用户名: 密码: 验证码:
Physicochemical and Biomechanical Stimuli in Cell-Based Articular Cartilage Repair
详细信息    查看全文
  • 作者:Holger Jahr (1) (2)
    Csaba Matta (2) (3) (4)
    Ali Mobasheri (2) (3) (5) (6)

    1. Department of Orthopaedic Surgery
    ; University Hospital RWTH Aachen University ; Pauwelsstra脽e 30 ; 52074 ; Aachen ; Germany
    2. The D-BOARD European Consortium for Biomarker Discovery
    ; Surrey ; UK
    3. Department of Veterinary Preclinical Sciences
    ; School of Veterinary Medicine ; Faculty of Health and Medical Sciences ; University of Surrey ; Duke of Kent Building ; Guildford ; Surrey ; GU2 7XH ; UK
    4. Department of Anatomy
    ; Histology and Embryology ; Faculty of Medicine ; University of Debrecen ; Nagyerdei krt. 98 ; Debrecen ; 4032 ; Hungary
    5. Arthritis Research UK Centre for Sport
    ; Exercise and Osteoarthritis ; Arthritis Research UK Pain Centre ; Medical Research Council and Arthritis Research UK Centre for Musculoskeletal Ageing Research ; University of Nottingham ; Queen鈥檚 Medical Centre ; Nottingham ; NG7 2UH ; UK
    6. Center of Excellence in Genomic Medicine Research (CEGMR)
    ; King Fahd Medical Research Center (KFMRC) ; King AbdulAziz University ; Jeddah ; 21589 ; Kingdom of Saudi Arabia
  • 关键词:Articular cartilage ; Cartilage repair ; Regenerative medicine ; Autologous chondrocyte implantation ; Chondrocyte metabolism ; Functional tissue engineering ; Cartilage bioengineering ; Mesenchymal stem cell ; Intracellular signaling pathways
  • 刊名:Current Rheumatology Reports
  • 出版年:2015
  • 出版时间:March 2015
  • 年:2015
  • 卷:17
  • 期:3
  • 全文大小:1,289 KB
  • 参考文献:1. Mow, VC, Ratcliffe, A, Poole, AR (1992) Cartilage and diarthrodial joints as paradigms for hierarchical materials and structures. Biomaterials 13: pp. 67-97
    2. Lesperance, LM, Gray, ML, Burstein, D (1992) Determination of fixed charge density in cartilage using nuclear magnetic resonance. J Orthop Res 10: pp. 1-13
    3. Mow, VCYGW, Hui, CF (2005) Structure and function of articular cartilage and meniscus. Basic orthopaedic biomechanics and mechanobiology. Lippincott Williams & Wilkins, Philadelphia (PA)
    4. Mow, VC, Ateshian, GA, Spilker, RL (1993) Biomechanics of diarthrodial joints: a review of twenty years of progress. J Biomech Eng 115: pp. 460-7
    5. Maroudas, AI (1976) Balance between swelling pressure and collagen tension in normal and degenerate cartilage. Nature 260: pp. 808-9
    6. Lai, WM, Mow, VC, Sun, DD, Ateshian, GA (2000) On the electric potentials inside a charged soft hydrated biological tissue: streaming potential versus diffusion potential. J Biomech Eng 122: pp. 336-46
    7. Bassett, CA, Pawluk, RJ (1972) Electrical behavior of cartilage during loading. Science 178: pp. 982-3
    8. Buschmann, MD, Grodzinsky, AJ (1995) A molecular model of proteoglycan-associated electrostatic forces in cartilage mechanics. J Biomech Eng 117: pp. 179-92
    9. Chen, AC, Nguyen, TT, Sah, RL (1997) Streaming potentials during the confined compression creep test of normal and proteoglycan-depleted cartilage. Ann Biomed Eng 25: pp. 269-77
    10. Mow, VC, Guo, XE (2002) Mechano-electrochemical properties of articular cartilage: their inhomogeneities and anisotropies. Annu Rev Biomed Eng 4: pp. 175-209
    11. Mow, VC, Wang, CC, Hung, CT (1999) The extracellular matrix, interstitial fluid and ions as a mechanical signal transducer in articular cartilage. Osteoarthr Cartil 7: pp. 41-58
    12. Little, CJ, Bawolin, NK, Chen, X (2011) Mechanical properties of natural cartilage and tissue-engineered constructs. Tissue Eng B Rev 17: pp. 213-27
    13. Smith, GD, Knutsen, G, Richardson, JB (2005) A clinical review of cartilage repair techniques. J Bone Joint Surg (Br) 87: pp. 445-9
    14. Domarad, BR, Buschmann, MT (1995) Interviewing older adults: increasing the credibility of interview data. J Gerontol Nurs 21: pp. 14-20
    15. Jordan, MA, Thiel, GS, Chahal, J, Nho, SJ (2012) Operative treatment of chondral defects in the hip joint: a systematic review. Curr Rev Musculoskelet Med 5: pp. 244-53
    16. Zhang, W, Moskowitz, RW, Nuki, G, Abramson, S, Altman, RD, Arden, N (2007) OARSI recommendations for the management of hip and knee osteoarthritis, part I: critical appraisal of existing treatment guidelines and systematic review of current research evidence. Osteoarthr Cart 15: pp. 981-1000
    17. Heinegard, D, Saxne, T (2011) The role of the cartilage matrix in osteoarthritis. Nat Rev Rheumatol 7: pp. 50-6
    18. Brady MA, Waldman SD, Ethier CR. The Application of multiple biophysical cues to engineer functional neo-cartilage for treatment of osteoarthritis (part I: cellular response). Tissue Eng Part B Rev. 2014. / This recent paper highlights the fact that tissue engineering strategies and the development of 鈥渟mart鈥?functionalized biomaterials that enable the delivery of growth factors or integration of conjugated nanoparticles may benefit from targeting known signal transduction pathways in combination with external biophysical cues.
