X. Chevalier and P. Richette, Cartilage articulaire normal : anatomie, physiologie, métabolisme, vieillissement, EMC -Rhumatol.-Orthopédie, vol.2, issue.1, pp.41-58, 2005.

A. J. Hayes, A. Hall, L. Brown, R. Tubo, and B. Caterson, Macromolecular organization and in vitro growth characteristics of scaffold-free neocartilage grafts, J. Histochem. Cytochem, vol.55, issue.8, pp.853-866, 2007.

J. E. Lafont, Lack of oxygen in articular cartilage: consequences for chondrocyte biology, Int. J. Exp. Pathol, vol.91, issue.2, pp.99-106, 2010.

D. Pfander and K. Gelse, Hypoxia and osteoarthritis: how chondrocytes survive hypoxic environments, Curr. Opin. Rheumatol, vol.19, issue.5, pp.457-462, 2007.

B. V. Shlopov, W. R. Lie, C. L. Mainardi, A. A. Cole, S. Chubinskaya et al., Osteoarthritic lesions: involvement of three different collagenases, Arthritis Rheum, vol.40, issue.11, pp.2065-2074, 1997.

C. R. Flannery, C. B. Little, C. E. Hughes, and B. Caterson, Expression and activity of articular cartilage hyaluronidases, Biochem. Biophys. Res. Commun, vol.251, issue.3, pp.824-833, 1998.

J. Marcelino and C. A. Mcdevitt, Attachment of articular cartilage chondrocytes to the tissue form of type VI collagen, Biochim. Biophys. Acta, vol.1249, issue.2, pp.180-188, 1995.

R. Schneiderman, D. Keret, and A. Maroudas, Effects of mechanical and osmotic pressure on the rate of glycosaminoglycan synthesis in the human adult femoral head cartilage: an in vitro study, J. Orthop. Res, vol.4, issue.4, pp.393-408, 1986.

R. J. Wilkins and A. C. Hall, Control of matrix synthesis in isolated bovine chondrocytes by extracellular and intracellular pH, J. Cell. Physiol, vol.164, issue.3, pp.474-481, 1995.

Y. Sommarin, T. Larsson, and D. Heinegård, Chondrocyte-matrix interactions. Attachment to proteins isolated from cartilage, Exp. Cell Res, vol.184, issue.1, pp.181-192, 1989.

C. Kiani, L. Chen, Y. J. Wu, A. J. Yee, and B. B. Yang, Structure and function of aggrecan, Cell Res, vol.12, issue.1, pp.19-32, 2002.

J. Vincourt, E. S. Cottet, and J. , C-propeptides of procollagens I alpha 1 and II that differentially accumulate in enchondromas versus chondrosarcomas regulate tumor cell survival and migration, Cancer Res, vol.70, issue.11, pp.4739-4748, 2010.

B. Alberts, A. Johnson, J. Lewis, M. Raff, K. Roberts et al., The Extracellular Matrix of Animals, 2002.

D. Heinegard, Proteoglycans and more -from molecules to biology, Int. J. Exp. Pathol, vol.90, issue.6, pp.575-586, 2009.

A. Hildebrand, M. Romarís, and L. M. Rasmussen, Interaction of the small interstitial proteoglycans biglycan, decorin and fibromodulin with transforming growth factor beta, Biochem. J, vol.302, issue.2, p.527, 1994.

F. H. Chen, K. T. Rousche, and R. S. Tuan, Technology Insight: adult stem cells in cartilage regeneration and tissue engineering, Nat. Rev. Rheumatol, vol.2, issue.7, pp.373-382, 2006.

N. Shintani, . Siebenrock-k-a, and E. B. Hunziker, TGF-ß1 Enhances the BMP-2-Induced Chondrogenesis of

, Bovine Synovial Explants and Arrests Downstream Differentiation at an Early Stage of Hypertrophy, PloS One, vol.8, issue.1, p.53086, 2013.

Y. Matsukura, T. Muneta, K. Tsuji, H. Koga, and I. Sekiya, Mesenchymal stem cells in synovial fluid increase after meniscus injury, Clin. Orthop, vol.472, issue.5, pp.1357-1364, 2014.

T. Iwanaga, M. Shikichi, H. Kitamura, H. Yanase, and K. Nozawa-inoue, Morphology and functional roles of synoviocytes in the joint, Arch. Histol. Cytol, vol.63, issue.1, pp.17-31, 2000.

G. M. Campo, A. Avenoso, D. 'ascola, and A. , Hyaluronan in part mediates IL-1beta-induced inflammation in mouse chondrocytes by up-regulating CD44 receptors, Gene, vol.494, issue.1, pp.24-35, 2012.

A. Cantagrel and A. Constantin, Membrane synoviale : de la physiologie à l'inflammaton rhumatoïde. Implications thérapeutiques, 2013.

V. Strand, R. Kimberly, and J. D. Isaacs, Biologic therapies in rheumatology: lessons learned, future directions, Nat. Rev. Drug Discov, vol.6, issue.1, pp.75-92, 2007.

M. Cutolo, B. Seriolo, B. Villaggio, C. Pizzorni, C. Craviotto et al., Androgens and estrogens modulate the immune and inflammatory responses in rheumatoid arthritis, Ann. N. Y. Acad. Sci, vol.966, pp.131-142, 2002.

E. Choy, Understanding the dynamics: pathways involved in the pathogenesis of rheumatoid arthritis, Rheumatol. Oxf. Engl, vol.51, issue.5, pp.3-11, 2012.

J. S. Smolen, D. Aletaha, M. Koeller, M. H. Weisman, and P. Emery, New therapies for treatment of rheumatoid arthritis, Lancet, vol.370, issue.9602, pp.1861-1874, 2007.

E. Choy and G. S. Panayi, Cytokine pathways and joint inflammation in rheumatoid arthritis, N. Engl. J. Medec, vol.344, issue.12, pp.907-916, 2001.

I. B. Mcinnes and G. Schett, Cytokines in the pathogenesis of rheumatoid arthritis, Nat. Rev. Immunol, vol.7, issue.6, pp.429-471, 2007.

D. T. Felson, R. C. Lawrence, and P. A. Dieppe, Osteoarthritis: new insights. Part 1 : the disease and its risk factors, Ann. Intern. Med, vol.133, issue.8, pp.635-646, 2000.

F. Eckstein, M. Hudelmaier, and R. Putz, The effects of exercise on human articular cartilage, J. Anat, vol.208, issue.4, pp.491-512, 2006.

L. Galois, S. Etienne, and L. Grossin, Dose-response relationship for exercise on severity of experimental osteoarthritis in rats: a pilot study, Osteoarthritis Cartilage, vol.12, issue.10, pp.779-86, 2004.

H. A. Wieland, M. Michaelis, B. J. Kirschbaum, and K. A. Rudolphi, Osteoarthritis -an untreatable disease?, Nat. Rev. Drug Discov, vol.4, issue.4, pp.331-344, 2005.

J. Pelletier and J. Martel-pelletier, Osteoarthritis: from molecule to man, The Novartis-ILAR Rheumatology Prize, vol.4, pp.13-19, 2001.

M. K. Lotz, S. Otsuki, S. P. Grogan, R. Sah, R. Terkeltaub et al., Cartilage cell clusters. Arthritis Rheum, vol.62, issue.8, pp.2206-2218, 2010.

S. B. Abramson, Osteoarthritis and nitric oxide, vol.16, pp.15-20, 2008.

E. Maneiro, M. J. López-armada, and A. Mc-de, Effect of nitric oxide on mitochondrial respiratory activity of human articular chondrocytes, Ann. Rheum. Dis, vol.64, issue.3, pp.388-395, 2005.

S. B. Abramson, Nitric oxide in inflammation and pain associated with osteoarthritis, Arthritis Res. Ther, vol.10, issue.2, p.1, 2008.

J. S. Smolen, R. Landewé, and F. C. Breedveld, EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs : 2013 update, Ann. Rheum. Dis, vol.73, issue.3, pp.492-509, 2014.

D. L. Scott, Biologics-Based Therapy for the Treatment of Rheumatoid Arthritis, Clin. Pharmacol. Ther, vol.91, issue.1, pp.30-43, 2012.

R. S. Woodrick and E. M. Ruderman, Safety of biologic therapy in rheumatoid arthritis, Nat. Rev. Rheumatol, vol.7, issue.11, pp.639-652, 2011.

F. S. Paula and J. D. Alves, Non-tumor necrosis factor-based biologic therapies for rheumatoid arthritis: present, future, and insights into pathogenesis, Biol. Targets Ther, vol.8, pp.1-12, 2014.

D. A. Pappas, L. Geraldino-pardilla, and J. M. Bathon, Immune modulation of rheumatoid arthritis, Best Pract. Res. Clin. Rheumatol, vol.25, issue.6, pp.873-889, 2011.

Y. Henrotin, M. Mathy, C. Sanchez, and C. Lambert, Chondroitin sulfate in the treatment of osteoarthritis: from in vitro studies to clinical recommendations, Ther. Adv. Musculoskelet. Dis, vol.2, issue.6, pp.335-383, 2010.

Y. Henrotin and X. Chevalier, Guidelines for the management of knee and hip osteoarthritis: For whom? Why? To do what? Presse Médicale, vol.39, pp.1180-1188, 2010.

M. Brittberg, A. Lindahl, A. Nilsson, C. Ohlsson, O. Isaksson et al., Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation, N. Engl. J. Med, vol.331, issue.14, pp.889-895, 1994.

C. Chung and J. Burdick, Engineering cartilage tissue, Adv. Drug Deliv. Rev, vol.60, issue.2, pp.243-62, 2008.

E. Roeder, C. Henrionnet, and J. C. Goebel, Dose-response of superparamagnetic iron oxide labeling on mesenchymal stem cells chondrogenic differentiation: a multi-scale in vitro study, PloS One, vol.9, issue.5, p.98451, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02364264

C. Gerard, C. Catuogno, and C. Amargier-huin, The effect of alginate, hyaluronate and hyaluronate derivatives biomaterials on synthesis of non-articular chondrocyte extracellular matrix, J. Mater. Sci. Mater. Med, vol.16, issue.6, pp.541-551, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00079365

Y. Chen, H. Lee, H. Chan, L. Sung, H. Chen et al.,

, sulfate/dermatan sulfate/chitosan scaffolds for cartilage tissue engineering, Biomaterials, vol.28, issue.14, pp.2294-305, 2007.

Y. Gong, Y. Zhu, Y. Liu, Z. Ma, C. Gao et al., Layer-by-layer assembly of chondroitin sulfate and collagen on aminolyzed poly(L-lactic acid) porous scaffolds to enhance their chondrogenesis, Acta Biomater, vol.3, issue.5, pp.677-685, 2007.

R. El-omar, J. Beroud, J. Stoltz, P. Menu, E. Velot et al., Umbilical cord mesenchymal stem cells: the new gold standard for mesenchymal stem cell-based therapies?, Tissue Eng. Part B Rev, vol.20, issue.5, pp.523-544, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01777817

C. Jorgensen, J. Gordeladze, and D. Noel, Tissue engineering through autologous mesenchymal stem cells, Curr. Opin. Biotechnol, vol.15, issue.5, pp.406-410, 2004.

