LJ, Wilder RL, Ristimaki AP, Sano H, Remmers EF, Epps HR, et al. Cyclooxygenase-l and -2 expression in rheumatoid synovial tissues. Effects of interleukin-1 beta, phorbol ester, and corticosteroids. J Clin Invest 1994; 93: 1095-101. 133. Morisset S, Patry C, Lora M, de Brum-Femandes AJ. Regulation of cyclooxygenase-2 expression in bovine chondrocytes in culture by interleukin lalpha, tumor necrosis factor-alpha, glucocorticoids, and l7beta-estradiol. J Rheumatol 1998; 25:1146-53. 134. Smith WL, Garavito RM, DeWitt DL. Prostaglandin endoperoxide H synthases (cyclooxygenases)-l and -2. J Biol Chem 1996; 271:33157-60. 46 135. Kirker-Head CA, Chandna VK, Agarwal RK, Morris EA, Tidwell A, O'Callaghan MW, et al. Concentrations of substance P and prostaglandin E2 in synovial fluid of normal and abnormal joints of horses. Am J Vet Res 2000; 61 :7 14-8. 136. Amin AR, Attur M, Patel RN, Thakker GD, Marshall PJ, Rediske J, et al. Superinduction of cyclooxygenase-2 activity in human osteoarthritis-affected cartilage. Influence of nitric oxide. J Clin Invest 1997; 99: 123 1-7. 137. Trudel G, Desaulniers N, Uhthoff HK, Laneuville 0. Different levels of COX-1 and COX-2 enzymes in synoviocytes and chondrocytes during joint contracture formation. J Rheumatol 2001; 28:2066-74. 138. Geng Y, Blanco FJ, Comelisson M, Lotz M. Regulation of cyclooxygenase-2 expression in normal human articular chondrocytes. J Immunol 1995; 155:796-801. 139. Habib A, Créminon C, Frobert Y, Grassi J, Pradelles P, Maclouf J. Demonstration of an inducible cyclooxygenase in human endothelial cells using antibodies raised against the carboxyl-terminal region of the cyclooxygenase-2. J Biol Chem 1993; 268:23448-54. 140. O'Sullivan MG, Huggins EM, Jr., Meade EA, DeWitt DL, McCall CE. Lipopolysaccharide induces prostaglandin H synthase-2 in alveolar macrophages. Biochem Biophys Res Commun 1992; 187:1123-7. 141. Tung JT, Venta PJ, Eberhart SW, Yuzbasiyan-Gurkan V, Alexander L, Caron JP. Effects of anti-arthritis preparations on gene expression and enzyme activity of cyclooxygenase-2 in cultured equine chondrocytes. Am J Vet Res 2002; 63:1134-9. 142. Masferrer JL, Zweifel BS, Manning PT, Hauser SD, Leahy KM, Smith WG, et al. Selective inhibition of inducible cyclooxygenase 2 in vivo is antiinflammatory and nonulcerogenic. Proc Natl Acad Sci U S A 1994; 91 :3228-32. 143. Seibert K, Zhang Y, Leahy K, Hauser S, Masferrer J, Perkins W, et al. Pharmacological and biochemical demonstration of the role of cyclooxygenase 2 in inflammation and pain. Proc Natl Acad Sci U S A 1994; 91:12013-7. 144. Simon LS, Lanza FL, Lipsky PE, Hubbard RC, Talwalker S, Schwartz BD, et al. Preliminary study of the safety and efficacy of SC-58635, a novel cyclooxygenase 2 inhibitor: efficacy and safety in two placebo-controlled trials in osteoarthritis and rheumatoid arthritis, and studies of gastrointestinal and platelet effects. Arthritis Rheum 1998; 41:1591-602. 145. Anderson GD, Hauser SD, McGarity KL, Bremer ME, Isakson PC, Gregory SA. Selective inhibition of cyclooxygenase (COX)-2 reverses inflammation and expression of COX-2 and interleukin 6 in rat adjuvant arthritis. J Clin Invest 1996; 97:2672-9. 47 146. Stichtenoth DO, Thorén S, Bian H, Peters-Golden M, Jakobsson PJ, Crofford LJ. Microsomal prostaglandin E synthase is regulated by proinflammatory cytokines and glucocorticoids in primary rheumatoid synovial cells. J Immunol 2001; 167:469-74. - 147. Masuko-Hongo K, Berenbaum F, Humbert L, Salvat C, Goldring MB, Thirion S. Up—regulation of microsomal prostaglandin E synthase 1 in osteoarthritic human cartilage: critical roles of the ERK-1/2 and p38 signaling pathways. Arthritis Rheum 2004; 50:2829-38. 148. Kojima F, Naraba H, Miyamoto S, Beppu M, Aoki H, Kawai S. Membrane- associated prostaglandin E synthase-1 is upregulated by proinflammatory cytokines in chondrocytes from patients with osteoarthritis. Arthritis Res Ther 2004; 62R355-65. 149. Murakami M, Naraba H, Tanioka T, Semmyo N, Nakatani Y, Kojima F, et al. Regulation of prostaglandin E2 biosynthesis by inducible membrane-associated prostaglandin E2 synthase that acts in concert with cyclooxygenase-2. J Biol Chem 2000; 275:32783-92. 150. Farley J, Mac Farlane P-H, Kombe A, Sirois J, Laverty S. Interleukin-l beta increases cyclooxygenase-2 and prostaglandin E synthase expression in equine chondrocytes. Proc 13th Amer Col Vet Surgeons 2003; Abstractz7-8. 151. Hardy MM, Seibert K, Manning PT, Currie MG, Woemer BM, Edwards D, et al. Cyclooxygenase 2-dependent prostaglandin E2 modulates cartilage proteoglycan degradation in human osteoarthritis explants. Arthritis Rheum 2002; 46: 1789-803. 152. Takafuji VA, McIlwraith CW, Howard RD. Effects of equine recombinant interleukin-lalpha and interleukin-lbeta on proteoglycan metabolism and prostaglandin E2 synthesis in equine articular cartilage explants. Am J Vet Res 2002; 63:551-8. 153. Thomas B, Thirion S, Humbert L, Tan L, Goldring MB, Bereziat G, et al. Differentiation regulates interleukin-lbeta-indueed cyclo—oxygenase-2 in human articular chondrocytes: role of p38 mitogen-activated protein kinase. Biochem J 2002; 362:367-73. 154. Badger AM, Griswold DE, Kapadia R, Blake S, Swift BA, Hoffman SJ, et al. Disease-modifying activity of SB 242235, a selective inhibitor of p38 mitogen-activated protein kinase, in rat adjuvant-induced arthritis. Arthritis Rheum 2000; 43:175-83. 155. Ridley SH, Sarsfield SJ, Lee JC, Bigg HF, Cawston TE, Taylor DJ, et al. Actions of IL-1 are selectively controlled by p38 mitogen-activated protein kinase: regulation of prostaglandin H synthase-2, metalloproteinases, and IL-6 at different levels. J Immunol 1997; 158:3165-73. 156. Ridley SH, Dean JL, Sarsfield SJ, Brook M, Clark AR, Saklatvala J. A p38 MAP kinase inhibitor regulates stability of interleukin-l-induced cyclooxygenase-2 mRNA. FEBS Lett 1998; 439:75-80. 48 157. Lasa M, Mahtani KR, Finch A, Brewer G, Saklatvala J, Clark AR. Regulation of cyclooxygenase 2 mRNA stability by the mitogen-activated protein kinase p38 signaling cascade. Mol Cell Biol 2000; 20:4265-74. 158. Newton R, Kuitert LM, Bergmann M, Adcock 1M, Barnes PJ. Evidence for involvement of NF-kappaB in the transcriptional control of COX-2 gene expression by IL-lbeta. Biochem Biophys Res Commun 1997; 237:28-32. 159. Crofford LJ, Tan B, McCarthy CJ, Hla T. Involvement of nuclear factor kappa B in the regulation of cyclooxygenase-2 expression by interleukin-1 in rheumatoid synoviocytes. Arthritis Rheum 1997; 40:226—36. 160. Kim SJ, Im DS, Kim SH, Ryu JH, Hwang SG, Seong JK, et al. Beta-catenin regulates expression of cyclooxygenase-2 in articular chondrocytes. Biochem Biophys Res Commun 2002; 296:221-6. 161. Copeland RA, Williams JM, Giannaras J, Numberg S, Covington M, Pinto D, et al. Mechanism of selective inhibition of the inducible isoform of prostaglandin G/H synthase. Proc Natl Acad Sci U S A 1994; 91:11202-6. 162. Crofford LJ. COX-2 in synovial tissues. Osteoarthritis Cart 1999; 7:406-8. 163. Thomas B, Berenbaum F, Humbert L, Bian H, Béréziat G, Crofford L, et al. Critical role of C/EBPdelta and C/EBPbeta factors in the stimulation of the cyclooxygenase-2 gene transcription by interleukin-lbeta in articular chondrocytes. Eur J Biochem 2000; 267:6798-809. 164. Dean DD. Proteinase-mediated cartilage degradation in osteoarthritis. Semin Arthritis Rheum 1991; 20:2-11. 165. Vincenti MP, Brinckerhoff CE. Transcriptional regulation of collagenase (MMP-1, MMP-13) genes in arthritis: integration of complex signaling pathways for the recruitment of gene-specific transcription factors. Arthritis Res 2002; 4: 157-64. 166. Cawston T. Matrix metalloproteinases and TIMPs: properties and implications for the rheumatic diseases. Mol Med Today 1998; 4: 130-7. 167. Arend WP, Dayer JM. Cytokines and cytokine inhibitors or antagonists in rheumatoid arthritis. Arthritis Rheum 1990; 33:305-15. 168. Seed MP, Ismaiel S, Cheung CY, Thomson TA, Gardner CR, Atkins RM, et al. Inhibition of interleukin 1 beta induced rat and human cartilage degradation in vitro by the metalloproteinase inhibitor U27391. Ann Rheum Dis 1993; 52:37-43. 49 169. Steinmeyer J, Daufeldt S, Kalbhen DA. The proteoglycan metabolism, morphology and viability of articular cartilage treated with a synthetic matrix metalloproteinase inhibitor. Res Exp Med 1997; 197:63-79. 170. Cawston TE, Galloway WA, Mercer E, Murphy G, Reynolds JJ. Purification of rabbit bone inhibitor of collagenase. Biochem J 1981; 195:159-65. 171. Blair HC, Dean DD, Howell DS, Teitelbaum SL, Jeffrey JJ. Hypertrophic chondrocytes produce immunoreactive collagenase in vivo. Connect Tissue Res 1989; 23:65-73. 172. Knauper V, Cowell S, Smith B, Lopez-Otin C, O'Shea M, Morris H, et al. The role of the C-terrninal domain of human collagenase-3 (MMP-13) in the activation of procollagenase-3, substrate specificity, and tissue inhibitor of metalloproteinase interaction. J Biol Chem 1997; 272:7608-16. 173. Fosang A], Last K, Knauper V, Murphy G, Neame PJ. Degradation of cartilage aggrecan by collagenase-3 MMP-13). FEBS Lett 1996; 380: 17-20. 174. Lauer-Fields JL, Tuzinski KA, Shimokawa K, Nagase H, Fields GB. Hydrolysis of triple-helical collagen peptide models by matrix metalloproteinases. J Biol Chem 2000; 275: 13282-90. 175. Ahmed S, Rahman A, Hasnain A, Goldberg VM, Haqqi TM. Phenyl N-tert- butylnitrone down-regulates interleukin-1 beta-stimulated matrix metalloproteinase-13 gene expression in human chondrocytes: suppression of c-Jun NH2-terminal kinase, p3 8- mitogen-activated protein kinase and activating protein-l . J Pharmacol Exp Ther 2003; 305 :98 1-8. 176. Stremme S, Duerr S, Bau B, Schmid E, Aigner T. MMP-8 is only a minor gene product of human adult articular chondrocytes of the knee. Clin Exp Rheumatol 2003; 21 :205-9. 177. Kanyama M, Kuboki T, Kojima S, F ujisawa T, Hattori T, Takigawa M, et al. Matrix metalloproteinases and tissue inhibitors of metalloproteinases in synovial fluids of patients with temporomandibular joint osteoarthritis. J Orofac Pain 2000; 14:20-30. 178. Murphy G, Cockett MI, Ward RV, Docherty AJ. Matrix metalloproteinase degradation of elastin, type IV collagen and proteoglycan. A quantitative comparison of the activities of 95 kDa and 72 kDa gelatinases, stromelysins-l and -2 and punctuated metalloproteinase (PUMP). Biochem J 1991; 277:277-9. 179. Irnai K, Ohta S, Matsumoto T, Fujimoto N, Sato H, Seiki M, et al. Expression of membrane-type 1 matrix metalloproteinase and activation of progelatinase A in human osteoarthritic cartilage. Am J Pathol 1997; 151 :245-56. 50 180. Clegg PD, Carter SD. Matrix metalloproteinase-2 and -9 are activated in joint diseases. Equine Vet J 1999; 31:324-30. 181. Takei I, Takagi M, Santavirta S, Ida H, Hamasaki M, Ishii M, et al. Matrix metalloproteinases and tissue inhibitors of metalloproteinases in joint fluid of the patients with loose artificial hip joints. J Biomed Mater Res 1999; 45:175-83. 182. Matrisian LM. Metalloproteinases and their inhibitors in matrix remodeling. Trends Genet 1990; 62121-5. 183. F ini ME, Karnrilowicz MJ, Ruby PL, Beeman AM, Borges KA, Brinckerhoff CE. Cloning of a complementary DNA for rabbit proactivator. A metalloproteinase that activates synovial cell collagenase, shares homology with stromelysin and transin, and is coordinately regulated with collagenase. Arthritis Rheum 1987; 30: 1254—64. 184. Steve J, Huch K, Gunther KP, Scharf HP. Interleukin-lbeta induces different gene expression of stromelysin, aggrecan and tumor-necrosis-factor-stimulated gene 6 in human osteoarthritic chondrocytes in vitro. Pathobiology 2000; 68: 144-9. 185. Stove J, Schoniger R, Huch K, Brenner R, Gunther KP, Puhl W, et al. Effects of dexamethasone on proteoglycan content and gene expression of IL-lbeta-stimulated osteoarthrotic chondrocytes in vitro. Acta Orthop Scand 2002; 73:562-7. 186. Okada Y, Shinmei M, Tanaka O, Naka K, Kimura A, Nakanishi I, et al. Localization of matrix metalloproteinase 3 (stromelysin) in osteoarthritic cartilage and synovium. Lab Invest 1992; 66:680-90. 187. Ohuchi E, Imai K, Fujii Y, Sato H, Seiki M, Okada Y. Membrane type 1 matrix metalloproteinase digests interstitial collagens and other extracellular matrix macromolecules. J Biol Chem 1997; 272:2446-51. 188. Koshy PJ, Lundy CJ, Rowan AD, Porter S, Edwards DR, Hogan A, et al. The modulation of matrix metalloproteinase and ADAM gene expression in human chondrocytes by interleukin-1 and oncostatin M: a time-course study using real-time quantitative reverse transcription-polymerase chain reaction. Arthritis Rheum 2002; 46:961 -7. 189. Pap T, Shigeyama Y, Kuchen S, Femihough JK, Simmen B, Gay RB, et al. Differential expression pattern of membrane-type matrix metalloproteinases in rheumatoid arthritis. Arthritis Rheum 2000; 43:1226-32. 190. Murphy G, Knauper V, Cowell S, Hembry R, Stanton H, Butler G, et al. Evaluation of some newer matrix metalloproteinases. Ann N Y Acad Sci 1999; 878:25-39. 51 191. Holmbeck K, Bianco P, Caterina J, Yamada S, Kromer M, Kuznetsov SA, et al. MTl-MMP-deficient mice develop dwarfism, osteopenia, arthritis, and connective tissue disease due to inadequate collagen turnover. Cell 1999; 99:81-92. 192. Sasaki K, Hattori T, Fujisawa T, Takahashi K, Inoue H, Takigawa M. Nitric oxide mediates interleukin-l-induced gene expression of matrix metalloproteinases and basic fibroblast growth factor in cultured rabbit articular chondrocytes. J Biochem (Tokyo) 1998; 123:431-9. 193. Tortorella MD, Malfait AM, Deccico C, Amer E. The role of ADAM-T84 (aggrecanase-1) and ADAM-TS5 (aggrecanase-2) in a model of cartilage degradation. Osteoarthritis Cart 2001; 9:539-52. 194. Tang BL. ADAMT S: a novel family of extracellular matrix proteases. Int J Biochem Cell Biol 2001; 33:33-44. 195. Tortorella MD, Pratta M, Liu RQ, Austin J, Ross OH, Abbaszade I, et al. Sites of aggrecan cleavage by recombinant human aggrecanase-1 (ADAMTS-4). J Biol Chem 2000; 275:18566-73. 196. Tortorella M, Pratta M, Liu RQ, Abbaszade 1, Ross H, Burn T, et al. The thrombospondin motif of aggrecanase-1 (ADAMTS-4) is critical for aggrecan substrate recognition and cleavage. J Biol Chem 2000; 275:25791-7. 197. Tortorella MD, Burn TC, Pratta MA, Abbaszade I, Hollis JM, Liu R, et al. Purification and cloning of aggrecanase-1: a member of the ADAMTS family of proteins. Science 1999; 284:1664-6. 198. Abbaszade 1, Liu RQ, Yang F, Rosenfeld SA, Ross OH, Link JR, et al. Cloning and characterization of ADAMTSl 1, an aggrecanase fiom the ADAMT S family. J Biol Chem 1999; 274:23443-50. 199. Amer EC. Aggrecanase-mediated cartilage degradation. Curr Opin Pharmacol 2002; 2:322-9. 200. Sandy JD, F lannery CR, Neame PJ, Lohmander LS. The structure of aggrecan fragments in human synovial fluid. Evidence for the involvement in osteoarthritis of a novel proteinase which cleaves the Glu 373 -Ala 374 bond of the interglobular domain. J Clin Invest 1992; 89:1512-6. 201. Lark MW, Gordy J T, Weidner JR, Ayala J, Kimura JH, Williams HR, et al. Cell- mediated catabolism of aggrecan. Evidence that cleavage at the "aggrecanase" site (Glu3 73-Ala3 74) is a primary event in proteolysis of the interglobular domain. J Biol Chem 1995; 270:2550-6. 52 202. Westling J, F osang AJ, Last K, Thompson VP, Tomkinson KN, Hebert T, et al. ADAMTS4 cleaves at the aggrecanase site (Glu373-Ala374) and secondarily at the matrix metalloproteinase site (Asn341-Phe342) in the aggrecan interglobular domain. J Biol Chem 2002; 277: 16059-66. 203. Tsuzaki M, Guyton G, Garrett W, Archambault JM, Herzog W, Almekinders L, et al. IL-1 beta induces COX2, MMP-1, -3 and -13, ADAMTS-4, IL-1 beta and IL-6 in human tendon cells. J Orthop Res 2003; 21 :256-64. 204. Lohmander LS, Neame PJ, Sandy JD. The structure of aggrecan fragments in human synovial fluid. Evidence that aggrecanase mediates cartilage degradation in inflammatory joint disease, joint injury, and osteoarthritis. Arthritis Rheum 1993; 36: 1214-22. 205. Malfait AM, Liu RQ, Ijiri K, Komiya S, Tortorella MD. Inhibition of ADAM-T84 and ADAM-T85 prevents aggrecan degradation in osteoarthritic cartilage. J Biol Chem 2002; 277:22201-8. 206. Little CB, Flannery CR, Hughes CE, Mort JS, Roughley PJ, Dent C, et al. Aggrecanase versus matrix metalloproteinases in the catabolism of the interglobular domain of aggrecan in vitro. Biochem J 1999; 344:61-8. 207. Vincenti MP, Clark TM, Brinckerhoff CE. Using inhibitors of metalloproteinases to treat arthritis. Easier said than done? Arthritis Rheum 1994; 37:1115-26. 208. Cawston T, Billington C, Cleaver C, Elliott S, Hui W, Koshy P, et al. The regulation of MMPs and TIMPs in cartilage turnover. Ann N Y Acad Sci 1999; 878: 120-9. 209. Woessner JF, Jr. Matrix metalloproteinases and their inhibitors in connective tissue remodeling. F ASEB J 1991; 522145-54. 210. Sadowski T, Steinmeyer J. Effects of non-steroidal antiinflammatory drugs and dexamethasone on the activity and expression of matrix metalloproteinase-1, matrix metalloproteinase-3 and tissue inhibitor of metalloproteinases-1 by bovine articular chondrocytes. Osteoarthritis Cart 2001; 9:407-15. 211. Bonassar LJ, Sandy JD, Lark MW, Plaas AH, Frank EH, Grodzinsky AJ. Inhibition of cartilage degradation and changes in physical properties induced by IL-lbeta and retinoic acid using matrix metalloproteinase inhibitors. Arch Biochem Biophys 1997; 344:404-12. 212. Gendron C, Kashiwagi M, Hughes C, Caterson B, Nagase H. TIMP-3 inhibits aggrecanase-mediated glycosaminoglycan release from cartilage explants stimulated by 'catabolic factors. F EBS letters 2003; 555:431-6. 213. Martel-Pelletier J, McCollum R, F uj imoto N, Obata K, Cloutier JM, Pelletier JP. Excess of metalloproteases over tissue inhibitor of metalloprotease may contribute to 53 cartilage degradation in osteoarthritis and rheumatoid arthritis. Lab Invest 1994; 70:807- 15. 214. Pavloff N, Staskus PW, Kishnani NS, Hawkes SP. A new inhibitor of metalloproteinases from chicken: ChIMP-3. A third member of the TIMP family. J Biol Chem 1992; 267:17321-6. 215. Apte SS, Olsen BR, Murphy G. The gene structure of tissue inhibitor of metalloproteinases (TIMP)-3 and its inhibitory activities define the distinct TIMP gene family. J Biol Chem 1996; 271:2874. 216. Kashiwagi M, Tortorella M, Nagase H, Brew K. TIMP-3 is a potent inhibitor of aggrecanase 1 (ADAM-T84) and aggrecanase 2 (ADAM-T85). J Biol Chem 2001; 276: 12501-4. 217. Hashimoto G, Aoki T, Nakamura H, Tanzawa K, Okada Y. Inhibition of ADAMTS4 (aggrecanase-1) by tissue inhibitors of metalloproteinases (TIMP-1, 2, 3 and 4). FEBS Lett 2001; 494:192-5. 218. Singh G. Recent considerations in nonsteroidal anti-inflammatory drug gastropathy. Am J Med 1998; 105:31S-388. 219. Bresalier RS, Sandler RS, Quan H, Bolognese JA, Oxenius B, Horgan K, et al. Cardiovascular events associated with rofecoxib in a colorectal adenoma chemoprevention trial. N Engl J Med 2005; 352: 1092-102. 220. Palrnoski MJ, Brandt KD. Effects of some nonsteroidal antiinflammatory drugs on proteoglycan metabolism and organization in canine articular cartilage. Arthritis Rheum 1980; 23:1010-20. 221. Rashad S, Revell P, Hemingway A, Low F, Rainsford K, Walker F. Effect of non- steroidal anti-inflammatory drugs on the course of osteoarthritis. Lancet 1989; 2:519-22. 222. Setnikar I, Rovati LC. Absorption, distribution, metabolism and excretion of glucosamine sulfate. A review. Arzneimittelforschung 2001; 51:699-725. 223. Hamerman D. The biology of osteoarthritis. N Engl J Med 1989; 320:1322-30. 224. Setnikar l, Giachetti C, Zanolo G. Absorption, distribution and excretion of radioactivity after a single intravenous or oral administration of [14C] glucosamine to the rat. Pharmatherapeutica 1984; 3:538-50. 225. Noyszewski EA, Wroblewski K, Dodge GR, Kudchodkar S, Beers J, Sarina AV, et al. Preferential incorporation of glucosamine into the galactosamine moieties of chondroitin sulfates in articular cartilage explants. Arthritis Rheum 2001; 44: 1089-95. 54 226. Conte A, Palmieri L, Segnini D, Ronca G. Metabolic fate of partially depolymerized chondroitin sulfate administered to the rat. Drugs Exp Clin Res 1991; 17:27-33. 227. Tesoriere G, Dones F, Magistro D, Castagnetta L. Intestinal absorption of glucosamine and N-acetylglucosamine. Experientia 1972; 28:770-1. 228. Setnikar I, Giacchetti C, Zanolo G. Pharmacokinetics of glucosamine in the dog and in man. Arzneimittelforschung 1986; 36:729-35. 229. Liang Z, Leslie J, Adebowale A, Ashraf M, Eddington ND. Determination of the nutraceutical, glucosamine hydrochloride, in raw materials, dosage forms and plasma using pre-column derivatization with ultraviolet HPLC. J Pharm Biomed Anal 1999; 20:807- 14. 230. Laverty. Synovial fluid levels and serum pharmacokinetics in a large animal model following treatment with oral glucosamineat clinically relevant doses. Arthritis Rheum 2005; 52:181-91. 231. Kelly GS. The role of glucosamine sulfate and chondroitin sulfates in the treatment of degenerative joint disease. Altem Med Rev 1998; 3:27-39. 232. Du J, Eddington N. Determination of the chondroitin sulfate disaccharides in dog and horse plasma by HPLC using chondroitinase digestion, precolumn derivatization, and fluorescence detection. Anal Biochem 2002; 306:252-8. 233. Conte A, de Bemardi M, Palmieri L, Lualdi P, Mautone G, Ronca G. Metabolic fate of exogenous chondroitin sulfate in man. Arzneimittelforschung 1991; 41:768-72. 234. Palmieri L, Conte A, Giovannini L, Lualdi P, Ronca G. Metabolic fate of exogenous chondroitin sulfate in the experimental animal. Arzneimittelforschung 1990; 40:319-23. 235. Volpi N. Oral bioavailability of chondroitin sulfate (Condrosulf) and its constituents in healthy male volunteers. Osteoarthritis Cart 2002; 10:768-77. 236. Volpi N. Oral absorption and bioavailability of ichthyic origin chondroitin sulfate in healthy male volunteers. Osteoarthritis Cart 2003; 11:433-41. 237. Conte A, Volpi N, Palmieri L, Bahous I, Ronca G. Biochemical and pharmacokinetic aspects of oral treatment with chondroitin sulfate. Arzneimittelforschung 1995; 45:918-25. 238. Palmieri L, Conte A, Giovannini L, Lualdi P, Ronca G. Metabolic fate of exogenous chondroitin sulfate in the experimental animal. Arzneimittelforschung 1990; 40:319-23. 239. Adebowale A, Du J, Liang Z, Leslie JL, Eddington ND. The bioavailability and pharmacokinetics of glucosamine hydrochloride and low molecular weight chondroitin 55 sulfate after single and multiple doses to beagle dogs. Biopharm Drug Dispos 2002; 23:217-25. 240. Du J, White N, Eddington ND. The bioavailability and pharmacokinetics of glucosamine hydrochloride and chondroitin sulfate after oral and intravenous single dose administration in the horse. Biopharm Drug Dispos 2004; 25:109-16. 241. Nguyen P, Mohamed SE, Gardiner D, Salinas T. A randomized double-blind clinical trial of the effect of chondroitin sulfate and glucosamine hydrochloride on temporomandibular joint disorders: a pilot study. Cranio 2001; 19:130-9. 242. Noack W, Fischer M, Forster KK, Rovati LC, Setnikar I. Glucosamine sulfate in osteoarthritis of the knee. Osteoarthritis Cart 1994; 2251—9. 243. Milller-Fassbender H, Bach GL, Haase W, Rovati LC, Setnikar I. Glucosamine sulfate compared to ibuprofen in osteoarthritis of the knee. Osteoarthritis Cart 1994; 2261-9. 244. Ruane R, Griffiths P. Glucosamine therapy compared to ibuprofen for joint pain. Br J Community Nurs 2002; 7: 148-52. 245. Reginster JY, Deroisy R, Rovati LC, Lee RL, Lejeune E, Bruyere O, et al. Long- terrn effects of glucosamine sulphate on osteoarthritis progression: a randomised, placebo-controlled clinical trial. Lancet 2001; 357:251-6. 246. Ronca F, Palmieri L, Panicucci P, Ronca G. Anti-inflammatory activity of chondroitin sulfate. Osteoarthritis Cart 1998; 6 Suppl A: 14-21. 247. Bourgeois P, Chales G, Dehais J, Delcambre B, Kuntz JL, Rozenberg S. Efficacy and tolerability of chondroitin sulfate 1200 mg/day vs chondroitin sulfate 3 x 400 mg/day vs placebo. Osteoarthritis Cart 1998; 6 Suppl A:25-30. 248. Bucsi L, Poor G. Efficacy and tolerability of oral chondroitin sulfate as a symptomatic slow-acting drug for osteoarthritis (SYSADOA) in the treatment of knee osteoarthritis. Osteoarthritis Cart 1998; 6 Suppl A:31-6. 249. Setnikar I, Palumbo R, Canali S, Zanolo G. Pharmacokinetics of glucosamine in man. Arzneimittelforschung 1993; 43:1 109-13. 250. Setnikar I, Pacini MA, Revel L. Antiarthritic effects of glucosamine sulfate studied in animal models. Arzneimittelforschung 1991; 41 :542-5. 251. Pujalte JM, Llavore EP, Ylescupidez FR. Double-blind clinical evaluation of oral glucosamine sulphate in the basic treatment of osteoarthrosis. Curr Med Res Opin 1980; 7: 1 10-14. 56 252. Tapadinhas MJ, Rivera 1C, Bignamini AA. Oral glucosamine sulphate in the management of arthrosis: report on a multi-centre open investigation in Portugal. Pharmatherapeutica 1982; 3: 157-68. 253. Lopes Vaz A. Double-blind clinical evaluation of the relative efficacy of ibuprofen and glucosamine sulphate in the management of osteoarthrosis of the knee in out- patients. Curr Med Res Opin 1982; 8:145-9. 254. Rovati LC. Clinical research in osteoarthritis: design and results of short-term and long-term trials with disease-modifying drugs. Int J Tissue React 1992; 14:243-51. 255. Jordan KM, Arden NK, Doherty M, Bannwarth B, Bijlsma JW, Dieppe P, et al. EULAR Recommendations 2003: an evidence based approach to the management of knee osteoarthritis: Report of a Task F orce of the Standing Committee for International Clinical Studies Including Therapeutic Trials (ESCISIT). Ann Rheum Dis 2003; 62:1145-55. 256. Patti ME, Virkamaki A, Landaker EJ, Kahn CR, Yki-Jarvinen H. Activation of the hexosamine pathway by glucosamine in vivo induces insulin resistance of early postreceptor insulin signaling events in skeletal muscle. Diabetes 1999; 48: 1562-71. 257. Monauni T, Zenti MG, Cretti A, Daniels MC, Targher G, Caruso B, et al. Effects of glucosamine infusion on insulin secretion and insulin action in humans. Diabetes 2000; 49:926-35. 258. Pouwels MJ, Jacobs JR, Span PN, Lutterman J A, Smits P, Tack CJ. Short-terrn glucosamine infusion does not affect insulin sensitivity in humans. J Clin Endocrinol Metab 2001; 86:2099-103. 259. Scroggie DA, Albright A, Harris MD. The effect of glucosamine-chondroitin supplementation on glycosylated hemoglobin levels in patients with type 2 diabetes mellitus: a placebo-controlled, double-blinded, randomized clinical trial. Arch Intern Med 2003; 163:1587-90. 260. Lee DY, Choi IS, Han JH, Yoo HS. Chitosan and D-glucosamine induce expression of Th1 cytokine genes in porcine spleen cells. J Vet Med Sci 2002; 64:645-8. 261. Matheu V, Gracia Bara MT, Pelta R, Vivas E, Rubio M. Immediate-hypersensitivity reaction to glucosamine sulfate. Allergy 1999; 54:643. 262. Gray HC, Hutcheson PS, Slavin RG. Is glucosamine safe in patients with seafood allergy? J Allergy Clin Immun012004; 114:459-60. 263. Pavelka K, Gatterova J, Olej arova M, Machacek S, Giacovelli G, Rovati LC. Glucosamine sulfate use and delay of progression of knee osteoarthritis: a 3-year, 57 randomized, placebo-controlled, double-blind study. Arch Intern Med 2002; 16222113- 23. 264. Soldani G, Romagnoli J. Experimental and clinical pharmacology of glycosaminoglycans (GAGs). Drugs Exp Clin Res 1991; 17281-5. 265. McNamara PS, Barr SC, Erb HN. Hematologic, hemostatic, and biochemical effects in dogs receiving an oral chondroprotective agent for thirty days. Am J Vet Res 1996; 57:1390-4. 266. Drovanti A, Bignamini AA, Rovati AL. Therapeutic activity of oral glucosamine sulfate in osteoarthrosis: a placebo-controlled double-blind investigation. Clin Ther 1980; 32260-72. 267. Qiu GX, Gao SN, Giacovelli G, Rovati L, Setnikar 1. Efficacy and safety of glucosamine sulfate versus ibuprofen in patients with knee osteoarthritis. Arzneimittelforschung 1998; 482469-74. 268. Priebe D, McDiarmid T, Mackler L, Tudiver F. Do glucosamine or chondroitin cause regeneration of cartilage in osteoarthritis? J F am Pract 2003; 52:237-9. 269. Rovati LC, Annefeld M, Giacovelli G, Schmid K, Setnikar I. Glucosamine in osteoarthritis. Lancet 1999; 35421640; 41-2. 270. Delafuente JC. Glucosamine in the treatment of osteoarthritis. Rheum Dis Clin North Am 2000; 2621-11, vii. 271. Debi R, Robinson D, Agar G, Halperin N. [GAG for osteoarthritis of the knee--a prospective study]. Harefuah 2000; 1382451-3, 518. 272. Bruyere O, Honore A, Ethgen O, Rovati LC, Giacovelli G, Henrotin YE, et al. Correlation between radiographic severity of knee osteoarthritis and future disease progression. Results from a 3-year prospective, placebo-controlled study evaluating the effect of glucosamine sulfate. Osteoarthritis Cart 2003; 1121-5. 273. Braham R, Dawson B, Goodman C. The effect of glucosamine supplementation on people experiencing regular knee pain. Br J Sports Med 2003; 37245-9; discussion 49. 274. Richy F, Bruyere O, Ethgen O, Cucherat M, Henrotin Y, Reginster JY. Structural and symptomatic efficacy of glucosamine and chondroitin in knee osteoarthritis: a comprehensive meta-analysis. Arch Intern Med 2003; 16321514-22. 275. McAlindon TE, LaValley MP, Gulin JP, Felson DT. Glucosamine and chondroitin for treatment of osteoarthritis: a systematic quality assessment and meta-analysis. JAMA 2000; 283:1469-75. 58 276. Morreale P, Manopulo R, Galati M, Boccanera L, Saponati G, Bocchi L. Comparison of the antiinflammatory efficacy of chondroitin sulfate and diclofenac sodium in patients with knee osteoarthritis. J Rheumatol 1996; 23:1385-91. 277. Uebelhart D, Thonar EJ, Delmas PD, Chantraine A, Vignon E. Effects of oral chondroitin sulfate on the progression of knee osteoarthritis: a pilot study. Osteoarthritis Cart 1998; 6 Suppl A:39-46. 278. Verbruggen G, Goemaere S, Veys EM. Chondroitin sulfate: S/DMOAD (structure/disease modifying anti-osteoarthritis drug) in the treatment of finger joint OA. Osteoarthritis Cart 1998; 6 Suppl A:37-8. 279. Rovetta G, Monteforte P, Molfetta G, Balestra V. Chondroitin sulfate in erosive osteoarthritis of the hands. Int J Tissue React 2002; 24:29-32. 280. Conrozier T. Les chondroitines sulfates (CS 4&6): schema d'utilisation et impact économique. Presse Med 1998; 27:1866-8. 281. Alekseeva LI, Mednikov BL, Piiavskii SA, Nasonova VA, Soldatov DG. F armakoékonomicheskie aspekty primeneniia struktuma pri osteoartroze. Ter Arkh 2001; 73290-2. 282. Doma V, Guerrero R. Effects of oral and intramuscular use of chondroitin sulfate in induced equine aseptic arthritis. J Equine Vet Science 1998; 182548-55. 283. Leeb BF, Schweitzer H, Montag K, Smolen J S. A metaanalysis of chondroitin sulfate in the treatment of osteoarthritis. J Rheumatol 2000; 272205-11. 284. Das A, Jr., Hammad TA. Efficacy of a combination of FCHG49 glucosamine hydrochloride, TRH122 low molecular weight sodium chondroitin sulfate and manganese ascorbate in the management of knee osteoarthritis. Osteoarthritis Cart 2000; 82343-50. 285. Leffler CT, Philippi AF, Leffler SG, Mosure JC, Kim PD. Glucosamine, chondroitin, and manganese ascorbate for degenerative joint disease of the knee or low back: a randomized, double-blind, placebo-controlled pilot study. Mil Med 1999; 164285- 91. 286. Anderson MA, Slater MR, Hammad TA. Results of a survey of small-animal practitioners on the perceived clinical efficacy and safety of an oral nutraceutical. Prev Vet Med 1999; 38:65-73. 287. Canapp SO, Jr., McLaughlin RM, Jr., Hoskinson JJ, Roush JK, Butine MD. Scintigraphic evaluation of dogs with acute synovitis after treatment with glucosamine hydrochloride and chondroitin sulfate. Am J Vet Res 1999; 6021552-7. 59 288. Hanson R, Smalley L, Huff G. Oral treatment with a glucosamine-chondroitin sulfate compound for degenerative joint disease in horses: 25 cases. Equine Practice 1997; 19:16-22. 289. Hanson R, Brawner W, Blaik M. Oral treatment with a nutraceutical (Cosequin®) for ameliorating signs of navicular syndrome in horses. Vet Therap 2001; 22148-59. 290. Richards J, Rodan 1. Panel report on feline senior health care, Part II. Compend Cont Educ Pract Vet. 1999; 212612-21. 291. Lippiello L, Woodward J, Karpman R, Hammad TA. In vivo chondroprotection and metabolic synergy of glucosamine and chondroitin sulfate. Clin Orthop 2000; 381:229- 40. 292. Gaby AR. Natural treatments for osteoarthritis. Altem Med Rev 1999; 42330-41. 293. Bassleer C, Henrotin Y, Franchimont P. In-vitro evaluation of drugs proposed as chondroprotective agents. Int J Tissue React 1992; 142231-41. 294. Johnson KA, Hulse DA, Hart RC, Kochevar D, Chu Q. Effects of an orally administered mixture of chondroitin sulfate, glucosamine hydrochloride and manganese ascorbate on synovial fluid chondroitin sulfate 3B3 and 7D4 epitope in a canine cruciate ligament transection model of osteoarthritis. Osteoarthritis Cart 2001; 9214-21. 295. McCarty MF, Russell AL, Seed MP. Sulfated glycosaminoglycans and glucosamine may synergize in promoting synovial hyaluronic acid synthesis. Med Hypotheses 2000; 54:798-802. 296. Orth MW, Peters TL, Hawkins JN. Inhibition of articular cartilage degradation by glucosamine-BC] and chondroitin sulphate. Equine Vet J Suppl 2002; 34:224-9. 297. Lippiello L. Glucosamine and chondroitin sulfate: biological response modifiers of chondrocytes under simulated conditions of joint stress. Osteoarthritis Cart 2003; 11:335- 42. 298. Beren J, Hill SL, Diener-West M, Rose NR. Effect of pre-loading oral glucosamine HCl/chondroitin sulfate/manganese ascorbate combination on experimental arthritis in rats. Exp Biol Med 2001; 2262144-51. 299. Bassleer C, Rovati L, F ranchimont P. Stimulation of proteoglycan production by glucosamine sulfate in chondrocytes isolated from human osteoarthritic articular cartilage in vitro. Osteoarthritis Cart 1998; 62427-34. 300. Dodge GR, Jimenez SA. Glucosamine sulfate modulates the levels of aggrecan and matrix metalloproteinase-3 synthesized by cultured human osteoarthritis articular chondrocytes. Osteoarthritis Cart 2003; 112424-32. 60 301. McCarty MF. Enhanced synovial production of hyaluronic acid may explain rapid clinical response to high-dose glucosamine in osteoarthritis. Med Hypotheses 1998; 502507-10. 302. Mroz PJ, Silbert J E. Use of 3H-glucosamine and 3SS-sulfate with cultured human chondrocytes to determine the effect of glucosamine concentration on formation of chondroitin sulfate. Arthritis Rheum 2004; 50:3574-9. 303. Gouze JN, Gouze E, Palmer GD, Kaneto H, Ghivizzani SC, Grodzinsky AJ, et al. Adenovirus-mediated gene transfer of glutamine: fructose-6-phosphate amidotransferase antagonizes the effects of interleukin-lbeta on rat chondrocytes. Osteoarthritis Cart 2004; 122217-24. 304. Ilic MZ, Martinac B, Handley CJ. Effects of long-term exposure to glucosamine and mannosamine on aggrecan degradation in articular cartilage. Osteoarthritis Cart 2003; 112613-22. 305. F enton JI, Chlebek-Brown KA, Peters TL, Caron JP, Orth MW. Glucosamine HCl reduces equine articular cartilage degradation in explant culture. Osteoarthritis Cart 2000; 82258-65. 306. Fenton J I, Chlebek-Brown KA, Caron JP, Orth MW. Effect of glucosamine on interleukin-l-conditioned articular cartilage. Equine Vet J Suppl 2002; 342219-23. 307. Pipemo M, Reboul P, Hellio Le Graverand MP, Peschard MJ, Annefeld M, Richard M, et al. Glucosamine sulfate modulates dysregulated activities of human osteoarthritic chondrocytes in vitro. Osteoarthritis Cart 2000; 82207-12. 308. Sandy JD, Gamett D, Thompson V, Verscharen C. Chondrocyte-mediated catabolism of aggrecan: aggrecanase-dependent cleavage induced by interleukin-1 or retinoic acid can be inhibited by glucosamine. Biochem J 1998; 335259-66. 309. Byron CR, Orth MW, Venta PJ, Lloyd J W, Caron JP. Influence of glucosamine on matrix metalloproteinase expression and activity in lipopolysaccharide-stimulated equine chondrocytes. Am J Vet Res 2003; 642666-71. 310. Gouze JN, Bianchi A, Becuwe P, Dauca M, Netter P, Magdalou J, et al. Glucosamine modulates IL-l-induced activation of rat chondrocytes at a receptor level, and by inhibiting the NF-kappa B pathway. FEBS Lett 2002; 5102166-70. 311. Shikhman AR, Kuhn K, Alaaeddine N, Lotz M. N-acetylglucosamine prevents IL-1 beta-mediated activation of human chondrocytes. J Immunol 2001; 166:5155-60. 312. Fenton JI, Chlebek-Brown KA, Peters TL, Caron JP, Orth MW. The effects of glucosamine derivatives on equine articular cartilage degradation in explant culture. Osteoarthritis Cart 2000; 82444-51. 61 313. Schlueter AE, Orth MW. Further studies on the ability of glucosamine and chondroitin sulphate to regulate catabolic mediators in vitro. Equine Vet J 2004; 36:634- 6. 314. Mello DM, Nielsen BD, Peters TL, Caron JP, Orth MW. Comparison of inhibitory effects of glucosamine and mannosamine on bovine articular cartilage degradation in vitro. Am J Vet Res 2004; 65:1440-5. 315. Meininger CJ, Kelly KA, Li H, Haynes TE, Wu G. Glucosamine inhibits inducible nitric oxide synthesis. Biochem Biophys Res Commun 2000; 279:234-9. 316. Largo R, Alvarez-Soria MA, Diez-Ortego I, Calvo E, Sénchez-Pemaute O, Egido J, et a1. Glucosamine inhibits IL-lbeta-induced NFkappaB activation in human osteoarthritic chondrocytes. Osteoarthritis Cart 2003; 11:290-8. 317. Nakamura H, Shibakawa A, Tanaka M, Kato T, Nishioka K. Effects of glucosamine hydrochloride on the production of prostaglandin E2, nitric oxide and metalloproteases by chondrocytes and synoviocytes in osteoarthritis. Clin Exp Rheumatol 2004; 222293-9. 318. Hua J, Sakamoto K, Nagaoka I. Inhibitory actions of glucosamine, a therapeutic agent for osteoarthritis, on the functions of neutrophils. J Leukoc Biol 2002; 712632-40. 319. Vacha J, Pesakova V, Krajickova J, Adam M. Effect of glycosaminoglycan polysulphate on the metabolism of cartilage ribonucleic acid. Arzneimittelforschung 1984; 34:607-9. 320. Lilja S, Barrach HJ. Normally sulphated and highly sulphated glycosaminoglycans (GAG) affecting fibrillogenesis of type I and type II collagen in vitro. Exp Pathol 1983; 232173-81. 321. Bassleer CT, Combal JP, Bougaret S, Malaise M. Effects of chondroitin sulfate and interleukin-1 beta on human articular chondrocytes cultivated in clusters. Osteoarthritis Cart 1998; 62196-204. 322. Nerucci F, Fioravanti A, Cicero MR, Collodel G, Marcolongo R. Effects of chondroitin sulfate and interleukin- 1 beta on human chondrocyte cultures exposed to pressurization: a biochemical and morphological study. Osteoarthritis Cart 2000; 8:279- 87. 323. Pipitone VR. Chondroprotection with chondroitin sulfate. Drugs Exp Clin Res 1991; 1723-7. 324. Omata T, Segawa Y, Itokazu Y, Inoue N, Tanaka Y. Effects of chondroitin sulfate-C on bradykinin-induced proteoglycan depletion in rats. Arzneimittelforschung 1999; 49:577-8 1 . 62 325. Baici A, Bradamante P. Interaction between human leukocyte elastase and chondroitin sulfate. Chem Biol Interact 1984; 5121-11. 326. Sugimoto K, Takahashi M, Yamamoto Y, Shimada K, Tanzawa K. Identification of aggrecanase activity in medium of cartilage culture. J Biochem (Tokyo) 1999; 1262449- 55. 327. Campo GM, Avenoso A, Campo S, Ferlazzo A, Altavilla D, Micali C, et al. Aromatic trap analysis of free radicals production in experimental collagen-induced arthritis in the rat: protective effect of glycosaminoglycans treatment. Free Radic Res 2003; 372257-68. 328. Campo GM, Avenoso A, Campo S, Ferlazzo AM, Altavilla D, Calatroni A. Efficacy of treatment with glycosaminoglycans on experimental collagen-induced arthritis in rats. Arthritis Res Ther 2003; 52R122-31. 63 CHAPTER 2 Chan PS, Caron JP, Rosa GJM, Orth MW. Glucosamine and chondroitin sulfate regulate gene expression and synthesis of nitric oxide and prostaglandin E2 in articular cartilage explants. Osteoarthritis Cart 2005; 132387-94. 64 Summary Objective: Glucosamine (GLN) and chondroitin sulfate (CS) are widely used to alleviate symptoms of osteoarthritis (0A). However, the mechanism(s) of action of these nutraceuticals remains unresolved. In the present study, we determined the effect of physiologically relevant concentrations of GLN and CS on gene expression and synthesis of nitric oxide (NO) and prostaglandin E2 (PGE2) in cytokine-stimulated articular cartilage explants. Methods: Using bovine articular cartilage explants in culture stimulated with IL-1, the effects of physiologically relevant concentrations of GLN and CS on gene expression of inducible nitric oxide synthase (iNOS), endothelial nitric oxide synthase (eNOS), cyclooxygenase-2 (COX-2) and microsomal prostaglandin E synthase 1 (mPGEsl) were assessed with quantitative real-time polymerase chain reaction (Q-RT-PCR). The production of NO and PGE2 was also quantified. Results: Chondroitin sulfate and the GLN and CS combination at concentrations attainable in the blood down-regulated IL-l induced mRNA expression of iNOS at 24 and 48 hours post-culture. Up-regulated iNOS expression at 24 hours by IL-1 was also suppressed by GLN. Glucosamine and CS transiently repressed the cytokine-stirnulated mPGEsl transcript. Synthesis of NO was reduced with CS alone and the combination after 24 hours of culture. Repression of COX-2 transcripts by GLN and CS was accompanied by concomitant reduction in PGE2 release. Conclusion: Our results indicate that physiologically relevant concentrations of GLN and CS can regulate gene expression and synthesis of NO and PGE2, providing a plausible explanation for their purported anti-inflammatory properties. 65 Keywords: osteoarthritis, interleukin-l, nitric oxide, prostaglandin, glucosamine, chondroitin sulfate Running title: Glucosamine and chondroitin sulfate regulate inflammatory mediators Introduction Osteoarthritis is characterized by the degeneration of matrix components of articular cartilage accompanied with an excess production of proinflammatory cytokines.1 Interleukin-1 beta is widely accepted as one of the proinflammatory cytokines that plays a pivotal role in the pathophysiology of 0A.2 It induces a cascade of catabolic events in chondrocytes including the upregulation in genes of matrix metalloproteinases (MMPs), iNOS, COX-2 and mPGEsl and the release of NO and PGE2.“5 Interleukin-1 also retards anabolic activities of the chondrocytes leading to declines in proteoglycan synthesis and collagen synthesis.7’ 8 Nitric oxide is produced from L-arginine catalyzed by a family of NO synthases.9' H The constitutive isoforms are eNOS and neuronal nitric oxide synthase (nNOS). An elevated level of iNOS is responsible for the secretion of NO, an inorganic free radical, in large amount from CA cartilage. Nitric oxide promotes inflammation and articular cartilage degeneration by increasing cytokine production, suppressing matrix synthesis, elevating MMPs and inactivating tissue inhibitor of metalloproteinases (TIMPs).'2'l4 Prostaglandin E2 mediates inflammatory and pain responses, and is the most abundant prostanoid found in the diseased joint.15 Cyclooxgenases and prostaglandin E synthases are rate-limiting enzymes responsible for generating PGE2. The inducible form of this enzyme is COX-2, which catalyzes the conversion of arachidonic acid to 66 prostaglandin H2 (PGH2) and mPGEsl, which isomerizes PGH2 to PGE2. The synthesis of PGE2 correlates with up-regulation of COX-2 and mPGEsl .6’ '6 Prostaglandin E2 elicits its catabolic effect on articular cartilage by enhancing MMP production and proteoglycan degradation and by inhibiting synthesis of TIMPs.'7‘ ‘8 Various compounds have been used in palliative treatment of OA. Two compounds that are increasing in favor as a treatment option for joint diseases both in humans and animals are glucosamine and chondroitin sulfate. These compounds are termed nutraceuticals because they have both nutrient and pharmaceutical properties. They have been used over the past 30 years for the treatment of OA and shown in several animal and human studies to be beneficial in reducing articular cartilage degeneration, pain, and inflammation. In animals, the combination reduces lameness in dogs and horses and is effective in managing pain in cats. ”'21 Glucosamine and CS used alone or in combination can decrease joint space narrowing, alleviate pain, and improve joint mobility in humans affected with knee OA.”'25 Elucidating the mechanism(s) of action of GLN and CS in vivo is an active area of research. Pain amelioration with GLN and CS may be attributed to a reduction in inflammatory mediators. In vitro, cytokine-stressed cartilage explants treated with GLN demonstrated a decline in iNOS transcript and NO release into the media.”30 Parallel with the inhibition of COX-2, PGE2 production and release was also inhibited with GLN.26' 28’ 31 Chondroitin sulfate possesses anti-inflammatory properties.32 To date, the majority of in vitro research has been performed with concentrations of GLN and CS that exceed those likely to be obtained by oral administration.26'28’ 3" 33‘” This was one of the reasons given by rheumatologists in their objection to recommending 67 GLN to patients.36 The concentrations of GLN in serum after oral and intravenous administration range from 1 to 20 ug/ml while CS concentrations are in the 5 to 200 ug/ml range depending on the route of administration, species and the molecular weight of CS.3742 Thus, the aim of the present study was to determine the effect of concentrations of GLN (5 rig/ml) and CS (20 ug/ml) that have been attained in blood on mRNA expression of iNOS, eNOS, COX-2 and mPGEsl and the production of NO and PGE2 in articular cartilage explants stimulated with IL-1, a cytokine associated with cartilage catabolism. Materials and methods EXPLANT CULTURES Articular cartilage was isolated from the carpal joints of Holstein steers (18-24 months old) obtained from a local abattoir within 3 hours of slaughter. Cartilage discs (6 mm) were biopsied from the articular surface and did not include cartilage with gross characteristics of OA and calcified cartilage. Two explant discs (approximately 60 mg total wet weight) were randomly picked and cultured in each well of a 24-well Falcon culture plate (Fisher Scientific, Pittsburgh, PA, U.S.A.) containing 1 ml of 1:1 modified version of Dulbecco’s modified Eagle’s medium: nutrient mixture F-12 (Ham) (Gibco, Grand Island, NY, U.S.A.), as previously described.27 The media was supplemented with 20 amino acids (Sigma, St. Louis, MO, U.S.A.), 50 rig/ml ascorbic acid, and 100 units/ml penicillin/streptomycin (Gibco). The concentrations of amino acids added were 50% of those previously reported.43 Cartilage explants were housed in a humidified incubator at 37°C with 7% CO2. 