    19. Grodzinsky, AJ, Levenston, ME, Jin, M, Frank, EH (2000) Cartilage tissue remodeling in response to mechanical forces. Annu Rev Biomed Eng 2: pp. 691-713
    20. Bondeson, J, Wainwright, S, Hughes, C, Caterson, B (2008) The regulation of the ADAMTS4 and ADAMTS5 aggrecanases in osteoarthritis: a review. Clin Exp Rheumatol 26: pp. 139-45
    21. Malemud, CJ, Papay, RS, Hering, TM, Holderbaum, D, Goldberg, VM, Haqqi, TM (1995) Phenotypic modulation of newly synthesized proteoglycans in human cartilage and chondrocytes. Osteoarthr Cart 3: pp. 227-38
    22. Nelson, F, Billinghurst, RC, Pidoux, I, Reiner, A, Langworthy, M, McDermott, M (2006) Early post-traumatic osteoarthritis-like changes in human articular cartilage following rupture of the anterior cruciate ligament. Osteoarthr Cart 14: pp. 114-9
    23. Williamson, AK, Chen, AC, Sah, RL (2001) Compressive properties and function-composition relationships of developing bovine articular cartilage. J Orthop Res 19: pp. 1113-21
    24. Bank, RA, Soudry, M, Maroudas, A, Mizrahi, J, TeKoppele, JM (2000) The increased swelling and instantaneous deformation of osteoarthritic cartilage is highly correlated with collagen degradation. Arthritis Rheum 43: pp. 2202-10
    25. Brittberg, M, Lindahl, A, Nilsson, A, Ohlsson, C, Isaksson, O, Peterson, L (1994) Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation. N Engl J Med 331: pp. 889-95
    26. Saris, DB, Vanlauwe, J, Victor, J, Haspl, M, Bohnsack, M, Fortems, Y (2008) Characterized chondrocyte implantation results in better structural repair when treating symptomatic cartilage defects of the knee in a randomized controlled trial versus microfracture. Am J Sports Med 36: pp. 235-46
    27. Kock, L, Donkelaar, CC, Ito, K (2012) Tissue engineering of functional articular cartilage: the current status. Cell Tissue Res 347: pp. 613-27
    28. Mansour, JM, Welter, JF (2013) Multimodal evaluation of tissue-engineered cartilage. J Med Biol Eng 33: pp. 1-16
    29. Miot, S, Scandiucci de Freitas, P, Wirz, D, Daniels, AU, Sims, TJ, Hollander, AP (2006) Cartilage tissue engineering by expanded goat articular chondrocytes. J Orthop Res 24: pp. 1078-85
    30. Waldman, SD, Grynpas, MD, Pilliar, RM, Kandel, RA (2003) The use of specific chondrocyte populations to modulate the properties of tissue-engineered cartilage. J Orthop Res 21: pp. 132-8
    31. Darling, EM, Athanasiou, KA (2005) Retaining zonal chondrocyte phenotype by means of novel growth environments. Tissue Eng 11: pp. 395-403
    32. Darling, EM, Hu, JC, Athanasiou, KA (2004) Zonal and topographical differences in articular cartilage gene expression. J Orthop Res 22: pp. 1182-7
    33. Khan, IM, Gilbert, SJ, Singhrao, SK, Duance, VC, Archer, CW (2008) Cartilage integration: evaluation of the reasons for failure of integration during cartilage repair. A review. Eur Cell Mater 16: pp. 26-39
    34. Gilbert, SJ, Singhrao, SK, Khan, IM, Gonzalez, LG, Thomson, BM, Burdon, D (2009) Enhanced tissue integration during cartilage repair in vitro can be achieved by inhibiting chondrocyte death at the wound edge. Tissue Eng A 15: pp. 1739-49
    35. Snyder, MJ, Wilensky, JA, Fortin, JD (2002) Current applications of electrotherapeutics in collagen healing. Pain Physician 5: pp. 172-81
    36. Zuzzi, DC, Ciccone Cde, C, Neves, LM, Mendonca, JS, Joazeiro, PP, Esquisatto, MA (2012) Evaluation of the effects of electrical stimulation on cartilage repair in adult male rats. Tissue Cell 45: pp. 275-81
    37. Balint, R, Cassidy, NJ, Cartmell, SH (2013) Electrical stimulation: a novel tool for tissue engineering. Tissue Eng B Rev 19: pp. 48-57
    38. Armstrong, PF, Brighton, CT, Star, AM (1988) Capacitively coupled electrical stimulation of bovine growth plate chondrocytes grown in pellet form. J Orthop Res 6: pp. 265-71
