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    2011, Vol. 5 Issue (3) : 266-281    https://doi.org/10.1007/s11706-011-0145-1
REVIEW ARTICLE
Biosilica-glass formation using enzymes from sponges [silicatein]: Basic aspects and application in biomedicine [bone reconstitution material and osteoporosis]
Shun-Feng WANG1, Xiao-Hong WANG1(), Lu GAN1, Matthias WIENS2, Heinz C. SCHR?DER2, Werner E. G. MüLLER2()
1. National Research Center for Geoanalysis, Chinese Academy of Geological Sciences, 26 Baiwanzhuang Dajie, Beijing 100037, China; 2. Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55128 Mainz, Germany
 Download: PDF(1577 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

In the last 15 years biomineralization, in particular biosilicification (i.e., the formation of biogenic silica, SiO2), has become an exciting source of inspiration for the development of novel bionic approaches, following “Nature as model”. Among the silica forming organisms there are the sponges that have the unique property to catalyze their silica skeletons by a specific enzyme termed silicatein. In the present review we summarize the present state of knowledge on silicatein-mediated “biosilica” formation in marine sponges, the involvement of further molecules in silica metabolism and their potential application in biomedicine. Recent advancements in the production of bone replacement material and in the potential use as a component in the treatment of osteoporosis are highlighted.

Keywords biomineralization      biosilica      medicine      biomaterials      osteoporosis     
Corresponding Author(s): WANG Xiao-Hong,Email:wxh0408@hotmail.com (X.H.W.); E. G. MüLLER Werner,Email:wmueller@uni-mainz.de (W.E.G.M.)   
Issue Date: 05 September 2011
 Cite this article:   
Shun-Feng WANG,Xiao-Hong WANG,Lu GAN, et al. Biosilica-glass formation using enzymes from sponges [silicatein]: Basic aspects and application in biomedicine [bone reconstitution material and osteoporosis][J]. Front Mater Sci, 2011, 5(3): 266-281.
 URL:  
https://academic.hep.com.cn/foms/EN/10.1007/s11706-011-0145-1
https://academic.hep.com.cn/foms/EN/Y2011/V5/I3/266
Fig.1  Spicules of . SEM analysis. Inset: Broken spicule with an empty axial canal; higher magnification. Silicatein filaments () - micrograph (visual light).() SEM analysis of the axial filament. (Reproduced with permission from Ref. [], Copyright 2007 Springer-Verlag)
Fig.2  Grouping of the deduced protein sequences of silicateins within the cathepsin/cysteine protease family. The deduced silicatein-α sequences from (SILCAa_SUBDO; accession number CAI46305.1) and (SILCAa_GEOCY; CAM57981.1), as well as the silicateins from : alpha (SILCAa_LUBAI; CAI43319.1), form a2 (SILCAa2_LUBAI; CAI91571.1), a3 (SILCAa3_LUBAI; |CAI91572.1), and a4 (SILCAa4_LUBAI; |CAI91573.1) had been aligned with silicatein-β from (SILCAb_SUBDO; CAI46304.1) and with cathepsin L from (CATHL_SUBDO; CAH04632.1) and (CATHL_LUBAI; CAI43320.1). The papain [proteinase IV] sequence from (PAPAIN_CARPA; CAA54974.1) was included and then used as outgroup in the phylogenetic tree. Residues conserved (similar or related with respect to their physico-chemical properties) in all sequences are shown in white on black, and those in at least two sequences in white on gray. The characteristic sites in the sequences, i.e. the catalytic triad amino acids, Ser (S; #) in silicateins and Cys in cathepsin, as well as His (H; #) and Asn (#) [all in red], and the segments of the propeptide and of the mature peptide are marked. Finally, the serine cluster ([*Ser*]) and the cleavage site of the signal peptide are marked (><). The consensus amino acids, characteristic for the sequences are in blue. A tree was constructed after aligning these sponge proteins. The tree was rooted with the papain sequence. (Reproduced with permission from Ref. [], Copyright 2007 Springer-Verlag)
