جلد 26، شماره 8 - ( 8-1398 )                   جلد 26 شماره 8 صفحات 55-42 | برگشت به فهرست نسخه ها

XML English Abstract Print


Download citation:
BibTeX | RIS | EndNote | Medlars | ProCite | Reference Manager | RefWorks
Send citation to:

Sahraei S S, Kalhor N, Sheykhhasan M. Application of scaffolds in cartilage tissue engineering: a review paper. RJMS 2019; 26 (8) :42-55
URL: http://rjms.iums.ac.ir/article-1-5591-fa.html
صحرایی سیده سعیده، کلهر ناصر، شیخ حسن محسن. کاربرد داربست‌ها در مهندسی بافت غضروف. مجله علوم پزشکی رازی. 1398; 26 (8) :42-55

URL: http://rjms.iums.ac.ir/article-1-5591-fa.html


دانشجوی دکتری زیست فناوری پزشکی، گروه سلول بنیادی مزانشیمی، جهاد دانشگاهی قم، قم، ایران ، m.sheykhhasan@acecr.ac.ir
چکیده:   (4580 مشاهده)
بافت غضروف به دلیل ماهیت خود که فاقد عروق خونی و اعصاب است، با یک چالش بالینی جهت ترمیم و بازسازی غضروف آسیب دیده روبرو می­باشد. آسیب به غضروف و بافت­های استئوکندرال می­تواند به علت استئوآرتریت، ورزش، سرطان­های تهاجمی، استرس­های تکراری و التهاب بافت باشد که با توجه به ظرفیت محدود آن برای بازسازی و یا ترمیم، نیاز به سیستم­های مناسب جایگزین است که بتواند عملکرد طبیعی بافت را از لحاظ فیزیکی، مکانیکی، بافت شناسی و بیولوژیکی دارا باشد. یکی از مهم ترین روش های جایگزین که به عنوان راه حلی کارا در این زمینه معرفی شده است، استراتژی مهندسی بافت می­باشد. هدف اصلی مهندسی بافت و پزشکی بازساختی در حوزه ارتوپدی، توسعه جایگزین­های زیستی به منظور بازسازی، حفظ و یا بهبود بافت آسیب دیده و عملکرد اندام غضروفی می­باشد. عناصر اصلی در استراتژی مهندسی بافت متشکل از سلول­های ترمیم کننده، فاکتورهای رشد، سیتوکین­ها و داربست­های مناسب می باشند. مواد زیستی مورد استفاده در مهندسی بافت به عنوان داربست به دو دسته کلی طبیعی و مصنوعی تقسیم بندی می­شوند. یک داربست ایده آل، باید دارای خواص زیست سازگاری و مکانیکی مطلوب همراه با زیست تخریب پذیری مناسب بوده و بتواند همراه با تعاملات موثر بین سلولی، فرآیند غضروف سازی را نیز القا نماید. در دهه­های اخیر، از فناوری نانو نیز به عنوان یک ابزار قدرتمند جهت کمک به مهندسی بافت غضروفی استفاده شده است. علم نانومواد روش­های جدیدی برای بهبود و تقویت مهندسی بافت ارائه کرده است. هدف از این مقاله ارائه یک مروری دقیق از بازسازی و ترمیم بافت غضروف  با استفاده از استراتژی های مهندسی بافت می­باشدو ارزشیابی یادگیری دانشجویان پرستاری و همچنین در زمان پذیرش مسئولیت‌های حرفه‌ای افراد در علوم پزشکی استفاده شود.
 
متن کامل [PDF 810 kb]   (4025 دریافت)    
نوع مطالعه: مروري | موضوع مقاله: بیولوژی (زیست شناسی)