    39. Brighton, CT, Jensen, L, Pollack, SR, Tolin, BS, Clark, CC (1989) Proliferative and synthetic response of bovine growth plate chondrocytes to various capacitively coupled electrical fields. J Orthop Res 7: pp. 759-65
    40. Brighton, CT, Wang, W, Clark, CC (2008) The effect of electrical fields on gene and protein expression in human osteoarthritic cartilage explants. J Bone Joint Surg Am 90: pp. 833-48
    41. Assiotis, A, Sachinis, NP, Chalidis, BE (2012) Pulsed electromagnetic fields for the treatment of tibial delayed unions and nonunions. A prospective clinical study and review of the literature. J Orthop Surg Res 7: pp. 24
    42. Hannemann, PF, Mommers, EH, Schots, JP, Brink, PR, Poeze, M (2014) The effects of low-intensity pulsed ultrasound and pulsed electromagnetic fields bone growth stimulation in acute fractures: a systematic review and meta-analysis of randomized controlled trials. Arch Orthop Trauma Surg 134: pp. 1093-106
    43. Chalidis, B, Sachinis, N, Assiotis, A, Maccauro, G (2011) Stimulation of bone formation and fracture healing with pulsed electromagnetic fields: biologic responses and clinical implications. Int J Immunopathol Pharmacol 24: pp. 17-20
    44. Jansen, JH, Jagt, OP, Punt, BJ, Verhaar, JA, Leeuwen, JP, Weinans, H (2010) Stimulation of osteogenic differentiation in human osteoprogenitor cells by pulsed electromagnetic fields: an in vitro study. BMC Musculoskelet Disord 11: pp. 188
    45. Dini, L, Abbro, L (2005) Bioeffects of moderate-intensity static magnetic fields on cell cultures. Micron 36: pp. 195-217
    46. Fini, M, Giavaresi, G, Carpi, A, Nicolini, A, Setti, S, Giardino, R (2005) Effects of pulsed electromagnetic fields on articular hyaline cartilage: review of experimental and clinical studies. Biomed Pharmacother 59: pp. 388-94
    47. Vavken, P, Arrich, F, Schuhfried, O, Dorotka, R (2009) Effectiveness of pulsed electromagnetic field therapy in the management of osteoarthritis of the knee: a meta-analysis of randomized controlled trials. J Rehabil Med 41: pp. 406-11
    48. Sadoghi, P, Leithner, A, Dorotka, R, Vavken, P (2013) Effect of pulsed electromagnetic fields on the bioactivity of human osteoarthritic chondrocytes. Orthopedics 36: pp. e360-5
    49. Fioravanti, A, Nerucci, F, Collodel, G, Markoll, R, Marcolongo, R (2002) Biochemical and morphological study of human articular chondrocytes cultivated in the presence of pulsed signal therapy. Ann Rheum Dis 61: pp. 1032-3
    50. Vincenzi, F, Targa, M, Corciulo, C, Gessi, S, Merighi, S, Setti, S (2013) Pulsed electromagnetic fields increased the anti-inflammatory effect of A(2)A and A(3) adenosine receptors in human T/C-28a2 chondrocytes and hFOB 1.19 osteoblasts. PLoS One 8: pp. e65561
    51. Ongaro, A, Pellati, A, Masieri, FF, Caruso, A, Setti, S, Cadossi, R (2011) Chondroprotective effects of pulsed electromagnetic fields on human cartilage explants. Bioelectromagnetics 32: pp. 543-51
    52. Luo, Q, Li, SS, He, C, He, H, Yang, L, Deng, L (2009) Pulse electromagnetic fields effects on serum E2 levels, chondrocyte apoptosis, and matrix metalloproteinase-13 expression in ovariectomized rats. Rheumatol Int 29: pp. 927-35
    53. Mizuno, S, Ogawa, R (2011) Using changes in hydrostatic and osmotic pressure to manipulate metabolic function in chondrocytes. Am J Physiol Cell Physiol 300: pp. C1234-45
    54. Urban, JP (1994) The chondrocyte: a cell under pressure. Br J Rheumatol 33: pp. 901-8
    55. Holmvall, K, Camper, L, Johansson, S, Kimura, JH, Lundgren-Akerlund, E (1995) Chondrocyte and chondrosarcoma cell integrins with affinity for collagen type II and their response to mechanical stress. Exp Cell Res 221: pp. 496-503
    56. Vunjak-Novakovic G, Goldstein S. Biomechanical principles of cartilage and bone tissue engineering. Basic orthopaedic biomechanics and mechano-biology. Lippincott Williams and Wilkins; 2005.
