.. Modèles-expérimentaux-d-'arthrose, 15 a. Modèle par section du ligament croisé antérieur, p.17

.. Thérapie-génique-et-articulation, A. Pourquoi-utiliser-la-thérapie-génique, and ?. , 20 C. Les différentes méthodes de transfert de gènes, Electroporation, vol.21, p.23

M. , .. Au, and M. , 36 a. Modèle expérimental d'arthrose, Mycobactéries, p.37

.. Mode-de-transfection, 38 a. Injection simple, Polyplexes : in vivo jetPEI?, vol.38, p.39

.. Vérification-du-marquage-de-la-sonde, 43 a. Par retard sur gel 43 b. Par immunodétection via un, Anticorp anti-DIG, p.43

I. Culture-cellulaire and .. , 46 A. Culture de chondrocytes et synoviocytes de rat, p.46

A. Vérification-de-la-qualité-des and .. , 55 a. Dosage spectrophotométrique 55 b. Migration sur gel d'agarose, p.55

.. Électriques-par-le-cliniporator, 74 A. Applications des impulsions sur le muscle squelettique de rat, p.75

H. , A. Aspect-général, and .. , 76 II. Applications des impulsions sur cartilage articulaire d'animaux sains

.. Vérification-du-marquage-de-la-sonde, 87 a. Par la technique de retard sur gel, p.88

.. Localisation-du-plasmide-pcdna-eyfp-par-hybridation-in-situ, 89 III. Applications des impulsions sur cartilage articulaire d'animaux pathologiques, p.90

C. Du, M. Dans-du, D. , and .. , 105 a, p.106
URL : https://hal.archives-ouvertes.fr/in2p3-00003985

.. Le and M. , 119 B. Effet de l'Induction d'Hsps par, p.119

M. Hsp70-par-le, 120 a. Vérification de l'innocuité d'une injection intra-articulaire de MG132 par semaine pendant 1 mois, p.121

J. Adams, M. Behnke, S. Chen, A. A. Cruickshank, L. R. Dick et al., Potent and selective inhibitors of the, 1998.

H. Nagase and M. Kashiwagi, Aggrecanases and cartilage matrix degradation, Arthritis Research & Therapy, vol.5, issue.2, pp.94-103, 2003.
DOI : 10.1186/ar630

E. Neumann, M. Schaefer-ridder, Y. Wang, and P. H. Hofschneider, Gene transfer into mouse lyoma cells by electroporation in high electric fields, Embo J, vol.1, pp.841-846, 1982.

S. Ohashi, T. Kubo, T. Kishida, T. Ikeda, K. Takahashi et al., Successful genetic transduction in vivo into synovium by means of electroporation, Biochemical and Biophysical Research Communications, vol.293, issue.5, pp.1530-1535, 2002.
DOI : 10.1016/S0006-291X(02)00386-8

S. Orlowski, J. Belehradek, . Jr, C. Paoletti, and L. M. Mir, Transient electropermeabilization of cells in culture, Biochemical Pharmacology, vol.37, issue.24, pp.4727-4760, 1988.
DOI : 10.1016/0006-2952(88)90344-9

T. Pazmany, S. P. Murphy, S. O. Gollnick, S. P. Brooks, and T. B. Tomasi, Activation of Multiple Transcription Factors and fos and jun Gene Family Expression in Cells Exposed to a Single Electric Pulse, Experimental Cell Research, vol.221, issue.1, pp.103-113, 1995.
DOI : 10.1006/excr.1995.1357

C. M. Pearson, Development of Arthritis, Periarthritis and Periostitis in Rats Given Adjuvants., Experimental Biology and Medicine, vol.91, issue.1, pp.95-101, 1956.
DOI : 10.3181/00379727-91-22179

B. Petit, M. C. Ronzière, D. J. Hartmann, and D. Herbage, Ultrastructural organization of type XI collagen in fetal bovine epiphyseal cartilage, Histochemistry, vol.265, issue.27, pp.231-240, 1993.
DOI : 10.1007/BF00269096

URL : https://hal.archives-ouvertes.fr/hal-00313334

M. J. Pond and G. Nuki, Experimentally-induced osteoarthritis in the dog., Annals of the Rheumatic Diseases, vol.32, issue.4, pp.387-395, 1973.
DOI : 10.1136/ard.32.4.387

K. P. Pritzker, Animal models for osteoarthritis: processes, problems and prospects., Annals of the Rheumatic Diseases, vol.53, issue.6, pp.406-426, 1994.
DOI : 10.1136/ard.53.6.406

S. Rajdev and F. R. Sharp, Stress Proteins as Molecular Markers of Neurotoxicity, Toxicologic Pathology, vol.30, issue.1, pp.105-117, 2000.
DOI : 10.1002/ana.410440403

A. D. Rowan, Cartilage catabolism in arthritis: factors that influence homeostasis, Expert Reviews in Molecular Medicine, vol.3, issue.17, pp.1-20, 2001.
DOI : 10.1017/S1462399401003209

J. Sambrook, E. F. Fritsch, and T. Maniatis, Molecular cloning : a laboratory manual, 1989.

P. Sarzi-puttini, M. A. Cimmino, R. Scarpa, R. Caporali, F. Parazzini et al., Osteoarthritis: An Overview of the Disease and Its Treatment Strategies, Seminars in Arthritis and Rheumatism, vol.35, issue.1, pp.1-10, 2005.
DOI : 10.1016/j.semarthrit.2005.01.013

M. P. Seed, F. L. Parker, S. Johns, A. P. Curnock, A. Bowden et al., Mycobacterium tuberculosis -induced monoarticular arthritis in the rat, a new in vivo model for the assessment of anti-rheumatic drugs, Clinical Rheumatology, vol.10, pp.461-462, 1991.

R. Stoop, P. M. Van-der-kraan, P. Buma, A. P. Hollander, R. C. Billinghurst et al., Type II collagen degradation in spontaneous osteoarthritis in C57BL/6 and BALB/c mice, Arthritis & Rheumatism, vol.331, issue.11, pp.2381-2390, 1999.
DOI : 10.1042/bj3310727

V. Strand, R. Kimberly, and J. D. Isaacs, Biologic therapies in rheumatology: lessons learned, future directions, Nature Reviews Drug Discovery, vol.256, issue.Suppl. 2, pp.75-92, 2007.
DOI : 10.1038/256495a0

S. Takayama and J. C. Reed, Molecular chaperone targeting and regulation by BAG family proteins, Nature Cell Biology, vol.275, issue.10, pp.237-278, 2001.
DOI : 10.1126/science.275.5302.983

R. M. Tanguay, Y. Wu, and E. W. Khandjian, Tissue-specific expression of heat shock proteins of the mouse in the absence of stress, Developmental Genetics, vol.32, issue.2, pp.112-120, 1993.
DOI : 10.1128/MCB.8.7.2925

A. V. Titomirov, S. Sukharev, and E. Kistanova, In vivo electroporation and stable transformation of skin cells of newborn mice by plasmid DNA, Biochimica et Biophysica Acta (BBA) - Gene Structure and Expression, vol.1088, issue.1, pp.131-135, 1991.
DOI : 10.1016/0167-4781(91)90162-F

D. E. Trentham, A. S. Townes, and A. H. Kang, Autoimmunity to type II collagen an experimental model of arthritis, Journal of Experimental Medicine, vol.146, issue.3, pp.857-68, 1977.
DOI : 10.1084/jem.146.3.857

P. M. Voorhees and R. Z. Orlowski, THE PROTEASOME AND PROTEASOME INHIBITORS IN CANCER THERAPY, Annual Review of Pharmacology and Toxicology, vol.46, issue.1, pp.189-213, 2006.
DOI : 10.1146/annurev.pharmtox.46.120604.141300

H. A. Wieland, M. Michaelis, B. J. Kirschbaum, and K. A. Rudolphi, Osteoarthritis ??? an untreatable disease?, Nature Reviews Drug Discovery, vol.3, issue.4, pp.331-375, 2005.
DOI : 10.1002/art.20365

S. Wilk and M. Orlowski, Evidence that Pituitary Cation-Sensitive Neutral Endopeptidase Is a Multicatalytic Protease Complex, Journal of Neurochemistry, vol.35, issue.3, pp.842-851, 1983.
DOI : 10.1016/0024-3205(79)90218-2

J. M. Williams, D. L. Felten, R. G. Peterson, and B. L. O-'connor, Effects of surgically induced instability on rat knee articular cartilage, J Anat, vol.134, pp.103-112, 1982.

R. O. Williams, Rodent models of arthritis: relevance for human disease, Clinical and Experimental Immunology, vol.25, issue.3, pp.330-332, 1998.
DOI : 10.1016/S0140-6736(89)90430-3

D. Wirth, P. Gustin, P. V. Drion, C. Dessy-doize, and E. S. Christians, Les protéines de choc thermique (heat choc proteins). I : Classification, structure, fonctions et implications dans les processus pathologiques, Ann. Méd. Vèt, vol.146, pp.201-216, 2002.

J. A. Wolff, R. W. Malone, P. Williams, W. Chong, G. Acsadi et al., Direct gene transfer into mouse muscle in vivo, Science, vol.247, issue.4949, pp.1465-1473, 1990.
DOI : 10.1126/science.1690918

T. K. Wong and E. Neumann, Electric field mediated gene transfer, Biochemical and Biophysical Research Communications, vol.107, issue.2, pp.584-591, 1982.
DOI : 10.1016/0006-291X(82)91531-5

URL : https://pub.uni-bielefeld.de/download/1774483/2311503

G. Y. Wu and C. H. Wu, Receptor-mediated gene delivery and expression in vivo, J. Biol. Chem, vol.263, pp.14621-14625, 1988.

M. Yamashita, K. Hirayoshi, and K. Nagata, Characterization of multiple members of the HSP70 family in platyfish culture cells: molecular evolution of stress protein HSP70 in vertebrates, Gene, vol.336, issue.2, pp.207-225, 2004.
DOI : 10.1016/j.gene.2004.04.023

]. W. Denk, J. H. Stricklen, and W. W. Webb, Two-photon laser scanning fluorescence microscopy, Science, vol.248, issue.4951, pp.248-273, 1990.
DOI : 10.1126/science.2321027

D. Dumas, L. Grossin, G. Cauchois, M. Gentils, and J. F. Stoltz, Comparison of wide-field/deconvolution and confocal microscopy in bioengineering. Interest of multi-photon microscopy in the study of articular cartilage, Biorheology, vol.40, pp.253-259, 2003.
URL : https://hal.archives-ouvertes.fr/hal-00559075

A. Diaspro and M. Robello, Two-photon excitation of fluorescence for three-dimensional optical imaging of biological structures, Journal of Photochemistry and Photobiology B: Biology, vol.55, issue.1, pp.1-8, 2000.
DOI : 10.1016/S1011-1344(00)00028-2

H. J. Koester, D. Baur, R. Uhl, and S. W. , Hell, Ca 2+ fluorescence imaging with pico-anf femtosecond two-photon excitation: signal and photodamage, Biophys. J, pp.77-2226, 1999.

D. Dumas, S. Muller, J. J. Padilla, V. Latger, S. Woodard et al., New trends in opical bioengineering: applications to cell biology, Recent Res. Devel. Optical Engg, vol.2, pp.295-315, 1999.

B. Pinsky, J. Ladasky, J. R. Lakowicz, K. Berndt, and R. A. Hoffman, Phase-resolved fluorescence lifetime measurements for flow cytometry, Cytometry, vol.30, issue.2, pp.123-135, 1993.
DOI : 10.1007/978-3-642-49204-4

R. Sanders, A. Draaijer, H. C. Gerritsen, P. M. Houpt, and Y. K. Levine, Quantitative pH Imaging in Cells Using Confocal Fluorescence Lifetime Imaging Microscopy, Analytical Biochemistry, vol.227, issue.2, pp.302-308, 1995.
DOI : 10.1006/abio.1995.1285

T. French, P. T. So, D. J. Weaver, T. Coelho-sampaio, E. Gratton et al., Two-photon fluorescence lifetime imaging microscopy of macrophage-mediated antigen processing, Journal of Microscopy, vol.185, issue.3, pp.339-353, 1997.
DOI : 10.1046/j.1365-2818.1997.d01-632.x

L. Grossin, C. Cournil-henrionnet, L. M. Mir, B. Liagre, D. Dumas et al., Direct gene transfer into rat articular cartilage by in vivo electroporation, The FASEB Journal, vol.17, issue.8, pp.829-835, 2003.
DOI : 10.3109/00016488909127491

URL : https://hal.archives-ouvertes.fr/hal-00319726

I. Majoul, M. Straub, R. Dufen, S. H. Hell, and H. Söling, Fluorescence resonance energy transfer analysis of protein???protein interactions in single living cells by multifocal multiphoton microscopy, Reviews in Molecular Biotechnology, vol.82, issue.3, pp.267-277, 2002.
DOI : 10.1016/S1389-0352(01)00042-3