J. A. Kode, S. Mukherjee, M. V. Joglekar, and A. A. Hardikar, Mesenchymal stem cells: immunobiology and role in immunomodulation and tissue regeneration, Cytotherapy, vol.11, issue.4, pp.377-391, 2009.

R. F. Schelbergen, S. Van-dalen, and M. Ter-huurne, Treatment efficacy of adipose-derived stem cells in experimental osteoarthritis is driven by high synovial activation and reflected by S100A8/A9 serum levels, Osteoarthritis Cartilage, vol.22, issue.8, pp.1158-1166, 2014.

G. Desando, C. Cavallo, and F. Sartoni, Intra-articular delivery of adipose derived stromal cells attenuates osteoarthritis progression in an experimental rabbit model, Arthritis Res. Ther, vol.15, issue.1, p.22, 2013.

M. Maumus, C. Jorgensen, and D. Noël, Mesenchymal stem cells in regenerative medicine applied to rheumatic diseases: role of secretome and exosomes, Biochimie, vol.95, issue.12, pp.2229-2234, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00832507

B. O. Diekman, N. Christoforou, and V. P. Willard, Cartilage tissue engineering using differentiated and purified induced pluripotent stem cells, Proc. Natl. Acad. Sci. U. S. A, vol.109, issue.47, pp.19172-19177, 2012.

R. M. Lauder, Chondroitin sulphate: a complex molecule with potential impacts on a wide range of biological systems, Complement. Ther. Med, vol.17, issue.1, pp.56-62, 2009.

F. Ronca, L. Palmieri, P. Panicucci, and G. Ronca, Anti-inflammatory activity of chondroitin sulfate, Osteoarthritis Cartilage. 6 Suppl A, pp.14-21, 1998.

N. Gerwin, C. Hops, and A. Lucke, Intraarticular drug delivery in osteoarthritis, Adv. Drug Deliv. Rev, vol.58, issue.2, pp.226-268, 2006.

S. Edwards, Intra-articular drug delivery: the challenge to extend drug residence time within the joint, Vet. J. Lond. Engl, vol.190, issue.1, pp.15-21, 1997.

T. Iannitti, D. Lodi, and B. Palmieri, Intra-articular injections for the treatment of osteoarthritis: focus on the clinical use of hyaluronic acid, Drugs RD, vol.11, issue.1, pp.13-27, 2011.

W. L. Sibbitt, A. Peisajovich, and A. A. Michael, Does Sonographic Needle Guidance Affect the Clinical Outcome of Intraarticular Injections?, J. Rheumatol, vol.36, issue.9, pp.1892-1902, 2009.

C. Vauthier and P. Couvreur, Nanotechnologies pour la thérapeutique et le diagnostic, 2008.

H. Hillaireau and P. Couvreur, Nanocarriers' entry into the cell: relevance to drug delivery, Cell. Mol. Life Sci, vol.66, issue.17, pp.2873-2896, 2009.

P. Milla, F. Dosio, and L. Cattel, PEGylation of proteins and liposomes: a powerful and flexible strategy to improve the drug delivery, Curr. Drug Metab, vol.13, issue.1, pp.105-119, 2012.

D. Papahadjopoulos, T. M. Allen, and A. Gabizon, Sterically stabilized liposomes: improvements in pharmacokinetics and antitumor therapeutic efficacy, Proc. Natl. Acad. Sci. U. S. A, vol.88, issue.24, pp.11460-11464, 1991.

N. Obermajer, P. Kocbek, and U. Repnik, Immunonanoparticles--an effective tool to impair harmful proteolysis in invasive breast tumor cells, FEBS J, vol.274, issue.17, pp.4416-4427, 2007.

S. Valetti, F. Maione, and S. Mura, Peptide-functionalized nanoparticles for selective targeting of pancreatic tumor, J. Controlled Release, vol.192, pp.29-39, 2014.

F. Caputo, D. Nicola, M. Ghibelli, and L. , Pharmacological potential of bioactive engineered nanomaterials, Biochem. Pharmacol, vol.92, issue.1, pp.112-130, 2014.

S. Bhattacharyya, R. A. Kudgus, R. Bhattacharya, and P. Mukherjee, Inorganic nanoparticles in cancer therapy, Pharm. Res, vol.28, issue.2, pp.237-259, 2011.

M. Janssen, G. Mihov, T. Welting, J. Thies, and P. Emans, Drugs and Polymers for Delivery Systems in OA Joints: Clinical Needs and Opportunities, Polymers, vol.6, issue.3, pp.799-819, 2014.

N. Butoescu, O. Jordan, and E. Doelker, Intra-articular drug delivery systems for the treatment of rheumatic diseases: a review of the factors influencing their performance, Eur. J. Pharm. Biopharm, vol.73, issue.2, pp.205-223, 2009.

. Rothenfluh-d-a, H. Bermudez, O. Neil, and C. P. , Biofunctional polymer nanoparticles for intraarticular targeting and retention in cartilage, Nat. Mater, vol.7, issue.3, pp.248-54, 2008.

H. Laroui, L. Grossin, and M. Léonard, Hyaluronate-covered nanoparticles for the therapeutic targeting of cartilage, Biomacromolecules, vol.8, issue.12, pp.3879-85, 2007.

Z. Chen, J. Chen, and L. Wu, Hyaluronic acid-coated bovine serum albumin nanoparticles loaded with brucine as selective nanovectors for intra-articular injection, Int. J. Nanomedicine, vol.8, pp.3843-3853, 2013.

N. Butoescu, M. Seemayer-c-a,-foti, O. Jordan, and E. Doelker, Dexamethasone-containing PLGA superparamagnetic microparticles as carriers for the local treatment of arthritis, Biomaterials, vol.30, issue.9, pp.1772-80, 2009.