    57. Lee C, Grad S, Wimmer M, Alini M. mechanical stimuli in articular cartilage tissue engineering. Topics in Tissue Engineering. 2006.
    58. Vanderploeg, EJ, Wilson, CG, Levenston, ME (2008) Articular chondrocytes derived from distinct tissue zones differentially respond to in vitro oscillatory tensile loading. Osteoarthr Cart 16: pp. 1228-36
    59. Li, Z, Yao, S, Alini, M, Grad, S (2007) Different response of articular chondrocyte subpopulations to surface motion. Osteoarthr Cart 15: pp. 1034-41
    60. Grad, S, Eglin, D, Alini, M, Stoddart, MJ (2011) Physical stimulation of chondrogenic cells in vitro: a review. Clin Orthop Relat Res 469: pp. 2764-72
    61. Unadkat, HV, Hulsman, M, Cornelissen, K, Papenburg, BJ, Truckenmuller, RK, Carpenter, AE (2011) An algorithm-based topographical biomaterials library to instruct cell fate. Proc Natl Acad Sci U S A 108: pp. 16565-70
    62. Park S, Im GI. Stem cell responses to nanotopography. J Biomed Mater Res A. 2014. / This review provides an overview of the effects of nanotopography on cell behavior in the context of musculoskeletal regeneration.
    63. Yim, EK, Sheetz, MP (2012) Force-dependent cell signaling in stem cell differentiation. Stem Cell Res Ther 3: pp. 41
    64. Discher, DE, Janmey, P, Wang, YL (2005) Tissue cells feel and respond to the stiffness of their substrate. Science 310: pp. 1139-43
    65. Janmey, PA, Wells, RG, Assoian, RK, McCulloch, CA (2013) From tissue mechanics to transcription factors. Differentiation 86: pp. 112-20
    66. Cigognini, D, Lomas, A, Kumar, P, Satyam, A, English, A, Azeem, A (2013) Engineering in vitro microenvironments for cell based therapies and drug discovery. Drug Discov Today 18: pp. 1099-108
    67. Orsi G, Fagnano M, De Maria C, Montemurro F, Vozzi G. A new 3D concentration gradient maker and its application in building hydrogels with a 3D stiffness gradient. J Tissue Eng Regen Med. 2014.
    68. Schuh, E, Hofmann, S, Stok, K, Notbohm, H, Muller, R, Rotter, N (2012) Chondrocyte redifferentiation in 3D: the effect of adhesion site density and substrate elasticity. J Biomed Mater Res A 100: pp. 38-47
    69. Jiang, F, Horber, H, Howard, J, Muller, DJ (2004) Assembly of collagen into microribbons: effects of pH and electrolytes. J Struct Biol 148: pp. 268-78
    70. Little, WC, Smith, ML, Ebneter, U, Vogel, V (2008) Assay to mechanically tune and optically probe fibrillar fibronectin conformations from fully relaxed to breakage. Matrix Biol 27: pp. 451-61
    71. Smith, ML, Gourdon, D, Little, WC, Kubow, KE, Eguiluz, RA, Luna-Morris, S (2007) Force-induced unfolding of fibronectin in the extracellular matrix of living cells. PLoS Biol 5: pp. e268
    72. Lee J, Abdeen AA, Kilian KA. Rewiring mesenchymal stem cell lineage specification by switching the biophysical microenvironment. Sci Rep. 2014;4:5188. doi:10.1038/srep05188 . / This important new study demonstrates that MSCs remain susceptible to the biophysical properties of the extracellular matrix鈥攅ven after several weeks of culture鈥攁nd can redirect lineage specification in response to changes in the microenvironment.