R. Pepperkok, A. Squire, S. Geley, and P. I. Bastiaens, Simultaneous detection of multiple green fluorescent proteins in live cells by fluorescence lifetime imaging microscopy, Current Biology, vol.9, issue.5, pp.269-272, 1999.
DOI : 10.1016/S0960-9822(99)80117-1

D. T. Felson, Y. J. Kim, J. F. , W. Kim, F. J. Blanco et al., Cell death and apoptosis in osteoarthritic cartilage Heat-shock proteins and their role in chondrocyte protection, an application for autologous transplantation Glutamine protects articular chondrocytes from heat stress and NO-induced apoptosis with HSP70 expression, Osteoarthritis Cartilage Gene transfer with HSP 70 in rat chondrocytes confers cytoprotection in vitro and during experimental osteoarthritis The proteasome: a new target for novel drug therapies Proteomic expression analysis of cardiomyocytes subjected to proteasome inhibition Upregulation of heat shock protein expression by proteasome inhibition: an antiapoptotic mechanism in the lens Modulation of inflammatory response in sepsis by proteasome inhibition Induction of heat shock protein 70 (Hsp70) by proteasome inhibitor MG 132 protects articular chondrocytes from cellular death in vitro and in vivo Dose-response relationship for exercise on severity of experimental osteoarthritis in rats: a pilot study, Increased apoptosis in rat osteoarthritic cartilage corresponds to degenerative chondral lesions and concomitant expression of caspase-314] S. Meiners, A. Ludwig, V. Stangl and K. Stangl, Proteasome inhibitors: Poisons and remedies, pp.1378-1383, 2001.

G. W. Shi, R. Z. Small, . Z. Orlowski16-]-x, C. M. Ding, A. K. Fernandez-prada et al., Proteasome Inhibitors Induce a p38 Mitogen-activated Protein Kinase (MAPK)-dependent Anti-apoptotic Program Involving MAPK Phosphatase-1 and Akt in Models of Breast Cancer, Proteasome inhibition: a new anti-inflammatory strategy, pp.33-47, 2001.
DOI : 10.1042/0264-6021:3440477

T. Niidome and L. Huang, Gene Therapy Progress and Prospects: Nonviral vectors, Gene Therapy, vol.184, issue.24, pp.1647-52, 2002.
DOI : 10.1128/JB.184.13.3657-3663.2002

J. Gehl, Electroporation: theory and methods, perspectives for drug delivery, gene therapy and research, Acta Physiologica Scandinavica, vol.129, issue.4, pp.437-484, 2003.
DOI : 10.1016/S0169-409X(98)00061-1

V. Preat, Administration de médicaments et de gènes par électrotransfert, Ann Pharm Fr, vol.55, pp.239-283, 2001.

P. Bigey, M. Bureau, and D. Scherman, In vivo plasmid DNA electrotransfer, Current Opinion in Biotechnology, vol.13, issue.5, pp.443-450, 2002.
DOI : 10.1016/S0958-1669(02)00377-4

L. Mir, M. Bureau, J. Gehl, R. Rangara, D. Rouy et al., High-efficiency gene transfer into skeletal muscle mediated by electric pulses, Proceedings of the National Academy of Sciences, vol.63, issue.5, pp.4262-4269, 1999.
DOI : 10.1016/S0006-3495(92)81709-5

N. Saidenberg-kermanac-'h, N. Bessis, V. Deleuze, C. Bloquel, M. Bureau et al., Efficacy of interleukin-10 gene electrotransfer into skeletal muscle in mice with collagen-induced arthritis, The Journal of Gene Medicine, vol.159, issue.2, pp.164-71, 2003.
DOI : 10.1002/eji.1830261020

J. Kim, S. Ho, W. Hahn, J. Jeong, E. Park et al., Electro-gene therapy of collagen-induced arthritis by using an expression plasmid for the soluble p75 tumor necrosis factor receptor-Fc fusion protein, Gene Therapy, vol.163, issue.15, pp.1216-1240, 2003.
DOI : 10.1002/jgm.275

L. Grossin, C. Cournil-henrionnet, L. Mir, B. Liagre, D. Dumas et al., Direct gene transfer into rat articular cartilage by in vivo electroporation, The FASEB Journal, vol.17, issue.8, pp.829-864, 2003.
DOI : 10.3109/00016488909127491

URL : https://hal.archives-ouvertes.fr/hal-00319726

R. Terauchi, K. Takahashi, Y. Arai, T. Ikeda, S. Ohashi et al., Hsp70 prevents nitric oxide-induced apoptosis in articular chondrocytes, Arthritis & Rheumatism, vol.5, issue.6, pp.1562-1570, 2003.
DOI : 10.1097/00001756-199412000-00064

L. Grossin, C. Henrionnet, B. Liagre, L. Mir, D. Dumas et al., Electric gene transfer of Hsp70 in rat patella protects in vivo from iodoacetate induced chondrotoxicity, Arthritis Rheum, vol.46, p.221, 2002.

. Fig, Transgene expression kinetics in cartilage after electroporation in vivo in the rat knee. Biodistribution of GFP after transfection of the plasmid pcDNA3, GFP, vol.31, issue.70

L. Grossin, Gene therapy in cartilage using electroporation, Joint Bone Spine, vol.70, issue.6, pp.480-482, 2003.
DOI : 10.1016/j.jbspin.2003.07.003

T. Aigner, K. , and H. A. , Apoptosis and cellular vitality: Issues in osteoarthritic cartilage degeneration, Arthritis & Rheumatism, vol.11, issue.8, 1986.
DOI : 10.1002/1529-0131(200006)43:6<1290::AID-ANR11>3.0.CO;2-R

W. B. Van-den-berg, Lessons from animal models of osteoarthritis, Current Opinion in Rheumatology, vol.13, issue.5, pp.452-456, 2001.
DOI : 10.1097/00002281-200109000-00019

T. Aigner, Apoptosis, necrosis, or whatever: how to find out what really happens?, The Journal of Pathology, vol.98, issue.1, pp.1-4, 2002.
DOI : 10.1083/jcb.98.1.267

C. J. Malemud, N. Islam, and T. M. Haqqi, Pathophysiological Mechanisms in Osteoarthritis Lead to Novel Therapeutic Strategies, Cells Tissues Organs, vol.174, issue.1-2, pp.34-48, 2003.
DOI : 10.1159/000070573