Fig.3  Silicatein: Model showing the activation of the enzyme. Immature silicatein. Mature silicatein. Amino acids of the catalytic center (cc): red; amino acids of the serine cluster [Ser, His and Asn]: green; signal sequence: orange; sequence cleaved from pro-silicatein molecule: yellow. (Reproduced with permission from Ref. [], Copyright 2007 Springer-Verlag)
Fig.4  Silicase. Alignment of the silicase deduced amino acid sequence from (SIA_SUBDO) with the human carbonic anhydrase II (CAH2_HUMAN). The carbonic anhydrase domain is framed (● e-CAdom ●). The similar amino acid residues in both sequences are shown in white on black. The three zinc-binding histidine residues (Z) and the characteristic amino acids forming the eukaryotic-type carbonic anhydrase signature (#, found in both sequences; -, present only in the carbonic anhydrases but not in the silicase) are indicated; +, residues forming the active-site hydrogen network. (Reproduced with permission from Ref. [], Copyright 2007 Springer-Verlag)
Fig.5  Silicase. Model showing the three histidine residues (red; His, His and His) in the catalytic center (cc) of the enzyme, that bind to a zinc ion. Proposed mechanism of action. (Reproduced with permission from Ref. [], Copyright 2007 Springer-Verlag)
Fig.6  application of the functional implant material [FIM] as potential bone implant material. SEM image of the implant material, depicting SSM within the PMSA matrix. Radiograph of the molded implant material to be used for implantation into rabbit femurs. Lateral view and top view of μCT analysis of the implant material, implanted in a rabbit femur. The implant material can be distinguished from hydroxyapatite of bone by application of pseudocolor contrasting. (Reproduced with permission from Ref. [], Copyright 2010 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim)
Fig.7  Role of biosilica during bone formation (scheme). Spatial relationship between silicon accumulation and calcium composition during early stages of bone formation in rats (modified according to Ref. []). Schematic representation of the effect of silica-based components on the expression of the three marker genes (amelogenin, ameloblastin, enamelin) in ameloblasts. The silica-based components stimulate the expression of amelogenin, resulting in the formation of nanospherical hydroxyapatite around which hydroxyapatite crystals are deposited. (Reproduced with permission from Ref. [], Copyright 2009 Springer-Verlag)
Fig.8  Biomedical application of biosilica and silicatein. Formation of biosilica layers on pig molars. After tissue removal, the teeth were treated with phosphate buffered saline, supplemented with protease and phosphatase inhibitors, according to Ref. []. Subsequently, dental hydroxyapatite was incubated with sodium metasilicate (100 μmol/L) in the absence (A) or presence (B) of recombinant silicatein (4 μg/mL PBS) for 12 h at 20°C. Then, the samples were examined by HR-SEM. In parallel, biosilica formation on femur bone samples was examined: untreated control (C) or silicatein-treated (D). The biosilica layers are marked (bs). (Reproduced with permission from Ref. [], Copyright 2009 Springer-Verlag)
Fig.9  Fine structure of the HA crystallites grown on SaOS-2 cell surfaces. Cells were grown in McCoy’s medium supplemented with AA, DEX and 100 μmol/L polyP (Ca salt) and/or biosilica for 4 d. On the surfaces of the cells (c) nodules (no) of HA crystallites are seen that are surrounded by clusters of cells. In all cases the nodules are entangled by processes or lobes originating from the underlying cells. At higher magnification ((C) and (D)) it becomes apparent that the cell lobes surround and embed the HA crystallites. In one area, the cell lobes that fuse around the crystallites are highlighted (><). (Reproduced with permission from Ref. [], Copyright 2011 Elsevier)
Fig.10  Proposed effects of biosilica on osteoblasts, osteoclasts, and their progenitors. Biosilica enhances expression of OPG in osteoblasts. Osteoblasts have the potential to differentiate to hydroxyapatite-forming osteocytes and lining cells. OPG counteracts various effects of RANKL, a cytokine that induces preosteoclast maturation and osteoclast activation. In concert with bio-silica polyphosphate (PolyP) was found to contribute to the anabolic bone metabolism. Scheme. (Reproduced with permission from Ref. [], Copyright 2011 Elsevier)
1 St?ber W, Fink A, Bohn E. Controlled growth of monodisperse silica spheres in the micron size range. Journal of Colloid and Interface Science, 1968, 26(1): 62–69
doi: 10.1016/0021-9797(68)90272-5
2 Brinker C J, Scherrer G W. Sol-Gel Science: The Physics and Chemistry of Sol-Gel Processing.London: Academic Press, 1990
3 Hench L L, West J K. The sol-gel process. Chemical Reviews, 1990, 90(1): 33–72
4 B?uerlein E. Biomineralization. Cambridge: Wiley-VCH, 2004
doi: 10.1002/3527604138
5 Müller W E G. Molecular phylogeny of metazoa (animals): Monophyletic origin. Naturwissenschaften, 1995, 82(7): 321–329
6 Müller W E G. Molecular phylogeny of Eumetazoa: genes in sponges (Porifera) give evidence for monophyly of animals. Progress in Molecular and Subcellular Biology, 1998, 19: 89–132
7 Müller W E G.Review: How was metazoan threshold crossed? The hypothetical Urmetazoa. Comparative Biochemistry and Physiology, 2001, 129(2-3): 433–460
8 Wang X H, Zhang X H, Schr?der H C, . Giant basal spicule from the deep-sea glass sponge Monorhaphis chuni: synthesis of the largest bio-silica structure on Earth by silicatein. Frontiers of Materials Science in China, 2009, 3(3): 226–240
9 Wang X, Wiens M, Schr?der H C, . Morphology of sponge spicules: silicatein a structural protein for bio-silica formation. Advanced Engineering Materials, 2010, 12(9): B422–B437
10 Simpson T L. The Cell Biology of Sponges.New York: Springer-Verlag, 1984
11 Sandford F. Physical and chemical analysis of the siliceous skeleton in six sponges of two groups (demospongiae and hexactinellida). Microscopy Research and Technique , 2003, 62(4): 336–355
12 Shimizu K, Cha J, Stucky G D, . Silicatein α: Cathepsin L-like protein in sponge biosilica. Proceedings of the National Academy of Sciences of the United States of America, 1998, 95(11): 6234–6238
13 Perry C C, Belton D, Shafran K. Studies of biosilicas; structural aspects, chemical principles, model studies and the future. Progress in Molecular and Subcellular Biology, 2003, 33: 269–299
14 Iler R K. The Chemistry of Silica.New York: John Wiley &amp; Sons, 1979
15 Perry C C. Silicification: the process by which organisms capture and mineralize silica. Reviews in Mineralogy and Geochemistry, 2003, 54(1): 291–327
16 Cha J N, Shimizu K, Zhou Y, . Silicatein filaments and subunits from a marine sponge direct the polymerization of silica and silicones in vitro. Proceedings of the National Academy of Sciences of the United States of America, 1999, 96(2): 361–365
17 Weaver J, Morse D E. Molecular biology of demosponge axial filaments and their roles in biosilification. Microscopy Research and Technique, 2003, 62(4): 356–367
18 Krasko A, Lorenz B, Batel R, . Expression of silicatein and collagen genes in the marine sponge Suberites domuncula is controlled by silicate and myotrophin. European Journal of Biochemistry, 2000, 267(15): 4878–4887
19 Krasko A, Gamulin V, Seack J, . Cathepsin, a major protease of the marine sponge Geodia cydonium: purification of the enzyme and molecular cloning of cDNA.Molecular Marine Biology and Biotechnology , 1997, 6(4): 296–307
20 Schr?der H C, Perovi?-Ottstadt S, Wiens M, . Differentiation capacity of the epithelial cells in the sponge Suberites domuncula. Cell and Tissue Research, 2004, 316(2): 271–280
21 Schr?der H C, Perovi?-Ottstadt S, Grebenjuk V A, .. Biosilica formation in spicules of the sponge Suberites domuncula: synchronous expression of a gene cluster. Genomics, 2005, 85(6): 666–678
22 Müller W E G, Belikov S I, Tremel W, . Siliceous spicules in marine demosponges (example Suberites domuncula). Micron, 2006, 37(2): 107–120
23 Kaluzhnaya O V, Belikov S I, Schr?der H C, . Dynamics of skeleton formation in the Lake Baikal sponge Lubomirskia baicalensis. Part II. Molecular biological studies. Naturwissenschaften , 2005, 92(3): 134–138
24 Wiens M, Belikov S I, Kaluzhnaya O V, . Molecular control of serial module formation along the apical-basal axis in the sponge Lubomirskia baicalensis: silicateins, mannose-binding lectin and mago nashi. Development Genes and Evolution, 2006, 216(5): 229–242
25 Krasko A, Schr?der H C, Batel R, . Iron induces proliferation and morphogenesis in primmorphs from the marine sponge Suberites domuncula.DNA and Cell Biology , 2002, 21(1): 67–80
26 Müller W E G, Krasko A, Le Pennec G, . Biochemistry and cell biology of silica formation in sponges. Microscopy Research and Technique, 2003, 62: 368–377
27 Müller W E G, Rothenberger M, Boreiko A, . Formation of siliceous spicules in the marine demosponge Suberites domuncula. Cell and Tissue Research, 2005, 321(2): 285–297
28 Tao K, Stearns N A, Dong J, . The proregion of cathepsin L is required for proper folding, stability and ER exit. Archives of Biochemistry and Biophysics, 1994, 311(1): 19–27
29 Schr?der H C, Wiens M, Schlo?macher U, . Silicatein-mediated polycondensation of orthosilicic acid: modeling of catalytic mechanism involving ring formation. Silicon, 2011,
doi: 10.1007/s12633-010-9057-4 pmid: (in press)
30 Schr?der H C,Krasko A, Le Pennec G, . Silicase, an enzyme which degrades biogenous amorphous silica: contribution to the metabolism of silica deposition in the demosponge Suberites domuncula. Progress in Molecular and Subcellular Biology, 2003, 33: 249–268
31 Sly W S, Hu P Y. Human carbonic anhydrases and carbonic anhydrase deficiencies. Annual Review of Biochemistry, 1995, 64: 375–401
32 Müller W E G, Schr?der H C, Loren B, . Silicatein-mediated synthesis of amorphous silicates and siloxanes and use thereof. European Patent, No. EP 1320624, 2000-07-28
33 Müller W E G, Schr?der H C, Krakso A. Decomposition and modification of silicate and silicone by silicase and use of the reversible enzyme. US Patent, No. US 2007218044, 2007-09-20
34 Sun Q, Vrieling E G, van Santen R A, . Bioinspired synthesis of mesoporous silicas. Current Opinion in Solid State and Materials Science , 2004, 8(2): 111–120
35 Schr?der H C, Brandt D, Schlo?macher U, . Enzymatic production of biosilica glass using enzymes from sponges: basic aspects and application in nanobiotechnology (material sciences and medicine). Naturwissenschaften, 2007, 94(5): 339–359
36 Müller W E G, Wang X M, Belikov S I, . Formation of siliceous spicules in demosponges: example Suberites domuncula. In: B?uerlein E, ed. Handbook of Biomineralization, Vol. 1: Biological Aspects and Structure Formation. Weinheim: Wiley-VCH, 2007, 59–82
37 Schr?der H C, Wang X H, Tremel W, . Biofabrication of biosilica-glass by living organisms. Natural Product Reports, 2008, 25(3): 455–474
38 Müller W E G, Wang X H, Cui F Z, . Sponge spicules as blueprints for the biofabrication of inorganic-organic composites and biomaterials. Applied Microbiology and Biotechnology, 2009, 83(3): 397–413
39 Wiens M, Wang X, Natalio F, . Bioinspired fabrication of bio-silica-based bone-substitution materials. Advanced Engineering Materials, 2010, 12(9): B438–B450
40 Hench L L, Wilson J.Surface-active biomaterials. Science, 1984, 226(4675): 630–636
41 Yamamuro T, Hench L L, Wilson J. Handbook on Bioactive Ceramics, vol. I: Bioactive Glasses and Glass-Ceramics. Boca Raton, FL: CRC Press, 1990
42 Schr?der H C, Boreiko O, Krasko A, . Mineralisation of SaOS-2 cells on enzymatically (silicatein) modified bioactive osteoblast-stimulating surfaces. Journal of Biomedical Materials Research Part B: Applied Biomaterials, 2005, 75B(2): 387–392
43 Curnow P, Kisailus D, Morse D E. Biocatalytic synthesis of poly(l-lactide) by native and recombinant forms of the silicatein enzymes. Angewandte Chemie International Edition, 2006, 45(4): 613–616
44 Wiens M, Wang X H, Schr?der H C, . The role of biosilica in the osteoprotegerin/RANKL ratio in human osteoblast-like cells. Biomaterials, 2010, 31(30): 7716–7725
45 Wiens M, Wang X H, Schlo?macher U, . Osteogenic potential of bio-silica on human osteoblast-like (SaOS-2) cells. Calcified Tissue International, 2010, 87(6): 513–524
46 Struyf E, Conley D J. Silica: an essential nutrient in wetland biogeochemistry. Frontiers in Ecology and the Environment, 2009, 7(2): 88–94
47 Carlisle E M.In vivo requirement for silicon in articular cartilage and connective tissue formation in the chick. The Journal of Nutrition, 1976, 106: 478–484
48 Van Dyck K, Van Cauwenbergh R, Robberecht H, . Bioavailability of silicon from food and food supplements. Fresenius’ Journal of Analytical Chemistry, 1999, 363(5-6): 541–544
49 Carlisle E M.Silicon: an essential element for the chick. Science, 1972, 178(4061): 619–621
50 Müller W E G, Boreiko A, Wang X H, . Morphogenetic activity of silica and bio-silica on the expression of genes controlling biomineralization using SaOS-2 cells. Calcified Tissue International, 2007, 81(5): 382–393
51 Aldinger G, Herr G, Küsswetter W, . Bone morphogenetic protein: a review. International Orthopaedics, 1991, 15(2): 169–177
52 Kamegai A, Tanabe T, Nagahara K, . Pathologic and enzyme histochemical studies on bone formation induced by bone morphogenetic protein in mouse muscle tissue. Acta Histochemica, 1990, 89(1): 25–35
53 Chung C-H, Golub E E, Forbes E, . Mechanism of action of β-glycerophosphate on bone cell mineralization. Calcified Tissue International, 1992, 51(4): 305–311
54 Schwarz K, Milne D B. Growth-promoting effects of silicon in rats. Nature , 1972, 239(5371): 333–334
55 Borsje M A, Ren Y, de Haan-Visser H W, . Comparison of low-intensity pulsed ultrasound and pulsed electromagnetic field treatments on OPG and RANKL expression in human osteoblast-like cells. The Angle Orthodontist, 2010, 80(3): 498–503
56 Simonet W S, Lacey D L, Dunstan C R, . Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell, 1997, 89(2): 309–319
57 Wang J C, Hemavathy K, Charles W, . Osteosclerosis in idiopathic myelofibrosis is related to the overproduction of osteoprotegerin (OPG). Experimental Hematology, 2004, 32(10): 905–910
58 Lane N E, Yao W. Developments in the scientific understanding of osteoporosis. Arthritis Research &amp; Therapy, 2009, 11(3): 228 (8 pages)
59 W?hler F. Ueber künstliche Bildung des Harnstoffs. Annalen der Physik, 1828 , 88(2): 253–256 (in German)
60 Pasteur L. Mémoire sur la fermentation appelée lactique. Mémoires de la Société (Royale) des Sciences, de l’Agriculture et des Arts à Lille, 1857, 5: 13–26 (in French)
doi: 10.1016/j.exphem.2004.07.006
61 Hoppe-Seyler E F. Preface. Zeitschrift für Physiologische Chemie, 1877, 1: 1 (in German)
62 Spallanzani L, Senebier J. Experiences sur la Digestion de l’Homme et de Différentes especes d’Animaux. Geneve: Chez Barthe?lemi Chirol, 1784 (in French)
doi: 10.1002/andp.18280880206
63 Müller W E G, Wang X H, Diehl-Seifert B, . Inorganic polymeric phosphate/polyphosphate as an inducer of alkaline phosphatase and a modulator of intracellular Ca2+ level in osteoblasts (SaOS-2 cells) in vitro. Acta Biomaterialia, 2011, 7(6): 2661–2671
64 Aoba T, Fukae M, Tanabe T, . Selective adsorption of porcine-amelogenins onto hydroxyapatite and their inhibitory activity on hydroxyapatite growth in supersaturated solutions. Calcified Tissue International, 1987, 41(5): 281–289
65 Carlisle E M.Silicon as an essential trace element in animal nutrition. In: Ciba Foundation Symposium 121. Wiley, Chichester, UK , 1986, 123–139
[1] Ling-Yu LI, Bin LIU, Rong-Chang ZENG, Shuo-Qi LI, Fen ZHANG, Yu-Hong ZOU, Hongwei (George) JIANG, Xiao-Bo CHEN, Shao-Kang GUAN, Qing-Yun LIU. In vitro corrosion of magnesium alloy AZ31 --- a synergetic influence of glucose and Tris[J]. Front. Mater. Sci., 2018, 12(2): 184-197.
[2] 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.
[3] Xuran GUO,Kaile ZHANG,Mohamed EL-AASSAR,Nanping WANG,Hany EL-HAMSHARY,Mohamed EL-NEWEHY,Qiang FU,Xiumei MO. The comparison of the Wnt signaling pathway inhibitor delivered electrospun nanoyarn fabricated with two methods for the application of urethroplasty[J]. Front. Mater. Sci., 2016, 10(4): 346-357.
[4] Su-Ju XU,Fu-Zhai CUI,Xiang-Dong KONG. Tumor-suppressor effects of chemical functional groups in an in vitro co-culture system[J]. Front. Mater. Sci., 2014, 8(2): 136-141.
[5] Lei YANG, Chao ZHONG. Advanced engineering and biomimetic materials for bone repair and regeneration[J]. Front Mater Sci, 2013, 7(4): 313-334.
[6] 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.
[7] Hua DENG, Xing-Can SHEN, Xiu-Mei WANG, Chang DU. Calcium carbonate crystallization controlled by functional groups: A mini-review[J]. Front Mater Sci, 2013, 7(1): 62-68.
[8] Zhen-Hua CHEN, Xiu-Li REN, Hui-Hui ZHOU, Xu-Dong LI. The role of hyaluronic acid in biomineralization[J]. Front Mater Sci, 2012, 6(4): 283-296.
[9] Xiao-Hong WANG, Ute SCHLO?MACHER, Shun-Feng WANG, Heinz C. SCHR?DER, Matthias WIENS, Renato BATEL, Werner E. G. MüLLER. From nanoparticles via microtemplates and milliparticles to deep-sea nodules: biogenically driven mineral formation[J]. Front Mater Sci, 2012, 6(2): 97-115.
[10] Punuri Jayasekhar BABU, Pragya SHARMA, Mohan Chandra KALITA, Utpal BORA. Green synthesis of biocompatible gold nanoparticles using Fagopyrum esculentum leaf extract[J]. Front Mater Sci, 2011, 5(4): 379-387.
[11] Jia-Cheng GAO, Li-Ying QIAO, Ren-Long XIN, . Effect of Mg 2+ concentration on biocompatibility of pure magnesium[J]. Front. Mater. Sci., 2010, 4(2): 126-131.
[12] Jia-Cheng GAO, Li-Ying QIAO, Ren-Long XIN, . Corrosion and bone response of magnesium implants after surface modification by heat-self-assembled monolayer[J]. Front. Mater. Sci., 2010, 4(2): 120-125.
[13] Xin-Yu YE, Min-Fang CHEN, Chen YOU, De-Bao LIU, . The influence of HF treatment on corrosion resistance and in vitro biocompatibility of Mg-Zn-Zr alloy[J]. Front. Mater. Sci., 2010, 4(2): 132-138.
[14] Xue-Nan GU, Yu-Feng ZHENG, . A review on magnesium alloys as biodegradable materials[J]. Front. Mater. Sci., 2010, 4(2): 111-115.
[15] Susan LIAO, Casey K. CHAN, S. RAMAKRISHNA, . Electrospun nanofibers: Work for medicine?[J]. Front. Mater. Sci., 2010, 4(1): 29-33.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed