Please wait a minute...
Frontiers of Materials Science

ISSN 2095-025X

ISSN 2095-0268(Online)

CN 11-5985/TB

Postal Subscription Code 80-974

2018 Impact Factor: 1.701

Front Mater Sci    2013, Vol. 7 Issue (4) : 313-334    https://doi.org/10.1007/s11706-013-0226-4
REVIEW ARTICLE
Advanced engineering and biomimetic materials for bone repair and regeneration
Lei YANG1(), Chao ZHONG2,3()
1. Institute of Orthopaedics, Department of Orthopaedic Surgery at the First Affiliated Hospital, Soochow University, Suzhou 215006, China; 2. Research Laboratory of Electronics (RLE), Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139-4307, USA; 3. Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139-4307, USA
 Download: PDF(1803 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Over the past decade, there has been tremendous progress in developing advanced biomaterials for tissue repair and regeneration. This article reviews the frontiers of this field from two closely related areas, new engineering materials for bone substitution and biomimetic mineralization for bone-like nanocomposites. Rather than providing an exhaustive overview of the literature, we focus on several representative directions. We also discuss likely future trends in these areas, including synthetic biology-enabled biomaterials design and multifunctional implant materials for bone repair and regeneration.

Keywords bone      engineering material      biomimetic material      implant      biomineralization     
Corresponding Author(s): YANG Lei,Email:yleibrown@gmail.com; ZHONG Chao,Email:zhongc@mit.edu   
Issue Date: 05 December 2013
 Cite this article:   
Lei YANG,Chao ZHONG. Advanced engineering and biomimetic materials for bone repair and regeneration[J]. Front Mater Sci, 2013, 7(4): 313-334.
 URL:  
https://academic.hep.com.cn/foms/EN/10.1007/s11706-013-0226-4
https://academic.hep.com.cn/foms/EN/Y2013/V7/I4/313
Fig.1  Low-elastic modulus porous metals for bone implants: Net shape, functional hip stems with designed porosity fabricated using LENS, scanning electron microscopy (SEM) image of entangled Ti wires, SEM image of porous Ta, and SEM image of porous NiTi. (Reproduced with permissions from Refs. [,-]).
Fig.2  Several samples of Ti–Zr–V–Cu–Al–Be alloys demonstrating large glass-forming ability and bending ductility (Reproduced with permission from Ref. []). biocompatibility tests of Zr–Ti–Co–Be alloy: Micrograph of fibroblast cell attachment on the amorphous metal surface after 7 d (arrows point to cell-layer buildup at the interface) and cell proliferation on the amorphous metal and high-density polyethylene (HDPE) surfaces. (Reproduced with permission from Ref. []).
Fig.3  SEM images showing anodized Ti nanotubular surface and nano to micron porous surface of anodized CoCrMo. (Reproduced with permissions from Ref. [] and Ref. [], respectively).
Fig.4  Histological results from rat amputee models showing lack of bone (blue stain) growth with untreated Ti screw and increased bone growth with anodized Ti screw, after 28 d of implantation. (Reproduced with permission from Prof. Thomas J. Webster and Nanovis, Inc.)
Fig.5  Altering NCD topography to control osteoblast adhesion and proliferation : SEM images of (a) NCD and (b) submicron crystalline diamond (SMCD) films; enhanced osteoblast density, proliferation and spreading on (c) NCD films compared to (d) SMCD films. (Reproduced with permission from Ref. []).
Fig.6  SEM images of uncoated Ti and Se nanoparticle coated Ti; Non-cancerous osteoblast and cancerous osteoblast densities on Se coated Ti after 1 and 3 d of culture, respectively (Data= mean±SEM; = 3, *<0.05 compared to uncoated Ti); Inhibited growths of in the presence of Se nanoparticles at all three Se suspension concentrations: 7.8, 15.5, and 31 μg/mL at all tested time points (3, 4, and 5 h) (Data= mean±SEM; = 3, *<0.01 compared to other concentrations). (Reproduced with permissions from Refs. [,-]).