S. Takayama, J. C. Reed, and S. Homma, Heat-shock proteins as regulators of apoptosis, Oncogene, vol.22, issue.56, pp.9041-9047, 2003.
DOI : 10.1016/S0959-440X(02)00315-9

R. K. Sironen, H. M. Karjalainen, M. A. Elo, K. Kaarniranta, K. Torronen et al., cDNA array reveals mechanosensitive genes in chondrocytic cells under hydrostatic pressure, Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, vol.1591, issue.1-3, pp.45-54, 2002.
DOI : 10.1016/S0167-4889(02)00247-1

L. Ravagnan, S. Gurbuxani, S. A. Susin, C. Maisse, E. Daugas et al., Heat-shock protein 70 antagonizes apoptosis-inducing factor, Nature Cell Biology, vol.12, issue.9, pp.839-843, 2001.
DOI : 10.3109/02656739609023695

URL : https://hal.archives-ouvertes.fr/hal-01606079

K. Takahashi, T. Kubo, R. S. Goomer, D. Amiel, K. Kobayashi et al., Analysis of heat shock proteins and cytokines expressed during early stages of osteoarthritis in a mouse model, Osteoarthritis and Cartilage, vol.5, issue.5, pp.321-329, 1997.
DOI : 10.1016/S1063-4584(97)80036-2

R. Terauchi, K. A. Takahashi, Y. Arai, T. Ikeda, S. Ohashi et al., Hsp70 prevents nitric oxide-induced apoptosis in articular chondrocytes, Arthritis & Rheumatism, vol.5, issue.6, pp.1562-1568, 2003.
DOI : 10.1097/00001756-199412000-00064

L. Grossin, C. Cournil-henrionnet, A. Watrin-pinzano, B. Terlain, J. Jouzeau et al., Overexpression and induction of heat shock protein 70 protect chondrocytes from cell death in vitro and in vivo, Arthritis Research & Therapy, vol.5, issue.Suppl 3, p.53, 2003.
DOI : 10.1186/ar854

O. Sperling, Y. Bromberg, H. Oelsner, and E. Zoref-shani, Reactive oxygen species play an important role in iodoacetate-induced neurotoxicity in primary rat neuronal cultures and in differentiated PC12 cells, Neuroscience Letters, vol.351, issue.3, pp.137-140, 2003.
DOI : 10.1016/S0304-3940(03)00858-9

S. W. Lee, H. J. Lee, W. T. Chung, S. M. Choi, S. H. Rhyu et al., TRAIL induces apoptosis of chondrocytes and influences the pathogenesis of experimentally induced rat osteoarthritis, Arthritis & Rheumatism, vol.416, issue.2, pp.534-542, 2004.
DOI : 10.1016/S0014-5793(97)01231-3

C. Guingamp, P. Gegout-pottie, L. Philippe, B. Terlain, P. Netter et al., Mono-iodoacetate-induced experimental osteoarthritis. A dose-response study of loss of mobility, morphology, and biochemistry, Arthritis & Rheumatism, vol.24, issue.9, pp.1670-1679, 1997.
DOI : 10.1016/0304-4165(86)90306-5

C. G. Concannon, A. M. Gorman, and A. Samali, On the role of Hsp27 in regulating apoptosis, APOPTOSIS, vol.8, issue.1, pp.61-70, 2003.
DOI : 10.1023/A:1021601103096

L. Grossin, C. Cournil-henrionnet, L. M. Mir, B. Liagre, D. Dumas et al., Direct gene transfer into rat articular cartilage by in vivo electroporation, The FASEB Journal, vol.17, issue.8, pp.829-835, 2003.
DOI : 10.3109/00016488909127491

URL : https://hal.archives-ouvertes.fr/hal-00319726

L. M. Mir, M. F. Bureau, J. Gehl, R. Rangara, D. Rouy et al., High-efficiency gene transfer into skeletal muscle mediated by electric pulses, Proc. Natl. Acad. Sci. USA 96, pp.4262-4267, 1999.
DOI : 10.1016/S0006-3495(92)81709-5

H. J. Mankin, H. Dorfman, L. Lippiello, and A. Zarins, Biochemical and Metabolic Abnormalities in Articular Cartilage from Osteo-Arthritic Human Hips, The Journal of Bone & Joint Surgery, vol.53, issue.3, pp.523-537, 1971.
DOI : 10.2106/00004623-197153030-00009

L. Galois, S. Etienne, L. Grossin, A. Watrin-pinzano, C. Cournil-henrionnet et al., Dose-response relationship for exercise on severity of 74, 2004.

A. Saied, E. Cherin, H. Gaucher, P. Laugier, P. Gillet et al., Assessment of Articular Cartilage and Subchondral Bone: Subtle and Progressive Changes in Experimental Osteoarthritis Using 50 MHz Echography In Vitro, Journal of Bone and Mineral Research, vol.69, issue.6, pp.1378-1386, 1997.
DOI : 10.2214/ajr.159.3.1503029

H. Dumond, N. Presle, P. Pottie, S. Pacquelet, B. Terlain et al., Site specific changes in gene expression and cartilage metabolism during early experimental osteoarthritis, Osteoarthritis and Cartilage, vol.12, issue.4, pp.284-295, 2004.
DOI : 10.1016/j.joca.2003.11.008

URL : https://hal.archives-ouvertes.fr/hal-01778159

L. Grossin, S. Etienne, N. Gaborit, A. Pinzano, C. Cournil-henrionnet et al., Induction of heat shock protein 70 (Hsp70) by proteasome inhibitor MG 132 protects articular chondrocytes from cellular death in vitro and in vivo, Biorheology, vol.41, pp.521-534, 2004.

K. Kuhn, D. D. Lima, S. Hashimoto, and M. Lotz, Cell death in cartilage, Osteoarthritis and Cartilage, vol.12, issue.1, pp.1-16, 2004.
DOI : 10.1016/j.joca.2003.09.015

M. Sharif, A. Whitehouse, P. Sharman, M. Perry, A. et al., Increased apoptosis in human osteoarthritic cartilage corresponds to reduced cell density and expression of caspase-3, Arthritis & Rheumatism, vol.146, issue.2, 2004.
DOI : 10.1053/joca.1999.0259

S. Hashimoto, K. Takahashi, D. Amiel, R. D. Coutts, and M. Lotz, Chondrocyte apoptosis and nitric oxide production during experimentally induced osteoarthritis, Arthritis & Rheumatism, vol.35, issue.7, pp.1266-1274, 1998.
DOI : 10.1083/jcb.41.1.59

J. P. Pelletier, F. Mineau, C. Boileau, and J. Martel-pelletier, Diacerein reduces the level of cartilage chondrocyte DNA fragmentation and death in experimental dog osteoarthritic cartilage at the same time that it inhibits caspase-3 and inducible nitric oxide synthase, Clin. Exp. Rheumatol, vol.21, pp.171-177, 2003.

J. Y. Jouzeau, P. Gillet, and P. Netter, Interest of animal models in the preclinical screening of anti-osteoarthritic drugs, Joint Bone Spine, vol.67, issue.6, pp.565-569, 2000.
DOI : 10.1016/S1297-319X(00)00214-1

M. J. Janusz, E. B. Hookfin, S. A. Heitmeyer, J. F. Woessner, A. J. Freemont et al., Moderation of iodoacetate-induced experimental osteoarthritis in rats by matrix metalloproteinase inhibitors, Osteoarthritis and Cartilage, vol.9, issue.8, pp.751-760, 2001.
DOI : 10.1053/joca.2001.0472

S. E. Bove, S. L. Calcaterra, R. M. Brooker, C. M. Huber, R. E. Guzman et al., Weight bearing as a measure of disease progression and efficacy of anti-inflammatory compounds in a model of monosodium iodoacetate-induced osteoarthritis, Osteoarthritis and Cartilage, vol.11, issue.11, pp.821-830, 2003.
DOI : 10.1016/S1063-4584(03)00163-8

D. Loeuille, P. Gonord, C. Guingamp, P. Gillet, A. Blum et al., In vitro magnetic resonance microimaging of experimental osteoarthritis in the rat knee joint, J. Rheumatol, vol.24, pp.133-139, 1997.

B. Gutmann, B. Hutter-paier, G. Skofitsch, M. Windisch, and R. Gmeinbauer, In vitro models of brain ischemia: The peptidergic drug cerebrolysin protects cultured chick cortical neurons from cell death, Neurotoxicity Research, vol.63, issue.2, pp.59-65, 2002.
DOI : 10.1016/S0024-3205(98)00236-7

B. Beltran, A. Mathur, M. R. Duchen, J. D. Erusalimsky, and S. Moncada, The effect of nitric oxide on cell respiration: A key to understanding its role in cell survival or death, Proc. Natl. Acad. Sci. USA 97, pp.14602-14607, 2000.
DOI : 10.1006/abbi.2000.1716

R. Malhotra and F. C. Brosius, Glucose Uptake and Glycolysis Reduce Hypoxia-induced Apoptosis in Cultured Neonatal Rat Cardiac Myocytes, Journal of Biological Chemistry, vol.1196, issue.18, pp.12567-12575
DOI : 10.1161/01.CIR.97.24.2454

J. E. Albina, S. Cui, R. B. Mateo, and J. S. Reichner, Nitric oxide-mediated apoptosis in murine peritoneal macrophages, J. Immunol, vol.150, pp.5080-5085, 1993.

C. Boileau, H. Dumond, N. Presle, S. Etienne, P. Gegout-pottie et al., Effect of selective COX2 inhibitor on cartilage lesion and chondrocyte apoptosis during experimental osteoarthritis in rat. Fund, Clin. Pharm, vol.16, p.47, 2002.

B. Grasl-kraupp, B. Ruttkay-nedecky, H. Koudelka, K. Bukowska, W. Bursch et al., In situ detection of fragmented DNA (TUNEL assay) fails to discriminate among apoptosis, necrosis, and autolytic cell death: a cautionary note, Hepatology, vol.21, pp.1465-1468, 1995.

J. P. Pelletier, D. V. Jovanovic, V. Lascau-coman, J. C. Fernandes, P. T. Manning et al., Selective inhibition of inducible nitric oxide synthase reduces progression of experimental osteoarthritis in vivo: Possible link with the reduction in chondrocyte apoptosis and caspase 3 level, Arthritis & Rheumatism, vol.43, issue.6, pp.1290-1299, 2000.
DOI : 10.1002/1529-0131(200006)43:6<1290::AID-ANR11>3.0.CO;2-R

S. P. Grogan, B. Aklin, M. Frenz, T. Brunner, T. Schaffner et al., In vitro model for the study of necrosis and apoptosis in native cartilage, The Journal of Pathology, vol.196, issue.1, pp.5-13, 2002.
DOI : 10.1053/joca.1998.0220

M. Matsuo, K. Nishida, A. Yoshida, T. Murakami, and H. Inoue, Expression of caspase-3 and -9 relevant to cartilage destruction and chondrocyte apoptosis in human osteoarthritic cartilage, Acta Med. Okayama, vol.55, pp.333-340, 2001.

J. G. Kiang, T. , and G. C. , Heat Shock Protein 70 kDa Molecular Biology, Biochemistry, and Physiology, Pharmacology & Therapeutics, vol.80, issue.2, pp.183-201, 1998.
DOI : 10.1016/S0163-7258(98)00028-X

Y. G. Weiss, A. Maloyan, J. Tazelaar, N. Raj, and C. S. Deutschman, Adenoviral transfer of HSP-70 into pulmonary epithelium ameliorates experimental acute respiratory distress syndrome, Journal of Clinical Investigation, vol.110, issue.6, pp.801-806, 2002.
DOI : 10.1172/JCI0215888

K. Suzuki, Y. Sawa, Y. Kaneda, H. Ichikawa, R. Shirakura et al., In vivo gene transfection with heat shock protein 70 enhances myocardial tolerance to ischemia-reperfusion injury in rat., Journal of Clinical Investigation, vol.99, issue.7, pp.1645-1650, 1997.
DOI : 10.1172/JCI119327

S. Okubo, O. Wildner, M. R. Shah, J. C. Chelliah, M. L. Hess et al., Gene Transfer of Heat-Shock Protein 70 Reduces Infarct Size In Vivo After Ischemia/Reperfusion in the Rabbit Heart, Circulation, vol.103, issue.6, pp.877-881, 2001.
DOI : 10.1161/01.CIR.103.6.877

B. Hoehn, T. M. Ringer, L. Xu, R. G. Giffard, R. M. Sapolsky et al., Overexpression of HSP72 after Induction of Experimental Stroke Protects Neurons from Ischemic Damage, Journal of Cerebral Blood Flow & Metabolism, vol.30, issue.11, pp.1303-1309, 2001.
DOI : 10.1161/01.STR.32.4.1028

J. C. Tiffee, J. P. Griffin, C. , and L. F. , Immunolocalization of stress proteins and extracellular matrix proteins in the rat tibia, Tissue and Cell, vol.32, issue.2, pp.141-147, 2000.
DOI : 10.1054/tice.2000.0097

J. B. Kouri-flores, K. A. Abbud-lozoya, and L. Roja-morales, Kinetics of the Ultrastructural Changes in Apoptotic Chondrocytes from an Osteoarthrosis Rat Model: A Window of Comparison to the Cellular Mechanism of Apoptosis in Human Chondrocytes, Ultrastructural Pathology, vol.26, issue.1, pp.33-40, 2002.
DOI : 10.1080/01913120252934314

M. E. Nuttall, D. P. Nadeau, P. W. Fisher, F. Wang, P. M. Keller et al., Inhibition of caspase-3-like activity prevents apoptosis while retaining functionality of human chondrocytesin vitro, Journal of Orthopaedic Research, vol.30, issue.3, pp.356-363, 2000.
DOI : 10.1074/jbc.270.6.2550

C. Bloquel, E. Fabre, M. F. Bureau, and D. Scherman, Plasmid DNA electrotransfer for intracellular and secreted proteins expression: new methodological developments and applications, The Journal of Gene Medicine, vol.6, issue.S1, pp.11-23, 2004.
DOI : 10.1002/jgm.508

H. Gene, . Is, . In, ]. T. Oa-references, H. A. Aigner et al., Apoptosis and cellular vitality: issues in osteoarthritic cartilage degeneration, Arthritis Rheum, vol.46, issue.1, pp.1986-1996, 2002.

F. J. Blanco, R. Guitian, E. Vazquez-martul, F. J. De-toro, and F. Galdo, Osteoarthritis chondrocytes die by apoptosis: A possible pathway for osteoarthritis pathology, Arthritis & Rheumatism, vol.74, issue.2, pp.41-284, 1998.
DOI : 10.1002/aja.1002030409

M. M. Bradford, A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding, Analytical Biochemistry, vol.72, issue.1-2, pp.248-254, 1976.
DOI : 10.1016/0003-2697(76)90527-3

J. M. Bruey, C. Ducasse, P. Bonniaud, L. Ravagnan, S. A. Susin et al., Hsp27 negatively regulates cell death by interacting with cytochrome c, Hsp27 negatively regulates cell death by interacting with cytochrome c, pp.645-652, 2000.
DOI : 10.1038/bjc.1994.140

J. A. Buckwalter and H. J. Mankin, Articular cartilage repair and transplantation, Arthritis & Rheumatism, vol.274, issue.8, pp.1331-1342, 1998.
DOI : 10.3109/17453679209154757

URL : http://onlinelibrary.wiley.com/doi/10.1002/1529-0131(199808)41:8<1331::AID-ART2>3.0.CO;2-J/pdf

K. T. Bush, A. L. Goldberg, and S. K. Nigam, Proteasome Inhibition Leads to a Heat-shock Response, Induction of Endoplasmic Reticulum Chaperones, and Thermotolerance, Journal of Biological Chemistry, vol.13, issue.14, pp.272-9086, 1997.
DOI : 10.1016/0006-291X(89)91804-4

P. Chomczynski and N. Sacchi, Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction, Analytical Biochemistry, vol.162, issue.1, pp.156-159, 1987.
DOI : 10.1016/0003-2697(87)90021-2

C. G. Concannon, A. M. Gorman, and A. Samali, On the role of Hsp27 in regulating apoptosis, APOPTOSIS, vol.8, issue.1, pp.61-70, 2003.
DOI : 10.1023/A:1021601103096

X. Z. Ding, C. M. Fernandez-prada, A. K. Bhattacharjee, and D. L. Hoover, OVER-EXPRESSION OF HSP-70 INHIBITS BACTERIAL LIPOPOLYSACCHARIDE-INDUCED PRODUCTION OF CYTOKINES IN HUMAN MONOCYTE-DERIVED MACROPHAGES, Cytokine, vol.16, issue.6, pp.210-219, 2001.
DOI : 10.1006/cyto.2001.0959

H. C. Drexler, Activation of the cell death program by inhibition of proteasome function, Proc. Natl. Acad. Sci. USA 94, pp.855-860, 1997.
DOI : 10.1002/jcp.1041590206

M. G. Ehrlich, H. J. Mankin, H. Jones, A. Grossman, C. Crispen et al., Biochemical confirmation of an experimental osteoarthritis model, The Journal of Bone & Joint Surgery, vol.57, issue.3, pp.392-396, 1975.
DOI : 10.2106/00004623-197557030-00018

C. Garrido, J. M. Bruey, A. Fromentin, A. Hammann, A. P. Arrigo et al., -dependent activation of procaspase-9, The FASEB Journal, vol.13, issue.14, pp.2061-2070, 1999.
DOI : 10.1038/bjc.1994.140

C. Guingamp, P. Gegout-pottie, L. Philippe, B. Terlain, P. Netter et al., Mono-iodoacetate-induced experimental osteoarthritis. A dose-response study of loss of mobility, morphology, and biochemistry, Arthritis & Rheumatism, vol.24, issue.9, pp.40-1670, 1997.
DOI : 10.1016/0304-4165(86)90306-5

R. E. Guzman, M. G. Evans, S. Bove, B. Morenko, and K. Kilgore, Mono-iodoacetate-induced histologic changes in subchondral bone and articular cartilage of rat femorotibial joints: an animal model of osteoarthritis, Toxicol. Pathol, pp.31-619, 2003.

L. Grossin, MG 132 is chondroprotective in vitro and in vivo Development and regulation of osteophyte formation during experimental osteoarthritis. Osteoarthritis Cartilage, pp.10-180, 2002.

A. Kawakami, T. Nakashima, H. Sakai, A. Hida, S. Urayama et al., Regulation of synovial cell apoptosis by proteasome inhibitor, Arthritis & Rheumatism, vol.444, issue.11, pp.42-2440, 1999.
DOI : 10.1016/S0014-5793(99)00032-0

D. Kim, S. H. Kim, and G. C. Li, Proteasome Inhibitors MG132 and Lactacystin Hyperphosphorylate HSF1 and Induce hsp70 and hsp27 Expression, Biochemical and Biophysical Research Communications, vol.254, issue.1, pp.264-268, 1999.
DOI : 10.1006/bbrc.1998.9840

T. Kubo, Y. Arai, K. Takahashi, T. Ikeda, S. Ohashi et al., Expression of transduced HSP70 gene protects chondrocytes from stress, J. Rheumatol, pp.28-330, 2001.

U. K. Laemmli, F. Beguin, and G. Gujer-kellenberger, A factor preventing the major head protein of bacteriophage T4 from random aggregation, Journal of Molecular Biology, vol.47, issue.1, pp.47-69, 1970.
DOI : 10.1016/0022-2836(70)90402-X

A. B. Meriin, V. L. Gabai, J. Yaglom, V. I. Shifrin, and M. Y. Sherman, Proteasome inhibitors activate stress kinases and induce Hsp72. Diverse effects on apoptosis, J. Biol. Chem, pp.273-6373, 1998.

D. D. Mosser, A. W. Caron, L. Bourget, A. B. Meriin, M. Y. Sherman et al., The Chaperone Function of hsp70 Is Required for Protection against Stress-Induced Apoptosis, Molecular and Cellular Biology, vol.20, issue.19, pp.7146-7159, 2000.
DOI : 10.1128/MCB.20.19.7146-7159.2000

B. S. Polla, S. Kantengwa, D. Francois, S. Salvioli, C. Franceschi et al., Mitochondria are selective targets for the protective effects of heat shock against oxidative injury., Proc. Natl. Acad. Sci. USA 93, pp.6458-6463, 1996.
DOI : 10.1073/pnas.93.13.6458

L. Ravagnan, S. Gurbuxani, S. A. Susin, C. Maisse, E. Daugas et al., Heat-shock protein 70 antagonizes apoptosis-inducing factor, Nature Cell Biology, vol.12, issue.9, pp.839-843, 2001.
DOI : 10.3109/02656739609023695

URL : https://hal.archives-ouvertes.fr/hal-01606079

J. D. Robertson, K. Datta, S. S. Biswal, and J. P. Kehrer, Heat-shock protein 70 antisense oligomers enhance proteasome inhibitor-induced apoptosis, Biochemical Journal, vol.344, issue.2, pp.477-485, 1999.
DOI : 10.1042/bj3440477

R. Sadoul, P. A. Fernandez, A. L. Quiquerez, I. Martinou, M. Maki et al., Involvement of the proteasome in the programmed cell death of NGF-deprived sympathetic neurons, EMBO J, vol.15, pp.3845-3852, 1996.

A. Samali, J. D. Robertson, E. Peterson, F. Manero, L. Van-zeijl et al., Hsp27 protects mitochondria of thermotolerant cells against apoptotic stimuli, Cell Stress & Chaperones, vol.90, issue.1, pp.49-58, 2001.
DOI : 10.1379/1466-1268(2001)006<0049:HPMOTC>2.0.CO;2

C. Stefanelli, F. Bonavita, I. Stanic, C. Pignatti, G. Farruggia et al., Inhibition of etoposide-induced apoptosis with peptide aldehyde inhibitors of proteasome, Biochemical Journal, vol.332, issue.3, pp.332-661, 1998.
DOI : 10.1042/bj3320661

K. Takahashi, T. Kubo, Y. Arai, J. Imanishi, M. Kawata et al., Localization of Heat Shock Protein in Osteoarthritic Cartilage, Scandinavian Journal of Rheumatology, vol.71, issue.5, pp.368-375, 1997.
DOI : 10.1139/o93-054

K. Takahashi, T. Kubo, R. S. Goomer, D. Amiel, K. Kobayashi et al., Analysis of heat shock proteins and cytokines expressed during early stages of osteoarthritis in a mouse model, Osteoarthritis and Cartilage, vol.5, issue.5, pp.321-329, 1997.
DOI : 10.1016/S1063-4584(97)80036-2

P. M. Van-der-kraan, E. L. Vitters, H. M. Van-beuningen, W. B. Van-den, and . Berg, Proteoglycan synthesis and osteophyte formation in 'metabolically' and 'mechanically' induced murine degenerative joint disease: an in-vivo autoradiographic study, Int. J. Exp. Pathol, pp.73-335, 1992.

M. Vayssier, N. Banzet, D. Francois, K. Bellmann, and B. S. Polla, Tobacco smoke induces both apoptosis and necrosis in mammalian cells: differential effects of HSP70, American Journal of Physiology-Lung Cellular and Molecular Physiology, vol.1, issue.4, pp.275-771, 1998.
DOI : 10.1126/science.275.5303.1129

U. Mixte-de, B. , F. Vandoeuvre-lès-nancy, F. Service-de-chirurgie-orthopédique-et-traumatologique, C. et al., Avenue de Lattre de Tassigny, 54035 Nancy Cedex, France Keywords: Cartilage, osteoarthritis, apoptosis, caspase, experimental model 1. Introduction Articular cartilage is a specialized and uniquely structured tissue that forms the smooth, gliding surface of the diarthrodial joints. It consists of an extracellular matrix (ECM) that is synthesized by the sparsely distributed resident cells, the chondrocytes. Osteoarthritis (OA) is a typical slow degenerative disease of the joints characterized by fibrillation and erosion of hyaline cartilage, subchondral bone sclerosis and osteophyte formation at the joint margins, leading to a gradual loss of articular cartilage, Recherche, vol.756120

H. Dumond, N. Presle, P. Pottie, S. Pacquelet, B. Terlain et al., Site specific changes in gene expression and cartilage metabolism during early experimental osteoarthritis, Osteoarthritis and Cartilage, vol.12, issue.4, pp.284-295, 2004.
DOI : 10.1016/j.joca.2003.11.008

URL : https://hal.archives-ouvertes.fr/hal-01778159

L. Galois, S. Etienne, L. Grossin, C. Cournil, A. Pinzano et al., Moderate-impact exercise is associated with decreased severity of experimental osteoarthritis in rats, Rheumatology, vol.42, issue.5, pp.692-693, 2003.
DOI : 10.1093/rheumatology/keg094

L. Galois, S. Etienne, L. Grossin, A. Watrin-pinzano, C. Cournil-henrionnet et al., Dose???response relationship for exercise on severity of experimental osteoarthritis in rats: a pilot study, Osteoarthritis and Cartilage, vol.12, issue.10, pp.779-786, 2004.
DOI : 10.1016/j.joca.2004.06.008

R. Goggs, S. D. Carter, G. Schulze-tanzil, M. Shakibaei, and A. Mobasheri, Apoptosis and the loss of chondrocyte survival signals contribute to articular cartilage degradation in osteoarthritis, The Veterinary Journal, vol.166, issue.2, pp.140-158, 2003.
DOI : 10.1016/S1090-0233(02)00331-3

S. P. Grogan, B. Aklin, M. Frenz, T. Brunner, T. Schaffner et al., In vitro model for the study of necrosis and apoptosis in native cartilage, The Journal of Pathology, vol.196, issue.1, pp.198-203, 2002.
DOI : 10.1053/joca.1998.0220

L. Grossin, S. Etienne, N. Gaborit, A. Pinzano, C. Cournil-henrionnet et al., Induction of heat shock protein 70 (Hsp70) by proteasome inhibitor MG 132 protects articular chondrocytes from cellular death in vitro and in vivo, Biorheology, vol.41, pp.521-534, 2004.

C. Guingamp, P. Gegout-pottie, L. Philippe, B. Terlain, P. Netter et al., Mono-iodoacetate-induced experimental osteoarthritis. A dose-response study of loss of mobility, morphology, and biochemistry, Arthritis & Rheumatism, vol.24, issue.9, pp.40-1670, 1997.
DOI : 10.1016/0304-4165(86)90306-5

S. Hashimoto, K. Takahashi, D. Amiel, R. D. Coutts, and M. Lotz, Chondrocyte apoptosis and nitric oxide production during experimentally induced osteoarthritis, Arthritis & Rheumatism, vol.35, issue.7, pp.1266-1274, 1998.
DOI : 10.1083/jcb.41.1.59

R. U. Janicke, M. L. Sprengart, M. R. Wati, and A. G. Porter, Caspase-3 Is Required for DNA Fragmentation and Morphological Changes Associated with Apoptosis, Journal of Biological Chemistry, vol.15, issue.16, pp.273-9357, 1998.
DOI : 10.1038/384368a0

J. Y. Jouzeau, P. Gillet, and P. Netter, Interest of animal models in the preclinical screening of anti-osteoarthritic drugs, Joint Bone Spine, vol.67, issue.6, pp.565-569, 2000.
DOI : 10.1016/S1297-319X(00)00214-1

I. N. Lavrik, A. Golks, and P. H. Krammer, Caspases: pharmacological manipulation of cell death, Journal of Clinical Investigation, vol.115, issue.10, pp.2665-2672, 2005.
DOI : 10.1172/JCI26252

S. W. Lee, H. J. Lee, W. T. Chung, S. M. Choi, S. H. Rhyu et al., TRAIL induces apoptosis of chondrocytes and influences the pathogenesis of experimentally induced rat osteoarthritis, Arthritis & Rheumatism, vol.416, issue.2, pp.534-542, 2004.
DOI : 10.1016/S0014-5793(97)01231-3

M. Matsuo, K. Nishida, A. Yoshida, T. Murakami, and H. Inoue, Expression of caspase-3 and -9 relevant to cartilage destruction and chondrocyte apoptosis in human osteoarthritic cartilage, Acta Med. Okayama, vol.55, pp.333-340, 2001.

D. Mistry, Y. Oue, M. G. Chambers, M. V. Kayser, and R. M. Mason, Chondrocyte death during murine osteoarthritis, Osteoarthritis and Cartilage, vol.12, issue.2, pp.131-141, 2004.
DOI : 10.1016/j.joca.2003.10.006

J. P. Pelletier, D. V. Jovanovic, V. Lascau-coman, J. C. Fernandes, P. T. Manning et al., Selective inhibition of inducible nitric oxide synthase reduces progression of experimental osteoarthritis in vivo: Possible link with the reduction in chondrocyte apoptosis and caspase 3 level, Arthritis & Rheumatism, vol.43, issue.6, pp.43-1290, 2000.
DOI : 10.1002/1529-0131(200006)43:6<1290::AID-ANR11>3.0.CO;2-R

A. R. Resendes, N. Majo, J. Segales, J. Espadamala, E. Mateu et al., Apoptosis in normal lymphoid organs from healthy normal, conventional pigs at different ages detected by TUNEL and cleaved caspase-3 immunohistochemistry in paraffin-embedded tissues, Veterinary Immunology and Immunopathology, vol.99, issue.3-4, pp.99-203, 2004.
DOI : 10.1016/j.vetimm.2004.02.001

H. I. Roach, T. Aigner, and J. B. Kouri, Chondroptosis: A variant of apoptotic cell death in chondrocytes?, Apoptosis, vol.9, issue.3, pp.265-277, 2004.
DOI : 10.1023/B:APPT.0000025803.17498.26

P. Sarzi-puttini, M. A. Cimmino, R. Scarpa, R. Caporali, F. Parazzini et al., Osteoarthritis: An Overview of the Disease and Its Treatment Strategies, Seminars in Arthritis and Rheumatism, vol.35, issue.1, pp.1-10, 2005.
DOI : 10.1016/j.semarthrit.2005.01.013

D. R. Schultz, W. J. Harrington, and J. , Apoptosis: Programmed cell death at a molecular level, Seminars in Arthritis and Rheumatism, vol.32, issue.6, pp.345-369, 2003.
DOI : 10.1053/sarh.2003.50005

D. J. Schurman and R. L. Smith, Osteoarthritis, Clinical Orthopaedics and Related Research, vol.427, pp.183-189, 2004.
DOI : 10.1097/01.blo.0000143555.33848.c4

M. Sharif, A. Whitehouse, P. Sharman, M. Perry, and M. Adams, Increased apoptosis in human osteoarthritic cartilage corresponds to reduced cell density and expression of caspase-3, Arthritis & Rheumatism, vol.146, issue.2, pp.507-515, 2004.
DOI : 10.1053/joca.1999.0259

R. Stoop, P. Buma, P. M. Van-der-kraan, A. P. Hollander, R. C. Billinghurst et al., Type II collagen degradation in articular cartilage fibrillation after anterior cruciate ligament transection in rats, Osteoarthritis and Cartilage, vol.9, issue.4, pp.308-315, 2001.
DOI : 10.1053/joca.2000.0390

R. Todd-allen, C. M. Robertson, F. L. Harwood, T. Sasho, S. K. Williams et al., Characterization of mature vs aged rabbit articular cartilage: analysis of cell density, apoptosis-related gene expression and mechanisms controlling chondrocyte apoptosis, Osteoarthritis and Cartilage, vol.12, issue.11, pp.917-923, 2004.
DOI : 10.1016/j.joca.2004.08.003

J. M. Williams, D. L. Felten, R. G. Peterson, and B. L. Connor, Effects of surgically induced instability on rat knee articular cartilage, J. Anat, vol.134, pp.103-109, 1982.