H. Zille, J. Paquet, and C. Henrionnet, Evaluation of intra-articular delivery of hyaluronic acid functionalized biopolymeric nanoparticles in healthy rat knees, Biomed. Mater. Eng, vol.20, issue.3, pp.235-242, 2010.

M. Saravanan, K. Bhaskar, G. Maharajan, and K. S. Pillai, Development of gelatin microspheres loaded with diclofenac sodium for intra-articular administration, J. Drug Target, vol.19, issue.2, pp.96-103, 2011.

Z. Zhang and G. Huang, Intra-articular lornoxicam loaded PLGA microspheres: enhanced therapeutic efficiency and decreased systemic toxicity in the treatment of osteoarthritis, Drug Deliv, vol.19, issue.5, pp.255-263, 2012.

A. Panusa, F. Selmin, G. Rossoni, M. Carini, F. Cilurzo et al., Methylprednisolone-loaded PLGA microspheres: a new formulation for sustained release via intra-articular administration. A comparison study with methylprednisolone acetate in rats, J. Pharm. Sci, vol.100, issue.11, pp.4580-4586, 2011.

R. Eswaramoorthy, C. Chang, S. Wu, G. Wang, C. Ho et al.,

, PLGA microspheres suppresses osteoarthritis progression in rats, Acta Biomater, vol.8, issue.6, pp.2254-62, 2012.

T. Boekhorst, B. Jensen, L. B. Colombo, and S. , MRI-assessed therapeutic effects of locally administered PLGA nanoparticles loaded with anti-inflammatory siRNA in a murine arthritis model, J. Controlled Release, vol.161, issue.3, pp.772-80, 2012.

J. Kawadkar, R. Jain, R. Kishore, A. Pathak, and M. K. Chauhan, Formulation and evaluation of flurbiprofenloaded genipin cross-linked gelatin microspheres for intra-articular delivery, J. Drug Target, vol.21, issue.2, pp.200-210, 2013.

R. E. Whitmire, D. S. Wilson, A. Singh, M. E. Levenston, N. Murthy et al., Self-assembling nanoparticles for intra-articular delivery of anti-inflammatory proteins, Biomaterials, vol.33, issue.30, pp.7665-75, 2012.

A. Gaignaux, J. Réeff, and F. Siepmann, Development and evaluation of sustained-release clonidineloaded PLGA microparticles, Int. J. Pharm, vol.437, issue.1-2, pp.20-28, 2012.

J. Présumey, G. Salzano, and G. Courties, PLGA microspheres encapsulating siRNA anti-TNFalpha: efficient RNAi-mediated treatment of arthritic joints, Eur. J. Pharm. Biopharm, vol.82, issue.3, pp.457-464, 2012.

Z. Chen, W. Liu, and D. Liu, Development of brucine-loaded microsphere/thermally responsive hydrogel combination system for intra-articular administration, J. Controlled Release, vol.162, issue.3, pp.628-635, 2012.

M. Morgen, D. Tung, B. Boras, W. Miller, A. Malfait et al., Nanoparticles for improved local retention after intra-articular injection into the knee joint, Pharm. Res, vol.30, issue.1, pp.257-68, 2013.

S. M. Ryan, J. Mcmorrow, and A. Umerska, An intra-articular salmon calcitonin-based nanocomplex reduces experimental inflammatory arthritis, J. Controlled Release, vol.167, issue.2, pp.120-129, 2013.

J. Ko, Y. Choi, G. Jeong, and G. Im, Sulforaphane-PLGA microspheres for the intra-articular treatment of osteoarthritis, Biomaterials, vol.34, issue.21, pp.5359-5368, 2013.

L. Bédouet, F. Pascale, M. Bonneau, and A. Laurent, In vitro evaluation of S-(+)-ibuprofen as drug candidate for intra-articular drug delivery system, Drug Dev. Ind. Pharm, vol.41, issue.1, pp.85-94, 2015.

L. Bédouet, L. Moine, F. Pascale, V. Nguyen, D. Labarre et al., Synthesis of hydrophilic intraarticular microspheres conjugated to ibuprofen and evaluation of anti-inflammatory activity on articular explants, Int. J. Pharm, vol.459, issue.1-2, pp.51-61, 2014.

M. Higaki, T. Ishihara, N. Izumo, M. Takatsu, and Y. Mizushima, Treatment of experimental arthritis with poly(D, L-lactic/glycolic acid) nanoparticles encapsulating betamethasone sodium phosphate, Ann. Rheum. Dis, vol.64, issue.8, pp.1132-1138, 2005.

N. Zhang, P. R. Wardwell, and R. A. Bader, In vitro efficacy of polysaccharide-based nanoparticles containing disease-modifying antirheumatic drugs, Pharm. Res, vol.31, issue.9, pp.2326-2334, 2014.

J. Dong, D. Jiang, Z. Wang, G. Wu, L. Miao et al., Intra-articular delivery of liposomal celecoxibhyaluronate combination for the treatment of osteoarthritis in rabbit model, Int. J. Pharm, vol.441, issue.1-2, pp.285-290, 2013.

C. Larsen, J. Ostergaard, and S. W. Larsen, Intra-articular depot formulation principles: role in the management of postoperative pain and arthritic disorders, J. Pharm. Sci, vol.97, issue.11, pp.4622-4654, 2008.

J. S. Park, H. N. Yang, and D. G. Woo, Chondrogenesis of human mesenchymal stem cells mediated by the combination of SOX trio SOX5, 6, and 9 genes complexed with PEI-modified PLGA nanoparticles, Biomaterials, vol.32, issue.14, pp.3679-88, 2011.

J. F. Schindler, J. B. Monahan, and W. G. Smith, p38 pathway kinases as anti-inflammatory drug targets, J. Dent. Res, vol.86, issue.9, pp.800-811, 2007.

C. Ding, Drug evaluation : VX-702, a MAP kinase inhibitor for rheumatoid arthritis and acute coronary syndrome, Curr. Opin. Investig. Drugs Lond. Engl, vol.7, issue.11, pp.1020-1025, 2000.

K. Rudolphi, N. Gerwin, N. Verzijl, P. Van-der-kraan, W. Van-den-berg et al., an inhibitor of interleukin-1beta converting enzyme, reduces joint damage in two murine models of osteoarthritis, Osteoarthritis Cartilage, vol.11, issue.10, pp.738-746, 2003.

H. K. Makadia and S. J. Siegel, Poly Lactic-co-Glycolic Acid (PLGA) as Biodegradable Controlled Drug Delivery Carrier, Polymers, vol.3, issue.3, pp.1377-1397, 2011.

F. Danhier, E. Ansorena, J. M. Silva, R. Coco, L. Breton et al., PLGA-based nanoparticles: an overview of biomedical applications, J. Controlled Release, vol.161, issue.2, pp.505-527, 2012.

J. M. Anderson and M. S. Shive, Biodegradation and biocompatibility of PLA and PLGA microspheres, Adv. Drug Deliv. Rev, vol.28, issue.1, pp.5-24, 1997.

H. S. Kwak, N. J. Lee, J. Kim, H. J. Yoo, and J. J. , Meniscal repair in vivo using human chondrocyte-seeded PLGA mesh scaffold pretreated with platelet-rich plasma, J. Tissue Eng. Regen. Med, 2014.

S. C. Ghosh, N. Alpay, S. Klostergaard, and J. , CD44: a validated target for improved delivery of cancer therapeutics, Expert Opin. Ther. Targets, vol.16, issue.7, pp.635-650, 2012.

H. Zhu, N. Mitsuhashi, and A. Klein, The role of the hyaluronan receptor CD44 in mesenchymal stem cell migration in the extracellular matrix, Stem Cells Dayt. Ohio, vol.24, issue.4, pp.928-963, 2006.

C. Christophis, I. Taubert, and G. R. Meseck, Shear Stress Regulates Adhesion and Rolling of CD44+

, Leukemic and Hematopoietic Progenitor Cells on Hyaluronan, Biophys. J, vol.101, issue.3, pp.585-593, 2011.

L. M. Negi, S. Talegaonkar, M. Jaggi, F. J. Ahmad, Z. Iqbal et al., Role of CD44 in tumour progression and strategies for targeting, J. Drug Target, vol.20, issue.7, pp.561-573, 2012.

D. J. Aguiar, W. Knudson, and C. B. Knudson, Internalization of the hyaluronan receptor CD44 by chondrocytes, Exp. Cell Res, vol.252, issue.2, pp.292-302, 1999.

A. Almalik, P. J. Day, and N. Tirelli, HA-coated chitosan nanoparticles for CD44-mediated nucleic acid delivery, Macromol. Biosci, vol.13, issue.12, pp.1671-1680, 2013.

S. Arpicco, C. Lerda, D. Pozza, and E. , Hyaluronic acid-coated liposomes for active targeting of gemcitabine, Eur. J. Pharm. Biopharm, vol.85, issue.3, pp.373-80, 2013.

H. Cho, I. Yoon, and H. Y. Yoon, Polyethylene glycol-conjugated hyaluronic acid-ceramide selfassembled nanoparticles for targeted delivery of doxorubicin, Biomaterials, vol.33, issue.4, pp.1190-1200, 2012.

Y. Gall, Hyaluronic acid: structure, metabolism and implication in cicatrisation, Ann. Dermatol. Vénéréologie, vol.137, issue.1, pp.30-39, 2010.

R. Stern, Hyaluronan catabolism: a new metabolic pathway, Eur. J. Cell Biol, vol.83, issue.7, pp.317-325, 2004.

O. Nagano and H. Saya, Mechanism and biological significance of CD44 cleavage, Cancer Sci, vol.95, issue.12, pp.930-935, 2004.

W. Knudson, G. Chow, and C. B. Knudson, CD44-mediated uptake and degradation of hyaluronan, Matrix Biol. J. Int. Soc. Matrix Biol, vol.21, issue.1, pp.15-23, 2002.

B. D. Lynn, E. A. Turley, and J. I. Nagy, Subcellular distribution, calmodulin interaction, and mitochondrial association of the hyaluronan-binding protein RHAMM in rat brain, J. Neurosci. Res, vol.65, issue.1, pp.6-16, 2001.

E. A. Turley, P. W. Noble, and L. Bourguignon, Signaling Properties of Hyaluronan Receptors, J. Biol. Chem, vol.277, issue.7, pp.4589-4592, 2002.

C. Tolg, R. Poon, R. Fodde, E. A. Turley, and B. A. Alman, Genetic deletion of receptor for hyaluronan-mediated motility (Rhamm) attenuates the formation of aggressive fibromatosis (desmoid tumor), Oncogene, vol.22, issue.44, pp.6873-6882, 2003.

G. M. Campo, A. Avenoso, S. Campo, D. 'ascola, A. Nastasi et al., Small hyaluronan oligosaccharides induce inflammation by engaging both toll-like-4 and CD44 receptors in human chondrocytes, Biochem. Pharmacol, vol.80, issue.4, pp.480-90, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00601166

S. Nedvetzki, E. Gonen, and N. Assayag, RHAMM, a receptor for hyaluronan-mediated motility, compensates for CD44 in inflamed CD44-knockout mice: a different interpretation of redundancy, Proc. Natl. Acad. Sci. U. S. A, vol.101, issue.52, pp.18081-18087, 2004.

D. G. Jackson, R. Prevo, S. Clasper, and S. Banerji, LYVE-1, the lymphatic system and tumor lymphangiogenesis, Trends Immunol, vol.22, issue.6, pp.317-321, 2001.

M. S. Pandey and P. H. Weigel, Hyaluronic Acid Receptor for Endocytosis (HARE)-mediated Endocytosis of

H. Hyaluronan and D. Sulfate, Acetylated Low Density Lipoprotein (AcLDL), but Not Chondroitin Sulfate Types A, C, D, or E, Activates NF-?B-regulated Gene Expression, J. Biol. Chem, vol.289, issue.3, pp.1756-1767, 2014.

S. Ohno, H. Im, C. B. Knudson, and W. Knudson, Hyaluronan oligosaccharides induce matrix metalloproteinase 13 via transcriptional activation of NFkappaB and p38 MAP kinase in articular chondrocytes, J. Biol. Chem, vol.281, issue.26, pp.17952-60, 2006.

N. Volpi, J. Schiller, R. Stern, and L. Soltés, Role, metabolism, chemical modifications and applications of hyaluronan, Curr. Med. Chem, vol.16, issue.14, pp.1718-1745, 2009.

C. E. Schanté, G. Zuber, C. Herlin, and T. F. Vandamme, Chemical modifications of hyaluronic acid for the synthesis of derivatives for a broad range of biomedical applications, Carbohydr. Polym, vol.85, issue.3, pp.469-489, 2011.

J. R. Glass, K. T. Dickerson, K. Stecker, and J. W. Polarek, Characterization of a hyaluronic acid-Arg-Gly-Asp peptide cell attachment matrix, Biomaterials, vol.17, issue.11, pp.1101-1108, 1996.

V. F. Della, V. Crescenzi, L. Callegaro, and . Gellan, Brevet, 1996.

F. D. Valle and A. Romeo, Polysaccharide esters and their salts, Brevet, 1990.

S. Pelletier, P. Hubert, F. Lapicque, E. Payan, and E. Dellacherie, Amphiphilic derivatives of sodium alginate and hyaluronate: synthesis and physico-chemical properties of aqueous dilute solutions, Carbohydr. Polym, vol.43, issue.4, pp.343-349, 2000.

L. W. Moreland, Intra-articular hyaluronan (hyaluronic acid) and hylans for the treatment of osteoarthritis: mechanisms of action, Arthritis Res. Ther, vol.5, issue.2, pp.54-67, 2003.

J. Velema and D. Kaplan, Biopolymer-based biomaterials as scaffolds for tissue engineering, Adv. Biochem. Eng. Biotechnol, vol.102, pp.187-238, 2006.

S. Van-vlierberghe, P. Dubruel, and E. Schacht, Biopolymer-based hydrogels as scaffolds for tissue engineering applications: a review, Biomacromolecules, vol.12, issue.5, pp.1387-1408, 2011.

M. B. Brown and S. A. Jones, Hyaluronic acid: a unique topical vehicle for the localized delivery of drugs to the skin, J. Eur. Acad. Dermatol. Venereol, vol.19, issue.3, pp.308-318, 2005.

D. Nashan, F. Meiss, and M. Müller, Therapeutic strategies for actinic keratoses--a systematic review, Eur. J. Dermatol. EJD, vol.23, issue.1, pp.14-32, 2013.

H. Zhang, S. Huang, X. Yang, and G. Zhai, Current research on hyaluronic acid-drug bioconjugates, Eur. J. Med. Chem, vol.86, pp.310-317, 2014.

G. Bajaj, M. R. Kim, S. I. Mohammed, and Y. Yeo, Hyaluronic acid-based hydrogel for regional delivery of paclitaxel to intraperitoneal tumors, J. Controlled Release, vol.158, issue.3, pp.386-392, 2012.

T. Nolan, R. E. Hands, and S. A. Bustin, Quantification of mRNA using real-time RT-PCR, Nat. Protoc, vol.1, issue.3, pp.1559-1582, 2006.

H. Laroui, Nanosphères polymères à couverture de hyaluronate pour la délivrance ciblée de molécules actives dans le traitement des affections du cartilage, 2007.

H. Zille, Nanoparticules recouvertes de dérivés amphiphiles de hyaluronate pour la vectorisation de molécules d'intérêt pour le traitement des pathologies de l'articulation, 2012.

M. I. Tammi, A. J. Day, and E. A. Turley, Hyaluronan and Homeostasis: A Balancing Act, J. Biol. Chem, vol.277, issue.7, pp.4581-4584, 2002.

J. Panyam, W. Zhou, S. Prabha, S. K. Sahoo, and V. Labhasetwar, Rapid endo-lysosomal escape of poly(DLlactide-co-glycolide) nanoparticles: implications for drug and gene delivery, FASEB J, vol.16, issue.10, pp.1217-1243, 2002.

D. Y. Park, B. Min, D. Kim, B. R. Song, M. Kim et al., Polyethylene wear particles play a role in development of osteoarthritis via detrimental effects on cartilage, meniscus, and synovium. Osteoarthritis Cartilage, vol.21, pp.2021-2029, 2013.

C. Chang, H. Fang, Y. Ho, and H. Huang, Chondrocyte acting as phagocyte to internalize polyethylene wear particles and leads to the elevations of osteoarthritis associated NO and PGE2, Biochem. Biophys. Res. Commun, vol.369, issue.3, pp.884-888, 2008.

M. Kapoor, J. Martel-pelletier, D. Lajeunesse, J. Pelletier, and H. Fahmi, Role of proinflammatory cytokines in the pathophysiology of osteoarthritis, Nat. Rev. Rheumatol, vol.7, issue.1, pp.33-42, 2011.

M. M. Hardy, K. Seibert, and P. T. Manning, Cyclooxygenase 2-dependent prostaglandin E2 modulates cartilage proteoglycan degradation in human osteoarthritis explants, Arthritis Rheum, vol.46, issue.7, pp.1789-1803, 2002.

J. Martel-pelletier, J. Pelletier, and H. Fahmi, Cyclooxygenase-2 and prostaglandins in articular tissues, Semin. Arthritis Rheum, vol.33, issue.3, pp.155-167, 2003.

L. Grossin, C. Cournil-henrionnet, and A. Pinzano, Gene transfer with HSP 70 in rat chondrocytes confers cytoprotection in vitro and during experimental osteoarthritis, FASEB J, vol.20, issue.1, pp.65-75, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00093343

M. Akmal, A. Singh, and A. Anand, The effects of hyaluronic acid on articular chondrocytes, J. Bone Joint Surg. Br, vol.87, issue.8, pp.1143-1149, 2005.

Y. Segawa, T. Muneta, and H. Makino, Mesenchymal stem cells derived from synovium, meniscus, anterior cruciate ligament, and articular chondrocytes share similar gene expression profiles, J. Orthop. Res, vol.27, issue.4, pp.435-441, 2009.

F. Gullo, D. Bari, and C. , Prospective purification of a subpopulation of human synovial mesenchymal stem cells with enhanced chondro-osteogenic potency, Rheumatol. Oxf. Engl, 2013.

I. Sekiya, M. Ojima, and S. Suzuki, Human mesenchymal stem cells in synovial fluid increase in the knee with degenerated cartilage and osteoarthritis, J. Orthop. Res, vol.30, issue.6, pp.943-949, 2012.

S. Koelling, J. Kruegel, and M. Irmer, Migratory chondrogenic progenitor cells from repair tissue during the later stages of human osteoarthritis, Cell Stem Cell, vol.4, issue.4, pp.324-335, 2009.

M. E. Candela, R. Yasuhara, M. Iwamoto, and M. Enomoto-iwamoto, Resident mesenchymal progenitors of articular cartilage, Matrix Biol, vol.39, pp.44-49, 2014.

H. Haleem-smith, E. Argintar, and C. Bush, Biological responses of human mesenchymal stem cells to titanium wear debris particles, J. Orthop. Res, vol.30, issue.6, pp.853-863, 2012.

C. C. Okafor, H. Haleem-smith, P. Laqueriere, P. A. Manner, and R. S. Tuan, Particulate endocytosis mediates biological responses of human mesenchymal stem cells to titanium wear debris, J. Orthop. Res, vol.24, issue.3, pp.461-473, 2006.

C. Vinatier, D. Mrugala, C. Jorgensen, J. Guicheux, and D. Noël, Cartilage engineering : a crucial combination of cells, biomaterials and biofactors, Trends Biotechnol, vol.27, issue.5, pp.307-314, 2009.

A. Rey-rico, J. Frisch, J. K. Venkatesan, G. Schmitt, H. Madry et al., Determination of effective rAAV-mediated gene transfer conditions to support chondrogenic differentiation processes in human primary bone marrow aspirates, Gene Ther, vol.22, issue.1, pp.50-57, 2015.

J. S. Park, H. N. Yang, D. G. Woo, S. Y. Jeon, and K. Park, SOX9 gene plus heparinized TGF-? 3 coated dexamethasone loaded PLGA microspheres for inducement of chondrogenesis of hMSCs, Biomaterials, vol.33, issue.29, pp.7151-63, 2012.

K. Park, J. Sy, J. S. Park, H. N. Yang, and D. G. Woo, Aggrecan-and COMP-loaded poly(L-lactic-coglycolic acid) nanoparticles stimulate chondrogenic differentiation of human mesenchymal stem cells, Stem Cells Dev, vol.23, issue.3, pp.305-322, 2013.

K. Toupet, M. Maumus, and J. Peyrafitte, Long-term detection of human adipose-derived mesenchymal stem cells after intraarticular injection in SCID mice, Arthritis Rheum, vol.65, issue.7, pp.1786-1794, 2013.

K. Toupet, M. Maumus, L. , and P. , Survival and biodistribution of xenogenic adipose mesenchymal stem cells is not affected by the degree of inflammation in arthritis, PloS One, vol.10, issue.1, p.114962, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01834412

C. Manferdini, M. Maumus, and E. Gabusi, Adipose-derived mesenchymal stem cells exert antiinflammatory effects on chondrocytes and synoviocytes from osteoarthritis patients through prostaglandin E2, Arthritis Rheum, vol.65, issue.5, pp.1271-1281, 2013.

E. Horisawa, T. Hirota, and S. Kawazoe, Prolonged anti-inflammatory action of DL-lactide/glycolide copolymer nanospheres containing betamethasone sodium phosphate for an intra-articular delivery system in antigen-induced arthritic rabbit, Pharm. Res, vol.19, issue.4, pp.403-410, 2002.

J. Grisar, M. Munk, and C. W. Steiner, Expression patterns of CD44 and CD44 splice variants in patients with rheumatoid arthritis, Clin. Exp. Rheumatol, vol.30, issue.1, pp.64-72, 2012.

D. W. Gilroy, A. Tomlinson, K. Greenslade, M. P. Seed, and D. A. Willoughby, The effects of cyclooxygenase 2 inhibitors on cartilage erosion and bone loss in a model of Mycobacterium tuberculosis-induced monoarticular arthritis in the rat, Inflammation, vol.22, issue.5, pp.509-519, 1998.

L. Jiang, L. Li, and C. Geng, Monosodium iodoacetate induces apoptosis via the mitochondrial pathway involving ROS production and caspase activation in rat chondrocytes in vitro, J. Orthop. Res, vol.31, issue.3, pp.364-369, 2013.

C. Guingamp, P. Gegout-pottie, P. L. Terlain, B. Netter, P. Gillet et al., Mono-iodoacetate-induced experimental osteoarthritis: a dose-response study of loss of mobility, morphology, and biochemistry, Arthritis Rheum, vol.40, issue.9, pp.1670-1679, 1997.

C. Cournil-henrionnet, Potentialités chondroprotectrices d'Hsp70 vis-à-vis des phénomènes apoptotiques : Études in vitro et in vivo chez le rat, 2003.

M. Pickarski, T. Hayami, Y. Zhuo, and L. T. Duong, Molecular changes in Articular Cartilage and Subchondral Bone in the Rat Anterior Cruciate Ligament Transection and Meniscectomized Models of Osteoarthritis, BMC Musculoskelet. Disord, vol.12, issue.1, p.197, 2011.

A. Braza-boïls, M. J. Alcaraz, and M. L. Ferrándiz, Regulation of the inflammatory response by tin protoporphyrin IX in the rat anterior cruciate ligament transection model of osteoarthritis, J. Orthop. Res, vol.29, issue.9, pp.1375-82, 2011.

V. Calamia, L. Lourido, and P. Fernández-puente, Secretome analysis of chondroitin sulfate-treated chondrocytes reveals anti-angiogenic, anti-inflammatory and anti-catabolic properties, Arthritis Res. Ther, vol.14, issue.5, p.202, 2012.

R. E. Wilusz, J. Sanchez-adams, and F. Guilak, The structure and function of the pericellular matrix of articular cartilage, Matrix Biol, vol.39, pp.25-32, 2014.

T. L. Vincent, Targeting mechanotransduction pathways in osteoarthritis: a focus on the pericellular matrix, Curr. Opin. Pharmacol, vol.13, issue.3, pp.449-454, 2013.

O. Horikawa, H. Nakajima, and T. Kikuchi, Distribution of type VI collagen in chondrocyte microenvironment: study of chondrons isolated from human normal and degenerative articular cartilage and cultured chondrocytes, J. Orthop. Sci, vol.9, issue.1, pp.29-36, 2004.

M. Afshari, K. Derakhshandeh, and L. Hosseinzadeh, Characterisation, cytotoxicity and apoptosis studies of methotrexate-loaded PLGA and PLGA-PEG nanoparticles, J. Microencapsul, vol.31, issue.3, pp.239-245, 2014.

S. S. Abolmaali, A. M. Tamaddon, and R. Dinarvand, A review of therapeutic challenges and achievements of methotrexate delivery systems for treatment of cancer and rheumatoid arthritis, Cancer Chemother. Pharmacol, vol.71, issue.5, pp.1115-1130, 2013.

D. A. Fox, Kinase inhibition--a new approach to the treatment of rheumatoid arthritis, N. Engl. J. Med, vol.367, issue.6, pp.565-567, 2012.

L. A. Macfarlane and D. J. Todd, Kinase inhibitors: the next generation of therapies in the treatment of rheumatoid arthritis, Int. J. Rheum. Dis, vol.17, issue.4, pp.359-368, 2014.

G. R. Burmester, R. Blanco, and C. Charles-schoeman, CP-690,550) in combination with methotrexate in patients with active rheumatoid arthritis with an inadequate response to tumour necrosis factor inhibitors: a randomised phase 3 trial, Lancet, vol.381, issue.9865, pp.451-460, 2013.

S. Lotz, S. Goderie, and N. Tokas, Sustained Levels of FGF2 Maintain Undifferentiated Stem Cell Cultures with Biweekly Feeding, PLoS ONE, vol.8, issue.2, p.56289, 2013.

J. S. Park, H. N. Yang, D. G. Woo, S. Y. Jeon, and K. Park, The promotion of chondrogenesis, osteogenesis, and adipogenesis of human mesenchymal stem cells by multiple growth factors incorporated into nanosphere-coated microspheres, Biomaterials, vol.32, issue.1, pp.28-38, 2011.

J. Kim, J. S. Park, and H. N. Yang, The use of biodegradable PLGA nanoparticles to mediate SOX9 gene delivery in human mesenchymal stem cells (hMSCs) and induce chondrogenesis, Biomaterials, vol.32, issue.1, pp.268-78, 2011.

S. Y. Jeon, J. S. Park, H. N. Yang, D. G. Woo, and K. Park, Co-delivery of SOX9 genes and anti-Cbfa-1 siRNA coated onto PLGA nanoparticles for chondrogenesis of human MSCs, Biomaterials, vol.33, issue.17, pp.4413-4436, 2012.

Y. Patil and J. Panyam, Polymeric nanoparticles for siRNA delivery and gene silencing, Int. J. Pharm, vol.367, issue.1-2, pp.195-203, 2009.

M. Gharagozloo, S. Majewski, and M. Foldvari, Therapeutic applications of nanomedicine in autoimmune diseases: From immunosuppression to tolerance induction, Nanomedicine Nanotechnol. Biol. Med, vol.11, issue.4, pp.1003-1018, 2015.

J. Paquet, Potentialités anti-inflammatoires de l'inhibition génomique et transcriptionnelle du TNF alpha par une approche de type oligonucléotidique, 2010.

J. Paquet, C. Henrionnet, and A. Pinzano, Alternative for anti-TNF antibodies for arthritis treatment, Mol. Ther, vol.19, issue.10, pp.1887-95, 2011.