Fig.7  SEM images of MWCNTs grown out of anodized nanotubular Ti (MWCNT-Ti) surface for biosensor purposes: currently conventional Ti; anodized nanotubular Ti; side and top views of MWCNT-Ti. Single arrows point to MWCNTs, whereas the double arrows point to the anodized nanotubular Ti substrate. (Reproduced with permission from Ref. []).
Fig.8  TEM and SAED correlated with SEM and ESB imaging of mineral freshly extracted from the distal end of the fin. TEM micrograph of mineral particle aggregates and the corresponding SAED patterns showing amorphous scatter of diffuse rings. Area marked with a rectangle produces poorly crystalline diffraction, and particle in produces a clear crystalline diffraction pattern, showing well defined reflections of the (002) and second order (004) apatite planes. SAED corresponds to the encircled area examined after storage for 1 week at room temperature: As the particles begin to crystallize, diffraction spots with spacing of the (002) plane appear (arrowheads), implying conversion into a crystalline apatite phase. High-resolution cryo-SEM of the same particle after examination in the TEM. (Scale bars 100 nm). (Reproduced with permission from Ref. []).
Fig.9  Cryo-electron tomography of collagen fibrils mineralized in the presence of pAsp for 72 h. Two-dimensional (2-D) cryo-TEM image. Slice from a section of the 3-D image along the plane (top-most inset), where crystals are visible edge-on (insets 1 and 2, white arrows). Black circle: ACP infiltrating the fibril (see below). Computer-generated 3-D visualization of mineralized collagen. The fibril is sectioned through the plane, revealing plate-shaped apatite crystals (colored in pink) embedded in the collagen matrix. Scale bars: 100 nm. (Reproduced with permission from Ref. []).
Fig.10  Self-assembling PAs used for biomimetic mineralization of hydroxyapatite. Chemical structure of the PA, comprising 5 regions: 1): a hydrophobic alkyl tail; 2) four cysteine residues that can form disulfide bonds to polymerize the self-assembled structure; 3) a flexible linker region of three glycine residues; 4) a single phosphorylated serine residue that was able to interact strongly with calcium ions and help direct mineralization of HA; 5) the cell adhesion ligand RGD. Molecular model of one single PA molecule. Schematic showing the self-assembly of PA molecules into a cylindrical micelle. (Reproduced with permission from Ref. []).
Fig.11  Schematic drawing showing that the predominance of polysaccharide (mainly GAG) exists at the interface of mineral and organic phase in bone. (Reproduced with permission from Ref. []).
Fig.12  Recapitulating the synthesis of bone-like composites through a biomimetic strategy inspired from recent progress about bone biomineralization. Biomimetic composites were obtained by first crosslinking maleic chitosan with poly (ethylene glycol) diacrylate (PEGDA) under ultraviolet (UV) light in water to form 3-D hydrogel networks, followed by biomimetic mineralization; Images of maleic chitosan/PEGDA hydrogel before and after mineralization. (Reproduced with permission from Ref. []).
Fig.13  Schematic of apatite-bound citrate (with oxygen of the carboxylates in red) interacting with Ca on two surfaces of high morphological importance of an idealized bone apatite nanocrystal. Calcium ions are blue filled circles on top and front surfaces, P is green (omitted on the top surfaces), OH ions are pink dots, while phosphate oxygen is omitted for clarity. The hexagonal crystal structure projected along the -axis (with greater depth of atoms indicated by lighter shading) shown in front reveals various layers of phosphate and calcium ions. (Reproduced with permission from Ref. []).
Fig.14  Citrate-mediated crystallization of apatite (AP) and carbonate apatite (CAP) phase over a time course from 5 min to 96 h. TEM images of AP crystallized after 5 min, 2 h and 96 h and CAP crystallized after 5 min, 2 h and 96 h. Insets show the SAED pattern collected for each sample. (Reproduced with permission from Ref. []).
1 McGowen J, Raisz L, Noonan A, . Bone health and osteoporosis: a report of the surgeon general. United States Department of Health and Human Services , 2004: 69–87
2 Bren L. Joint replacement: an inside look. FDA Consumer , 2004, 38(2): 12–19
3 American Academy of Orthopaedic Surgeons. [Internet] Rosemont. IL: AAOS , 2008
4 ATA Global Biomedical Materials Congress. Shanghai: Asia Technology Alliance, 2013
5 Balasundaram G. Nanomaterials for better orthopedics. In: Webster T J, ed. Nanotechnology for the Regeneration of Hard and Soft Tissues . Hackensack, New Jersey; London: World Scientific, 2007, 53–78
6 Krishna B V, Bose S, Bandyopadhyay A. Low stiffness porous Ti structures for load-bearing implants. Acta Biomaterialia , 2007, 3(6): 997–1006
7 He G, Liu P, Tan Q. Porous titanium materials with entangled wire structure for load-bearing biomedical applications. Journal of the Mechanical Behavior of Biomedical Materials , 2012, 5(1): 16–31
8 Kato K, Ochiai S, Yamamoto A, . Novel multilayer Ti foam with cortical bone strength and cytocompatibility. Acta Biomaterialia , 2013, 9(3): 5802–5809
9 Arciniegas M, Aparicio C, Manero J, . Low elastic modulus metals for joint prosthesis: Tantalum and nickel–titanium foams. Journal of the European Ceramic Society , 2007, 27(11): 3391–3398
10 Demetriou M D, Wiest A, Hofmann D C, . Amorphous metals for hard-tissue prosthesis. JOM , 2010, 62(2): 83–91
11 Ashby M, Greer A. Metallic glasses as structural materials. Scripta Materialia , 2006, 54(3): 321–326
12 Hofmann D C, Suh J Y, Wiest A, . Development of tough, low-density titanium-based bulk metallic glass matrix composites with tensile ductility. Proceedings of the National Academy of Sciences of the United States of America , 2008, 105(51): 20136–20140
13 Hofmann D C, Suh J Y, Wiest A, . Designing metallic glass matrix composites with high toughness and tensile ductility. Nature , 2008, 451(7182): 1085–1089
14 Hashimoto K. 2002 WR Whitney Award Lecture: In pursuit of new corrosion-resistant alloys. Corrosion , 2002, 58(9): 715–722
15 Chen Q, Liu L, Zhang S M. The potential of Zr-based bulk metallic glasses as biomaterials. Frontiers of Materials Science in China , 2010, 4(1): 34–44
16 Li J, Shi L-L, Zhu Z-D, . Zr61Ti2Cu25Al12 metallic glass for potential use in dental implants: Biocompatibility assessment by in vitro cellular responses. Materials Science and Engineering C , 2013, 33(4): 2113–2121
17 Li H F, Wang Y B, Zheng Y F, . Osteoblast response on Ti- and Zr-based bulk metallic glass surfaces after sand blasting modification. Journal of Biomedical Materials Research Part B: Applied Biomaterials , 2012, 100B(7): 1721–1728
18 Yan F, Liu G, Tao N, . Strength and ductility of 316L austenitic stainless steel strengthened by nano-scale twin bundles. Acta Materialia , 2012, 60(3): 1059–1071
19 Ye W, Li Y, Wang F. The improvement of the corrosion resistance of 309 stainless steel in the transpassive region by nano-crystallization. Electrochimica Acta , 2009, 54(4): 1339–1349
20 de Oliveira P T, Nanci A. Nanotexturing of titanium-based surfaces upregulates expression of bone sialoprotein and osteopontin by cultured osteogenic cells. Biomaterials , 2004, 25(3): 403–413
21 Cohen A, Liu-Synder P, Storey D, . Decreased fibroblast and increased osteoblast functions on ionic plasma deposited nanostructured Ti coatings. Nanoscale Research Letters , 2007, 2(8): 385–390
22 Ward B C, Webster T J. The effect of nanotopography on calcium and phosphorus deposition on metallic materials in vitro. Biomaterials , 2006, 27(16): 3064–3074
23 Webster T J, Ejiofor J U. Increased osteoblast adhesion on nanophase metals: Ti, Ti6Al4V, and CoCrMo. Biomaterials , 2004, 25(19): 4731–4739
24 Nakanishi Y, Miura H, Tokunaga K, . Nano-level surface texturing on Co–Cr–Mo alloy inhibits macrophage activation in joint prostheses. The Bone &amp; Joint Journal , 2013, 95-B(Supp 15): 278
25 Huang H H, Pan S J, Lai Y L, . Osteoblast-like cell initial adhesion onto a network-structured titanium oxide layer. Scripta Materialia , 2004, 51(11): 1017–1021
26 Yao C, Perla V, McKenzie J L, . Anodized Ti and Ti6Al4V possessing nanometer surface features enhances osteoblast adhesion. Journal of Biomedical Nanotechnology , 2005, 1(1): 68–73
27 Zhu X, Chen J, Scheideler L, . Effects of topography and composition of titanium surface oxides on osteoblast responses. Biomaterials , 2004, 25(18): 4087–4103
28 Das K, Bose S, Bandyopadhyay A. Surface modifications and cell–materials interactions with anodized Ti. Acta Biomaterialia , 2007, 3(4): 573–585
29 Yao C, Slamovich E B, Webster T J. Increased osteoblast adhesion on nano-rough anodized titanium and CoCrMo. NSTI Nanotechnology Conference and Trade Show – NSTI Nanotech, Technical Proceedings , 2006, 119–122
30 Popat K C, Eltgroth M, Latempa T J, . Decreased Staphylococcus epidermis adhesion and increased osteoblast functionality on antibiotic-loaded titania nanotubes. Biomaterials , 2007, 28(32): 4880–4888
31 Gruen D M. Nanocrystalline diamond films. Annual Review of Materials Science , 1999, 29(1): 211–259
32 Yang L, Zhang L, Webster T J. Carbon nanostructures for orthopedic medical applications. Nanomedicine , 2011, 6(7): 1231–1244
33 Bajaj P, Akin D, Gupta A, . Ultrananocrystalline diamond film as an optimal cell interface for biomedical applications. Biomedical Microdevices , 2007, 9(6): 787–794
34 Pareta R, Yang L, Kothari A, . Tailoring nanocrystalline diamond coated on titanium for osteoblast adhesion. Journal of Biomedical Materials Research Part A , 2010, 95A(1): 129–136
35 Yang L, Sheldon B W, Webster T J. The impact of diamond nanocrystallinity on osteoblast functions. Biomaterials , 2009, 30(20): 3458–3465
36 Yang L, Sheldon B W, Webster T J. Orthopedic nano diamond coatings: control of surface properties and their impact on osteoblast adhesion and proliferation. Journal of Biomedical Materials Research Part A , 2009, 91A(2): 548–556
37 Grausova L, Bacakova L, Kromka A, . Nanodiamond as promising material for bone tissue engineering. Journal of Nanoscience and Nanotechnology , 2009, 9(6): 3524–3534
38 Yang L, Chinthapenta V, Li Q, . Understanding osteoblast responses to stiff nanotopographies through experiments and computational simulations. Journal of Biomedical Materials Research Part A , 2011, 97A(4): 375–382
39 Rodrigues A A, Baranauskas V, Ceragioli H J, . In vivo preliminary evaluation of bone-microcrystalline and bone-nanocrystalline diamond interfaces. Diamond and Related Materials , 2010, 19(10): 1300–1306
40 Yang L, Li Y W, Sheldon B W, . Altering surface energy of nanocrystalline diamond to control osteoblast responses. Journal of Materials Chemistry , 2012, 22(1): 205–214
41 Klauser F, Hermann M, Steinmuller-Nethl D, . Direct and protein-mediated cell attachment on differently terminated nanocrystalline diamond. Chemical Vapor Deposition , 2010, 16(1–3): 42–49
42 Kromka A, Grausova L, Bacakova L, . Semiconducting to metallic-like boron doping of nanocrystalline diamond films and its effect on osteoblastic cells. Diamond and Related Materials , 2010, 19(2–3): 190–195
43 Kloss F R, Gassner R, Preiner J, . The role of oxygen termination of nanocrystalline diamond on immobilisation of BMP-2 and subsequent bone formation. Biomaterials , 2008, 29(16): 2433–2442
44 Steinmüller-Nethl D, Kloss F R, Najam-Ul-Haq M, . Strong binding of bioactive BMP-2 to nanocrystalline diamond by physisorption. Biomaterials , 2006, 27(26): 4547–4556
45 Jakubowski W, Bartosz G, Niedzielski P, . Nanocrystalline diamond surface is resistant to bacterial colonization. Diamond and Related Materials , 2004, 13(10): 1761–1763
46 Tran P, Webster T J. Enhanced osteoblast adhesion on nanostructured selenium compacts for anti-cancer orthopedic applications. International Journal of Nanomedicine , 2008, 3(3): 391–396
47 Tran P A, Sarin L, Hurt R H, . Opportunities for nanotechnology-enabled bioactive bone implants. Journal of Materials Chemistry , 2009, 19(18): 2653–2659
48 Navarro-Alarcón M, López-Martínez M C. Essentiality of selenium in the human body: relationship with different diseases. Science of the Total Environment , 2000, 249(1–3): 347–371
49 Kopeikin V V, Valueva S V, Kipper A I, . Synthesis of selenium nanoparticles in aqueous solutions of poly(vinylpyrrolidone) and morphological characteristics of the related nanocomposites. Polymer Science Series A , 2003, 45(4): 374–379
50 Tran P A, Sarin L, Hurt R H, . Titanium surfaces with adherent selenium nanoclusters as a novel anticancer orthopedic material. Journal of Biomedical Materials Research Part A , 2010, 93(4): 1417–1428
51 Tran P A, Webster T J. Selenium nanoparticles inhibit Staphylococcus aureus growth. International Journal of Nanomedicine , 2011, 6: 1553–1558
52 Wang Q, Webster T J. Nanostructured selenium for preventing biofilm formation on polycarbonate medical devices. Journal of Biomedical Materials Research Part A , 2012, 100A(12): 3205–3210
53 Holinka J, Pilz M, Kubista B, . Effects of selenium coating of orthopaedic implant surfaces on bacterial adherence and osteoblastic cell growth. The Bone &amp; Joint Journal , 2013, 95-B(5): 678–682
54 Sirivisoot S, Yao C, Xiao X, . Developing biosensors for monitoring orthopedic tissue growth. In: Firestone M, Schmidt J, Malmstadt N, eds. MRS Proceedings Volume 950, Symposium D: Biosurfaces and Biointerfaces . Materials Research Society , 2007
55 Sirivisoot S, Webster T J. Multiwalled carbon nanotubes enhance electrochemical properties of titanium to determine in situ bone formation. Nanotechnology , 2008, 19(29): 295101
56 Sirivisoot S, Webster T J. In situ bone growth detection using carbon nanotubes–titanium sensors. BioNanoScience , 2013, 3(2): 184–191
57 Sirivisoot S, Yao C, Xiao X, . Greater osteoblast functions on multiwalled carbon nanotubes grown from anodized nanotubular titanium for orthopedic applications. Nanotechnology , 2007, 18(36): 365102
58 Sirivisoot S, Pareta R, Webster T J. Electrically controlled drug release from nanostructured polypyrrole coated on titanium. Nanotechnology , 2011, 22(8): 085101
59 Song J, Malathong V, Bertozzi C R. Mineralization of synthetic polymer scaffolds: a bottom-up approach for the development of artificial bone. Journal of the American Chemical Society , 2005, 127(10): 3366–3372
60 Gkioni K, Leeuwenburgh S C G, Douglas T E L, . Mineralization of hydrogels for bone regeneration. Tissue Engineering Part B: Reviews , 2010, 16(6): 577–585
61 Huang J, Wong C, George A, . The effect of genetically engineered spider silk-dentin matrix protein 1 chimeric protein on hydroxyapatite nucleation. Biomaterials , 2007, 28(14): 2358–2367
62 Kim H J, Kim U J, Kim H S, . Bone tissue engineering with premineralized silk scaffolds. Bone , 2008, 42(6): 1226–1234
63 Segman-Magidovich S, Grisaru H, Gitli T, . Matrices of acidic β-sheet peptides as templates for calcium phosphate mineralization. Advanced Materials , 2008, 20(11): 2156–2161
64 Gungormus M, Branco M, Fong H, . Self assembled bi-functional peptide hydrogels with biomineralization-directing peptides. Biomaterials , 2010, 31(28): 7266–7274
65 George A, Ravindran S. Protein templates in hard tissue engineering. Nano Today , 2010, 5(4): 254–266
66 Wise E R, Maltsev S, Davies M E, . The organic-mineral interface in bone is predominantly polysaccharide. Chemistry of Materials , 2007, 19(21): 5055–5057
67 Hu Y Y, Rawal A, Schmidt-Rohr K. Strongly bound citrate stabilizes the apatite nanocrystals in bone. Proceedings of the National Academy of Sciences of the United States of America , 2010, 107(52): 22425–22429
68 Mahamid J, Addadi L, Weiner S. Crystallization pathways in bone. Cells, Tissues, Organs , 2011, 194(2–4): 92–97
69 Weiner S, Addadi L. Crystallization pathways in biomineralization. Annual Review of Materials Research , 2011, 41: 21–40
70 Termine J D, Posner A S. Infrared analysis of rat bone: age dependency of amorphous and crystalline mineral fractions. Science , 1966, 153(3743): 1523–1525
71 Mahamid J, Sharir A, Addadi L, . Amorphous calcium phosphate is a major component of the forming fin bones of zebrafish: Indications for an amorphous precursor phase. Proceedings of the National Academy of Sciences of the United States of America , 2008, 105(35): 12748–12753
72 Beniash E, Metzler R A, Lam R S K, . Transient amorphous calcium phosphate in forming enamel. Journal of Structural Biology , 2009, 166(2): 133–143
73 Mahamid J, Aichmayer B, Shimoni E, . Mapping amorphous calcium phosphate transformation into crystalline mineral from the cell to the bone in zebrafish fin rays. Proceedings of the National Academy of Sciences of the United States of America , 2010, 107(14): 6316–6321
74 Olszta M J, Cheng X G, Jee S S, . Bone structure and formation: A new perspective. Materials Science and Engineering R: Reports , 2007, 58(3-5): 77–116
75 Nudelman F, Pieterse K, George A, . The role of collagen in bone apatite formation in the presence of hydroxyapatite nucleation inhibitors. Nature Materials , 2010, 9(12): 1004–1009
76 Salgado A J, Coutinho O P, Reis R L. Bone tissue engineering: state of the art and future trends. Macromolecular Bioscience , 2004, 4(8): 743–765
77 Jee S S, Kasinath R K, DiMasi E, . Oriented hydroxyapatite in turkey tendon mineralized via the polymer-induced liquid-precursor (PILP) process. CrystEngComm , 2011, 13(6): 2077–2083
78 Liu Y, Kim Y K, Dai L, . Hierarchical and non-hierarchical mineralisation of collagen. Biomaterials , 2011, 32(5): 1291–1300
79 Liu Y, Li N, Qi Y P, . Intrafibrillar collagen mineralization produced by biomimetic hierarchical nanoapatite assembly. Advanced Materials , 2011, 23(8): 975–980
80 Maas M, Guo P, Keeney M, . Preparation of mineralized nanofibers: collagen fibrils containing calcium phosphate. Nano Letters , 2011, 11(3): 1383–1388
81 Wang Y, Aza?s T, Robin M, . The predominant role of collagen in the nucleation, growth, structure and orientation of bone apatite. Nature Materials , 2012, 11(8): 724–733
82 Palmer L C, Newcomb C J, Kaltz S R, . Biomimetic systems for hydroxyapatite mineralization inspired by bone and enamel. Chemical Reviews , 2008, 108(11): 4754–4783
83 Palmer L C, Stupp S I. Molecular self-assembly into one-dimensional nanostructures. Accounts of Chemical Research , 2008, 41(12): 1674–1684
84 Hartgerink J D, Beniash E, Stupp S I. Self-assembly and mineralization of peptide-amphiphile nanofibers. Science , 2001, 294(5547): 1684–1688
85 Spoerke E D, Anthony S G, Stupp S I. Enzyme directed templating of artificial bone mineral. Advanced Materials , 2009, 21(4): 425–430
86 Mata A, Geng Y B, Henrikson K J, . Bone regeneration mediated by biomimetic mineralization of a nanofiber matrix. Biomaterials , 2010, 31(23): 6004–6012
87 Boskey A L, Spevak L, Doty S B, . Effects of bone CS-proteoglycans, DS-decorin, and DS-biglycan on hydroxyapatite formation in a gelatin gel. Calcified Tissue International , 1997, 61(4): 298–305