    73. Liu, C, Baek, S, Kim, J, Vasko, E, Pyne, R, Chan, C (2014) Effect of static pre-stretch induced surface anisotropy on orientation of mesenchymal stem cells. Cell Mol Bioeng 7: pp. 106-21
    74. Li, Y, Chu, JS, Kurpinski, K, Li, X, Bautista, DM, Yang, L (2011) Biophysical regulation of histone acetylation in mesenchymal stem cells. Biophys J 100: pp. 1902-9
    75. Wang, N, Tytell, JD, Ingber, DE (2009) Mechanotransduction at a distance: mechanically coupling the extracellular matrix with the nucleus. Nat Rev Mol Cell Biol 10: pp. 75-82
    76. Gieni, RS, Hendzel, MJ (2008) Mechanotransduction from the ECM to the genome: are the pieces now in place?. J Cell Biochem 104: pp. 1964-87
    77. Crisp, M, Liu, Q, Roux, K, Rattner, JB, Shanahan, C, Burke, B (2006) Coupling of the nucleus and cytoplasm: role of the LINC complex. J Cell Biol 172: pp. 41-53
    78. Steinmetz, NJ, Bryant, SJ (2012) Chondroitin sulfate and dynamic loading alter chondrogenesis of human MSCs in PEG hydrogels. Biotechnol Bioeng 109: pp. 2671-82
    79. Mhanna, R, Kashyap, A, Palazzolo, G, Vallmajo-Martin, Q, Becher, J, Moller, S (2014) Chondrocyte culture in three dimensional alginate sulfate hydrogels promotes proliferation while maintaining expression of chondrogenic markers. Tissue Eng A 20: pp. 1454-64
    80. Govey, PM, Loiselle, AE, Donahue, HJ (2013) Biophysical regulation of stem cell differentiation. Curr Osteoporos Rep 11: pp. 83-91
    81. Trappmann, B, Chen, CS (2013) How cells sense extracellular matrix stiffness: a material鈥檚 perspective. Curr Opin Biotechnol 24: pp. 948-53
    82. Breuls, RG, Jiya, TU, Smit, TH (2008) Scaffold stiffness influences cell behavior: opportunities for skeletal tissue engineering. Open Orthop J 2: pp. 103-9
    83. Hing, WA, Sherwin, AF, Poole, CA (2002) The influence of the pericellular microenvironment on the chondrocyte response to osmotic challenge. Osteoarthr Cart 10: pp. 297-307
    84. Likhitpanichkul, M, Guo, XE, Mow, VC (2005) The effect of matrix tension-compression nonlinearity and fixed negative charges on chondrocyte responses in cartilage. Mol Cell Biomech 2: pp. 191-204
    85. Lai, WM, Gu, WY, Mow, VC (1998) On the conditional equivalence of chemical loading and mechanical loading on articular cartilage. J Biomech 31: pp. 1181-5
    86. Schneiderman, R, Keret, D, Maroudas, A (1986) 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 4: pp. 393-408
    87. Guilak, F, Erickson, GR, Ting-Beall, HP (2002) The effects of osmotic stress on the viscoelastic and physical properties of articular chondrocytes. Biophys J 82: pp. 720-7
    88. Cheung, CY, Ko, BC (2013) NFAT5 in cellular adaptation to hypertonic stress鈥攔egulations and functional significance. J Mol Signal 8: pp. 5
    89. Jeon, US, Kim, JA, Sheen, MR, Kwon, HM (2006) How tonicity regulates genes: story of TonEBP transcriptional activator. Acta Physiol (Oxf) 187: pp. 241-7
    90. Lee, SD, Choi, SY, Lim, SW, Lamitina, ST, Ho, SN, Go, WY (2011) TonEBP stimulates multiple cellular pathways for adaptation to hypertonic stress: organic osmolyte-dependent and -independent pathways. Am J Physiol Renal Physiol 300: pp. F707-15
    91. Halterman, JA, Kwon, HM, Wamhoff, BR (2012) Tonicity-independent regulation of the osmosensitive transcription factor TonEBP (NFAT5). Am J Physiol Cell Physiol 302: pp. C1-8
    92. Windt, AE, Haak, E, Das, RH, Kops, N, Welting, TJ, Caron, MM (2010) Physiological tonicity improves human chondrogenic marker expression through nuclear factor of activated T-cells 5 in vitro. Arthritis Res Ther 12: pp. R100
    93. Windt, AE, Haak, E, Kops, N, Verhaar, JA, Weinans, H, Jahr, H (2012) Inhibiting calcineurin activity under physiologic tonicity elevates anabolic but suppresses catabolic chondrocyte markers. Arthritis Rheum 64: pp. 1929-39
    94. Koo, J, Kim, KI, Min, BH, Lee, GM (2010) Controlling medium osmolality improves the expansion of human articular chondrocytes in serum-free media. Tissue Eng C Methods 16: pp. 957-63
    95. Koo, J, Kim, KI, Min, BH, Lee, GM (2010) Differential protein expression in human articular chondrocytes expanded in serum-free media of different medium osmolalities by DIGE. J Proteome Res 9: pp. 2480-7
    96. Peffers, MJ, Milner, PI, Tew, SR, Clegg, PD (2010) Regulation of SOX9 in normal and osteoarthritic equine articular chondrocytes by hyperosmotic loading. Osteoarthr Cart 18: pp. 1502-8
    97. Tew, SR, Peffers, MJ, McKay, TR, Lowe, ET, Khan, WS, Hardingham, TE (2009) Hyperosmolarity regulates SOX9 mRNA posttranscriptionally in human articular chondrocytes. Am J Physiol Cell Physiol 297: pp. C898-906
    98. Tsai TT, Danielson KG, Guttapalli A, Oguz E, Albert TJ, Shapiro IM, et al. TonEBP/OREBP is a regulator of nucleus pulposus cell function and survival in the intervertebral disc. J Biol Chem. 2006;281:25416鈥?5424.
    99. Gajghate, S, Hiyama, A, Shah, M, Sakai, D, Anderson, DG, Shapiro, IM (2009) Osmolarity and intracellular calcium regulate aquaporin2 expression through TonEBP in nucleus pulposus cells of the intervertebral disc. J Bone Miner Res 24: pp. 992-1001
    100. Tsai, TT, Guttapalli, A, Agrawal, A, Albert, TJ, Shapiro, IM, Risbud, MV (2007) MEK/ERK signaling controls osmoregulation of nucleus pulposus cells of the intervertebral disc by transactivation of TonEBP/OREBP. J Bone Miner Res 22: pp. 965-74
    101. Pingguan-Murphy, B, El-Azzeh, M, Bader, DL, Knight, MM (2006) Cyclic compression of chondrocytes modulates a purinergic calcium signalling pathway in a strain rate- and frequency-dependent manner. J Cell Physiol 209: pp. 389-97
    102. Hiyama, A, Gajghate, S, Sakai, D, Mochida, J, Shapiro, IM, Risbud, MV (2009) Activation of TonEBP by calcium controls {beta}1,3-glucuronosyltransferase-I expression, a key regulator of glycosaminoglycan synthesis in cells of the intervertebral disc. J Biol Chem 284: pp. 9824-34
    103. Erickson, GR, Alexopoulos, LG, Guilak, F (2001) Hyper-osmotic stress induces volume change and calcium transients in chondrocytes by transmembrane, phospholipid, and G-protein pathways. J Biomech 34: pp. 1527-35
    104. Pritchard, S, Votta, BJ, Kumar, S, Guilak, F (2008) Interleukin-1 inhibits osmotically induced calcium signaling and volume regulation in articular chondrocytes. Osteoarthr Cart 16: pp. 1466-73
    105. Sanchez, JC, Lopez-Zapata, DF (2010) Effects of osmotic challenges on membrane potential in human articular chondrocytes from healthy and osteoarthritic cartilage. Biorheology 47: pp. 321-31
    106. Korhonen, RK, Han, SK, Herzog, W (2010) Osmotic loading of articular cartilage modulates cell deformations along primary collagen fibril directions. J Biomech 43: pp. 783-7
    107. Finan, JD, Chalut, KJ, Wax, A, Guilak, F (2009) Nonlinear osmotic properties of the cell nucleus. Ann Biomed Eng 37: pp. 477-91
    108. Szafranski, JD, Grodzinsky, AJ, Burger, E, Gaschen, V, Hung, HH, Hunziker, EB (2004) Chondrocyte mechanotransduction: effects of compression on deformation of intracellular organelles and relevance to cellular biosynthesis. Osteoarthr Cart 12: pp. 937-46
    109. Oswald, ES, Chao, PH, Bulinski, JC, Ateshian, GA, Hung, CT (2008) Dependence of zonal chondrocyte water transport properties on osmotic environment. Cell Mol Bioeng 1: pp. 339-48
    110. Xu, X, Urban, JP, Tirlapur, UK, Cui, Z (2010) Osmolarity effects on bovine articular chondrocytes during three-dimensional culture in alginate beads. Osteoarthr Cart 18: pp. 433-9
    111. Sun, DD, Guo, XE, Likhitpanichkul, M, Lai, WM, Mow, VC (2004) The influence of the fixed negative charges on mechanical and electrical behaviors of articular cartilage under unconfined compression. J Biomech Eng 126: pp. 6-16
    112. Matta, C, Mobasheri, A, Gergely, P, Zakany, R (2014) Ser/Thr-phosphoprotein phosphatases in chondrogenesis: neglected components of a two-player game. Cell Signal 26: pp. 2175-85
    113. Windt, AE, Jahr, H, Farrell, E, Verhaar, JA, Weinans, H, Osch, GJ (2010) Calcineurin inhibitors promote chondrogenic marker expression of dedifferentiated human adult chondrocytes via stimulation of endogenous TGFbeta1 production. Tissue Eng A 16: pp. 1-10
    114. Yoo, SA, Park, BH, Yoon, HJ, Lee, JY, Song, JH, Kim, HA (2007) Calcineurin modulates the catabolic and anabolic activity of chondrocytes and participates in the progression of experimental osteoarthritis. Arthritis Rheum 56: pp. 2299-311
    115. Nakamura, Y, Takarada, T, Kodama, A, Hinoi, E, Yoneda, Y (2009) Predominant promotion by tacrolimus of chondrogenic differentiation to proliferating chondrocytes. J Pharmacol Sci 109: pp. 413-23
    116. Reinhold, MI, Abe, M, Kapadia, RM, Liao, Z, Naski, MC (2004) FGF18 represses noggin expression and is induced by calcineurin. J Biol Chem 279: pp. 38209-19
    117. Tomita, M, Reinhold, MI, Molkentin, JD, Naski, MC (2002) Calcineurin and NFAT4 induce chondrogenesis. J Biol Chem 277: pp. 42214-8
    118. Nishigaki, F, Sakuma, S, Ogawa, T, Miyata, S, Ohkubo, T, Goto, T (2002) FK506 induces chondrogenic differentiation of clonal mouse embryonic carcinoma cells, ATDC5. Eur J Pharmacol 437: pp. 123-8
    119. Obayashi, K, Tomonari, M, Yoshimatsu, H, Fukuyama, R, Ieiri, I, Higuchi, S (2011) Dosing time-dependency of the arthritis-inhibiting effect of tacrolimus in mice. J Pharmacol Sci 116: pp. 264-73
    120. Kang, KY, Ju, JH, Song, YW, Yoo, DH, Kim, HY, Park, SH (2013) Tacrolimus treatment increases bone formation in patients with rheumatoid arthritis. Rheumatol Int 33: pp. 2159-63
    121. Crabtree, GR (2001) Calcium, calcineurin, and the control of transcription. J Biol Chem 276: pp. 2313-6
    122. Wu, H, Peisley, A, Graef, IA, Crabtree, GR (2007) NFAT signaling and the invention of vertebrates. Trends Cell Biol 17: pp. 251-60
    123. Ranger, AM, Gerstenfeld, LC, Wang, J, Kon, T, Bae, H, Gravallese, EM (2000) The nuclear factor of activated T cells (NFAT) transcription factor NFATp (NFATc2) is a repressor of chondrogenesis. J Exp Med 191: pp. 9-22
    124. Wang, J, Gardner, BM, Lu, Q, Rodova, M, Woodbury, BG, Yost, JG (2009) Transcription factor Nfat1 deficiency causes osteoarthritis through dysfunction of adult articular chondrocytes. J Pathol 219: pp. 163-72
    125. Greenblatt, MB, Ritter, SY, Wright, J, Tsang, K, Hu, D, Glimcher, LH (2013) NFATc1 and NFATc2 repress spontaneous osteoarthritis. Proc Natl Acad Sci U S A 110: pp. 19914-9
    126. Sitara, D, Aliprantis, AO (2010) Transcriptional regulation of bone and joint remodeling by NFAT. Immunol Rev 233: pp. 286-300
    127. Yaykasli, KO, Oohashi, T, Hirohata, S, Hatipoglu, OF, Inagawa, K, Demircan, K (2009) ADAMTS9 activation by interleukin 1 beta via NFATc1 in OUMS-27 chondrosarcoma cells and in human chondrocytes. Mol Cell Biochem 323: pp. 69-79
    128. Thirunavukkarasu, K, Pei, Y, Moore, TL, Wang, H, Yu, XP, Geiser, AG (2006) Regulation of the human ADAMTS-4 promoter by transcription factors and cytokines. Biochem Biophys Res Commun 345: pp. 197-204
    129. Rodova, M, Lu, Q, Li, Y, Woodbury, BG, Crist, JD, Gardner, BM (2011) Nfat1 regulates adult articular chondrocyte function through its age-dependent expression mediated by epigenetic histone methylation. J Bone Miner Res 26: pp. 1974-86
    130. Lin, SS, Tzeng, BH, Lee, KR, Smith, RJ, Campbell, KP, Chen, CC (2014) Cav3.2 T-type calcium channel is required for the NFAT-dependent Sox9 expression in tracheal cartilage. Proc Natl Acad Sci U S A 111: pp. E1990-8
    131. Matta, C, Fodor, J, Szijgyarto, Z, Juhasz, T, Gergely, P, Csernoch, L (2008) Cytosolic free Ca2+ concentration exhibits a characteristic temporal pattern during in vitro cartilage differentiation: a possible regulatory role of calcineurin in Ca-signalling of chondrogenic cells. Cell Calcium 44: pp. 310-23
    132. Miclea, RL, Siebelt, M, Finos, L, Goeman, JJ, Lowik, CW, Oostdijk, W (2011) Inhibition of Gsk3beta in cartilage induces osteoarthritic features through activation of the canonical Wnt signaling pathway. Osteoarthr Cart 19: pp. 1363-72
    133. Siebelt, M, Windt, AE, Groen, HC, Sandker, M, Waarsing, JH, Muller, C (2014) FK506 protects against articular cartilage collagenous extra-cellular matrix degradation. Osteoarthr Cart 22: pp. 591-600
    134. Zanotti, S, Canalis, E (2013) Notch suppresses nuclear factor of activated T cells (NFAT) transactivation and Nfatc1 expression in chondrocytes. Endocrinology 154: pp. 762-72
    135. Bradley, EW, Drissi, MH (2010) WNT5A regulates chondrocyte differentiation through differential use of the CaN/NFAT and IKK/NF-kappaB pathways. Mol Endocrinol 24: pp. 1581-93
    136. Pitsillides, AA, Beier, F (2011) Cartilage biology in osteoarthritis鈥攍essons from developmental biology. Nat Rev Rheumatol 7: pp. 654-63
    137. Roddy, KA, Prendergast, PJ, Murphy, P (2011) Mechanical influences on morphogenesis of the knee joint revealed through morphological, molecular and computational analysis of immobilised embryos. PLoS One 6: pp. e17526
    138. Sarraf, CE, Otto, WR, Eastwood, M (2011) In vitro mesenchymal stem cell differentiation after mechanical stimulation. Cell Prolif 44: pp. 99-108
    139. Sun, HB (2010) Mechanical loading, cartilage degradation, and arthritis. Ann N Y Acad Sci 1211: pp. 37-50
    140. O鈥機onor, CJ, Case, N, Guilak, F (2013) Mechanical regulation of chondrogenesis. Stem Cell Res Ther 4: pp. 61
    141. Muhammad, H, Rais, Y, Miosge, N, Ornan, EM (2012) The primary cilium as a dual sensor of mechanochemical signals in chondrocytes. Cell Mol Life Sci 69: pp. 2101-7
    142. Wang, Y, Maciejewski, BS, Lee, N, Silbert, O, McKnight, NL, Frangos, JA (2006) Strain-induced fetal type II epithelial cell differentiation is mediated via cAMP-PKA-dependent signaling pathway. Am J Physiol Lung Cell Mol Physiol 291: pp. L820-7
    143. Lee, T, Kim, SJ, Sumpio, BE (2003) Role of PP2A in the regulation of p38 MAPK activation in bovine aortic endothelial cells exposed to cyclic strain. J Cell Physiol 194: pp. 349-55
    144. Juh谩sz T, Matta C, Somogyi C, Katona 脡, Tak谩cs R, Soha RF, et al. Mechanical loading stimulates chondrogenesis via the PKA/CREB-Sox9 and PP2A pathways in chicken micromass cultures. Cell Signal. 2014;26(3):468鈥?2. doi:10.1016/j.cellsig.2013.12.001
    145. Zakany, R, Szucs, K, Bako, E, Felszeghy, S, Czifra, G, Biro, T (2002) Protein phosphatase 2A is involved in the regulation of protein kinase A signaling pathway during in vitro chondrogenesis. Exp Cell Res 275: pp. 1-8
    146. Yoon, YM, Kim, SJ, Oh, CD, Ju, JW, Song, WK, Yoo, YJ (2002) Maintenance of differentiated phenotype of articular chondrocytes by protein kinase C and extracellular signal-regulated protein kinase. J Biol Chem 277: pp. 8412-20
    147. Lee, YA, Kang, SS, Baek, SH, Jung, JC, Jin, EJ, Tak, EN (2007) Redifferentiation of dedifferentiated chondrocytes on chitosan membranes and involvement of PKCalpha and P38 MAP kinase. Mol Cells 24: pp. 9-15
    148. Kawanishi, M, Oura, A, Furukawa, K, Fukubayashi, T, Nakamura, K, Tateishi, T (2007) Redifferentiation of dedifferentiated bovine articular chondrocytes enhanced by cyclic hydrostatic pressure under a gas-controlled system. Tissue Eng 13: pp. 957-64
    149. Matta, C, Zakany, R (2013) Calcium signalling in chondrogenesis: implications for cartilage repair. Front Biosci (Schol Ed) 5: pp. 305-24
    150. Mobasheri, A, Lewis, R, Maxwell, JE, Hill, C, Womack, M, Barrett-Jolley, R (2010) Characterization of a stretch-activated potassium channel in chondrocytes. J Cell Physiol 223: pp. 511-8
    151. O鈥機onor, CJ, Leddy, HA, Benefield, HC, Liedtke, WB, Guilak, F (2014) TRPV4-mediated mechanotransduction regulates the metabolic response of chondrocytes to dynamic loading. Proc Natl Acad Sci U S A 111: pp. 1316-21
    152. Roberts, SR, Knight, MM, Lee, DA, Bader, DL (2001) Mechanical compression influences intracellular Ca2+ signaling in chondrocytes seeded in agarose constructs. J Appl Physiol (1985) 90: pp. 1385-91
    153. Wilsman, NJ (1978) Cilia of adult canine articular chondrocytes. J Ultrastruct Res 64: pp. 270-81
    154. Wann, AK, Zuo, N, Haycraft, CJ, Jensen, CG, Poole, CA, McGlashan, SR (2012) Primary cilia mediate mechanotransduction through control of ATP-induced Ca2+ signaling in compressed chondrocytes. Faseb J 26: pp. 1663-71
  • 刊物主题:Rheumatology;
  • 出版者:Springer US
  • ISSN:1534-6307
文摘
Articular cartilage is a unique load-bearing connective tissue with a low intrinsic capacity for repair and regeneration. Its avascularity makes it relatively hypoxic and its unique extracellular matrix is enriched with cations, which increases the interstitial fluid osmolarity. Several physicochemical and biomechanical stimuli are reported to influence chondrocyte metabolism and may be utilized for regenerative medical approaches. In this review article, we summarize the most relevant stimuli and describe how ion channels may contribute to cartilage homeostasis, with special emphasis on intracellular signaling pathways. We specifically focus on the role of calcium signaling as an essential mechanotransduction component and highlight the role of phosphatase signaling in this context.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700