فهرست منابع
1. 1. Mooney DJ, Mikos AG. Growing new organs. Sci Am 1999; 280:60-65.
2. 2. Akmal M, Singh A, Anand A, Kesani A, Aslam N, Goodship A, et al. The effects of hyaluronic acid on articular chondrocytes. J Bone Joint Surg Br 2005; 87:1143-1149.
3. 3. Alhadlaq A, Elisseeff JH, Hong L, Williams CG, Caplan AI, Sharma B, et al. Adult stem cell driven genesis of human-shaped articular condyle. Ann Biomed Eng 2004; 32:911-923.
4. 4. Alhadlaq A, Mao JJ. Tissue-engineered osteochondral constructs in the shape of an articular condyle. J Bone Joint Surg Am 2005; 87:936-944.
5. 5. Concaro S, Gustavson F, Gatenholm P. Bioreactors for tissue engineering of cartilage. Adv
6. Biochem Eng Biotechnol 2009; 112:125-143.
7. 6. Isogai N, Kusuhara H, Ikada Y, Ohtani H, Jacquet R, Hillyer J, et al. Comparison of different chondrocytes for use in tissue engineering of cartilage model structures. Tissue Eng 2006; 12:691-703.
8. 7. Bomer N, den Hollander W, Suchiman H, Houtman E, Slieker R, Heijmans B, et al. Neo-cartilage engineered from primary chondrocytes is epigenetically similar to autologous cartilage, in contrast to using mesenchymal stem cells. Osteoarthritis and cartilage 2016; 24:1423-1430.
9. 8. Qomi RT, Sheykhhasan M. Adipose-derived stromal cell in regenerative medicine: a review. World journal of stem cells 2017; 9:107.
10. 9. Sheykhhasan M, Ghiasi MS. Advances in adipose-derived stem cells and cartilage regeneration. Tehran University Medical Journal TUMS Publications 2018; 76:295-303.
11. 10. Sheykhhasan M, Ghiasi M. Evaluation of the Production Methods of Induced Pluripotent Stem Cells: A Short Review. Journal of Rafsanjan University of Medical Sciences 2016; 15:355-376.
12. 11. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007; 131:861-872.
13. 12. Craft AM, Rockel JS, Nartiss Y, Kandel RA, Alman BA, Keller GM. Generation of articular chondrocytes from human pluripotent stem cells. Nat Biotechnol 2015; 33:638-645.
14. 13. Ren K, He C, Xiao C, Li G, Chen X. Injectable glycopolypeptide hydrogels as biomimetic scaffolds for cartilage tissue engineering. Biomaterials 2015; 51:238-249.
15. 14. Gong Y, Wang C, Lai RC, Su K, Zhang F, Wang D-a. An improved injectable polysaccharide hydrogel: modified gellan gum for long-term cartilage regeneration in vitro. Journal of Materials Chemistry 2009; 19:1968-1977.
16. 15. Bidarra SJ, Barrias CC, Granja PL. Injectable alginate hydrogels for cell delivery in tissue engineering. Acta biomaterialia 2014; 10:1646-1662.
17. 16. Liu M, Zeng X, Ma C, Yi H, Ali Z, Mou X, et al. Injectable hydrogels for cartilage and bone tissue engineering. Bone research 2017; 5:17014.
18. 17. Shen Z-S, Cui X, Hou R-X, Li Q, Deng H-X, Fu J. Tough biodegradable chitosan–gelatin hydrogels via in situ precipitation for potential cartilage tissue engineering. RSC Advances 2015; 5:55640-55647.
19. 18. Naderi‐Meshkin H, Andreas K, Matin MM, Sittinger M, Bidkhori HR, Ahmadiankia N, et al. Chitosan‐based injectable hydrogel as a promising in situ forming scaffold for cartilage tissue engineering. Cell biology international 2014; 38:72-84.
20. 19. Moreira CD, Carvalho SM, Mansur HS, Pereira MM. Thermogelling chitosan–collagen–bioactive glass nanoparticle hybrids as potential injectable systems for tissue engineering. Materials Science and Engineering: C 2016; 58:1207-1216.
21. 20. Kamoun EA. N-succinyl chitosan–dialdehyde starch hybrid hydrogels for biomedical applications. Journal of Advanced research 2016; 7:69-77.
22. 21. Kon E, Gobbi A, Filardo G, Delcogliano M, Zaffagnini S, Marcacci M. Arthroscopic second-generation autologous chondrocyte implantation compared with microfracture for chondral lesions of the knee: prospective nonrandomized study at 5 years. Am J Sports Med 2009; 37:33-41.
23. 22. Kontturi L-S, Järvinen E, Muhonen V, Collin EC, Pandit AS, Kiviranta I, et al. An injectable, in situ forming type II collagen/hyaluronic acid hydrogel vehicle for chondrocyte delivery in cartilage tissue engineering. Drug delivery and translational research 2014; 4:149-158.
24. 23. Oh BH, Bismarck A, Chan‐Park MB. Injectable, Interconnected, High‐Porosity Macroporous Biocompatible Gelatin Scaffolds Made by Surfactant‐Free Emulsion Templating. Macromolecular rapid communications 2015; 36:364-372.
25. 24. Geng X, Mo X, Fan L, Yin A, Fang J. Hierarchically designed injectable hydrogel from oxidized dextran, amino gelatin and 4-arm poly (ethylene glycol)-acrylate for tissue engineering application. Journal of Materials Chemistry 2012; 22:25130-25139.
26. 25. Yu F, Cao X, Li Y, Zeng L, Yuan B, Chen X. Correction: An injectable hyaluronic acid/PEG hydrogel for cartilage tissue engineering formed by integrating enzymatic crosslinking and Diels–Alder “click chemistry”. Polymer Chemistry 2018; 9:3959-3960.
27. 26. Eyre D. Collagen of articular cartilage. Arthritis Res 2002; 4:30-35.
28. 27. Eyre DR, Weis MA, Wu JJ. Articular cartilage collagen: an irreplaceable framework? Eur Cell Mater 2006; 12:57-63.
29. 28. Ko CS, Huang JP, Huang CW, Chu IM. Type II collagen-chondroitin sulfate-hyaluronan scaffold cross-linked by genipin for cartilage tissue engineering. J Biosci Bioeng 2009; 107:177-182.
30. 29. Wang DA, Varghese S, Sharma B, Strehin I, Fermanian S, Gorham J, et al. Multifunctional chondroitin sulphate for cartilage tissue-biomaterial integration. Nat Mater 2007; 6:385-392.
31. 30. Frenkel SR, Di Cesare PE. Scaffolds for articular cartilage repair. Ann Biomed Eng 2004; 32:26-34.
32. 31. Rhee S, Puetzer JL, Mason BN, Reinhart-King CA, Bonassar LJ. 3D bioprinting of spatially heterogeneous collagen constructs for cartilage tissue engineering. ACS Biomaterials Science & Engineering 2016; 2:1800-1805.
33. 32. Cherubino P, Grassi FA, Bulgheroni P, Ronga M. Autologous chondrocyte implantation using a bilayer collagen membrane: a preliminary report. J Orthop Surg (Hong Kong) 2003; 11:10-15.
34. 33. Gigante A, Enea D, Greco F, Bait C, Denti M, Schonhuber H, et al. Distal realignment and patellar autologous chondrocyte implantation: mid-term results in a selected population. Knee Surg Sports Traumatol Arthrosc 2009; 17:2-10.
35. 34. Ochi M, Uchio Y, Kawasaki K, Wakitani S, Iwasa J. Transplantation of cartilage-like tissue made by tissue engineering in the treatment of cartilage defects of the knee. J Bone Joint Surg Br 2002; 84:571-578.
36. 35. Funayama A, Niki Y, Matsumoto H, Maeno S, Yatabe T, Morioka H, et al. Repair of full-thickness articular cartilage defects using injectable type II collagen gel embedded with cultured chondrocytes in a rabbit model. Journal of Orthopaedic Science 2008; 13:225-232.
37. 36. Liao J, Qu Y, Chu B, Zhang X, Qian Z. Biodegradable CSMA/PECA/Graphene Porous Hybrid Scaffold for Cartilage Tissue Engineering. Sci Rep 2015; 5:9879.
38. 37. Chen F, Yu S, Liu B, Ni Y, Yu C, Su Y, et al. An Injectable Enzymatically Crosslinked Carboxymethylated Pullulan/Chondroitin Sulfate Hydrogel for Cartilage Tissue Engineering. Sci Rep 2016; 6:20014.
39. 38. Schiavone Panni A, Cerciello S, Vasso M. The manangement of knee cartilage defects with modified amic technique: preliminary results. Int J Immunopathol Pharmacol 2011; 24:149-152.
40. 39. Kusano T, Jakob RP, Gautier E, Magnussen RA, Hoogewoud H, Jacobi M. Treatment of isolated chondral and osteochondral defects in the knee by autologous matrix-induced chondrogenesis (AMIC). Knee Surg Sports Traumatol Arthrosc 2012; 20:2109-2115.
41. 40. Gille J, Behrens P, Volpi P, De Girolamo L, Reiss E, Zoch W, et al. Outcome of Autologous Matrix Induced Chondrogenesis (AMIC) in cartilage knee surgery: data of the AMIC Registry. Archives of orthopaedic and trauma surgery 2013; 133:87-93.
42. 41. Yuan L, Li B, Yang J, Ni Y, Teng Y, Guo L, et al. Effects of composition and mechanical property of injectable collagen I/II composite hydrogels on chondrocyte behaviors. Tissue Engineering Part A 2016; 22:899-906.
43. 42. Kim IL, Mauck RL, Burdick JA. Hydrogel design for cartilage tissue engineering: a case study with hyaluronic acid. Biomaterials 2011; 32:8771-8782.
44. 43. Kujawa MJ, Caplan AI. Hyaluronic acid bonded to cell-culture surfaces stimulates chondrogenesis in stage 24 limb mesenchyme cell cultures. Dev Biol 1986; 114:504-518.
45. 44. Strehin I, Nahas Z, Arora K, Nguyen T, Elisseeff J. A versatile pH sensitive chondroitin sulfate-PEG tissue adhesive and hydrogel. Biomaterials 2010; 31:2788-2797.
46. 45. Fan H, Hu Y, Li X, Lu R, Bai J, Wang J. [Experimental study on gelatin-chondroitin sulfate-sodium hyaluronate tri-copolymer as novel scaffolds for cartilage tissue engineering]. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi 2005; 19:473-477.
47. 46. Nehrer S, Domayer S, Dorotka R, Schatz K, Bindreiter U, Kotz R. Three-year clinical outcome after chondrocyte transplantation using a hyaluronan matrix for cartilage repair. Eur J Radiol 2006; 57:3-8.
48. 47. Ferruzzi A, Buda R, Faldini C, Vannini F, Di Caprio F, Luciani D, et al. Autologous chondrocyte implantation in the knee joint: open compared with arthroscopic technique. Comparison at a minimum follow-up of five years. J Bone Joint Surg Am 2008; 90 Suppl 4:90-101.
49. 48. Gobbi A, Kon E, Berruto M, Filardo G, Delcogliano M, Boldrini L, et al. Patellofemoral full-thickness chondral defects treated with second-generation autologous chondrocyte implantation: results at 5 years' follow-up. Am J Sports Med 2009; 37:1083-1092.
50. 49. Clar H, Pascher A, Kastner N, Gruber G, Robl T, Windhager R. Matrix-assisted autologous chondrocyte implantation into a 14cm(2) cartilage defect, caused by steroid-induced osteonecrosis. Knee 2010; 17:255-257.
51. 50. Kon E, Di Martino A, Filardo G, Tetta C, Busacca M, Iacono F, et al. Second-generation autologous chondrocyte transplantation: MRI findings and clinical correlations at a minimum 5-year follow-up. Eur J Radiol 2011; 79:382-388.
52. 51. Kon E, Filardo G, Berruto M, Benazzo F, Zanon G, Della Villa S, et al. Articular cartilage treatment in high-level male soccer players: a prospective comparative study of arthroscopic second-generation autologous chondrocyte implantation versus microfracture. Am J Sports Med 2011; 39:2549-2557.
53. 52. Kon E, Filardo G, Condello V, Collarile M, Di Martino A, Zorzi C, et al. Second-generation autologous chondrocyte implantation: results in patients older than 40 years. Am J Sports Med 2011; 39:1668-1675.
54. 53. Filardo G, Kon E, Berruto M, Di Martino A, Patella S, Marcheggiani Muccioli GM, et al. Arthroscopic second generation autologous chondrocytes implantation associated with bone grafting for the treatment of knee osteochondritis dissecans: Results at 6 years. Knee 2012; 19:658-663.
55. 54. Brix MO, Stelzeneder D, Chiari C, Koller U, Nehrer S, Dorotka R, et al. Treatment of Full-Thickness Chondral Defects With Hyalograft C in the Knee: Long-term Results. Am J Sports Med 2014; 42:1426-1432.
56. 55. Rowley JA, Madlambayan G, Mooney DJ. Alginate hydrogels as synthetic extracellular matrix materials. Biomaterials 1999; 20:45-53.
57. 56. Alsberg E, Anderson KW, Albeiruti A, Franceschi RT, Mooney DJ. Cell-interactive alginate hydrogels for bone tissue engineering. J Dent Res 2001; 80:2025-2029.
58. 57. Sultzbaugh K, Speaker T. A method to attach lectins to the surface of spermine alginate microcapsules based on the avidin biotin interaction. Journal of microencapsulation 1996; 13:363-376.
59. 58. Gerard C, Catuogno C, Amargier-Huin C, Grossin L, Hubert P, Gillet P, et al. The effect of alginate, hyaluronate and hyaluronate derivatives biomaterials on synthesis of non-articular chondrocyte extracellular matrix. J Mater Sci Mater Med 2005; 16:541-551.
60. 59. Sheykhhasan M, Qomi RT, Kalhor N, Mehdizadeh M, Ghiasi M. Evaluation of the ability of natural and synthetic scaffolds in providing an appropriate environment for growth and chondrogenic differentiation of adipose-derived mesenchymal stem cells. Indian journal of orthopaedics 2015.49:561.
61. 60. Ghiasi M, Kalhor N, Tabatabaei Qomi R, Sheykhhasan M. The effects of synthetic and natural scaffolds on viability and proliferation of adipose-derived stem cells. Frontiers in Life Science 2016; 9:32-43.
62. 61. Sheykhhasan M, Ghiasi M, Pak HB. The assessment of natural scaffolds ability in chondrogenic differentiation of human adipose-derived mesenchymal stem cells. Internet Journal of Medical Update-EJOURNAL 2016; 11:11-16.
63. 62. Li H, Hu C, Yu H, Chen C. Chitosan composite scaffolds for articular cartilage defect repair: a review. RSC Advances 2018; 8:3736-3749.
64. 63. Chenite A, Chaput C, Wang D, Combes C, Buschmann MD, Hoemann CD, et al. Novel injectable neutral solutions of chitosan form biodegradable gels in situ. Biomaterials 2000; 21:2155-2161.
65. 64. Gille J, Behrens P, Volpi P, de Girolamo L, Reiss E, Zoch W, et al. Outcome of Autologous Matrix Induced Chondrogenesis (AMIC) in cartilage knee surgery: data of the AMIC Registry. Arch Orthop Trauma Surg 2013; 133:87-93.
66. 65. Pascarella A, Ciatti R, Pascarella F, Latte C, Di Salvatore MG, Liguori L, et al. Treatment of articular cartilage lesions of the knee joint using a modified AMIC technique. Knee Surgery, Sports Traumatology, Arthroscopy 2010; 18:509-513.
67. 66. Wang T, Lai JH, Yang F. Effects of Hydrogel Stiffness and Extracellular Compositions on Modulating Cartilage Regeneration by Mixed Populations of Stem Cells and Chondrocytes In Vivo. Tissue Eng Part A 2016; 22:1348-1356.
68. 67. Drury JL, Mooney DJ. Hydrogels for tissue engineering: scaffold design variables and applications. Biomaterials 2003; 24:4337-4351.
69. 68. Sol P, Martins A, Reis R, Neves N. Advanced polymer composites and structures for bone and cartilage tissue engineering. Nanocomposites for Musculoskeletal Tissue Regeneration: Elsevier; 2016. p. 123-142.
70. 69. Zhang L, Webster TJ. Nanotechnology and nanomaterials: promises for improved tissue regeneration. Nano today 2009; 4:66-80.
71. 70. Li X, Wang L, Fan Y, Feng Q, Cui FZ, Watari F. Nanostructured scaffolds for bone tissue engineering. Journal of Biomedical Materials Research Part A 2013; 101:2424-2435.
72. 71. Madry H, Rey-Rico A, Venkatesan JK, Johnstone B, Cucchiarini M. Transforming growth factor beta-releasing scaffolds for cartilage tissue engineering. Tissue Engineering Part B: Reviews 2013;106:120-25.
73. 72. Biondi M, Borzacchiello A, Mayol L, Ambrosio L. Nanoparticle-Integrated Hydrogels as Multifunctional Composite Materials for Biomedical Applications. Gels 2015; 1:162-178.
74. 73. Gaharwar AK, Peppas NA, Khademhosseini A. Nanocomposite hydrogels for biomedical applications. Biotechnol Bioeng 2014; 111:441-453.
75. 74. Eslahi N, Abdorahim M, Simchi A. Smart Polymeric Hydrogels for Cartilage Tissue Engineering: A Review on the Chemistry and Biological Functions. Biomacromolecules 2016; 17:3441-3463.
76. 75. Toh WS, Loh XJ. Advances in hydrogel delivery systems for tissue regeneration. Mater Sci Eng C Mater Biol Appl 2014; 45:690-697.
77. 76. Asghari F, Samiei M, Adibkia K, Akbarzadeh A, Davaran S. Biodegradable and biocompatible polymers for tissue engineering application: a review. Artif Cells Nanomed Biotechnol 2017; 45:185-192.
78. 77. Carrow JK, Gaharwar AK. Bioinspired polymeric nanocomposites for regenerative medicine. Macromolecular Chemistry and Physics 2015; 216:248-264.
79. 78. Shabestari Khiabani S, Farshbaf M, Akbarzadeh A, Davaran S. Magnetic nanoparticles: preparation methods, applications in cancer diagnosis and cancer therapy. Artif Cells Nanomed Biotechnol 2017; 45:6-17.
80. 79. Wu S, Weng Z, Liu X, Yeung K, Chu PK. Functionalized TiO2 based nanomaterials for biomedical applications. Advanced functional materials 2014; 24:5464-5481.
81. 80. Lu PJ, Huang SC, Chen YP, Chiueh LC, Shih DYC. Analysis of titanium dioxide and zinc oxide nanoparticles in cosmetics. journal of food and drug analysis 2015; 23:587-594.
82. 81. Salarian M, Xu WZ, Wang Z, Sham TK, Charpentier PA. Hydroxyapatite-TiO(2)-based nanocomposites synthesized in supercritical CO(2) for bone tissue engineering: physical and mechanical properties. ACS Appl Mater Interfaces 2014; 6:16918-16931.
83. 82. Jayakumar R, Ramachandran R, Divyarani VV, Chennazhi KP, Tamura H, Nair SV. Fabrication of chitin-chitosan/nano TiO2-composite scaffolds for tissue engineering applications. Int J Biol Macromol 2011; 48:336-344.
84. 83. Cao L, Wu X, Wang Q, Wang J. Biocompatible nanocomposite of TiO2 incorporated bi-polymer for articular cartilage tissue regeneration: A facile material. J Photochem Photobiol B 2018; 178:440-446.
85. 84. Naranda J, Susec M, Maver U, Gradisnik L, Gorenjak M, Vukasovic A, et al. Polyester type polyHIPE scaffolds with an interconnected porous structure for cartilage regeneration. Sci Rep 2016; 6:28695.
86. 85. Yang J, Zhang YS, Yue K, Khademhosseini A. Cell-laden hydrogels for osteochondral and cartilage tissue engineering. Acta biomaterialia 2017; 57:1-25.
87. 86. Vyas C, Poologasundarampillai G, Hoyland J, Bartolo P. 3D printing of biocomposites for osteochondral tissue engineering. Biomedical Composites (Second Edition): Elsevier; 2017. p. 261-302.
88. 87. Rana D, Ratheesh G, Ramakrishna S, Ramalingam M. Nanofiber composites in cartilage tissue engineering. Nanofiber Composites for Biomedical Applications: Elsevier; 2017. p. 325-344.
89. 88. Yang X, Lu Z, Wu H, Li W, Zheng L, Zhao J. Collagen-alginate as bioink for three-dimensional (3D) cell printing based cartilage tissue engineering. Materials Science and Engineering: C 2018; 83:195-201.
90. 89. Aliakbarshirazi S, Talebian A. Electrospun gelatin nanofibrous scaffolds for cartilage tissue engineering. Materials Today: Proceedings 2017; 4:7059-7064.
91. 90. Wang C, Hou W, Guo X, Li J, Hu T, Qiu M, et al. Two-phase electrospinning to incorporate growth factors loaded chitosan nanoparticles into electrospun fibrous scaffolds for bioactivity retention and cartilage regeneration. Materials Science and Engineering: C 2017; 79:507-515.
92. 91. Agheb M, Dinari M, Rafienia M, Salehi H. Novel electrospun nanofibers of modified gelatin-tyrosine in cartilage tissue engineering. Materials Science and Engineering: C 2017; 71:240-251.
93. 92. Cao L, Zhang F, Wang Q, Wu X. Fabrication of chitosan/graphene oxide polymer nanofiber and its biocompatibility for cartilage tissue engineering. Materials Science and Engineering: C 2017; 79:697-701.

ارسال نظر درباره این مقاله : نام کاربری یا پست الکترونیک شما:
CAPTCHA

ارسال پیام به نویسنده مسئول


بازنشر اطلاعات
Creative Commons License این مقاله تحت شرایط Creative Commons Attribution-NonCommercial 4.0 International License قابل بازنشر است.

کلیه حقوق این وب سایت متعلق به مجله علوم پزشکی رازی می باشد.

طراحی و برنامه نویسی : یکتاوب افزار شرق

© 2024 CC BY-NC 4.0 | Razi Journal of Medical Sciences

Designed & Developed by : Yektaweb