88 Boskey A L, Stiner D, Binderman I, . Effects of proteoglycan modification on mineral formation in a differentiating chick limb-bud mesenchymal cell culture system. Journal of Cellular Biochemistry , 1997, 64(4): 632–643
89 Rees S G, Wassell D T H, Shellis R P, . Effect of serum albumin on glycosaminoglycan inhibition of hydroxyapatite formation. Biomaterials , 2004, 25(6): 971–977
90 Septier D, Hall R C, Lloyd D, . Quantitative immunohistochemical evidence of a functional gradient of chondroitin 4-sulphate/dermatan sulphate, developmentally regulated in the predentine of rat incisor. The Histochemical Journal , 1998, 30(4): 275–284
91 Takagi M, Maeno M, Yamada T, . Nature and distribution of chondroitin sulphate and dermatan sulphate proteoglycans in rabbit alveolar bone. The Histochemical Journal , 1996, 28(5): 341–351
92 Reid D G, Duer M J, Murray R C, . The organic-mineral interface in teeth is like that in bone and dominated by polysaccharides: Universal mediators of normal calcium phosphate biomineralization in vertebrates? Chemistry of Materials , 2008, 20(11): 3549–3550
93 Duer M J, Frisci? T, Proudfoot D, . Mineral surface in calcified plaque is like that of bone: further evidence for regulated mineralization. Arteriosclerosis, Thrombosis, and Vascular Biology , 2008, 28(11): 2030–2034
94 Duer M J, Frisci? T, Murray R C, . The mineral phase of calcified cartilage: its molecular structure and interface with the organic matrix. Biophysical Journal , 2009, 96(8): 3372–3378
95 Zhong C, Chu C C. Biomimetic mineralization of acid polysaccharide-based hydrogels: towards porous 3-dimensional bone-like biocomposites. Journal of Materials Chemistry , 2012, 22(13): 6080–6087
96 Zhong C, Wu J, Reinhart-King C A, . Synthesis, characterization and cytotoxicity of photo-crosslinked maleic chitosan–polyethylene glycol diacrylate hybrid hydrogels. Acta Biomaterialia , 2010, 6(10): 3908–3918
97 Li Q, Li M, Zhu P, . In vitro synthesis of bioactive hydroxyapatite using sodium hyaluronate as a template. Journal of Materials Chemistry , 2012, 22(38): 20257–20265
98 Deng Y, Wang H, Zhang L, . In situ synthesis and in vitro biocompatibility of needle-like nano-hydroxyapatite in agar–gelatin co-hydrogel. Materials Letters , 2013, 104: 8–12
99 Posner A S, Beebe R A. The surface chemistry of bone mineral and related calcium phosphates. Seminars in Arthritis and Rheumatism , 1975, 4(3): 267–291
100 Xie B Q, Nancollas G H. How to control the size and morphology of apatite nanocrystals in bone. Proceedings of the National Academy of Sciences of the United States of America , 2010, 107(52): 22369–22370
101 Delgado-López J M, Iafisco M, Rodríguez I, . Crystallization of bioinspired citrate-functionalized nanoapatite with tailored carbonate content. Acta Biomaterialia , 2012, 8(9): 3491–3499
102 Weber W, Fussenegger M. Emerging biomedical applications of synthetic biology. Nature Reviews Genetics , 2012, 13(1): 21–35
[1] Inamullah MAITLO, Safdar ALI, Muhammad Yasir AKRAM, Farooq Khurum SHEHZAD, Jun NIE. Binary phase solid-state photopolymerization of acrylates: design, characterization and biomineralization of 3D scaffolds for tissue engineering[J]. Front. Mater. Sci., 2017, 11(4): 307-317.
[2] Qin YANG, Yingying DU, Yifan WANG, Zhiying WANG, Jun MA, Jianglin WANG, Shengmin ZHANG. Si-doping bone composite based on protein template-mediated assembly for enhancing bone regeneration[J]. Front. Mater. Sci., 2017, 11(2): 106-119.
[3] Cheng-Bing CHU,Hong ZENG,Ding-Xia SHEN,Hui WANG,Ji-Fang WANG,Fu-Zhai CUI. A new rabbit model of implant-related biofilm infection: development and evaluation[J]. Front. Mater. Sci., 2016, 10(1): 80-89.