68 Explants were maintained in media without serum for 2 equilibration days prior to the addition of treatments. After equilibration, all treatments received 10% fetal bovine serum (F BS, Gibco) and 50 ng/ml of human recombinant interleukin-1 beta (rhIL-l B, R & D Systems, Minneapolis, MN, U.S.A.) added to induce cartilage catabolism. The concentrations of glucosamine HCl (5 rig/ml; F CHG49®, Nutramax Laboratories, Edgewood, MD, U.S.A.) and low molecular weight (16.9 kDa) CS (20 rig/ml; TRH122®, Nutramax Laboratories) chosen were well within the range of levels measured in the blood”‘ 41. There were 5 treatments per experiment (Table I), an F BS control (Ctrl), 50 ng/ml rhIL-lB (IL-1), 50 ng/ml rhIL-l B with the addition of 5 jig/ml GLN (IL-1 + GLN), 50 ng/ml rhIL-l B with the addition of 20 ug/ml CS (IL-1 + CS) and 50 ng/ml rhIL-l B with the addition of both GLN and CS (IL-l + GLN + CS). Each treatment consisted of 24 discs (12 wells) collected from a pool of 3 steers for an experiment. Cartilage discs were collected 6, 24 and 48 hours post-stimulation, frozen in liquid nitrogen and stored at -80°C until RNA isolation. Conditioned media were collected at 24 and 48 hours and stored at 4°C for nitrite and PGE2 analysis. Experiment was repeated a total of 3 times for each time point, each time using tissue from a pool of 3 different animals. NITRIC OXIDE ASSAY Nitrite is a stable end product of NO metabolism and was measured in conditioned media using the Griess reagent and sodium nitrite as standard.44 Briefly, 150 pl medium was incubated with 150 pl of 1.0% sulfanilamide, 0.1% N-l- napthylethylenediamide hydrochloride and 25% phosphoric acid at room temperature for 5 minutes. Due to some precipitation of reagents with CS, 96-well plates were spun at 3250 rpm for 3 minutes at 4°C. The remaining supernatant was transferred to a new plate. 69 Absorbance was measured at 540 nm using a Spectromax 300 plate reader (Molecular Devices, Sunnyvale, CA, U.S.A.). PROSTAGLANDIN E2 ASSAY Prostaglandin E2 release into conditioned media was quantified using a commercially available competitive enzyme linked immunosorbent assay kit (R & D Systems). Indomethacin (10 ug/ml) was added to conditioned media samples collected at 24 and 48 hours post-stimulation to inhibit any prostaglandin synthase present, and stored at -20°C until analysis. Samples were run according to the manufacturer’s instructions. Absorbance was determined at 405 nm with a wavelength correction set at 590 nm. TOTAL RNA ISOLATION The extraction of RNA from explants was performed following a modified protocol.45 Briefly, cartilage was homogenized in TRIzol® reagent (Invitrogen, Carlsbad, CA, U.S.A.), chloroform was added to extract total RNA followed by vigorous agitation and a 2 minute incubation. The aqueous phase containing RNA was collected after centrifugation and the RNA was precipitated with ethanol. Total RNA was then purified further with RNeasy mini columns (Qiagen, Valencia, CA, U.S.A.) and quantified by UV spetrophotometry (Beckman Coulter, Fullerton, CA, U.S.A.). Total chondrocyte RNA was resolved on 1.2% agarose gel to validate spectrophotometric determinations and RNA integrity. cDNA SYNTHESIS For each sample, 2 pg of RNA was treated with DNase I (Invitrogen) to degrade contaminating single and double standed DNA. Treated RNA was converted to single stranded cDNA using Superscript II reverse transcriptase (Invitrogen) as recommended 70 by the manufacturer. Single stranded cDNA was quantified with UV spectrophotometry (Beckman Coulter) and diluted to 50 ng/ul. QUANTITATIVE REAL-TIME POLYMERASE CHAIN REACTION (Q-RT-PCR) Primers for glyceraldehyde phosphate dehydrogenase (GAPDH, used as a housekeeping gene) and molecules from Table II were designed using the Primer Express software version 2.0 (Applied Biosystems, Foster City, CA, U.S.A.). Optimal concentrations of each set of primers were determined with a Primer Matrix (lowest standard deviation with no change in cycle to threshold (Ct)). Quantitative real-time PCR was performed with 50 ng cDNA templates in 96-well plates using the ABI PRISM 7000 sequence detection system (Applied Biosystems). The analysis of each sample was performed in duplicate. The cDNA templates were combined with optimal concentrations of primers and SYBR Green PCR dye mix (Applied Biosystems) in a total volume of 50 ul and the amplification conducted as recommended by manufacturer. The PCR conditions were 2 min at 50°C and 10 min at 95°C followed by 40 cycles of extension at 95°C for 15 sec and 1 min at 60°C, and data collected during the last 30 sec. Threshold lines were adjusted to intersect amplification lines in the linear portion of the amplification curves. The software automatically recorded the Ct. The GAPDH gene was used as an endogenous control and run together with the samples for each amplification reaction to allow for normalization of different samples for RNA loading, cDNA synthesis, and amplification efficiencies and for comparison of samples run at different times. The FBS control was used as a calibrator (i.e. the fold change for control is 1.0). Replicated data was normalized with GAPDH and the fold change in gene 71 expression relative to FBS control treatment was calculated using the delta delta Ct method.46 STATISTICAL ANALYSIS Data for NO and PGE2 release into conditioned media were analyzed using a linear mixed effects model, including the fixed effect of treatment and the random effects of pool, the interaction between treatment and pool, and repeated measures within each treatment and pool combination. Treatment effects were compared within each time point using the multiple comparisons approach of Tukey. The computations were performed using the MIXED procedure of SAS.47 Relative gene expression data determined using Q-RT-PCR was analyzed considering the nonparametric ANOVA approach of Friedman.48 Differences were declared statistically significant when P<0.05, unless otherwise noted. Spearman’s rank correlations (r) between gene expression data and biochemical data were computed using the CORR procedure of SAS.47 Results THE EFFECT OF GLN AND CS ON GENE EXPRESSION Bovine articular cartilage explants cultured with 50 ng/ml rhIL-lB for 6, 24 and 48 hours resulted in significant (P<0.01) up-regulation of iNOS expression relative to control (Table H1). The large standard error for both IL-1 and IL-1 plus CS treatments at 6 hours was attributed to variation between pools of animal tissue and not technical error within a replication. Glucosanrine (5 ug/ml) down-regulated iNOS mRNA expression from 11.1 fold to 4.5 fold in explants cultured for 24 hours (Table III). Chondroitin sulfate (20 ug/ml) added alone or in combination with GLN for 24 hours was as effective 72 as GLN in suppressing iNOS transcripts (Table III) but only the combination reduced iNOS expression to control levels. After 48 hours, cytokine stimulated explants treated with CS or GLN and CS repressed iNOS mRNA expression to control levels (Table III). The iNOS transcript of explants treated with GLN alone at the 48 hour time point was not different from IL-l stimulated explants (Table IH). Cartilage explants stimulated with 50 ng/ml rhIL-l B demonstrated significant elevation in COX-2 mRNA expression relative to control by 30 fold for 6 hours post- stimulation (Table 111). Like iNOS mRNA expression at 6 hours, variability of COX-2 level of transcript at this time point was due to differences between replications. At the 6 hour time point, GLN or CS used individually and used in combination resulted in a decline of COX-2 transcripts to control levels (Table III). The COX-2 gene for explants treated with 50 ng/ml rhIL-l B was significantly up-regulated by 8 fold and 13 fold for 24 and 48 hours post-stimulation respectively (Table 111). At both time points, GLN suppressed the activation of the COX-2 gene to control levels (Table III). Physiologically relevant concentration of CS at 20 rig/ml did not repress COX-2 transcript increased by IL-1 until 48 hours (Table III). The GLN and CS combination decreased COX-2 expression to 3.4 and 1.0 fold change for 24 and 48 hours respectively (Table HI), which is not different from control. The transcript for mPGEsl, the terminal enzyme involved in PGE2 synthesis was elevated by about 9 fold (P=0.0008) relative to control, in explants cultured with 50 ng/ml rhIL-l B over a 6 hour period (Table HI). At this time point, GLN alone and the combination nutraceuticals repressed mPGEsl mRNA expression to control levels (Table 73 111). No effect on mPGEsl expression was found for any of the treatments at 24 and 48 horns (Table III). THE EFFECT OF GLN AND CS ON NO AND PGE2 SYNTHESIS Nitric oxide released into conditioned media at 24 hours after the addition of treatment was increased with 50 ng/ml rhIL-lB to 21.39 uM fiom 5.98 uM for FBS control (Figure 1, Panel A). The addition of CS or GLN plus CS suppressed NO release to 16.93 and 17.27 uM respectively (Figure 1, Panel A). At 48 hours, the elevation in NO release with 50 ng/ml rhIL—l B was not reduced by either nutraceutical (Figure 1, Panel B). Explants stimulated with 50 ng/ml rhIL- 1 B released significantly more PGE2 into media when compared to control. After 24 hours stimulation, PGE2 release was elevated to 1986 pg/ml compared with basal PGE2 release at 169 pg/ml (Figure 2, Panel A). The addition of GLN and CS, or in combination reduced PGE2 release induced by cytokine to control levels (Figure 2, Panel A). The basal PGE2 release into media 48 hours post- treatment was 171 pg/ml (Figure 2, Panel B). This level was elevated to 1122 pg/ml when 50 ng/ml rhIL-lB was added (Figure 2, Panel B). Glucosamine decreased PGE2 release due to IL-1 by 68% (Figure 2, Panel B). Likewise, the combination resulted in a 76% decline in PGE2 release induced by IL-1 (Figure 2, Panel B). Chondroitin sulfate addition to the explants in vitro tended (P<0.10) to decrease PGE2 release (Figure 2, Panel B). 74 THE EFFECT OF GLN AND CS ON ENDOTHELIAL NO SYNTHASE GENE EXPRESSION Since the biochemical data on NO release conflicted with the Q-RT-PCR data on iNOS gene expression profile, we decided to investigate the expression of the eNOS gene, another isoform of the NO synthase enzyme. Only a transient pattern of eNOS regulation by CS was witnessed. The IL-l mediated increase in eNOS transcript of 3.3 fold at 24 hours was significantly (P<0.05) reduced to 0.3 fold with 20 rig/ml CS. No significant differences were seen with the other treatments (data not shown). Discussion To our knowledge, the present study is the first to describe the effect of GLN and CS attainable in blood on gene expression and synthesis of inflammatory mediators using a cartilage explant culture. The concentration chosen for GLN was 5 rig/ml while that for CS was 20 ug/ml. These concentrations were at the low end of the range found in blood after oral or intravenous administration.” 4" 42 Previous trials fiom our laboratory used concentrations which are higher than the range found in the plasma of animals administered GLN and CS.49 Another novelty of this research lies in the nutraceutical combination investigated. Few in vitro studies have been conducted with the GLN and CS combination although they are frequently marketed as a combination. One study reported that they are synergistic,50 while another suggested they may be complementary.49 Nitric oxide is implicated in the pathogenesis of arthritis. Normal cartilage explants produce little NO.51 On the contrary, chondrocytes and the synovial membrane 75 in OA and rheumatoid arthritis (RA) patients produce NO abundantly and so does cytokine-induced cartilage.5 "53 In fact, activated articular chondrocytes produce more NO than any other cells, including synoviocytes, hepatocytes and macrophages.3 Inducible forms of NO synthase are found in cartilage and produce large amounts of NO in response to IL-1.13 Both iNOS mRNA and protein concentrations are higher in OA cartilage relative to healthy articular cartilage and increase with severity of the degenerative joint disease.“ The ability of IL-1 to enhance iNOS transcript was also evident in the present study (Table III).3 Nitric oxide suppresses proteoglycan synthesis by inhibiting the process of sulfation on the glycosaminoglycan chains.”’ 55 Inhibition of prolyl hydroxylase coupled with the suppression of type II collagen synthesis was associated with the release of large amounts of NO.56 This inorganic free radical also mediates the degradation of matrix components via activation of MMPs.” Besides cartilage destruction, NO has also been implicated as a mediator of inflammatory responses.‘2 Thus, the inhibition of NO synthesis may be effective as a therapeutic route for DA. Glucosamine and CS have been used successfirlly for treating chronic inflammatory arthritides. One of the mechanisms of action of these compounds may be the suppression of NO production. Previous work fi'om our laboratory using explants from horses indicates decreased NO production with 250 ug/ml CS and with the combination of 500 ug/ml GLN and 250 jig/ml cs.49 Inhibition ofNO with GLN occurred at concentrations greater than 1mg/ml in equine explants and human chondrocytes.27’ 30’ 49 Suppression of NO synthesis with GLN was associated with iNOS mRNA and protein repression.29’ 30 In the current trial, CS alone and in combination with 76 I!” GLN at physiological concentrations were effective in reducing NO synthesis induced by IL-1 at 24 hours (Figure 1, Panel A). At 48 hours of culture, nitrite has accumulated to the same level for all treatments except for control (Figure 1, Panel B). These levels of NO in conditioned media may be explained by transcriptional regulation of genes pertaining to the enzymes responsible for synthesizing NO. Although the levels of NO were significantly reduced at 24 hours, these were marginal declines. Since NO and PGE2 can modulate the activity of the other”, even transient regulation of NO may affect the anti-inflammatory response to some extent. The inducible form of the NO synthases is iNOS and there are two constitutive isoforms, eNOS and nNOS. The constitutive form of eNOS was initially thought to be confined to endothelial cells while nN OS was localized to the central and autonomic nervous systems.9 However, eNOS and nN OS have increasingly been identified in a lot of different cell types including chondrocytes.58 Endothelial NO synthase may also be inducible since human eNOS gene contains activator protein-l (AP-l) site in its promoter region.59 The mitogen activated protein kinase (MAPK) pathway is activated by cytokines leading to AP-l phosphorylation and up-regulation of a number of genes associated with cartilage catabolism.60 Human OA chondrocytes express elevated levels of nNOS.61 To date, only the genes for iNOS and eNOS and not nNOS in the bovine species have been cloned and sequenced. Since eNOS mRNA levels were not markedly affected at 48 hours, the lack of treatment effect on NO production could be in part due to the activity of eNOS and potentially nN OS. Prostaglandin E2, another catabolic mediator in the pathogenesis of 0A is formed from a series of enzymatic reactions termed the arachidonic acid cascade. It mediates 77 synoviocyte proliferation and is responsible for pain and inflammatory responses. Prostaglandin E2 enhances the degradative processes of proteoglycans.” ‘8 It is found in diseased joints and is significantly elevated in synovial fluid of OA patients.” 18 Large amounts of PGE2 are synthesized by chondrocytes that are exposed to IL-1 in vitro.18 Bovine explant cultures induced with IL-1 demonstrated significant increase in PGE2 from basal levels (Figure 2, Panel A & B). Elevated levels of PGE2 depleted matrix components from intact articular cartilage cultured in vitro.62 The induction of COX-2 is largely responsible for elevated concentrations of PGE2.31’63 Cyclooxygenase-2 expression is elevated in cartilage specimens of OA and RA patients.“ 65 Findings from the present study demonstrated the significant up- regulation of the COX-2 gene relative to control in response to IL-1 for all time points (Table III) in accord with previous reports.3 1’ 66 Levels of PGE2 in our explant cultures were positively correlated with COX-2 mRNA (r = 0.81, P<0.0001) in agreement with Geng et. (11.66 Inhibiting PGE2 release is an increasingly common strategy for providing relief to patients affected with arthritis as evidenced by the development of COX-2 inhibitors. ' In clinical studies, GLN and CS have been consistently beneficial in improving the pain scores and inflammatory responses of patients affected with CA.”25 In vitro results from previous work in our laboratory and others demonstrated declines in the inflammatory mediator, PGE2, with GLN and CS supplementation.26’ 31’49’67 In the present study, GLN, CS alone or in cambination reduced IL-l induced PGE2 levels. The ability of GLN and CS to reduce PGE2 synthesis in the present study can be explained by their effect on COX-2 gene expression. Since there is a lag time of about 12 to 18 hours 78 from the time the gene is expressed to the time it is translated into protein, the COX-2 mRNA data at 6 hours does correspond to the PGE2 levels at 24 hours while mRNA data at 24 hours has a similar profile with PGE2 levels at 48 hours. The ability of GLN to suppress COX-2 mRNA expression by about 2 fold at 24 hours post-stimulation parallels the work by Largo et. al. in human chondrocytes.31 However, the concentration of GLN used in their study was 1 mg/ml which is 200 times the concentration used in our study. The present study substantiates the claim that repression of COX-2 transcripts and eventually PGE2 is at least one of the mechanisms GLN alone or together with CS exert their analgesic effects. The effect of these chondroprotective agents on terminal PGE enzymes involved in the formation of PGE2 has not been reported. In fact, we only know of one abstract that has investigated this enzyme in chondrocytes.68 There are reports of concomitant regulation of COX-2, mPGEsl and PGE2.'6’ 68 Transcripts of COX-2 and mPGEsl are correlated in the present study (r = 0.43, P<0.05). However, differences in the timing for COX-2 and mPGEsl induction may be due to dissimilar regulatory sequences in these two genes.6 Explants treated either with GLN alone or in combination with CS repressed IL-l induced mPGEsl expression at 6 hours post-culture (Table IH). Further studies on the contribution of this brief regulation on mPGEsl in chondrocytes by the nutraceuticals are warranted. Determining the mechanism of action behind GLN and CS is essential to elucidating the beneficial effects elicited by these compounds in combating arthritic symptoms. Understanding how these agents may regulate NO and PGE2 is an active area of research. Besides being pro-inflammatory, NO and PGE2 have the ability to increase 79 proteoglycan loss and decrease proteoglycan synthesis.”’ '8 Our results suggest a transient modulation of mPGEsl and NO synthase gene expression, and NO synthesis by the nutraceuticals. Physiologically relevant concentrations of GLN and CS repress COX- 2 expression that eventually translates into accompanying declines in PGE2 synthesis. Thus, the findings in the present study may explain at least in part how IL-l induced proteoglycan breakdown and decreased proteoglycan synthesis is prevented in vitroég’ 70. Results fiom the present study indicate that GLN and CS may regulate the expression and synthesis of NO and PGE2, providing a plausible explanation for the symptomatic relief attributed to the administration of these compounds. Acknowledgements The authors are grateful to Angela E. Schlueter for technical assistance and Bellingar’s Packing in Ashley, M1 for the provision of Holstein steers. This work was supported by the Michigan Agricultural Experiment Station and Nutramax Laboratories, Inc. 80 Table I Description of components found in treatment media of explant cultures Treatment FBS (%) rhIL-l B (IL-1) Glucosamine Chondroitin (GLbD sulfate (CS) Ctrl 10 - - - IL-1 10 50 ng/ml - - IL-l + GLN 10 50 ng/ml 5 jig/ml - IL-1 + CS 10 50 ng/ml - 20 rig/ml IL-l + GLN + CS 10 50 11ng 5 Hg/ml 20 ug/ml 81 A2: ooov EUEUUGéiUOOtOOO Oz: cog UH0m 3030:5058 208:: Heston ”8200.000: 0:00 0:50.50 :03000: 50:0 00808239083200: 03:82:56 :8 00m: amalhv 2082.2.» 0020.6: 0:0 0850a = 030,—. 82 Table III Relative gene expression of explants stimulated with rhIL-l B and physiologically relevant concentrations of glucosamine and chondroitin sulfate Mean fold change (:1: SE) in gene expression relative to control 6 hours post-stimulation Treatment Gene IL-1 IL-1 + GLN IL-l + cs IL-l + GLN + cs iNOS 87.7 i 73.6 a 4.1 :l: 1.0 a 26.7 a: 24.9 a 4.1 3: 2.8 ‘ cox-2 30.0 :1: 13.3 b 1.5 :1: 0.9 a 2.7 :l: 2.5 a 3.3 a: 3.1 ‘ mPGEsl 8.7 i 2.8 b 4.5 i 1.0 a 5.4 a: 1.5 b 4.9 i 1.5 a Mean fold change (0 SE) in gene expression relative to control 24 hours post-stimulation iNOS 11.1131“ 4.53:1.9a 4.400.6‘ 3.03:0.8a cox-2 7.7 :h 0.6 b 3.2 a; 2.0 a 7.1 :t 0.6 b 3.4 i 0.6 “ mPGEsl 0.7 :t 0.1 a 0.4 at 0.1 a 1.0 :t 0.3 a 0.6 3. 0.2 a Mean fold change (d: SE) in gene expression relative to control 48 hours post-stimulation iNOS 9.4027b 3.101.4" 2301.38 1.20103 cox-2 12.6 :t 4.0 b 3.4 :t 0.7 a 2.8 :l: 1.0 a 1.0 :l: 0.8 " mPGEsl 4.8 i 2.0 a 1.6 :h 0.8 a 1.5 :l: 0.8 ° 0.8 i 0.3 ' Different superscripts for values within a row (i.e. one gene) denote significant differences (P<0.05) between treatments. 83 Figure 1. Mean (iSE) nitrite release into conditioned media 24 hours (Panel A) and 48 hours (Panel B) afier treatments are added. Ctrl= fetal bovine serum control; IL-1= 50 ng/ml rhIL-l [3 (human recombinant interleukin-1 beta); IL-l + GLN= 50 ng/ml rhIL-l [3 and 5 ug/ml glucosamine; IL-l + CS= 50 ng/ml rhIL—lB and 20 ug/ml chondroitin sulfate; IL-l + GLN + CS= 50 ng/ml rhIL-l B, 5 ug/ml glucosamine and 20 pg/ml chondroitin sulfate. Different superscripts indicate significant differences at P<0.05. 84 25 l Panel A c 20- 10q Nitrite (uM) IL-l+GLN m1+cs m1+cm+cs Treatment 18 - Panel B 15- 12- Nitrite (uM) 34 o4 . cm 1 I IL-l+GLN IL-l+CS lIrl+GLN+CS Treatment 85 Figure 2. Mean (iSE) prostaglandin E2 (PGE2) release into conditioned media 24 hours (Panel A) and 48 hours (Panel B) afier the addition of treatments. Ctrl= fetal bovine serum control; IL-1= 50 ng/ml rhIL-l B (human recombinant interleukin-1 beta); IL-l + GLN= 50 ng/ml rhIL-l B and 5 pg/ml glucosamine; IL-l + CS= 50 ng/ml rhIL-l B and 20 ug/ml chondroitin sulfate; IL-l + GLN + CS= 50 ng/ml rhIL-l B, 5 itng glucosamine and 20 ug/ml chondroitin sulfate. Different superscripts indicate significant differences at P<0.05. * Tended to be different than the IL-1 treatment (P<0.10). 86 PGE2 (pg/ml) PGE2 (pg/ml) 87 3500 1 Panel A 3000 4 '3 l 2500 4 2000 - 1500 0 1000 1 a a 500 4 a I a T l 1 I 0 U j I Ctrl 11,1 IL—l + GLN 1L-1 + cs IL—l + GLN + cs Treatment 1800 - Panel B b 1500 a 1200 a 900 4 bit 600 1 a i 1 l a a 300 4 l T J 1 l 0 1 a . Ctrl IL-1 IL-1 + GLN IL-l + cs [L] + GLN + cs Treatment References 1. 10. ll. Pelletier JP, DiBattista JA, Roughley P, McCollum R, Martel-Pelletier J. Cytokines and inflammation in cartilage degradation Rheum Dis Clin North Am 1993;19:545-68. Dinarello CA. Interleukin-l. Ann N Y Acad Sci 1988;546:122-32. Stadler J, Stefanovic-Racic M, Billiar TR, Curran RD, McIntyre LA, Georgescu HI, et al. Articular chondrocytes synthesize nitric oxide in response to cytokines and lipopolysaccharide. J Immunol 1991;147:3915-20. Vincenti MP, Coon CI, Lee 0, Brinckerhoff CE. Regulation of collagenase gene expression by IL-1 beta requires transcriptional and post-transcriptional mechanisms. Nucleic Acids Res 1994;22:4818-27. Morisset S, Patry C, Lora M, de Brum-Fernandes AJ. Regulation of cyclooxygenase-2 expression in bovine chondrocytes in culture by interleukin lalpha, tumor necrosis factor-alpha, glucocorticoids, and ”beta-estradiol J Rheumatol 1998;25:1 146-53. Stichtenoth DO, Thorén S, Bian H, Peters-Golden M, J akobsson PJ, Crofford LJ, et al. Microsomal prostaglandin E synthase is regulated by proinflammatory cytokines and glucocorticoids in primary rheumatoid synovial cells. J Immunol 2001;167:469-74. Benton HP, Tyler J A. Inhibition of cartilage proteoglycan synthesis by interleukin 1. Biochem Biophys Res Commun 1988;154:421-8. Tyler JA, Benton HP. Synthesis of type II collagen is decreased in cartilage cultured with interleukin 1 while the rate of intracellular degradation remains unchanged Coll Relat Res 1988;8:393-405. Bredt DS, Hwang PM, Glatt CE, Lowenstein C, Reed RR, Snyder SH. Cloned and expressed nitric oxide synthase structurally resembles cytochrome P-450 reductase. Nature 1991;351:714-8. Marsden PA, Schappert KT, Chen HS, Flowers M, Sundell CL, Wilcox JN, et al. Molecular cloning and characterization of human endothelial nitric oxide synthase. FEBS Lett 1992;307:287-93. Xie QW, Cho HJ, Calaycay J, Mumford RA, Swiderek KM, Lee TD, et al. Cloning and characterization of inducible nitric oxide synthase from mouse macrophages Science 1992;256:225-8. 88 12. l3. 14. 15. 16. 17. 18. 19. 20. 21. 22. Evans CH. Nitric oxide: what role does it play in inflammation and tissue destruction? Agents Actions Suppl 1995;47:107-16. Murrell GA, Jang D, Williams RJ. Nitric oxide activates metalloprotease enzymes in articular cartilage. Biochem Biophys Res Commun 1995;206:15-21. Taskiran D, Stefanovic-Racic M, Georgescu H, Evans C. Nitric oxide mediates suppression of cartilage proteoglycan synthesis by interleukin-1. Biochem Biophys Res Commun 1994;200:142-8. Kirker-Head CA, Chandna VK, Agarwal RK, Morris EA, Tidwell A, O'Callaghan MW, et al. Concentrations of substance P and prostaglandin E2 in synovial fluid of normal and abnormal joints of horses Am J Vet Res 2000;61:714-8. Murakami M, Naraba H, Tanioka T, Semmyo N, Nakatani Y, Kojima F, et al. Regulation of prostaglandin E2 biosynthesis by inducible membrane-associated prostaglandin E2 synthase that acts in concert with cyclooxygenase-2. J Biol Chem 2000;275:32783-92. Yamada H, Kikuchi T, Nemoto O, Obata K, Sato H, Seiki M, et al. Effects of indomethacin on the production of matrix metalloproteinase-3 and tissue inhibitor of metalloproteinases-1 by human articular chondrocytes J Rheumatol 1996;23:1739-43. Hardy MM, Seibert K, Manning PT, Currie MG, Woemer BM, Edwards D, et al. Cyclooxygenase 2-dependent prostaglandin E2 modulates cartilage proteoglycan degradation in human osteoarthritis explants Arthritis Rheum 2002;46: 1789-803. Anderson MA, Slater MR, Hammad TA. Results of a survey of small-animal practitioners on the perceived clinical efficacy and safety of an oral nutraceutical. Prev Vet Med 1999;38:65-73. Hanson R, Brawner W, Blaik M. Oral treatment with a nutraceutical (Cosequin®) for ameliorating signs of navicular syndrome in horses. Veterinary Therapeutics 2001;2:148-159. Richards J, Rodan I. Panel report on feline senior health care, Part II. The Compendium on continuing education for the practicing veterinarian. 1999;21:612-621. Leffler CT, Philippi AF, Lefiler SG, Mosure JC, Kim PD. Glucosamine, chondroitin, and manganese ascorbate for degenerative joint disease of the knee or low back: a randomized, double-blind, placebo-controlled pilot study. Mil Med 1999;164:85-91. 89 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. Das A Jr, Hammad TA. Efficacy of a combination of F CHG49 glucosamine hydrochloride, TRH122 low molecular weight sodium chondroitin sulfate and manganese ascorbate in the management of knee osteoarthritis. Osteoarthritis Cart 2000;8z343-50. Houpt JB, McMillan R, Wein C, Paget-Dellio SD. Effect of glucosamine hydrochloride in the treatment of pain of osteoarthritis of the knee. J Rheumatol 1999;26:2423-30. Uebeth D, Thonar EJ, Delmas PD, Chantraine A, Vignon E. Effects of oral chondroitin sulfate on the progression of knee osteoarthritis: a pilot study. Osteoarthritis Cart l998;6 Suppl A:39-46. F enton J I, Chlebek-Brown KA, Caron JP, Orth MW. Effect of glucosamine on interleukin-l-conditioned articular cartilage. Equine Vet J Suppl 2002;34:219-23. Fenton JI, Chlebek-Brown KA, Peters TL, Caron JP, Orth MW. Glucosamine HCl reduces equine articular cartilage degradation in explant culture. Osteoarthritis Cart 2000;8z258-65. Gouze JN, Bordji K, Gulberti S, Terlain B, Netter P, Magdalou J, et al. Interleukin-lbeta down-regulates the expression of glucuronosyltransferase I, a key enzyme priming glycosaminoglycan biosynthesis: influence of glucosamine on interleukin-lbeta-mediated effects in rat chondrocytes Arthritis Rheum 2001 ;44:351—60. Meininger CJ, Kelly KA, Li H, Haynes TE, Wu G. Glucosamine inhibits inducible nitric oxide synthesis. Biochem Biophys Res Commun 2000;279:234-9. Shikhman AR, Kuhn K, Alaaeddine N, Lotz M. N-acetylglucosamine prevents IL-l beta-mediated activation of human chondrocytes. J Immunol 2001;166:5155- 60. Largo R, Alvarez-Soria MA, Diez-Ortego I, Calvo E, Sénchez-Pemaute O, Egido J, et al. Glucosamine inhibits IL-lbeta-induced NFkappaB activation in human osteoarthritic chondrocytes Osteoarthritis Cart 2003;11:290-8. Campo GM, Avenoso A, Campo S, F erlazzo A, Altavilla D, Micali C, et al. Aromatic trap analysis of fi'ee radicals production in experimental collagen- induced arthritis in the rat: protective effect of glycosaminoglycans treatment Free Radic Res 2003;37:257-68. F enton JI, Chlebek-Brown KA, Peters TL, Caron JP, Orth MW. The effects of glucosamine derivatives on equine articular cartilage degradation in explant culture. Osteoarthritis Cart 2000;8z444-51. 90 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 45. Gouze JN, Bianchi A, Becuwe P, Dauca M, Netter P, Magdalou J, et al. Glucosamine modulates IL-l -induced activation of rat chondrocytes at a receptor level, and by inhibiting the NF-kappa B pathway. F EBS Lett 2002;510:166-70. Hua J, Sakamoto K, Nagaoka I. Inhibitory actions of glucosamine, a therapeutic agent for osteoarthritis, on the functions of neutrophils. J Leukoc Biol 2002;71:632-40. Manson JJ, Rahman A. This house believes that we should advise our patients with osteoarthritis of the knee to take glucosamine. Rheum Oxford 2004;43:100- 1. Adebowale A, Du J, Liang Z, Leslie J L, Eddington ND. The bioavailability and pharmacokinetics of glucosamine hydrochloride and low molecular weight chondroitin sulfate after single and multiple doses to beagle dogs Biopharm Drug Dispos 2002;23:217-25. Du J, Eddington N. Determination of the chondroitin sulfate disaccharides in dog and horse plasma by HPLC using chondroitinase digestion, precolumn derivatization, and fluorescence detection Anal Biochem 2002;306:252-8. Setnikar I, Rovati LC. Absorption, distribution, metabolism and excretion of glucosamine sulfate. A review. Arzneimittelforschung 2001;51:699-725. Setnikar I, Palumbo R, Canali S, Zanolo G. Pharmacokinetics of glucosamine in man Arzneimittelforschung 1993 ;43 : 1 109—13. Du J, White N, Eddington ND. The bioavailability and pharmacokinetics of glucosamine hydrochloride and chondroitin sulfate after oral and intravenous single dose administration in the horse. Biopharm Drug Dispos 2004;25:109-16. Volpi N. Oral bioavailability of chondroitin sulfate (Condrosulf) and its constituents in healthy male volunteers Osteoarthritis Cart 2002;10:768-77. Rosselot G, Reginato AM, Leach RM. Development of a serum-free system to study the effect of growth hormone and insulinlike growth factor-I on cultured postembryonic grth plate chondrocytes. In Vitro Cell Dev Biol 1992;28Az235- 44. Blanco FJ, Ochs RL, Schwarz H, Lotz M. Chondrocyte apoptosis induced by nitric oxide. Am J Pathol 1995;146:75-85. Reno C, Marchuk L, Sciore P, Frank CB, Hart DA. Rapid isolation of total RNA from small samples of hypocellular, dense connective tissues Biotechniques 1997;22:1082-6. 91 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real- time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001;25:402-8. SAS. SAS/STAT User's Guide Version 8.2. Cary, NC: SAS Institute, Inc. 2001. Zar JH. Biostatistical Analysis. 3rd Ed ed: Prentice-Hall, Upper Saddle River, New Jersey 1996. Orth MW, Peters TL, Hawkins JN. Inhibition of articular cartilage degradation by glucosamine-HG] and chondroitin sulphate. Equine Vet J Suppl 2002;34:224-9. Lippiello L, Woodward J, Karpman R, Hammad TA. In vivo chondroprotection and metabolic synergy of glucosamine and chondroitin sulfate. Clin Orthop 2000;381:229—40. Pelletier JP, Mineau F, Ranger P, Tardif G, Martel-Pelletier J. The increased synthesis of inducible nitric oxide inhibits IL-lra synthesis by human articular chondrocytes: possible role in osteoarthritic cartilage degradation Osteoarthritis Cart 1996;4277-84. Vuolteenaho K, Moilanen T, Hamalainen M, Moilanen E. Regulation of nitric oxide production in osteoarthritic and rheumatoid cartilage. Role of endogenous IL-l inhibitors. Scand J Rheumatol 2003;32:19-24. Abramson SB, Attur M, Amin AR, Clancy R. Nitric oxide and inflammatory mediators in the perpetuation of osteoarthritis Curr Rheumatol Rep 2001;3z535- 41. Melchiorri C, Meliconi R, Frizziero L, Silvestri T, Pulsatelli L, Mazzetti I, et al. Enhanced and coordinated in vivo expression of inflammatory cytokines and nitric oxide synthase by chondrocytes from patients with osteoarthritis Arthritis Rheum 1998;41:2165-74. Tamura T, Nakanishi T, Kimura Y, Hattori T, Sasaki K, Norimatsu H, et al. Nitric oxide mediates interleukin-l-induced matrix degradation and basic fibroblast growth factor release in cultured rabbit articular chondrocytes: a possible mechanism of pathological neovascularization in arthritis Endocrinology 1996;137:3729-37. Cao M, Westerhausen-Larson A, Niyibizi C, Kavalkovich K, Georgescu HI, Rizzo CF, et al. Nitric oxide inhibits the synthesis of type-II collagen without altering Col2Al mRNA abundance: prolyl hydroxylase as a possible target Biochem J 1997;324:305-10. 92 57. 58. 59. 60. 61. 62. 63. 65. 66. 67. 68. Salvemini D. Regulation of cyclooxygenase enzymes by nitric oxide. Cell Mol Life Sci 1997;53:576-82. Evans CH, Stefanovic-Racic M, Lancaster J. Nitric oxide and its role in orthopaedic disease. Clin Orthop 1995;312:275-94. Marsden PA, Heng HH, Scherer SW, Stewart RJ, Hall AV, Shi XM, et al. Structure and chromosomal localization of the human constitutive endothelial nitric oxide synthase gene. J Biol Chem 1993;268:17478-88. Geng Y, Valbracht J, Lotz M. Selective activation of the mitogen-activated protein kinase subgroups c-Jun NH2 terminal kinase and p38 by IL-1 and TNF in human articular chondrocytes. J Clin Invest 1996;98:2425-30. Amin AR, Attur M, Vyas P, Leszczynska-Piziak J, Levartovsky D, Rediske J, et al. Expression of nitric oxide synthase in human peripheral blood mononuclear cells and neutrophils. J Inflamm 1995;47:190-205. F ulkerson JP, Ladenbauer-Bellis IM, Chrisman OD. In vitro hexosamine depletion of intact articular cartilage by E-prostaglandins: prevention by chloroquine. Arthritis Rheum 1979;22:1117-21. Tung JT, Venta PJ, Eberhart SW, Yuzbasiyan-Gurkan V, Alexander L, Caron JP. Effects of anti-arthritis preparations on gene expression and enzyme activity of cyclooxygenase-2 in cultured equine chondrocytes Am J Vet Res 2002;63zl 134-9. Crofford LJ, Wilder RL, Ristimiiki AP, et al. Cyclooxygenase-l and -2 expression in rheumatoid synovial tissues. Effects of interleukin-1 beta, phorbol ester, and corticosteroids. J Clin Invest 1994;93:1095-101. Amin AR, Attur M, Patel RN, Thakker GD, Marshall PJ, Rediske J, et al. Superinduction of cyclooxygenase-2 activity in human osteoarthritis-affected cartilage. Influence of nitric oxide. J Clin Invest 1997;99:1231-7. Geng Y, Blanco F J , Comelisson M, Lotz M. Regulation of cyclooxygenase-2 expression in normal human articular chondrocytes J Immunol 1995;155:796- 80]. Nakamura H, Shibakawa A, Tanaka M, Kato T, Nishioka K. Effects of glucosamine hydrochloride on the production of prostaglandin E2, nitric oxide and metalloproteases by chondrocytes and synoviocytes in osteoarthritis Clin Exp Rheumatol 2004;22:293-9. Farley J, Mac F arlane PH, Kombe A, Sirois J, Laverty S. Interleukin-1 beta increases cyclooxygenase-2 and prostaglandin E synthase expression in equine 93 69. 70. chondrocytes Proceedings of the 13th American College Veterinary Surgeons Symposium, 2003;Abstract:7-8. Lippiello L. Glucosamine and chondroitin sulfate: biological response modifiers of chondrocytes under simulated conditions of joint stress. Osteoarthritis Cart 2003;11:335-42. Bassleer C, Rovati L, Franchimont P. Stimulation of proteoglycan production by glucosamine sulfate in chondrocytes isolated from human osteoarthritic articular cartilage in vitro. Osteoarthritis Cart l998;6:427-34. 94 CHAPTER 3 Chan PS, Caron JP, Orth MW. Glucosamine and chondroitin sulfate regulate gene expression of proteolytic enzymes and their inhibitors in interleukin-l challenged bovine articular cartilage explants. Am J Vet Res 2005; in press. 95 Abstract Objective-To determine the effects of glucosamine (GLN) and chondroitin sulfate (CS), at concentrations attainable in vivo, on expression of genes encoding proteolytic enzymes, enzyme inhibitors and macromolecules of articular cartilage in interleukin-1 (IL-1) challenged bovine cartilage explants. Sample Population-Bovine articular cartilage explants. Procedures-Explants were cultured in a commercial medium for 48 hours. Cartilage was then exposed to media containing 10% fetal bovine serum (F BS), 50 ng/ml IL-1, and 5 ug/ml GLN, 20 ug/ml CS alone or in combination for 24 and 48 hours. Cartilage was frozen and RNA was extracted from the frozen cartilage. Gene expression of matrix metalloproteinase (MMP)-2, 3, 9, 13 and 14; aggrecanase (Agg)-l and 2; tissue inhibitor of metalloproteinase (TIMP)-1, 2 and 3; type II collagen (Col II) and aggrecan relative to F BS control were assessed with quantitative real-time polymerase chain reaction. Results-Up-regulated MMP-3, MMP-13 and Agg-l transcripts at 24 hours were repressed by the GLN and CS combination by at least about 6 fold. Glucosamine was effective in suppressing IL-l induced mRNA expression of MMP-1 3, Agg-l and Agg-2 while CS was effective in decreasing IL-l induced MMP-13 transcript at 24 hours. At 48 hours post-stimulation, GLN and CS added alone and in combination significantly abrogated Agg—l and Agg-2 gene induction. The combination also decreased IL-l stimulated MMP-13 transcript. Treatments had no effect on Col 1] or aggrecan expression. Conclusions and Clinical Relevance-Our results indicate that GLN and CS, at concentrations that fall well within the range measured in synovial fluid and blood after 96 oral administration, may regulate expression of matrix degrading enzymes and their inhibitors at the transcriptional level, providing a plausible mechanism for their purported chondroprotective properties. Introduction Progressive and permanent articular cartilage degeneration is the hallmark characteristic of osteoarthritis (OA). Biological and mechanical factors uncouple the normal balance between articular cartilage degradation and synthesis. Cartilage destruction primarily results from an imbalance between synthesis and degradation of the extracellular matrix, particularly aggrecan and type II collagen (Col II). The expression and activity of proteolytic enzymes such as matrix metalloproteinases (MMPs) and aggrecanases exceeds that of endogenous inhibitors like tissue inhibitor of metalloproteinases (TIMPs).l Excess production of matrix degrading enzymes is in large part induced by the release of interleukin-1 (IL-l).2 Interleukin-1 is widely accepted as one of the proinflammatory cytokines that plays a critical role in OA pathogenesis.3 It up-regulates proteolytic enzymes and retards anabolic activities of the chondrocyte leading to declines in synthesis of collagen and proteoglycan}5 Degradation of the extracellular matrix in vivo and in vitro with IL-1 stimulation is reported in the human, bovine, porcine, equine and lapine species.6 The nutraceuticals, glucosamine (GLN) and chondroitin sulfate (CS) have been used over the past 30 years for treatment of OA and have reduced articular cartilage degeneration in animal models and human clinical trials. In humans, both compounds have demonstrated therapeutic effects on symptoms of OA, alleviating pain and 97 improving rnobility.7’9 In animals, the combination reduces lameness in dogs and horses and is effective in managing pain in cats.”'2 Despite several studies with positive results, use of these nutraceuticals is still not widely accepted due to small sample sizes and short-term nature of the trials. Many of the commercially available nutraceutical products contain both GLN and CS. Glucosamine and CS were synergistic in protecting articular damage in vivo13 and another study suggested they were complementary. '4 Thus, the chondroprotective properties of each molecule may increase when taken together and combining them for explant studies is important. Most in vitro studies have used concentrations of the nutraceuticals that exceed those generally found in synovial fluid. '5 The concentrations of GLN in synovial fluid and blood after oral and intravenous administration range from 0.05 to 20 [lg/1111.15.19 Depending on the route of administration, species, and the source and molecular weight of CS, the concentration of CS in serum ranges from 5 to 200 rig/mlm'20 Thus, in the present study, we determined the effect of GLN (5 rig/ml) and CS (20 ug/ml) individually and in combination, at concentrations attainable in viva, on gene expression of IL-1 stimulated bovine articular cartilage explants. The genes of interest included were matrix metalloproteinase (MMP)-2, MMP-3, MMP-9, MMP-l3, MMP-14, aggrecanase (Agg)-1, Agg—2, tissue inhibitor of metalloproteinase (TIMP)-l, TIMP-2, TIMP-3, Col 11 and aggrecan. Materials and Methods Explant cultures-Articular cartilage was isolated from the carpal joints of Holstein steers (18-24 months old) obtained fiom a local abattoir within 3 hours of 98 slaughter. Cartilage discs (6 mm in diameter) were biopsied from the articular surface and did not include the calcified layer of the tissue and cartilage with gross characteristics of OA. Two explant discs (approximately 60 mg total wet weight) were selected at random and cultured in each well of a 24-well Falcon culture plate11 containing 1 ml of 1:1 modified version of Dulbecco’s modified Eagle’s medium (DMEM): nutrient mixture F - 12 (Ham)b, as previously described.” The media was supplemented with amino acids, 50 ug/ml ascorbic acid and 100 units/ml penicillin-streptomycin ”’21. Cartilage explants were maintained in a humidified incubator at 37°C with 7% C02. Explants were maintained in media without serum for 48 hours before the addition of treatments. Media in the wells were exchanged daily. After equilibration, all treatments received 10% fetal bovine serum (F BS) b. Human recombinant interleukin-1 beta (rhIL-l B)° was added at a concentration of 50 ng/ml to induce cartilage catabolism. To examine the effects of GLN and CS, glucosamine HCld and low molecular weight chondroitin sulfate6 were added to the wells at the same time as FBS and rhIL-l B. The concentrations of GLN and CS chosen were 5 rig/ml and 20 rig/ml respectively. These concentrations were well within the range of levels attainable in synovial fluid and blood after oral and intravenous administration.” '20 There were 5 treatments per experiment (Table I), an FBS control (Ctrl), 50 ng/ml rhIL-l B (IL-1), 50 ng/ml rhIL-lB with the addition of 5 ug/ml GLN (IL-l + GLN), 50 ng/ml rhIL-lB with the addition of 20 rig/ml CS (IL-1 + CS) and 50 ng/ml rhIL-lB with the addition of both GLN and CS (IL-l + GLN + CS). Each treatment consisted of 12 wells (24 discs) collected from a pool of 3 steers for an experiment. Cartilage explants were collected 24 and 48 hours after stimulation, frozen in liquid nitrogen and stored at -80°C until RNA isolation Experiment 99 was repeated a total of 3 times for each time point, each time using cartilage from a pool of 3 different animals. Total RNA isolation-Total RNA was extracted from cartilage explants following a modified protocol.22 Briefly, cartilage was homogenized in TRIzol® reagentf and chloroform was added to extract total RNA followed by vigorous agitation and a 2- minute incubation. The aqueous phase containing RNA was collected after centrifugation (4°C, 12,000 X g, 15 minutes) and RNA precipitated with an equal volume of 70% ethanol. Total RNA was then purified further with RN easy mini columns8 and quantified by UV spectrophotometryh. Total chondrocyte RNA was resolved on 1.2% agarose gel to validate spectrophotometric determination and RNA integrity. cDNA synthesis-F or each sample, 2 1.1g of total RNA was treated with DNase If to degrade contaminating single and double stranded DNA. Treated RNA was converted to single stranded cDNA using Superscript II reverse transcriptasef as recommended by the manufacturer. Single stranded cDNA was quantified by UV spectrophotometry", and diluted with RNase-free water to 50 ng/ul. Quantitative real-time polymerase chain reaction (Q-RT-PCR)-Primers for glyceraldehyde phosphate dehydrogenase (GAPDH, used as a housekeeping gene) and molecules from Table II were designed using the Primer Express software version 2.0‘. Nucleotide sequences used for primer design were obtained from public databases (Genbank’). Optimal concentrations of each set of primers were determined with a primer matrix [lowest standard deviation with no change in cycle to threshold (CT)]. Quantitative real-time PCR was performed with 50 ng cDNA templates in 96-well platesi using the ABI PRISM 7000 sequence detection systemi. The cDNA templates were combined with 100 optimal concentrations of primers and SYBR Green PCR dye mixi in a total volume of 50 111 and the amplification conducted as recommended by the manufacturer. The PCR conditions were 2 min at 50°C and 10min at 95°C followed by 40 cycles of extension at 95°C for 15 sec and l min at 60°C, and data collected during the last 30 sec. Threshold lines were adjusted to intersect amplification lines in the linear portion of the amplification curves. The software automatically recorded the CT. The analysis of each sample was performed in duplicate, and a standard deviation of S 0.5 between replicates was set as a criterion for inclusion of data. The GAPDH gene was used as an endogenous control and run together with the samples for each amplification reaction to allow for normalization of different samples for RNA loading, cDNA synthesis, and amplification efficiencies and for comparison of samples run at different times. The F BS control treatment was used as a calibrator (i.e. the fold change for control is 1.0). Replicated data was normalized with GAPDH and the fold change in gene expression relative to FBS control was calculated using the 2(-AACT) method.23 Statistical analysis-Relative gene expression data determined using Q-RT-PCR was analyzed with SAS", using the nonparametric ANOVA approach of Friedman. The p-values of the gene-specific analyses were corrected for a False Discovery Rate of 5% using the approach discussed by Benjarnini and Hochberg.24 Differences were declared statistically significant when P<0.05, unless otherwise noted. Results The effect of glucosamine and chondroitin sulfate on gene expression of proteolytic enzymes-Explants cultured with rhIL-l B for 24 hours resulted in significant 101 up—regulation of MMP-3, MMP-13, Agg—l and Agg-2 relative to control (Figure 1, Panel A). Glucosamine repressed mRNA expression of MMP—l3, Agg-l and Agg—2 at 24 hours after induction (Figure 1, Panel A). Chondroitin sulfate suppressed a 7.7 times resting level induction of MMP-13 by IL-1 to control levels (Figure 1, Panel A). The GLN and CS combination was effective in down-regulating IL-l induced gene expression of MMP-3, MMP-13 and Agg-l (Figure 1, Panel A). There were no treatment effects for MMP-2 and MMP-9 (data not shown), and MMP-l4 (Figure 1, Panel A & Panel B) at 24 and 48 hours post-culture. The induction of MMP-13, Agg-l and Agg—2 mRNA expression by IL-1 at 24 hours were sustained up to 48 hours post-stimulation (Figure 1, Panel B). Glucosamine and CS added alone or in combination for 48 hours significantly reduced IL-l stimulated Agg-l and Agg-2 mRNA transcripts (Figure 1, Panel B). At 48 hours, the GLN and CS combination decreased IL-l induced MMP-13 from 4.9 fold to 1.0 fold (Figure 1, Panel B). No treatment effects were found for MMP-3 (Figure 1, Panel B) at the 48-hour time point. The effect of glucosamine and chondroitin sulfate on gene expression of inhibitors of proteolytic enzymes-At 24 hours post-stimulation, the GLN and CS combination tended (P<0.10) to up-regulate TIMP-3 transcript relative to CS (Figure 2, Panel A). Treatment effect was not found for TIMP-1 (Figure 2, Panel A & Panel B) and TIMP-2 (data not shown) at both 24 and 48-hour time points and TIMP-3 at 48 hours (Figure 2, Panel B). 102 The effect of glucosamine and chondroitin sulfate on gene expression of cartilage macromolecules-No treatment effect was found for expression of genes encoding both Col 11 and aggrecan at all time points. Discussion Glucosamine and CS have been used for over 3 decades to treat OA. Clinical trials showed that GLN and CS used individually or in combination are effective in improving the symptoms of knee and hip OA.7'9’25’26 Their benefits also extend to animals suffering from OA, which is a common and natural occurrence in dogs, horses and catsm'12 Despite the fact they have been reported to be beneficial, their exact mechanism(s) of action remains to be resolved. Previous experiments have provided some clues about the chondroprotective properties of the nutraceuticals. However, one major limitation to these studies is that concentrations of the nutraceuticals employed have been quite high, ranging from 0.1 to 10 mg/ml.”'2 1’27'3 6 These concentrations are most probably not attainable in viva. This is one reason given by rheumatologists for not advocating the use of these nutraceuticals.” The current study represents an improved approach to study the effects of GLN and CS in that concentrations of the nutraceuticals were used at levels that have been reported in blood after oral and intravenous administration. Now that we have determined the beneficial effects at these concentrations, further experiments will be done looking at a range of concentrations within the reported values. We also studied the effect of GLN and CS as a combination since they are increasingly marketed as one entity and the combination has a putative synergistic effect in decreasing the severity of cartilage lesions.l3 103 The predominate groups of enzymes responsible for cartilage breakdown are MMPs and aggrecanases. The MMPs are a group of zinc containing proteinases that play a central role in the degeneration of the extracellular matrix. During matrix breakdown, the normal balance between MMPs and TIMPs is disrupted towards an increase in activity of MMPs and a decrease in TIMPs.l Chondrocytes and synoviocytes in OA specimens produce MMPs particularly MMP-3 and MMP-13.38 Cytokines stimulate the expression of MMP-3 and MMP-133942 in agreement with the current study. MMP-3 has a broad range of substrate specificity and deemed as one of the major enzymes responsible for matrix proteolysis.43 MMP-13 plays a critical role in OA by breaking down type II collagen preferentially.“ MMP-14 belongs to the membrane-type MMPs. It hydrolyzes collagens and is capable of activating latent MMP-2.43 Thus, suppressing the expression of these enzymes will likely be beneficial to OA patients. The ability of GLN and CS to protect cartilage from being degraded pathologically” may be explained at least in part by the regulation of MMP synthesis. Specifically, the GLN and CS combination inhibited the cytokine-induced expression of a number of the MMPs examined. Glucosamine and CS in combination exerted a transient influence on the MMP-3 transcript in that a significant reduction in cytokine-induced expression was observed at 24 but not at 48 hours. Previous reports have demonstrated suppression of MMP-3 gene expression and enzyme activity by GLN.27’29’3 "34 However, the concentration of GLN used was at least 100 ug/ml, far exceeding the concentration employed in the present study. One study that used glucosamine sulfate at low concentrations (~200 ng/ml) on human OA chondrocytes did show a decline in MMP-3 activity and production but not on mRN A expression.“ We observed that GLN and CS 104 used alone were effective in down-regulating MMP-13 for at least 24 hours and the repression was prolonged by the combination. These findings supplements previous studies that reported a decrease in MMP-13 protein and activity with the nutraceutical combination[4 and with GLN alone at higher concentrations than we employed.27 To date, the effects of GLN and CS on MMP-l4 mRNA expression has not been investigated. Neither the nutraceutical combination nor IL-l had a significant influence on its expression. The absence of an effect of IL-1 on MMP-14 and MMP-2 is supported by their reported constitutive expression in adult human cartilage.“46 Lack of an effect with GLN and CS on the expression and activity of MMP-2 paralleled previous studiesmiz"36 Expression patterns of MMP-2 also coincided with MMP-l4 since MMP- 14 activates it. The expression of MMP-9 is strongly inducible by inflammatory cytokines.“ The activity of MMP-9 was, however significantly reduced by 1 mg/ml GLN in equine cartilage explants.l4 The reason for an absence of IL-1 effect on MMP-9 mRN A expression in the present study is not known. Aggrecanases plays a pivotal role in normal and pathological turnover of aggrecan in cartilage.48 Together with MMPs, Agg-l and Agg-2 hydrolyzes the major proteoglycan of articular cartilage and are induced by IL— 1 .28’42’49'50 The suppression of IL-1 stimulated aggrecanase activity with GLN has been reported by Sandy et. al.28 and [lie et. al.51 The concentrations of GLN used in the present study is about 80 and 200 times lower than that employed in studies by Sandy et. al.28 and Ilic et. al.5 1 respectively. Chondroitin sulfate and the nutraceutical combination at pharmacological concentrations successfully repressed both aggrecanases by 48 hours post-stimulation. Chondroitin sulfate inhibited aggrecanase activity in culture.30 Simultaneous suppression of MMPs 105 and aggrecanases with GLN and CS supplementation may represent an effective way to protect matrix components from catabolic processes. Synthetic inhibitors of MMPs, TIMPs and agents that elevated TIMPs demonstrated effectiveness towards reversing or halting catabolic activities associated with OA.52 Addition of TIMP-1 and TIMP-2 reduced MMP-3 concentrations in canine chondrocytes culture.53 TIMP-3 retarded IL-l stimulated proteoglycan release that is mediated by aggrecanases4 Thus, upregulation of TIMPs may be one of many events to counter catabolic activity in OA pathogenesis. TIMP-3 was up—regulated 2.4 fold by the nutraceutical combination relative to IL-1 at 6 hours of culture (data not shown). In a different study conducted in our laboratory employing 10 pg/ml GLN in combination with 20 ug/ml CS found that TIMP-3 was significantly elevated relative to IL-1 at 8 hours post-stimulation (unpublished observation). Studies are ongoing in our laboratory to verify if TIMP-3 transcript up—regulation is accompanied by an elevation in TIMP-3 protein when explants are supplemented with the nutraceutical combination. Glucosamine and CS at these pharmacological concentrations did not affect the expression of gene encoding aggrecan and type II collagen. Glucosamine sulfate at concentrations of 10 ug/ml to 100 ug/ml had no effect on collagen production in vitra.55'56 Horses affected with OA established no association of mRNA expression of Ms with aggrecan and type II collagen gene expressions7 In normal human chondrocytes, GLN had no effect on proteoglycan synthesis.58 Our findings on aggrecan contrast the study by Dodge and co-worker45 that reported a dose dependent increase in aggrecan mRNA with GLN (from about 200 11ng to 30 ug/ml) and this may be attributed to the difference in species (bovine versus human), type of tissue (normal 106 versus OA), the source of GLN (GLN hydrochloride versus GLN sulfate), type of culture used (explants versus monolayers), different concentrations of IL-1 (50 11ng versus 5 ng/ml), time length of culture (a maximum of 48 hours versus 72 hours) and the absence versus the presence of preincubation with GLN. Lack of adequate stress may also prevent changes seen in macromolecules with GLN and CS supplementation.”59 The synthesis of these major macromolecules of cartilage is catalyzed by a number of enzymes. These nutraceuticals may exert their effects on mediators of aggrecan and collagen synthesis or affect macromolecule synthesis at a translational or post-translational level. Glucosamine and CS increased sulfate incorporation in stressed cartilage59 and GLN prevented IL-l mediated inhibition of galactose-betal, 3-glucuronosyltransferase I, an enzyme that catalyzes the addition of the initial glucuronic acid residue in glycosaminoglycan synthesis.29 Both GLN and cs also increase hyaluronic acid synthesis.“-61 With the recent controversy surrounding COX-2 inhibitors in humans and the deleterious effects of nonsteroidal anti-inflammatory drugs on matrix homeostasis, the use of alternative therapies for OA needs to be explored further. Since both GLN and CS exhibited good therapeutic efficacy, remarkable tolerability and safety coupled with effective symptomatic relief for humans and animals7'12’25’26’62, their continued use looks promising. The present study shows that GLN and CS at concentrations that are biologically relevant prevented some of the changes in gene expression associated with IL-1. The inhibition of proteoglycan degradation with GLN and C855'59 may be in part mediated by the repression of genes associated with some major cartilage-degrading enzymes and possibly up-regulation of metalloproteinase inhibitors. The combination was more effective in antagonizing some gene expression than if GLN and CS were used 107 individually. Further experiments should be conducted to determine if these changes in gene expression can be replicated using an animal model for OA. Footnotes aFisher Scientific, Pittsburgh, PA. I’Gibco, Grand Island, NY. c R & D Systems, Minneapolis, MN. dFCHG49®, Nutramax Laboratories, Edgewood, MD. ”TRH122®, Nutramax Laboratories, Edgewood, MD. fInvitrogen, Carlsbad, CA. gQiagen, Valencia, CA. hBeckrnan Coulter, Fullerton, CA. iPerkin-Elmer Applied Biosystems, Foster City, CA. jGenbank Database, National Centre Biotechnology Information. "SAs Institute Inc, Cary, NC. 108 Table I: Description of components found in treatment media of explant cultures Treatment FBS (%) rhIL—lB (IL-1) Glucosamine Chondroitin (GLN) sulfate (CS) FBS control 10 - - - IL-1 10 50 rig/ml - - IL-l + GLN 10 50 ng/ml 5 rig/ml - IL-l + CS 10 50 ng/ml - 20 rig/ml IL—l + GLN + CS 10 50 ng/ml 5 “g/ml 20 pg/ml F BS= fetal bovine serum; rhIL-1B= human recombinant interleukin-1 beta 109 05505550550000 55505005050550 3.380.... 5.20 0055005055550005 5550005505055 285 58000. 55505000050500 50050505000550 848x 0 60 50505055005000 55550500550000 53.2122 5-55 0505550550005005 55550050555000 53.2122 055 5050550000000 5050505055550 53.2122 :55 05505005505500 55000505000055 ESE... 0-0m? 55055500500000 55055505055000050 25052 70.0.... 55500505000000 05050505005050 02.41.? 2.02: 50505050055555. 05555550050050¢ 50.5.:— Eazeh 0:03:0U 0:00 56000.. 32.0 00208.nen oat-.00.. 02:35:50 .55 con: 500585 .«e non—om we 5.0.41.3 000:0:000 50.5.5 09.0.5.— ES 203.—eh "~— 030,—. 110 Figure 1. Mean i SE expression of genes encoding proteolytic enzymes relative to F BS control for cartilage explants stimulated for 24 hours (Panel A) and 48 hours (Panel B). FBS = fetal bovine serum. IL-1 = 50 11ng human recombinant interleukin-1. IL-1 + GLN = 50 ng/ml human recombinant interleukin-1 with the addition of 5 1.1ng glucosamine HCl. IL-l + CS = 50 ng/ml human recombinant interleukin-l with the addition of 20 ug/ml chondroitin sulfate. IL-l + GLN + CS = 50 ng/ml human recombinant interleukin-l with the addition of 5 pg/ml glucosamine HC1 and 20 pg/ml chondroitin sulfate. MMP-3 = matrix metalloproteinase-3. MMP-13 = matrix metalloproteinase-13. MMP-l4 = matrix metalloproteinase-l4. Agg-l = aggrecanase-l. Agg-Z = aggrecanase-2. Different letters within a gene indicate significant (P<0.05) differences between treatments. 111 Panel A D FBS control I IL-1 I lL-1 + GLN [D lL-1 + CS B lL-1 + GLN + CS 161 Mean relative gene expression Panel B 1“ N Mean relative gene expression OJ 112 Figure 2. Mean i SE expression of genes encoding inhibitors of proteolytic enzymes and cartilage macromolecules relative to F BS control for cartilage explants stimulated for 24 hours (Panel A) and 48 hours (Panel B). FBS = fetal bovine serum. IL-1 = 50 ng/ml human recombinant interleukin-1. IL-1 + GLN = 50 ng/ml human recombinant interleukin-l with the addition of 5 ug/ml glucosamine HCI. IL-l + CS = 50 ng/ml human recombinant interleukin—1 with the addition of 20 pg/ml chondroitin sulfate. IL-l + GLN + CS = 50 11ng human recombinant interleukin-1 with the addition of 5 ug/ml glucosamine HCl and 20 pg/ml chondroitin sulfate. TIMP-1 = tissue inhibitor of metalloproteinase-1. TIMP-3 = tissue inhibitor of metalloproteinase-3. Col II = type II collagen. Different letters within a gene indicate that treatments tended (P<0.10) to be different. 113 Panel A El FBS control I lL-1 I IL-1 + GLN ll] lL-1 + CS a lL-1 + GLN + CS ab Mean relative gene expression Panel B & u 21 Mean relative gene expression IMP-1 TIMP-3 Col ll Aggrocan 114 References l. Tchetverikov I, Ronday HK, Van El B, et al. MMP profile in paired serum and synovial fluid samples of patients with rheumatoid arthritis. Ann Rheum Dis 2004;63:881-883. Lefebvre V, Peeters-Joris C, Vacs G. Modulation by interleukin 1 and tumor necrosis factor alpha of production of collagenase, tissue inhibitor of metalloproteinases and collagen types in differentiated and dedifferentiated articular chondrocytes Biochim Biophys Acta 1990;10522366-378. Dinarello CA. Interleukin-1. Ann N Y Acad Sci 1988;546:122-132. Benton HP, Tyler J A. Inhibition of cartilage proteoglycan synthesis by interleukin 1. Biochem Biophys Res Comm 1988;154:421-428. Tyler JA, Benton HP. Synthesis of type II collagen is decreased in cartilage cultured with interleukin 1 while the rate of intracellular degradation remains unchanged Coll Relat Res 1988;8z393-405. Dingle JT, Page Thomas DP, Hazleman B. The role of cytokines in arthritic diseases: in vitro and in vivo measurements of cartilage degradation. Int J Tissue React l987;9:349-354. Lefiler CT, Philippi AF, Leffler SG, et al. Glucosamine, chondroitin, and manganese ascorbate for degenerative joint disease of the knee or low back: a randomized, double-blind, placebo-controlled pilot study. Mil Med 1999;164:85- 91. Das A Jr, Hammad TA. Efficacy of a combination of F CHG49 glucosamine hydrochloride, TRHIZZ low molecular weight sodium chondroitin sulfate and manganese ascorbate in the management of knee osteoarthritis Osteoarthritis Cartilage 2000;8:343-350. McAlindon TE, LaValley MP, Gulin JP, et al. Glucosamine and chondroitin for treatment of osteoarthritis: a systematic quality assessment and meta-analysis JAMA 2000;283:1469-1475. 10. Canapp SO Jr, McLaughlin RM Jr, Hoskinson JJ, et al. Scintigraphic evaluation of dogs with acute synovitis after treatment with glucosamine hydrochloride and chondroitin sulfate. Am J Vet Res 1999;60:1552-1557. 11. Hanson R, Brawner W, Blaik M. Oral treatment with a nutraceutical (Cosequin®) for ameliorating signs of navicular syndrome in horses. Vet Ther 2001;2zl48-159. 115 12. Richards J, Rodan 1. Panel report on feline senior health care, Part II. Compend Contin Educ Pract Vet 1999;21:612-621. 13. Lippiello L, Woodward J, Karpman R, et al. In vivo chondroprotection and metabolic synergy of glucosamine and chondroitin sulfate. Clin Orthop 2000;381 :229-240. 14. Orth MW, Peters TL, Hawkins JN. Inhibition of articular cartilage degradation by glucosamine-HCl and chondroitin sulphate. Equine Vet J 2002;3z224-229. 15. Laverty S, Sandy JD, Celeste C, et al. Synovial fluid levels and serum pharmacokinetics in a large animal model following treatment with oral glucosamine at clinically relevant doses. Arthritis Rheum 2005;52:181-191. 16. Setnikar I, Giacchetti C, Zanolo G. Pharmacokinetics of glucosamine in the dog and in man Arzneimittelforschung 1986;36:729-735. 17. Liang Z, Leslie J, Adebowale A, et al. Determination of the nutraceutical, glucosamine hydrochloride, in raw materials, dosage forms and plasma using pre- column derivatization with ultraviolet HPLC. J Pharm Biomed Analysis 1999;20:807-814. 18. Du J, Eddington N. Determination of the chondroitin sulfate disaccharides in dog and horse plasma by HPLC using chondroitinase digestion, precolumn derivatization, and fluorescence detection. Analytical Biochemistry 2002;3 06:25 2- 258. 19. Du J, White N, Eddington ND. The bioavailability and pharmacokinetics of glucosamine hydrochloride and chondroitin sulfate after oral and intravenous single dose administration in the horse. Biopharm Drug Dispos 2004;25:109-116. 20. Volpi N. Oral absorption and bioavailability of ichthyic origin chondroitin sulfate in healthy male volunteers Osteoarthritis Cartilage 2003;11:433-441. 21. Fenton J I, Chlebek-Brown KA, Peters TL, et al. Glucosamine HCl reduced equine articular cartilage degradation in explant culture. Osteoarthritis Cartilage 2000;8:258-265. 22. Reno C, Marchuk L, Sciore P, et al. Rapid isolation of total RNA from small samples of hypocellular, dense connective tissues. Biotechniques 1997 ;22: l 082- 1086. 23. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real- time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001; 25:402-408. 116 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 3.4. 35. Benjamini, V, Hochberg V. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Statist Soc B 1995;57:289-300. 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;Suppl 6Az39-46. Reginster J Y, Deroisy R, Rovatti LC, et al. Long—term effects of glucosamine sulphate on osteoarthritis progression: a randomized, placebo-controlled clinical trial. Lancet 2001;357:251-256. Byron CR, Orth MW, Venta PJ, et al. Influence of glucosamine on matrix metalloproteinase expression and activity in lipopolysaccharide-stimulated equine chondrocytes. Am J Vet Res 2003;64:666-671. Sandy JD, Gamett D, Thompson V, et al. Chondrocyte mediated catabolism of aggrecan: aggrecanase dependent cleavage induced by interleukin-1 or retinoic acid can be inhibited by glucosamine. Biochem J 1998;335:59-66. Gouze JN, Bordji K, Gulberti S, et al. Interleukin-10 down-regulates the expression of glucuronosyltransferase I, a key enzyme priming glycosaminoglycan biosynthesis: Influence of glucosamine on Interleukin-10- medicated effects in rat chondrocytes. Arthritis Rheum 2001;44:351-360. Sugimoto K, Takahashi M, Yamamoto Y, ct al. Identification of aggrecanase activity in medium of cartilage culture. J Biochem (Tokyo) 1999;126:449—455. Fenton J I, Chlebek KA, Caron JP, et al. Effect of glucosamine on interleukin-l conditioned articular cartilage. Equine Vet J 2002;Supp 34:219-223. Largo R, Alvarez-Soda MA, Dieportego I, et al. Glucosamine inhibits [Le-102- induced NFKB activation in human osteoarthritic chondrocytes. Osteoarthritis Cartilage 2003;] 1:290-298. Mello DM, Nielsen BD, Peters TL, et al. Comparison of inhibitory effects of glucosamine and mannosamine on bovine articular cartilage degradation in vitro. Am J Vet Res 2004;65:144021445. Nakamura H, Shibakawa A, Tanaka M, et al. Effects of glucosamine hydrochloride on the production of prostaglandin E2, nitric oxide and metalloproteases by chondrocytes and synoviocytes in osteoarthritis. Clin Exp Rheumat 2004;22:293-299. Hua J, Sakamoto K, Nagaoka I. Inhibitory actions of glucosamine, a therapeutic agent for osteoarthritis, on the function of neutrophils. J Leukoc Biol 2002;71:632=640. 117 36. 37. 38. 39. 40. 41. 42. 43. 45. 46. Schlueter AE, Orth MW. Further studies on the ability of glucosamine and chondroitin sulphate to regulate catabolic mediators in vitro. Equine Vet J 2004;36:634-636. Manson JJ, Rahman A. This house believes that we should advise our patients with osteoarthritis of the knee to take glucosamine. Rheum Oxford 2004;43:100- 1 01 . Zafarullah M, Pelletier JP, Cloutier JM, et al. Elevated metalloproteinase and tissue inhibitor of metalloproteinase mRNA in human osteoarthritic synovia. J Rheumatol 1993;20:693-697. Caron JP, Tardif G, Martel-Pelletier J, et a1. Modulation of matrix metalloprotease 13 (collagenase 3) gene expression in equine chondrocytes by IL-1 and corticosteroids. Am J Vet Res 1996;57:1631-1634. Morris EA, Treadwell BV. Effect of interleukin 1 on articular cartilage from young and aged horses and comparison with metabolism of osteoarthritic cartilage. Am J Vet Res 1994;55:138-146. Richardson D, Dodge GR. Effects of interleukin-10 and tumor necrosis factor-a on expression of matrix-related genes by cultured equine articular chondrocytes. Am J Vet Res 2000;61:624-630. Koshy P] T, Lundy CJ, Rowan AD, et al. The modulation of matrix metalloproteinase and ADAM gene expression in human chondrocytes by interleukin-1 and oncostatin M: A time course study using real time quantitative reverse transcription- polymerase chain reaction. Arthritis Rheum 2002;46:961- 967. Matrisian LM. Metalloproteinases and their inhibitors in matrix remodeling Trends Genet 1990;6:121-125. . Reboul P, Pelletier JP, Tardif G, et al. The new collagenase, collagenase 3, is expressed and synthesized by human chondrocytes but not by synoial fibroblasts: a role in osteoarthritis. J Clin Invest 1996;97:2011-2019. Dodge GR, Jimenez SA. Glucosamine sulfate modulates the levels of aggrecan and matrix metalloproteinase-3 synthesized by cultured human osteoarthritis articular chondrocytes. Osteoarthritis Cartilage 2003;11:424-432. Chubinskaya S, Kuettner KE, Cole AA. Expression of matrix metalloproteinases in normal and damaged articular cartilage from human knee and ankle joints. Lab Invest 1999;79:1669-1677. 118 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. Mengshol JA, Mix KS, Brinckerhoff CE. Matrix metalloproteinases as therapeutic targets in arthritic diseases: bull’s-eye or missing the mark? Arthritis Rheum 2002 ;46: 1 3-20. Lark MW, Bayne EK, Flanagan J, et al. Aggrecan degradation in human cartilage. Evidence for both matrix metalloproteinase and aggrecanase activity in normal, osteoarthritic, and rheumatoid joints. J Clin Invest 1997;100:93-106. Tortorella MD, Malfait AM, Deccico C, et al. The role of ADAM-T84 (aggrecanase-1) and ADAM-T85 (aggrecanase-2) in a model of cartilage degradation Osteoarthritis Cartilage 2001;92539-552. Sandy JD, Verscharen C. Analysis of aggrecan in human knee cartilage and synovial fluid indicates that aggrecanase (ADAMTS) activity is responsible for the catabolic turnover and loss of whole aggrecan whereas other protease activity is required for C-terrninal processing in vivo. Biochem J2001;358:615-626. Ilic MZ, Martinac B, Handley CJ. Effects of long-term exposure to glucosamine and mannosamine on aggrecan degradation in articular cartilage. Osteoarthritis Cartilage 2003 ;1 1 :613-622. Steinmeyer J, Daufeldt S, Kalbhen DA. The proteoglycan metabolism, morphology and viability of articular cartilage treated with a synthetic matrix metalloproteinase inhibitor. Res Exp Med 1997;197:63-79. Kuroki K, Cook JL, Kreeger JM, et al. The effects of TIMP-1 and -2 on canine chondrocytes cultured in three-dimensional agarose culture system. Osteoarthritis Cartilage 2003;1 1:625-635. Gendron C, Kashiwagi M, Hughes C, et al. TIMP-3 inhibits aggrecanase- mediated glycosaminoglycan release from cartilage explants stimulated by catabolic factors. FEBS Lett 2003;555:431-436. Bassleer C, Rovati L, F ranchimont P. Stimulation of proteoglycan production by glucosamine sulfate in chondrocytes isolated fiom human osteoarthritis articular cartilage in vitro. Osteoarthritis Cartilage l998;6:427-434. Bassleer C, Henrotin Y, Franchimont P. In vitro evaluation of drugs proposed as chondroprotective agents. Int J Tissue React 1992;14:231-241. Trumble TN, Trotter GW, Oxford JR, et al. Synovial fluid gelatinase concentrations and matrix metalloproteinase and cytokine expression in naturally occurring joint disease in horses. Am J Vet Res 2001;62:1467-1477. Mroz PJ, Silbert JE. Use of 3H-glucosamine and 35S-su1fate with cultured'human chondrocytes to determine the effect of glucosamine concentration on formation of chondroitin sulfate. Arthritis Rheum 50;3574-3579. 119 5 9. Lippiello L. Glucosamine and chondroitin sulfate: biological response modifiers of chondrocytes under simulated conditions of j oint stress. Osteoarthritis Cartilage 2003;] 1:335-342. 60. Ronca F, Palmieri L, Panicucci P, et al. Anti-inflammatory activity of chondroitin sulfate. Osteoarthritis Cartilage 1998;Suppl 6A: 14-21 . 61. McCarty MF. Enhanced synovial production of hyaluronic acid may explain rapid clinical response to high-dose glucosamine in osteoarthritis Med Hypo 1998;50:507-510. 62. McNamara PS, Barr SC, Erb HN, et al. Hematological, hemostatic, and biochemical effects in cats receiving an oral chondroprotective agent for thirty days. Vet T her 2000;]:108-117. 120 CHAPTER 4 Chan PS, Caron JP, Orth MW. Short term gene expression changes in interleukin- IB stimulated cartilage explants with glucosamine and chondroitin sulfate. Arthritis Rheum 2005; submitted. 121 ABSTRACT Objective. To determine the short-terrn effects of glucosamine (GLN) and chondroitin sulfate (CS) on expression of genes encoding inflammatory mediators and matrix enzymes in interleukin-1 (IL-1) induced bovine cartilage explants. Methods. Dose response experiments were conducted for IL-1, GLN and CS to select concentrations of each that will help improve the sensitivity of detecting treatment effects on cartilage explants. Based on the dose response experiments, treatments included F BS control, 15 ng/ml IL-1, and 15 ng/ml IL-l with the addition of 10 ug/ml GLN and 20 rig/m1 CS. Media were measured for nitric oxide (NO) and prostaglandin E2 (PGE2) while explants were frozen for RNA extraction at 8, l6 and 24 hours. Gene expression relative to FBS control for inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), microsomal prostaglandin E synthase-1 (mPGEs 1 ), nuclear factor kappa beta p65 subunit (NF KB), matrix metalloproteinase (MMP)-3, l3, aggrecanase (Agg)—1, 2 and tissue inhibitor of metalloproteinase-3 (TIMP-3) were assessed with quantitative real-time polymerase chain reaction (Q-RT-PCR). In a separate study using incubation of explants with the aforementioned treatments for 48 hours, proteoglycan release was measured with dimethylmethylene blue assay and TIMP-3 protein was evaluated with Western blots. Results. The GLN and CS combination abrogated IL-l induced gene expression of iNOS, COX-2, mPGEsl, NFKB at all time points. NO, PGE2 and proteoglycan release were reduced with the combination. Stimulated MMP-13, Agg-l and Agg-2 mRNA abundance were repressed whereas TIMP-3 was up-regulated by the combination at all time points. TIMP-3 protein abundance was increased by the combination relative to IL-1 at 48 hours. 122 Conclusion. Glucosamine and CS in combination suppress synthesis and expression of genes encoding inflammatory mediators and proteolytic enzymes while up regulating TIMP-3. This provides a plausible mechanism for GLN and CS purported mild anti- inflammatory and chondroprotective properties. INTRODUCTION Glucosamine (GLN) and chondroitin sulfate (CS) are the predominant nutraceutical supplements marketed for improving joint health. Glucosamine is an amino monosaccharide and a major building block of proteoglycans. Clinical trials show that GLN administered orally was effective in decreasing pain and improving joint mobility in patients with osteoarthritis (OA) (1). Chondroitin sulfate, a complex glycosaminoglycan, is a major component of aggrecan. Beneficial effects of CS include improved joint mobility, reduced rate of joint space narrowing and a reduction of erosive OA (2, 3). Glucosamine is commonly combined with CS in many of the commercially available nutraceutical products. The combination has been proven efficacious in reducing pain, improving joint function, and halting or reversing joint degeneration in humans with mild to moderate OA of the knee (4). Severe cartilage lesions in an in vivo rabbit instability model of CA were diminished with GLN and CS supplementation (5). Very few in vitro studies aimed at determining the mode of action of these nutraceuticals have employed the combination although clinical studies have reported enhanced efficacy with co- administration of GLN and CS. Despite several studies reporting favorable results, the exact mechanism of action of these nutraceuticals still remains to be resolved. The applicability of many in vitro 123 mechanistic studies has been questioned because concentrations of the nutraceuticals used greatly exceed those generally found in blood and synovial fluid (6, 7). The concentrations of GLN in blood and synovial fluid after oral and intravenous (IV) administration range from about 0.05 to 20 ug/ml (7-10). Depending on the route and frequency of administration, species, and the source and molecular weight of CS, the concentration of CS in serum ranges from 5 to 200 uglml (6, 8, 11, 12). In past studies, cartilage explants or cell cultures stimulated with IL-1 demonstrated suppression of nitric oxide (N O) and prostaglandin E2 (PGE2) release to the media with supplementation of GLN and CS in the concentration range of 0.1 to 10 mg/ml (13-22). Two other studies employed concentrations of GLN and CS that are attainable in vivo and have shown that these nutraceuticals were able to prevent a decline in proteoglycan synthesis typically induced by catabolic agents (23, 24). Recently, we provided preliminary data demonstrating that biologically relevant concentrations of GLN (5 rig/ml) and CS (20 ug/ml) to regulate both gene expression and protein synthesis of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in addition to a reduction in the microsomal prostaglandin E synthase-1 (mPGEsl) gene in 24 hour incubations (25). These concentrations of GLN and CS were also able to repress major cartilage proteolytic enzymes implicated in OA pathogenesis at the pre-translational level (26). Most in vitro studies have used IL-l as a catabolic agent in the range of 1 to 10 11ng (19, 24, 27-29). The objective of the current study is to further characterize the effects of biologically relevant concentrations of these nutraceuticals using a subsaturating IL—l induction of bovine cartilage explants. Specifically, we investigated 124 the effect of GLN and CS in combination on IL-1 induced proteoglycan degradation, gene expression and protein synthesis on an array of inflammatory mediators and cartilage matrix degrading enzymes and one of their inhibitors in short-terrn explant cultures. MATERIALS AND METHODS Explant cultures. Articular cartilage was isolated from the carpal joints of Holstein steers (18-24 months old) obtained fiom a local abattoir within 3 hours of slaughter. Cartilage discs (6 mm in diameter) were biopsied from the articular surface and did not include the calcified layer of the tissue nor cartilage with gross characteristics of OA. Two explant discs (approximately 60 mg total wet weight) were selected at random and cultured in each well of a 24-well Falcon culture plate (Fisher Scientific, Pittsburgh, PA) containing 1 ml of 1:1 modified version of Dulbecco’s modified Eagle’s medium (DMEM): nutrient mixture F -12 (Ham) (Gibco, Grand Island, NY), as previously described (14). The media was supplemented with 50 ug/ml ascorbic acid, 100 units/ml penicillin-streptomycin (Gibco) and 20 amino acids (Sigma, St. Louis, MO). The concentrations of amino acids added were 50% of those previously reported (30). Cartilage explants were maintained in a humidified incubator at 37°C with 7% CO2. Dose response experiment with IL-1. Explants were maintained in media without serum for 48 hours before the addition of treatments. Media in the wells were exchanged daily. Afier equilibration, all wells received 10% fetal bovine serum (F BS, Gibco) and varying concentrations (0, 5, 10, 20 and 50 ng/ml) of human recombinant interleukin-1 beta (rhIL-l B, R & D Systems, Minneapolis, MN) for 24 hours to determine 125 the subsaturating concentration of IL-1 that would result in stimulation of nitric oxide (NO) and PGE2 release. There were 8 wells per IL—l concentration. Conditioned media collected at 24 hours were stored at 4°C for NO and PGE2 analysis. Experiment was repeated a total of 3 times, each time using tissue from a different animal. Dose response experiments with GLN and CS. Similar to the methods above, afier a 48 hour equilibration with serum free media, all explants were treated with 10% FBS, 15 ng/ml IL-l (approximated subsaturating concentration of lL-l), and varying concentrations of glucosamine HCl (FCHG49®, Nutramax Laboratories, Edgewood, MD) for 24 hours. The concentrations of GLN chosen were 0, 1, 5, 10 and 20 rig/ml. These concentrations were within the range of levels attainable in blood after oral and IV administration (6-10). Conditioned media collected at 24 hours were stored at 4°C for NO and PGE2 analysis. There were 8 wells for each GLN concentration. The experiment was repeated a total of 3 times, each time using tissue from a different steer. These procedures for GLN were repeated for CS. Concentrations of low molecular weight (16.9 kDa) CS (TRH122®, Nutramax Laboratories) chosen were 0, 5, 20, 50 and 100 ug/ml. GLN and CS in combination. Explants were maintained in media without serum for 48 hours prior to addition of treatments. Afier equilibration, all treatments received ' 10% FBS and 15 ng/ml IL-1 for 8, 16 and 24 hours to induce inflammatory mediators and cartilage catabolism. To examine the effects of GLN and CS, they were added to the wells at the same time as F BS and IL-1. The concentrations of GLN and CS chosen were 10 ug/ml and 20 ug/ml respectively (discussed in results). There were 3 treatments per experiment, an FBS control, 15 ng/ml IL-1 (IL-1) and 15 ng/ml IL-l with the addition of 10 ug/ml GLN and 20 ug/ml CS (IL-1 + GLN + CS). Each treatment consisted of 12 126 wells (24 discs) per time point. Cartilage explants were collected at 8, 16 and 24 hours after stimulation, frozen in liquid nitrogen, and stored at -80°C until RNA isolation. Conditioned media were collected at all time points and stored at 4°C for NO (all time points) and PGE2 (only the 24-hour samples) analysis. The experiment was repeated a total of 3 times, each time using cartilage from a different animal. Proteoglycan and TIMP-3 study. Explants were maintained in media without serum for 48 hours prior to addition of treatments. After equilibration, all treatments received 10% FBS. To examine the effects of GLN and CS on proteoglycan release (an indicator of metalloproteinase and aggrecanase activity) and TIMP-3 protein, they were added to the wells at the same time as F BS and IL-1 . There were 4 treatments per experiment, an F BS control, 15 ng/ml lL-l (IL-1), 10 ug/ml GLN and 20 ug/ml CS (GLN + CS) and 15 ng/ml IL-l with the addition of 10 ug/ml GLN and 20 ug/ml CS (IL-1 + GLN + CS). Each treatment consisted of 6 wells (12 discs). Media was collected and replaced daily. Cartilage explants were collected 48 hours after stimulation, frozen in liquid nitrogen and stored at -80°C until protein extraction. Conditioned media were collected at 24 and 48 hours post-stimulation and stored at 4°C for dimethyhnethylene blue (DMB) analysis. The experiment was repeated a total of 3 times, each time using cartilage from a different animal. Nitric oxide assay. Nitrite was measured in conditioned media using the Griess reagent and sodium nitrite as standard (32). Briefly, 150 pl medium was incubated with 150 pl of 1.0% sulfanilamide, 0.1% N-l-napthylethylenediamide hydrochloride and 25% phosphoric acid at room temperature for 5 nrinutes. Due to some precipitation of reagents with CS, 96-well plates were spun at 1000 X g for 3 rrrinutes at 4°C. The remaining 127 supernatant was transferred to a new plate. Absorbance was measured at 540 nm using a Spectromax 300 plate reader (Molecular Devices, Sunnyvale, CA). Prostaglandin E2 assay. Prostaglandin E2 release into conditioned media was quantified using a commercially available competitive enzyme linked immunosorbent assay kit according to manufacturer’s instructions (R & D Systems). Conditioned media samples were stabilized with indomethacin (10 ug/ml) and stored at -20°C until analysis. Dimethylmethylene blue assay. Proteoglycan release into conditioned media was measured using a DMB assay as previously described (33). Proteoglycan concentration was determined by measuring sulfated glycosaminoglycan (GAG) content compared to a chondroitin sulfate standard. Absorbance was measured at 530 nm with a wavelength correction set at 590nm using a Spectromax 300 plate reader (Molecular Devices). For samples containing media with 20 ug/ml CS (GLN + CS and IL-1 + GLN + CS), GAG content was subtracted with DMB readings from media measured only with 20 ug/ml CS. Total RNA isolation. Total RNA was extracted from cartilage explants following a modified protocol (34). Briefly, cartilage was homogenized in TRIzol® reagent (Invitrogen, Carlsbad, CA) and chloroform was added to extract total RNA followed by vigorous agitation and a 2-minute incubation. The aqueous phase containing RNA was collected after centrifugation (4°C, 12,000 X g, 15 minutes) and RNA precipitated with an equal volume of 70% ethanol. Total RNA was then purified further with RNeasy mini columns (Qiagen, Valencia, CA) and quantified by UV spectrophotometry (Beckman Coulter, Fullerton, CA). Total chondrocyte RNA was resolved on 1.2% agarose gel to validate spectrophotometric determination and RNA integrity. 128 cDNA synthesis. For each sample, 2 ug of total RNA was treated with DNase I (Invitrogen) to degrade contaminating single and double stranded DNA. Treated RNA was converted to single stranded cDNA using Superscript II reverse transcriptase (Invitrogen) as recommended by the manufacturer. Single stranded cDNA was quantified by UV spectrophotometry (Beckman Coulter), and diluted with RNase-free water to 10 ng/ul. Quantitative real-time polymerase chain reaction (Q-RT-PCR). Glyceraldehyde phosphate dehydrogenase (GAPDH) was validated as an appropriate housekeeping gene. Primers for GAPDH and molecules from Table I were designed using the Primer Express software version 2.0 (Perkin-Ehner Applied Biosystems, Foster City, CA). These molecules were chosen from other studies that have demonstrated significant induction with higher concentrations of IL-1 (25, 26). Nucleotide sequences used for primer design were obtained from public databases (Genbank). Optimal concentrations of each set of primers were determined with a primer matrix [lowest standard deviation with no change in cycle to threshold (C7)]. Quantitative real-time PCR was performed with 50 ng cDNA templates in 96-well plates (Perkin-Elmer Applied Biosystems) using the ABI PRISM 7000 sequence detection system (Perkin-Elmer Applied Biosystems) as previously described (25). The FBS control treatment was used as a calibrator (i.e. the fold change for control is 1.0). Replicated data was normalized with GAPDH and the fold change in gene expression relative to FBS control was calculated using the 2(-AACT) method (3 5). Protein extraction. Protein was extracted from cartilage explants using a modified protocol (36). Explants were rinsed with sterile phosphate buffer solutions 129 (PBS) and homogenized. Pulverized explants were placed in microcentrifuge tubes with stir bars and 10 ul extraction buffer (50 mmol/l Tris HCl, 10 mmol/l CaCl2, 2 mol/l guanidine HCl and 0.05% Brij-35, pH 7.5) per mg tissue. The mixture was stirred overnight at 4°C and then centrifuged at 18, 000 g for 30 minutes at 4°C. The supernatant was dialyzed for 24 hours against assay buffer (50 mmon Tris HCl, 10 mmol/l CaCl2, 0.2 mol/l NaCl and 0.05% Brij-35, pH 7.5) using Spectrapor 3 dialysis tubing with a 3.5- kd cutoff (Spectrum Medical Industries, Los Angeles, CA). Dialysis was continued for 24. hours with distilled water. Western blots. The amount of protein in the explant extract was quantified using the Pierce Micro BCA Protein Assay (Pierce, Rockford, IL) with bovine serum albumin as the standard. Protein extracts (80 rig/lane) were loaded on 12.5% sodium dodecyl sulfate (SDS)-polyacrylamide gels following denaturation by boiling for 5 minutes in SDS loading buffer and electrophoresed. Following electrophoresis, proteins were transferred to polyvinylidene difluoride membranes (Bio-Rad Laboratories, Hercules, CA). Protein transfer and size determinations were validated using prestained protein markers. Membranes were blocked with 5% nonfat dry milk in TTBS (lOmmol/l Tris- HCl, pH 8, 0.05% Tween-20, 150mmol/l NaCl) for 1 hour at room temperature and subsequently probed with antibodies directed against TIMP-3 (Chenricon International, Temecula, CA). Si gnals were detected using horseradish peroxidase-conjugated secondary antibodies and an enhanced chemiluminescence detection kit (ECL, Amersham Biosciences, Piscataway, NJ). Blots were developed (Futura Model B film processor), stripped (62.5 mmol/l Tris-HCl, pH 6.8, 100 mmol/l B-mercaptoethanol, and 2% SDS; 50°C for 30 minutes), and reprobed with anti-B-actin antibody (Abcam Inc., 130 Cambridge, MA). Films were scanned after development and the density of each band quantified using computer-aided densitometry. Amounts of TIMP-3 proteins were normalized relative to amounts of B-actin detected in each sample. Statistical analysis. Data for NO, PGE2, inhibition of NO and PGE2 (expressed as percentage inhibition relative to negative controls), and proteoglycan release into conditioned media were analyzed using a linear mixed effects model, including the fixed effect of concentration! treatment and the random effect of steer. Concentration/ treatment effects were compared within each time point using the multiple comparisons approach of Tukey. The computations were performed using the MD(ED procedure of SAS (3 7). Relative gene expression data determined using Q-RT-PCR and densitometry measurements from Western blots were analyzed using the nonparametric ANOVA approach of Friedman using SAS (3 7). Differences were declared statistically significant when P<0.05, unless otherwise noted. Spearman’s rank correlations (r) between Q-RT- PCR data and data for NO and PGE2 release were computed using the CORR procedure of SAS (37). RESULTS The effect of increasing IL-l concentration on NO and PGE2 release. There were dose dependent increases in NO and PGE2 release with increasing IL-l concentration. Nitrite release increased from 12.84 M to 65.95 M with increasing IL-l concentrations from 0 ng/ml to 50 ng/ml respectively (data not shown). The release of PGE2 increased from its basal level of 65.56 pg/ml in the absence of IL-1 to 1501.03 131 pg/ml with 50 ng/ml IL-l (Figure 1A). Based on these data, a subsaturating concentration of IL-1 (15 ng/ml) was selected for subsequent experiments. The effect of increasing GLN and CS concentrations on NO and PGE2 release. Increasing GLN concentration abrogated IL-l stimulated release of PGE2. The release of PGE2 was inhibited by about 36 %, 47 %, 49 % and 50 % with 1, 5, 10 and 20 ug/ml GLN respectively (F igurelB). Increasing CS concentration inhibited PGE2 release hour 18 %, 20 %, 30 % and 34 % with 5, 20, 50 and 100 ug/ml CS respectively (Figure 1B). There was no significant effect of GLN or CS at any concentration on NO (data not shown). Numerically, maximal reduction of NO occurred with 10 ug/ml GLN (16%) and 20 ug/ml CS (11%). Based on these results, we selected 10 ug/ml of GLN and 20 rig/ml of CS for the subsequent experiments. The GLN and CS combination suppressed inflammatory mediators. Interleukin-l stimulated increases in iNOS, COX—2, mPGEsl and NFKB transcripts at all time points were down-regulated by the GLN and CS combination (Figures 2A, 2B and 2C). Nitrite release was not different between treatments at 8 hour post-stimulation. Both NO and PGE2 release were significantly induced by IL-1 at 16 (N 0 only) and 24 hours post-culture. The elevation in these inflammatory mediators was effectively reduced by the GLN and CS combination (T able II). The GLN and CS combination repressed gene expression of matrix enzymes and reduced proteoglycan release. There was significant up-regulation of MMP-3 mRN A abundance by about 4 fold and 42 fold at 8 and 24 hours after IL-1 stimulation respectively. Glucosamine and CS suppressed IL-l induced expression of the MMP-3 gene by 35 fold at 24 hours. Cytokine induced mRNA expression of MMP-1 3, Agg-l and 132 Agg-2 were repressed at all time points by the combination (Figures 3A, 3B and 3C). Induction of proteoglycan release with IL-1 was significantly (P<0.01) reduced by the GLN and CS combination at both 24 and 48 hours post-stimulation (Figure 4A). The GLN and CS combination up-regulated TIMP-3. Gene expression of TIMP-3 was elevated with the GLN and CS combination relative to IL-1 at all time points and relative to FBS control at 8 and 24 hours post-stimulation (Figures 3A, 3B and 3C). TIMP-3 protein abundance was increased by the combination relative to IL-1 at 48 hours after culture (Figure 4B and 4C). DISCUSSION We performed the present study to expand on previous studies (25, 26) involving the effects of GLN and CS on the expression and synthesis of putative mediators of OA. Specifically, in this series of experiments, GLN and CS were employed at concentrations achievable in blood after oral and IV administration. Moreover, a subsaturating dose of the cytokine was used in this study to ensure the possibility of a two-tailed response to the arthritogenic stimulus and gene expression was examined at additional time points following stimulation and treatment. Pooling of animals in this experiment was avoided to prevent masking of outliers that may contribute to large variation in the results. Our results indicate that clinically relevant concentrations of this nutraceutical combination retain cartilage-sparing effects in this model. As expected IL-l significantly induced expression of the iN OS gene at all time points in the current study, an event that has been associated with cartilage degradation and suppression of ECM synthesis (3 8-40). Hence, limiting NO synthesis may be critical 133 to retarding OA progression and finds support in the documented beneficial effects of iNOS inhibitors in a canine OA model (36). In the present experiment, GLN and CS suppressed iNOS mRNA abundance and NO release. Individually, neither significantly decreased NO production, suggesting the two had a synergistic effect (5). At concentrations 10 fold or more than those employed in the present study, GLN and CS, alone or in combination are capable of repressing iNOS mRNA expression and reducing cytokine-induced release of NO (14, 15, 18-22, 27). The current study complements previous reports supporting co-administration of GLN and CS (5, 22). Prostaglandin E2 is the most abundant prostanoid found in diseased joints (41). Formed from the arachidonic acid cascade, PGE2 mediates synoviocyte proliferation, inflammatory and pain responses. Rate limiting enzymes responsible for making PGE2 include the cyclooxygenases and prostaglandin E synthases. The inducible forms of these enzymes are COX-2 and mPGEsl respectively. The synthesis of PGE2 correlates well with the elevation of COX-2 (r = 0.93, P<0.0002) and mPGEsl (r = 0.95, P<0.0001) transcripts in agreement with other studies (25, 42, 43). The concomitant regulation of COX-2, mPGEsl and PGE2 in the current study parallels other reports (25, 43). At biologically relevant concentrations, GLN and CS in combination effectively decreased IL-l induced gene expression of COX-2 and mPGEsl at all time points, and eventually PGE2 synthesis. A difference observed for 24 hours post-culture in the current study and in Chan et. al. (25) is that marginal reduction in mPGEsl transcript with GLN and CS is now significantly detected. These findings of modulation of iNOS, COX-2 and mPGEsl activities are in keeping with reductions in articular inflammation in OA patients and may help explain the analgesic effects of GLN and CS (4, 44). 134 The expression and activity of catabolic enzymes such as MMPs and aggrecanases exceeds those of endogenous inhibitors like TIMPs in OA (45). The ability of GLN and CS in combination to inhibit cartilage erosion (5) and prevent proteoglycan release in the current study (Figure 4) may be partially attributed to regulation of these enzymes. Concentrations of MMPs are elevated in OA cartilage specimens and they are localized at the site of OA lesions (46, 47). The nutraceutical combination demonstrated effectiveness in mitigating IL-l elevation of MMP-3 only at 24 hours post culture, consistent with a previous report where there was no treatment effect at the 6 hour time point (26). Past experiments with GLN, at higher concentrations, showed suppression of MMP-3 gene expression and enzyme activity (15, 17, 19, 48). Suppression of the MMP- 13 transcript with the combination at 24 hours also agrees with Chan et. al. (26). However, at a higher lL—l concentration, the ability of GLN and CS to reduce expression of the MMP-13 gene at 6 hours was not detected. In this study, induction of MMP-13 occurred at all time points and was suppressed at all time points by the combination. The findings on MMP-13 are also in agreement with other studies that reported a decrease in MMP-13 protein and activity in equine cartilage with the nutraceutical combination (22) and with GLN alone (17). Concentrations employed in the current study were at least 30 times lower for GLN and 6 times lower for CS than those reported in previous studies. The aggrecanases have been implicated as the primary proteins responsible for initiating aggrecan release from OA and injured joints (49). The present study demonstrated that GLN and CS in combination repressed IL-l up-regulation of Agg-l and Agg—2 at all time points. Our current findings roughly parallel those of Chan et. al. (26). The effect of GLN and CS in combination on aggrecanases has not been extensively 135 studied. Glucosamine supplemented alone suppressed IL-l stimulated aggrecanase activity at concentrations that were at least 40 times higher than the current study (28, 50). Chondroitin sulfate used individually in culture inhibited aggrecanase activity (51). The TIMPs have the potential to reduce proteoglycan destruction. Localized in the extracellular matrix, TIMP-3 exhibits potent inhibitory activities against MMPs and aggrecanases (52). Interleukin-l stimulated glycosaminoglycan release via aggrecanase was reversed by TIMP-3 (53). Our current study detected elevation in the TIMP-3 transcript with the combination relative to IL-1 throughout the 24 hour culture period. Simultaneous suppression of MMPs and aggrecanases coupled with up-regulation of TIMP-3 with GLN and CS supplementation may represent an effective way to protect matrix components fiom being degraded, as evidenced by the decline in proteoglycan release. Interleukin-l is upstream of the activation of a number of phosphorylation dependent signaling pathways leading to the nuclear translocation of transcription factors and activation of proteins participating in translation of mRNA. NFKB stimulates expression of iNOS and COX-2, and their end products, which contribute to the inflammatory process in arthritis (54). There was simultaneous expression of NFKB with iNOS (r = 0.48, P<0.01) and COX-2 (r = 0.55, P<0.003) in the present study. The down- regulation of certain MMPs with GLN and CS is perhaps also a downstream event of the repression of IL-1 signaling molecules. NFKB is one of two early response genes needed for MMP transcription (5 5). The activation of MMP-3 and MMP-13 relies on NFKB (56, 57). The mRNA expression of NFKB in the present study is highly correlated with MMP-13 (r = 0.76, P<0.0001) and IL-1 induction transcripts of both NFKB and MMP-13 136 increased significantly (P<0.05) with time of stimulation. Glucosamine, at 1 mg/ml, prevented IL-l induced IKB degradation, NFKB activation and nuclear translocation of p50 and p65 NFKB subunits and PGE2 release in human chondrocytes (13). In rat chondrocytes, GLN at concentration 450 times higher than the current study decreased NFKB activation (16). Our study shows the ability of GLN and CS in combination to repress IL-l stimulated mRNA abundance of the p65 NFKB subunit. This is essential since NFKB is able to regulate its own gene expression whereby IL-l induced increases in NFKB translocation to the nucleus stimulates continuous synthesis of NFKB to replace those that were translocated (5 8). Thus, the effect of GLN and CS on genes of iNOS, COX-2, and the MMPs could be explained at least, in part, via the inhibition of NF KB. Further studies are needed to substantiate the effect of these nutraceuticals on signaling events in the chondrocytes. Considering the adverse effects elicited by NSAIDs and COX-2 inhibitors, availability of other compounds that can help those suffering from joint pain is essential. The benefits of GLN and CS for symptomatic relief of OA have been clearly documented (1-4). These nutraceuticals are safer alternatives judging from the paucity of reports of adverse events (59). In vitro studies are revealing the mechanisms that underlie the in vivo effects of GLN and CS. From the present study, suppression by GLN and CS in combination of NO and PGE2 production may partially contribute to the anti- inflammatory effects experienced by CA patients consuming these compounds. The purported cartilage protective feature of GLN and CS (5) and the ability of these nutraceuticals to prevent IL-l induced proteoglycan degradation, and decrease proteoglycan synthesis (23, 24) is attributed, at least in part, to suppression of catabolic 137 matrix enzymes and up-regulation of TIMP-3, an important enzyme inhibitor. GLN and CS may regulate signaling pathways upstream of the production of inflammatory mediators and matrix enzymes, which translates into beneficial effects thereafter. Glucosamine and CS may also impact positively the interactions that occur between all these molecules that are associated with CA pathogenesis. Further experiments are being planned to confirm these findings in vivo. 138 0HOHHOH0<0<00H<00000 <0H0m 508500 083.85 023003.. 058500 0000 0000005 530 03508325 080-305 030.8055 5.5 00.5 305008 .5 30% mo 09!.0 3000503 5055.5 02055 058 083.85 ”5 030.5. 139 Table II: Nitrite and PGE2 release from explants at 8, 16 and 24 hours post-stimulation Treatment Variable FBS control IL—1 IL-1 + GLN + CS Nitrite release (uM t SE) 1.80 i 0.37‘1 7.67 i- 1.251‘ 5.82 5: 0.44‘ll at 8 hours post-stimulation Nitrite release (uM i SE) 5.56 i 0.99a 28.17 i 386" 15.61 i 1.873 at 16 hours post- stimulation Nitrite release (uM 3: SE) 8.04 i 0.94“ 52.60 i 555° 22.54 i 2.315 at 24 hours post- stimulation PGE2 release (pg/ml t SE) 70.03 i 9.113 1159.16 i 248.09c 675.29 1 106.84” at 24 hours post- stimulation F BS = fetal bovine serum; IL-1 = 15 11ng human recombinant interleukin-1 beta (rhIL- IB); IL-l + GLN + CS = 15 ng/ml rhIL-IB with the addition of 10 pig/ml glucosamine and 20 ug/ml chondroitin sulfate; PGE2 = prostaglandin E2 Different superscripts for values within a row (i.e. one variable) denote significant (P<0.05) differences between treatments. 140 Figure l: Prostaglandin E2 (PGE2) release 3: SE in conditioned media with increasing interleukin—1 (IL-1) concentration (A). Percentage inhibition of PGE2 release i SE from 0 ug/ml glucosamine (GLN) or chondroitin sulfate (CS) in conditioned media with 15 ng/ml IL-1 and increasing GLN or CS concentration (B). Different letters for values within each line indicate significant (P<0.05) differences between concentrations of IL-1, GLN or CS. 141 PGEZ (pg/ml) Inhibition of PGE2 (%) 20001 b b 1500- 1000- 5004 o I I I I o 5 1o 20 so lL-1 concentration (nglml) B Chondroitin sulfate concentration (uglml) 5 20 50 100 so : : : : 4 1’ W 60 b T I) ab 40" ll .fl- ' - - ?L 204 ,- _ - . . u l. ' t J. b b ab ab 0 4 : : : 4 0 1 5 10 20 Glucosamine concentration (uglml) -°-Gluoosamine - 0- Chondroitin sulfate 142 Figure 2: Mean relative gene expression i SE of inflammatory mediators at 8 hours (A), 16 hours (B) and 24 hours (C) post-stimulation. Different letters for values within each gene indicate significant (P<0.05) differences between treatments. iNOS = inducible nitric oxide synthase; COX-2 = cyclooxygenase-2; mPGEsl = microsomal prostaglandin E synthase-1;NFKB = nuclear factor kappa beta p65 subunit; F BS = fetal bovine serum; IL-1 = 15 ng/ml human recombinant interleukin-1 beta (rhlL-IB); IL-l + GLN + CS = 15 ng/ml rhIL-lfi with the addition of 10 ug/ml glucosamine and 20 ug/ml chondroitin sulfate. 143 w m m w 5.3298 023.8 cues. COX-2 mPGEs1 NFKB iNOS cofinoaxo «>532 cams. COX-2 mPGEs‘I NFKB iNOS 5.3298 3:52 :32 NFKB mPGEs1 INOS Gene [:1 FBS control I lL-1 l lL-1 + GLN + CS 144 Figure 3: Mean relative gene expression 1: SE of enzymes at 8 hours (A), 16 hours (B) and 24 hours (C) post-stimulation. Different letters for values within each gene indicate significant (P<0.05) differences between treatments. MMP-3 = matrix metalloproteinase- 3; MMP-13 = matrix metalloproteinase-13; Agg—l = aggrecanase-1; Agg-2 = aggrecanase-2; TIMP-3 = tissue inhibitor of metalloproteinase-3; F BS = fetal bovine serum; IL-1 = 15 ng/ml human recombinant interleukin-1 beta (rhIL-l [3); IL—1 + GLN + CS = 15 ng/ml rhIL-l B with the addition of 10 ug/ml glucosamine and 20 ug/ml chondroitin sulfate. 145 Mean relative expression Mean relative expression n. Mean relative expression J; I G MMP-3 MMP-13 A994 A994 TIMN MMPJ MMP-13 Agg-z TIMP-3 _ a: TIMP-3 MMP-3 MMP-13 Agg-1 A99-2 Gene UFBS control I lL-1 a IL-1 + GLN + CS 146 Figure 4: Proteoglycan release i SE in conditioned media at 24 and 48 hours (A) post- stimulation. Different letters for values within each time point indicate significant (P<0.01) differences between treatments. Representative western blots of TIMP-3 and B- actin protein expression at 48 hours post-culture (B). Mean relative abundance of TIMP-3 proteins 1: SE, as determined by densitometry (C). Different letters indicate significant (P<0.05) differences between treatments. (1) FBS = fetal bovine serum; (2) IL-1 = 15 ng/ml human recombinant interleukin-1 beta (thL-l B); (3) GLN + CS = 10 ug/ml glucosamine and 20 ug/ml chondroitin sulfate; (4) IL-1 + GLN + CS = 15 ng/ml rhIL-l B with the addition of 10 ug/ml glucosamine and 20 rig/ml chondroitin sulfate. 147 £250 - c E O 3200 - d) m ‘8 7d 150 - C 8 2.100 4 U) o 8 2 5° ‘ m o . El FBS control B 46 kDa 42 kDa C .° 9 9 f“ . at O Q n l I a Relative density (TlMP-3Iactin) G 'u P O L D FBS control 24 48 Time post-stimulation (h) I lL-1 El GLN + CS [3 lL-1 + GLN + C8 —> - ~I... B—actin I lL-1 ElGLN + CS ElL-1 + GLN + CS 148 REFERENCES 1. Reginster JY, Deroisy R, Rovati LC, Lee RL, Lejeune E, Bruyere O, et al. Long-term effects of glucosamine sulphate on osteoarthritis progression: a randomised, placebo- controlled clinical trial. Lancet 2001; 357:251-6. 2. Uebelhart D, Thonar EJ, Delmas PD, Chantraine A, Vignon E. Effects of oral chondroitin sulfate on the progression of knee osteoarthritis: a pilot study. Osteoarthritis Cart 1998; 6 Suppl A:39-46. 3. Rovetta G, Monteforte P, Molfetta G, Balestra V. Chondroitin sulfate in erosive osteoarthritis of the hands. Int J Tissue React 2002; 24:29-32. 4. Leffler CT, Philippi AF, Leffler SG, Mosure JC, Kim PD. Glucosamine, chondroitin, and manganese ascorbate for degenerative joint disease of the knee or low back: a randomized, double-blind, placebo-controlled pilot study. Mil Med 1999; 164:85-91. 5. Lippiello L, Woodward J, Karpman R, Hammad TA. In vivo chondroprotection and metabolic synergy of glucosamine and chondroitin sulfate. Clin Orthop 2000; 381:229- 40. 6. Adebowale A, Du J, Liang Z, Leslie J L, Eddington ND. The bioavailability and pharmacokinetics of glucosamine hydrochloride and low molecular weight chondroitin sulfate after single and multiple doses to beagle dogs. Biopharm Drug Dispos 2002; 23:217-25. 7. Laverty S, Sandy JD, Celeste C, Vachon P, Marier JF, Plass AH. Synovial fluid levels and serum pharmacokinetics in a large animal model following treatment with oral glucosamineat clinically relevant doses. Arthritis Rheum 2005; 52:181-91. 8. Du J, White N, Eddington ND. The bioavailability and pharmacokinetics of glucosamine hydrochloride and chondroitin sulfate after oral and intravenous single dose administration in the horse. Biopharm Drug Dispos 2004; 25:109-16. 9. Setnikar I, Palumbo R, Canali S, Zanolo G. Pharmacokinetics of glucosamine in man. Arzneimittelforschung 1993; 43:1 109-13. 10. Liang Z, Leslie J, Adebowale A, Ashraf M, Eddington ND. Determination of the nutraceutical, glucosamine hydrochloride, in raw materials, dosage forms and plasma using pre-column derivatization with ultraviolet HPLC. J Pharm Biomed Anal 1999; 20:807-14. 11. Du J, Eddington N. Determination of the chondroitin sulfate disaccharides in dog and horse plasma by HPLC using chondroitinase digestion, precolumn derivatization, and fluorescence detection. Anal Biochem 2002; 306:252-8. 149 12. Volpi N. Oral absorption and bioavailability of ichthyic origin chondroitin sulfate in healthy male volunteers. Osteoarthritis Cart 2003; 11:433-41. 13. Largo R, Alvarez-Sofia MA, Diez-Ortego I, Calvo E, Sénchez-Pemaute O, Egido J, et al. Glucosamine inhibits IL-lbeta-induced NF kappaB activation in human osteoarthritic chondrocytes. Osteoarthritis Cart 2003; 11:290-8. l4. Fenton JI, Chlebek-Brown KA, Peters TL, Caron JP, Orth MW. Glucosamine HCl reduces equine articular cartilage degradation in explant culture. Osteoarthritis Cart 2000; 8:258-65. 15. Fenton JI, Chlebek-Brown KA, Caron JP, Orth MW. Effect of glucosamine on interleukin-l-conditioned articular cartilage. Equine Vet J Suppl 2002; 34:219-23. 16. Gouze JN, Bianchi A, Becuwe P, Dauca M, Netter P, Magdalou J, et al. Glucosamine modulates IL-l-induced activation of rat chondrocytes at a receptor level, and by inhibiting the NF-kappa B pathway. FEBS Lett 2002; 510:166-70. 17. Byron CR, Orth MW, Venta PJ, Lloyd J W, Caron JP. Influence of glucosamine on matrix metalloproteinase expression and activity in lipopolysaccharide-stimulated equine chondrocytes. Am J Vet Res 2003; 64:666-71. 18. Meininger CJ, Kelly KA, Li H, Haynes TE, Wu G. Glucosamine inhibits inducible nitric oxide synthesis. Biochem Biophys Res Commun 2000; 279:234-9. l9. Nakamura H, Shibakawa A, Tanaka M, Kato T, Nishioka K. Effects of glucosamine hydrochloride on the production of prostaglandin E2, nitric oxide and metalloproteases by chondrocytes and synoviocytes in osteoarthritis. Clin Exp Rheumatol 2004; 22:293-9. 20. Mello DM, Nielsen BD, Peters TL, Caron JP, Orth MW. Comparison of inhibitory effects of glucosamine and mannosamine on bovine articular cartilage degradation in vitro. Am J Vet Res 2004; 65: 1440-5. 21. Schlueter AE, Orth MW, Further studies on the ability of glucosamine and chondroitin sulphate to regulate catabolic mediators in vitro. Equine Vet J 2004; 36:634- 6. 22. Orth MW, Peters TL, Hawkins JN. Inhibition of articular cartilage degradation by glucosamine-HCl and chondroitin sulphate. Equine Vet J Suppl 2002; 34:224-9. 23. Bassleer C, Rovati L, Franchimont P. Stimulation of proteoglycan production by glucosamine sulfate in chondrocytes isolated from human osteoarthritic articular cartilage in vitro. Osteoarthritis Cart 1998; 6:427-34. 150 24. Lippiello L. Glucosamine and chondroitin sulfate: biological response modifiers of chondrocytes under simulated conditions of joint stress. Osteoarthritis Cart 2003; 11:335- 42. 25. Chan PS, Caron JP, Rosa GJ M, Orth MW. Glucosamine and chondroitin sulfate regulate gene expression and synthesis of nitric oxide and prostaglandin E2 in articular cartilage explants. Osteoarthritis Cart 2005; 13:3 86-93. 26. Chan PS, Caron JP, Orth MW. Glucosamine and chondroitin sulfate regulate gene expression of proteolytic enzymes and their inhibitors in interleukin-l induced bovine articular cartilage explants. Am J Vet Res 2005; in press 27. Shikhman AR, Kuhn K, Alaaeddine N, Lotz M. N-acetylglucosamine prevents IL-l beta-mediated activation of human chondrocytes. J Immunol 2001; 166:5155-60. 28. Sandy JD, Gamett D, Thompson V, Verscharen C. Chondrocyte-mediated catabolism of aggrecan: aggrecanase-dependent cleavage induced by interleukin-1 or retinoic acid can be inhibited by glucosamine. Biochem J 1998; 335:59-66. 29. Dodge GR, Jimenez SA. Glucosamine sulfate modulates the levels of aggrecan and matrix metalloproteinase-3 synthesized by cultured human osteoarthritis articular chondrocytes. Osteoarthritis Cart 2003; 11:424-32. 30. Rosselot G, Reginato AM, Leach RM. Development of a serum-free system to study the effect of growth hormone and insulinlike growth factor-I on cultured postembryonic growth plate chondrocytes. In Vitro Cell Dev Biol 1992; 28A:235-44. 31. Volpi N. Oral bioavailability of chondroitin sulfate (Condrosulf) and its constituents in healthy male volunteers. Osteoarthritis Cart 2002; 10:768-77. 32. Blanco FJ, Ochs RL, Schwarz H, Lotz M. Chondrocyte apoptosis induced by nitric oxide. Am J Pathol 1995; 146:75-85. 33. Chandrasekhar S, Esterman MA, Hoffman HA. Microdetermination of proteoglycans and glycosaminoglycans in the presence of guanidine hydrochloride. Anal Biochem 1987; 161:103-8. 34. Reno C, Marchuk L, Sciore P, Frank CB, Hart DA. Rapid isolation of total RNA from small samples of hypocellular, dense connective tissues. Biotechniques 1997; 22: 1082-6. 35 . Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001; 25:402-8. 36. Pelletier JP, Jovanovic DV, Lascau-Coman V, Femandes JC, Manning PT, Connor JR, et al. Selective inhibition of inducible nitric oxide synthase reduces progression of 151 experimental osteoarthritis in vivo: possible link with the reduction in chondrocyte apoptosis and caspase 3 level. Arthritis Rheum 2000; 43: 1290-9. 37. SAS, SAS/STAT User's Guide Version 8.2, ed., SAS Institute, Inc., Cary, NC 2001. 38. Evans CH. Nitric oxide: what role does it play in inflammation and tissue destruction?, Agents Actions Suppl 1995; 47:107-16. 39. Murrell GA, Jang D, Williams RJ. Nitric oxide activates metalloprotease enzymes in articular cartilage. Biochem Biophys Res Commun 1995; 206:15-21. 40. Taskiran D, Stefanovic-Racic M, Georgescu H, Evans C. Nitric oxide mediates suppression of cartilage proteoglycan synthesis by interleukin-1. Biochem Biophys Res Commun 1994; 200:142-8. 41. Kirker-Head CA, Chandna VK, Agarwal RK, Morris EA, Tidwell A, O'Callaghan MW, et al. Concentrations of substance P and prostaglandin E2 in synovial fluid of normal and abnormal joints of horses. Am J Vet Res 2000; 61 :714-8. 42. Murakami M, Naraba H, Tanioka T, Semmyo N, Nakatani Y, Kojima F, et al. Regulation of prostaglandin E2 biosynthesis by inducible membrane-associated prostaglandin E2 synthase that acts in concert with cyclooxygenase-2. J Biol Chem 2000; 275:32783-92. 43. Stichtenoth DO, Thorén S, Bian H, Peters-Golden M, Jakobsson PJ, Crofford LJ. Microsomal prostaglandin E synthase is regulated by proinflammatory cytokines and glucocorticoids in primary rheumatoid synovial cells. J Immunol 2001; 167:469-74. 44. Das A, J r., Hammad TA. Efficacy of a combination of FCHG49 glucosamine hydrochloride, TRHl 22 low molecular weight sodium chondroitin sulfate and manganese ascorbate in the management of knee osteoarthritis. Osteoarthritis Cart 2000; 8:343-50. 45. Dean DD, Martel-Pelletier J, Pelletier JP, Howell DS, Woessner JF, Jr. Evidence for metalloproteinase and metalloproteinase inhibitor imbalance in human osteoarthritic cartilage. J Clin Invest 1989; 84:678-85. 46. Dean DD. Proteinase-mediated cartilage degradation in osteoarthritis. Sem Arthritis and Rheum 1991; 20:2-11. 47. Tetlow LC, Adlam DJ, Woolley DE. Matrix metalloproteinase and proinflammatory cytokine production by chondrocytes of human osteoarthritic cartilage: associations with degenerative changes. Arthritis Rheum 2001; 44:585-94. 48. Gouze JN, Bordji K, Gulberti S, Terlain B, Netter P, Magdalou J, et al. Interleukin- lbeta down-regulates the expression of glucuronosyltransferase I, a key enzyme priming 152 glycosaminoglycan biosynthesis: influence of glucosamine on interleukin-1beta-mediated effects in rat chondrocytes. Arthritis Rheum 2001; 44:351-60. 49. Lohmander LS, Neame PJ, Sandy JD. The structure of aggrecan fragments in human synovial fluid. Evidence that aggrecanase mediates cartilage degradation in inflammatory joint disease, joint injury, and osteoarthritis. Arthritis Rheum 1993; 36:1214-22. 50. Ilic MZ, Martinac B, Handley CJ. Effects of long-term exposure to glucosamine and mannosamine on aggrecan degradation in articular cartilage. Osteoarthritis Cart 2003; 11:613-22. 51. Sugimoto K, Takahashi M, Yamamoto Y, Shimada K, Tanzawa K. Identification of aggrecanase activity in medium of cartilage culture. Tokyo 1999; 126:449-55. 52. Kashiwagi M, Tortorella M, Nagase H, Brew K. TIMP-3 is a potent inhibitor of aggrecanase l (ADAM-T84) and aggrecanase 2 (ADAM-T85). J Biol Chem 2001; 276:12501-4. 53. Gendron C, Kashiwagi M, Hughes C, Caterson B, Nagase H. TIMP-3 inhibits aggrecanase-mediated glycosaminoglycan release from cartilage explants stimulated by catabolic factors. F EBS Lett 2003; 555:431-6. 54. Yamamoto Y, Gaynor RB. Therapeutic potential of inhibition of the NF-kappaB pathway in the treatment of inflammation and cancer. J Clin Invest 2001; 107:135-42. 55. Vincenti MP, Brinckerhoff CE. Transcriptional regulation of collagenase (MMP-1, MMP-l 3) genes in arthritis: integration of complex signaling pathways for the recruitment of gene-specific transcription factors. Arthritis Res 2002; 4:157-64. 56. Liacini A, Sylvester J, Li WQ, Zafarullah M. Inhibition of interleukin-l-stimulated MAP kinases, activating protein-1 (AP-1) and nuclear factor kappa B (N F-kappa B) transcription factors down-regulates matrix metalloproteinase gene expression in articular chondrocytes. Matrix Biol 2002; 21 :251-62. 57. Mengshol JA, Vincenti MP, Coon CI, Barchowsky A, Brinckerhofl~ CE. Interleukin-1 induction of collagenase 3 (matrix metalloproteinase 13) gene expression in chondrocytes requires p3 8, c-Jun N-terminal kinase, and nuclear factor kappaB: differential regulation of collagenase 1 and collagenase 3. Arthritis Rheum 2000; 43:801-11. 58. Agarwal S, Deschner J, Long P, Verma A, Hofman C, Evans CH, et al. Role of NF- kappaB transcription factors in antiinflammatory and proinflammatory actions of mechanical signals. Arthritis Rheum 2004; 50:3541-8. 59. Jordan KM, Arden NK, Doherty M, Bannwarth B, Bijlsma JW, Dieppe P, et al. EULAR Recommendations 2003: an evidence based approach to the management of knee osteoarthritis: Report of a Task Force of the Standing Committee for International 153 Clinical Studies Including Therapeutic Trials (ESCISIT). Ann Rheum Dis 2003; 62:1145-55. 154 CONCLUSION Previous research findings with glucosamine and CS in our laboratory and others provided some clues about the mechanism(s) of action for these nutraceuticals although the concentrations used were far from relevant. Most of these studies were conducted with glucosamine and very few with CS and the majority mainly assessed biochemical parameters including NO, PGE2, and proteoglycan synthesis. Only a handful have looked at enzymatic activity. Data from these studies point to the possibility of cartilage-sparing properties and suppression of molecules that participate in the inflammation process. The present research was targeted at expanding on previous results using concentrations of glucosamine and CS that are achievable in vivo in the blood of humans, dogs, and horses after oral administration of these nutraceuticals. Specifically, the mRN A abundance of an array of proteolytic enzymes and inflammatory mediators were determined since iNOS, COX-2, and several MMPs in particular are regulated at the transcriptional level. Moreover, the effects of these nutraceuticals on chondrocyte gene expression have rarely been studied. Current research with biologically relevant concentrations of glucosamine (5 rig/ml) and CS (20 ug/ml) demonstrated their ability of pre-translational regulation of genes encoding inflammatory mediators and cartilage matrix catabolic enzymes. Using an lL-l concentration (50 ng/ml) reported in previous trials in our laboratory to stimulate cartilage catabolism, explants supplemented with glucosamine and CS showed transient suppression of molecules implicated in the progression of OA such as NO and genes of iNOS, mPGEsl and MMPs. Glucosamine and CS down-regulation of COX-2 was 155 accompanied by reductions in PGE2 production. The ability of these nutraceuticals to suppress NO and PGE2 is critical since both inflammatory mediators facilitate cartilage degradation. Both NO and PGE2 are important regulatory molecules of MMPs. NO and PGE2 also interact with one another. Studies have indicated that an NO-dependent pathway mediates IL-l stimulation and inhibition of PGE2 and the opposite may also be true. More studies are needed to determine how these two molecules affect each other in the process of inflammation and cartilage destruction. The mRNA abundance of enzymes (iNOS, COX-2 and mPGEsl) that catalyze the production of these inflammatory mediators serves as another point of regulation affected by glucosamine and CS. Researchers in the field of cartilage have just started studying mPGEsl since it was only recently detected in chondrocytes. The ability of glucosamine and CS to affect mPGEsl is pertinent since it is the terminal enzyme involved in PGE2 synthesis. The positive effect of glucosamine and CS on MMPs and aggrecanases at the gene level is important information. MMPs are the primary enzymes responsible for ECM remodeling and found in elevated concentrations in the sites of OA lesions while aggrecanases participate in pathological turnover of aggrecan. Moreover, the majority of studies with biologically irrelevant concentrations of glucosamine and CS have only assessed general activities of these enzymes. Lack of treatment effect on genes of matrix components; type II collagen and aggrecan keeps up with current literature on the possibility that glucosamine and CS may exert their effects on matrix constituents at a translational and post-translational level. F urthennore, the synthesis of these macromolecules is catalyzed by a number of enzymes that have yet to be investigated. The nutraceutical combination antagonized some gene expression induced by IL- 156 1 better than when they were used individually. This finding is in support of past studies that reported enhanced symptomatic efficacy in glucosamine and CS when taken together. We are confident that these responses are highly specific to the nutraceuticals added in culture since previous research in our laboratory with glucosamine has proven that its effects on the release of inflammatory mediators is specific and not due to an artifact of high sugar moieties in the culture media. Given that most in vitro studies that used IL-l as a stimulant on cartilage explants employ it at a much lower concentration than what was used in chapters 2 and 3, detection in modulation of genes especially those with subtle changes was probably limited by this factor. Another limitation was attributed to pooling of animals for tissue sampling that caused much variability in the results. Preliminary studies also demonstrated that chondrocytes become refiactory to IL-1 after 48 hours. Thus, a subsaturating dose of IL-1 (15 ng/ml) with cartilage explants from individual animals was used for a separate study with the duration of regulation characterized within 24 hours. The data from this research provided further evidence for the proposed mechanism(s) of action for glucosamine (10 rig/ml) and CS (20 uglml). Figure 1 depicts a summary of this evidence. Glucosamine and CS in combination effectively repressed genes of selected molecules associated with inflammation and cartilage degradation. The NFKB p65 subunit that is downstream of IL-1 signaling and an important factor driving transcription of most of the selected genes in this study was negatively regulated by glucosamine and CS. This is an interesting finding as NF KB regulates its own expression. Decreasing its mRN A abundance will probably interrupt transcription of some of the critical mediators 157 of OA, in particular iNOS, COX-2, MMP-3 and MMP-13. Future studies focusing on the effects of the nutraceuticals on nuclear translocation and activation of this signaling pathway are warranted. Some research suggests that glucosamine may regulate proteins such as the NF KB via the glycosylation process. Glycosylation may offset the ability of the transcription factors to be phosphorylated, preventing their translocation to the nucleus to affect transcription of other genes containing enhancer elements in their promoter regions. This postulation remains to be proven in chondrocytes. Other studies reported that glucosamine and CS are beneficial for CA patients by providing building blocks for cartilage matrix synthesis. Our research suggests that these nutraceuticals do more than just serve as precursors for matrix synthesis. The present research demonstrated the ability of these nutraceuticals to regulate mediators of OA at the pre-translational level, decreasing mRNA abundance of iNOS, COX-2, mPGEsl , NF KB, MMP-3, MMP-13, Agg-l and Agg-2. Repression of most of these genes may be mediated by inhibition of NF KB. Another novel finding of this research pertains to up- regulation of mRNA and protein abundance of TIMP-3. Since TIMP-3 has potent affinity for many of the matrix proteolytic enzymes, the ability of glucosamine and CS to elevate it is important. Moreover, these effects on the putative mediators of OA are coupled with a reduction in proteoglycan loss and, production of NO and PGE2. Hence, these research findings may explain, in part, the possible mechanisms for mild anti-inflammatory and chondroprotective features of glucosamine and CS. Further studies with the nutraceuticals should determine in depth how the selected OA mediators are regulated. The drawback to the present study is the fact that only abundance of genes was measured although a few biochemical variables were included, 158 and they did match up well with the gene data. Future studies should investigate the effects of glucosamine and CS on rates of transcription and translation, and the stability of the transcript and protein. Parameters such as enzymatic activities, binding and substrate assays would be useful to study post-transcriptional regulation. Studies are also needed to determine how and if CS accesses the chondrocytes. The anabolic aspects affected by glucosamine and CS should be considered as well by exploring molecules such as the grth factors; TGFB, lGF-l , and osteogenic protein-l , recently implicated to mediate proteoglycan synthesis. Other variables associated with pain such as bradykinin, and neurotransmitters including serotonin and dopamine should also be investigated. Bradykinin is capable of inducing proteoglycan degradation. A comparison in responses of normal and healthy cartilage with CA cartilage to glucosamine and CS will also be interesting. The concentrations of glucosamine and CS chosen should be closer to those in the synovial fluid as more pharmacokinetic and pharmacodynamic studies become available reporting on the concentrations of the nutraceuticals measured in the synovial fluid after oral and intravenous administration. Since cartilage does not exist in isolation, these in vitro findings would be more convincing when confirmed in viva using for example, a rabbit instability model or some other model of OA. Overall, results from the present research complement those found in clinical trials. Studies in vivo with animals and clinical trials with humans have reported on the ability of glucosamine and CS to attenuate cartilage loss. Current findings with relevant concentrations of the nutraceuticals demonstrated an association between reduced mRNA abundance of catabolic enzymes and up—regulation of an important enzyme inhibitor with 159 the inhibition of proteoglycan loss. Reductions in pain assessments also coincide with suppression of inflammatory mediators measured in the present experiments. This finding is useful information for physicians and veterinarians in their consideration whether to recommend or not recommend these nutraceuticals to their patients. No longer can these compounds be dismissed because their concentrations investigated in vitro are irrelevant. Furthermore, millions of people suffer from OA and with the recent controversy surrounding COX-2 inhibitors, the availability of safer alternatives such as glucosamine and CS may be critical to a patient’s health and well-being. However, these nutraceuticals are no “magic bullet”. Clinical trials have shown that they appear only to be beneficial to those with mild to moderate OA. Until more studies are perform, we cannot say definitively that glucosamine and CS is the answer to treating OA. Nevertheless, the present research does provide additional information pointing to possible mechanism(s) of action of glucosamine and CS that could explain the results seen from clinical trials. 160 Figure 1: Proposed mechanism(s) of action for glucosamine and CS as indicated by results from the present in vitro experiments. X indicates suppression by the glucosamine and CS combination while / indicates up-regulation by the glucosamine and CS combination. 161 162 Millillillliiillli11111111 1293 02736 1280