[4] Zonggang CHEN,Xiuli ZHANG,Lingzhi KANG,Fei XU,Zhaoling WANG,Fu-Zhai CUI,Zhongwu GUO. Recent progress in injectable bone repair materials research[J]. Front. Mater. Sci., 2015, 9(4): 332-345.
[5] Peng WANG,Bin PI,Jin-Ning WANG,Xue-Song ZHU,Hui-Lin YANG. Preparation and properties of calcium sulfate bone cement incorporated with silk fibroin and Sema3A-loaded chitosan microspheres[J]. Front. Mater. Sci., 2015, 9(1): 51-65.
[6] Ya-Wei DU,Li-Nan ZHANG,Xin YE,He-Min NIE,Zeng-Tao HOU,Teng-Hui ZENG,Guo-Ping YAN,Peng SHANG. In vitro and in vivo evaluation of bone morphogenetic protein-2 (BMP-2) immobilized collagen-coated polyetheretherketone (PEEK)[J]. Front. Mater. Sci., 2015, 9(1): 38-50.
[7] Hong-Man WANG,Fu-Yao LI. Bibliometric analysis of the literature from the mainland of China on animal-derived regenerative implantable medical devices[J]. Front. Mater. Sci., 2014, 8(4): 403-408.
[8] Ya-Wei DU,Li-Nan ZHANG,Zeng-Tao HOU,Xin YE,Hong-Sheng GU,Guo-Ping YAN,Peng SHANG. Physical modification of polyetheretherketone for orthopedic implants[J]. Front. Mater. Sci., 2014, 8(4): 313-324.
[9] Zhen-Tao YU,Ming-Hua ZHANG,Yu-Xing TIAN,Jun CHENG,Xi-Qun MA,Han-Yuan LIU,Chang WANG. Designation and development of biomedical Ti alloys with finer biomechanical compatibility in long-term surgical implants[J]. Front. Mater. Sci., 2014, 8(3): 219-229.
[10] Hai-Yan XU,Ning GU. Magnetic responsive scaffolds and magnetic fields in bone repair and regeneration[J]. Front. Mater. Sci., 2014, 8(1): 20-31.
[11] Da-Fu CHEN,Zhi-Yu ZHOU,Xue-Jun DAI,Man-Man GAO,Bao-Ding HUANG,Tang-Zhao LIANG,Rui SHI,Li-Jin ZOU,Hai-Sheng LI,Cody BÜNGER,Wei TIAN,Xue-Nong ZOU. Time-sequential changes of differentially expressed miRNAs during the process of anterior lumbar interbody fusion using equine bone protein extract, rhBMP-2 and autograft[J]. Front. Mater. Sci., 2014, 8(1): 72-86.
[12] Ke-Bin LIU,Kui HUANG,Yu TENG,Yan-Zheng QU,Wei CUI,Zhen-Fei HUANG,Ting-Fang SUN,Xiao-Dong GUO. Use of mineralized collagen bone graft substitutes and dorsal locking plate in treatment of elder metaphyseal comminuted distal radius fracture[J]. Front. Mater. Sci., 2014, 8(1): 87-94.
[13] Yi-Xia YIN,Ji-Ling YI,Li-Juan XIE,Qiong-Jiao YAN,Hong-Lian DAI,Shi-Pu LI. Synthesis, characterization and biological evaluation of poly [LA-co-(Glc-alt-Lys)] for nerve regeneration scaffold[J]. Front. Mater. Sci., 2014, 8(1): 95-101.
[14] Min-Fang CHEN, Jing ZHANG, Chen YOU. Ultraviolet-accelerated formation of bone-like apatite on oxidized Ti–24Nb–4Zr–7.9Sn alloy[J]. Front Mater Sci, 2013, 7(4): 362-369.
[15] Rui-Bo ZHAO, Hua-Feng HAN, Shao DING, Ze-Hao LI, Xiang-Dong KONG. Effect of silk sericin on morphology and structure of calcium carbonate crystal[J]. Front Mater Sci, 2013, 7(2): 177-183.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed