AccScience Publishing / IJB / Volume 4 / Issue 1 / DOI: 10.18063/ijb.v4i1.120
Cite this article
8
Download
583
Views
Journal Browser
Volume | Year
Issue
Search
News and Announcements
View All
RESEARCH ARTICLE

An nMgO containing scaffold: Antibacterial activity, degradation properties and cell responses

Cijun Shuai1,2,3 Wang Guo1 Chengde Gao1 Youwen Yang1 Ping Wu4 Pei Feng1*
Show Less
1 State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
2 Jiangxi University of Science and Technology, Ganzhou, China
3 Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
4 College of Chemistry, Xiangtan University, Xiangtan, China
© Invalid date by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

Bone repair failure caused by implant-related infections is a common and troublesome problem. In this study, an antibacterial scaffold was developed via selective laser sintering with incorporating nano magnesium oxide (nMgO) to poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV). The results indicated the scaffold exerted high antibacterial activity. The antibacterial mechanism was that nMgO could cause oxidative damage and mechanical damage to bacteria through the production of reactive oxygen species (ROS) and direct contact action, respectively, which resulted in the damage of their structures and functions. Besides, nMgO significantly increased the compressive properties of the scaffold including strength and modulus, due to its excellent mechanical properties and uniform dispersion in the PHBV matrix. Moreover, the degradation tests indicated nMgO neutralized the acid degradation products of PHBV and benefited the degradation of the scaffold. The cell culture demonstrated that nMgO promoted the cellular adhesion and proliferation, as well as osteogenic differentiation. The present work may open the door to exploring nMgO as a promising antibacterial material for tissue engineering. 

Keywords
Nano magnesium oxide
antibacterial scaffolds
degradation properties
cytocompatibility
mechanical properties
References

[1]Zimmerli W, 2014, Clinical presentation and treatment of orthopaedic implant-associated infection. J Intern Med, 276(2): 111–119. http://dx.doi.org/10.1111/joim.12233
[2]Saidin S, Chevallier P, Abdul Kadir M R, et al., 2013, Polydopamine as an intermediate layer for silver and hydroxyapatite immobilisation on metallic biomaterials surface. Mater Sci Eng C Mater Biol Appl, 33(8): 4715–4724. http://dx.doi.org/10.1016/j.msec.2013.07.026
[3]Lorenzetti M, Dogsa I, Stosicki T, et al., 2015, The influence of surface modification on bacterial adhesion to titanium-based substrates. ACS Appl Mater Interfaces, 7(3): 1644–1651. http://dx.doi.org/10.1021/am507148n
[4]Overbye K and Barrett J, 2005, Antibiotics: Where did we go wrong? Drug Discov Today, 10(1): 45–52. http://dx.doi.org/10.1016/s1359-6446(04)03285-4
[5]Londonkar R L, Madire Kattegouga U, Shivsharanappa K, et al., 2013, Phytochemical screening and in vitro antimicrobial activity of Typha angustifolia Linn leaves extract against pathogenic gram negative micro organisms. J Pharm Res, 6(2): 280–283. http://dx.doi.org/10.1016/j.jopr.2013.02.010
[6]Trampuz A and Zimmerli W, 2006, Antimicrobial agents in orthopaedic surgery. Drugs, 66(8): 1089–1106. http://dx.doi.org/10.2165/00003495-200666080-00005
[7]Goodman S B, Yao Z, Keeney M, et al., 2013, The future of biologic coatings for orthopaedic implants. Biomaterials, 34(13): 3174–3183. http://dx.doi.org/10.1016/j.biomaterials.2013.01.074
[8]Yang S, Zhang Y, Yu J, et al., 2014, Antibacterial and mechanical properties of honeycomb ceramic materials incorporated with silver and zinc. Mater Des, 59: 461–465. http://dx.doi.org/10.1016/j.matdes.2014.03.025
[9]Yazdimamaghani M, Vashaee D, Assefa S, et al., 2014, Hybrid macroporous gelatin/bioactive-glass/nanosilver scaffolds with controlled degradation behavior and antimicrobial activity for bone tissue engineering. J Biomed Nanotechnol, 10(6): 911–931. http://dx.doi.org/10.1166/jbn.2014.1783
[10]Sánchez-Salcedo S, Shruti S, Salinas A J, et al., 2014, In vitro antibacterial capacity and cytocompatibility of SiO2–CaO–P2O5 meso-macroporous glass scaffolds enriched with ZnO. J Mater Chem B, 2(30): 4836–4847. http://dx.doi.org/10.1039/c4tb00403e
[11]Vargas-Reus M A, Memarzadeh K, Huang J, et al., 2012, Antimicrobial activity of nanoparticulate metal oxides against peri-implantitis pathogens. Int J Antimicrob Agents, 40(2): 135–139. http://dx.doi.org/10.1016/j.ijantimicag.2012.04.012
[12]Dizaj S M, Lotfipour F, Barzegar-Jalali M, et al., 2014, Antimicrobial activity of the metals and metal oxide nanoparticles. Mater Sci Eng C Mater Biol Appl, 44: 278–284. http://dx.doi.org/10.1016/j.msec.2014.08.031
[13]Li Y, Zhang W, Niu J, et al., 2012, Mechanism of photogenerated reactive oxygen species and correlation with the antibacterial properties of engineered metal-oxide nanoparticles. ACS Nano, 6(6): 5164–5173. http://dx.doi.org/10.1021/nn300934k
[14]Krishnamoorthy K, Moon J Y, Hyun H B, et al., 2012, Mechanistic investigation on the toxicity of MgO nanoparticles toward cancer cells. J Mater Chem, 22(47): 24610–24617. http://dx.doi.org/10.1039/c2jm35087d
[15]Staiger M P, Pietak A M, Huadmai J, et al., 2006, Magnesium and its alloys as orthopedic biomaterials: A review. Biomaterials, 27(9): 1728–1734. http://dx.doi.org/10.1016/j.biomaterials.2005.10.003
[16]De Silva R T, Mantilaka M M, Ratnayake S P, et al., 2017, Nano-MgO reinforced chitosan nanocomposites for high performance packaging applications with improved mechanical, thermal and barrier properties. Carbohydr Polym, 157: 739–747. http://dx.doi.org/10.1016/j.carbpol.2016.10.038
[17]Zhao Y, Liu B, You C, et al., 2016, Effects of MgO whiskers on mechanical properties and crystallization behavior of PLLA/MgO composites. Mater Des, 89: 573–581. http://dx.doi.org/10.1016/j.matdes.2015.09.157
[18]Haldorai Y and Shim J-J, 2014, An efficient removal of methyl orange dye from aqueous solution by adsorption onto chitosan/MgO composite: A novel reusable adsorbent. Appl Surf Sci, 292: 447–453. http://dx.doi.org/10.1016/j.apsusc.2013.11.158
[19]Yamamoto O, Ohira T, Alvarez K, et al., 2010, Antibacterial characteristics of CaCO3–MgO composites. Mater Sci Eng B, 173(1–3): 208–212. http://dx.doi.org/10.1016/j.mseb.2009.12.007
[20]Ma F, Lu X, Wang Z, et al., 2011, Nanocomposites of poly(ʟ-lactide) and surface modified magnesia nanoparticles: Fabrication, mechanical property and biodegradability. J Phys Chem Solids, 72(2): 111–116. http://dx.doi.org/10.1016/j.jpcs.2010.11.008
[21]Feng P, Peng S, Wu P, et al., 2016, A space network structure constructed by tetraneedlelike ZnO whiskers supporting boron nitride nanosheets to enhance comprehensive properties of poly (ʟ-lacti acid) scaffolds. Sci Rep, 6: 33385. http://dx.doi.org/10.1038/srep33385
[22]Lee J M, Sing S L, Tan E Y S, et al., 2016, Bioprinting in cardiovascular tissue engineering: A review. Int J Bioprint, 2(2): 27–36. http://dx.doi.org/10.18063/IJB.2016.02.006 
[23]Murphy C, Kolan K, Li W, et al., 2017, 3D bioprinting of stem cells and polymer/bioactive glass composite scaffolds for bone tissue engineering. Int J Bioprint, 3(1): 54–64. http://dx.doi.org/10.18063/IJB.2017.01.005
[24]Eshraghi S and Das S, 2010, Mechanical and microstructural properties of polycaprolactone scaffolds with 1-D, 2-D, and 3-D orthogonally oriented porous architectures produced by selective laser sintering. Acta Biomater, 6(7): 2467–2476. http://dx.doi.org/10.1016/j.actbio.2010.02.002
[25]Eshraghi S and Das S, 2012, Micromechanical finite-element modeling and experimental characterization of the compressive mechanical properties of polycaprolactone–hydroxyapatite composite scaffolds prepared by selective laser sintering for bone tissue engineering. Acta Biomater, 8(8): 3138–3143. http://dx.doi.org/10.1016/j.actbio.2012.04.022
[26]Amalric J, Mutin P H, Guerrero G, et al., 2009, Phosphonate monolayers functionalized by silver thiolate species as antibacterial nanocoatings on titanium and stainless steel. J Mater Chem, 19(1): 141–149. http://dx.doi.org/10.1039/b813344a
[27]Simchi A, Tamjid E, Pishbin F, et al., 2011, Recent progress in inorganic and composite coatings with bactericidal capability for orthopaedic applications. Nanomedicine, 7(1): 22–39. http://dx.doi.org/10.1016/j.nano.2010.10.005
[28]Ye L, Liu J, Jiang Z, et al., 2013, Facets coupling of BiOBr-g-C3N4 composite photocatalyst for enhanced visible-light-driven photocatalytic activity. Appl Catal B, 142–143: 1–7. http://dx.doi.org/10.1016/j.apcatb.2013.04.058
[29]Wu D, Wang B, Wang W, et al., 2015, Visible-light-driven BiOBr nanosheets for highly facet-dependent photocatalytic inactivation of Escherichia coli. J Mater Chem A, 3(29): 15148–15155. http://dx.doi.org/10.1039/c5ta02757h
[30]Bruzauskaite I, Bironaite D, Bagdonas E, et al., 2016, Scaffolds and cells for tissue regeneration: Different scaffold pore sizes-different cell effects. Cytotechnology, 68(3): 355–369. http://dx.doi.org/10.1007/s10616-015-9895-4
[31]Roosa S M, Kemppainen J M, Moffitt E N, et al., 2010, The pore size of polycaprolactone scaffolds has limited influence on bone regeneration in an in vivo model. J Biomed Mater Res A, 92(1): 359–368. http://dx.doi.org/10.1002/jbm.a.32381
[32]Schek R M, Wilke E N, Hollister S J, et al., 2006, Combined use of designed scaffolds and adenoviral gene therapy for skeletal tissue engineering. Biomaterials, 27(7): 1160–1166. http://dx.doi.org/10.1016/j.biomaterials.2005.07.029
[33]Ten E, Jiang L and Wolcott M P, 2012, Crystallization kinetics of poly(3-hydroxybutyrate-co-3-hydroxyvalerate)/cellulose nanowhiskers composites. Carbohydr Polym, 90(1): 541. http://dx.doi.org/10.1016/j.carbpol.2012.05.076
[34]Shuai C, Guo W, Gao C, et al., 2017, Calcium silicate improved bioactivity and mechanical properties of poly(3-hydroxybutyrate-co-3-hydroxyvalerate) scaffolds. Polymers, 9(5): 175. http://dx.doi.org/10.3390/polym9050175
[35]Yin Y, Zhang G and Xia Y, 2002, Synthesis and characterization of MgO nanowires through a vapor-phase precursor method. Adv Funct Mater, 12(4): 293–298. http://dx.doi.org/10.1002/1616-3028(20020418)12:4<293::aid-adfm293>3.0.co;2-u
[36]Hutmacher D W, Schantz J T, Lam C X, et al., 2007, State of the art and future directions of scaffold-based bone engineering from a biomaterials perspective. J Tissue Eng Regen Med, 1(4): 245–260. http://dx.doi.org/10.1002/term.24
[37]Ning N-y, Yin Q-j, Luo F, et al., 2007, Crystallization behavior and mechanical properties of polypropylene/halloysite composites. Polymer, 48(25): 7374–7384. http://dx.doi.org/10.1016/j.polymer.2007.10.005
[38]Li H Y, Tan Y Q, Zhang L, et al., 2012, Bio-filler from waste shellfish shell: Preparation, characterization, and its effect on the mechanical properties on polypropylene composites. J Hazard Mater, 217–218: 256–262. http://dx.doi.org/10.1016/j.jhazmat.2012.03.028
[39]He F, Fan J and Lau S, 2008, Thermal, mechanical, and dielectric properties of graphite reinforced poly(vinylidene fluoride) composites. Polym Test, 27(8): 964–970. http://dx.doi.org/10.1016/j.polymertesting.2008.08.010
[40]Maity J, Jacob C, Das C K, et al., 2008, Direct fluorination of Twaron fiber and the mechanical, thermal and crystallization behaviour of short Twaron fiber reinforced polypropylene composites. Compos Part A Appl Sci Manuf, 39(5): 825–833. http://dx.doi.org/10.1016/j.compositesa.2008.01.009
[41]Peng D, Qin W, Wu X, et al., 2015, Improvement of the resistance performance of carbon/cyanate ester composites during vacuum electron radiation by reduced graphene oxide modified TiO2. RSC Adv, 5(94): 77138–77146. http://dx.doi.org/10.1039/c5ra11113g
[42]Liu G, Zhou T, Liu W, et al., 2014, Enhanced desulfurization performance of PDMS membranes by incorporating silver decorated dopamine nanoparticles. J Mater Chem A, 2(32): 12907. http://dx.doi.org/10.1039/c4ta01778a
[43]Lee S-W, Han S M and Nix W D, 2009, Uniaxial compression of fcc Au nanopillars on an MgO substrate: The effects of prestraining and annealing. Acta Mater, 57(15): 4404–4415. http://dx.doi.org/10.1016/j.actamat.2009.06.002
[44]Applerot G, Lellouche J, Lipovsky A, et al., 2012, Understanding the antibacterial mechanism of CuO nanoparticles: Revealing the route of induced oxidative stress. Small, 8(21): 3326–3337. http://dx.doi.org/10.1002/smll.201200772
[45]Applerot G, Lipovsky A, Dror R, et al., 2009, Enhanced antibacterial activity of nanocrystalline ZnO due to increased ROS-mediated cell injury. Adv Funct Mater, 19(6): 842–852. http://dx.doi.org/10.1002/adfm.200801081
[46]Sawai J, Kojima H, Igarashi H, et al., 2000, Antibacterial characteristics of magnesium oxide powder. World J Microbiol Biotechnol, 16(2): 187–194. http://dx.doi.org/10.1023/A:1008916209784
[47]Krishnamoorthy K, Manivannan G, Kim S J, et al., 2012, Antibacterial activity of MgO nanoparticles based on lipid peroxidation by oxygen vacancy. J Nanopart Res, 14(9): 1063. http://dx.doi.org/10.1007/s11051-012-1063-6
[48]Sterrer M, Diwald O and Knözinger E, 2000, Vacancies and electron deficient surface anions on the surface of MgO nanoparticles. J Phys Chem B, 104(15): 3601–3607. http://dx.doi.org/10.1021/jp993924l 
[49]Berger T, Sterrer M, Stankic S, et al., 2005, Trapping of photogenerated charges in oxide nanoparticles. Mater Sci Eng C, 25(5–8): 664–668. http://dx.doi.org/10.1016/j.msec.2005.06.013
[50]Sterrer M, Berger T, Diwald O, et al., 2003, Energy transfer on the MgO surface, monitored by UV-induced H2 chemisorption. J Am Chem Soc, 125(1): 195–199. http://dx.doi.org/10.1021/ja028059o
[51]Long T C, Saleh N, Tilton R D, et al., 2006, Titanium dioxide (P25) produces reactive oxygen species in immortalized brain microglia (BV2): Implications for nanoparticle neurotoxicity. Environ Sci Technol, 40(14): 4346–4352. http://dx.doi.org/10.1021/es060589n
[52]Xia T, Kovochich M, Brant J, et al., 2006, Comparison of the abilities of ambient and manufactured nanoparticles to induce cellular toxicity according to an oxidative stress paradigm. Nano Lett, 6(8): 1794–1807. http://dx.doi.org/10.1021/nl061025k
[53]Jin T and He Y, 2011, Antibacterial activities of magnesium oxide (MgO) nanoparticles against foodborne pathogens. J Nanopart Res, 13(12): 6877–6885. http://dx.doi.org/10.1007/s11051-011-0595-5
[54]Yamamoto O, Sawai J, Kojima H, et al., 2002, Effect of mixing ratio on bactericidal action of MgO–CaO powders. J Mater Sci Mater Med, 13(8): 789–792. http://dx.doi.org/10.1023/A:1016179225955
[55]Jeevanandam P and Klabunde K, 2002, A study on adsorption of surfactant molecules on magnesium oxide nanocrystals prepared by an aerogel route. Langmuir, 18(13): 5309–5313. http://dx.doi.org/10.1021/la0200921
[56]He Y, Ingudam S, Reed S, et al., 2016, Study on the mechanism of antibacterial action of magnesium oxide nanoparticles against foodborne pathogens. J Nanobiotechnol, 14(1): 54. http://dx.doi.org/10.1186/s12951-016-0202-0
[57]Salomao R, Bittencourt L and Pandolfelli V, 2007, A novel approach for magnesia hydration assessment in refractory castables. Ceram Int, 33(5): 803–810. http://dx.doi.org/10.1016/j.ceramint.2006.01.004
[58]Mo L, Deng M, Tang M, et al., 2014, MgO expansive cement and concrete in China: Past, present and future. Cem Concr Res, 57: 1–12. http://dx.doi.org/10.1016/j.cemconres.2013.12.007
[59]Shan D, Shi Y, Duan S, et al., 2013, Electrospun magnetic poly (ʟ-lactide) (PLLA) nanofibers by incorporating PLLA-stabilized Fe3O4 nanoparticles. Mater Sci Eng C, 33(6): 3498–3505. http://dx.doi.org/10.1016/j.msec.2013.04.040
[60]Marom R, Shur I, Solomon R, et al., 2005, Characterization of adhesion and differentiation markers of osteogenic marrow stromal cells. J Cell Physiol, 202(1): 41–48. http://dx.doi.org/10.1002/jcp.20109
[61]Wang F, Zhai D, Wu C, et al., 2016, Multifunctional mesoporous bioactive glass/upconversion nanoparticle nanocomposites with strong red emission to monitor drug delivery and stimulate osteogenic differentiation of stem cells. Nano Res, 9(4): 1193–1208. http://dx.doi.org/10.1007/s12274-016-1015-z
[62]Zhang J and Zhu Y, 2014, Synthesis and characterization of CeO2-incorporated mesoporous calcium-silicate materials. Microporous Mesoporous Mater, 197: 244–251. http://dx.doi.org/10.1016/j.micromeso.2014.06.018
[63]Hoppe A, Guldal N S and Boccaccini A R, 2011, A review of the biological response to ionic dissolution products from bioactive glasses and glass-ceramics. Biomaterials, 32(11): 2757–2774. http://dx.doi.org/10.1016/j.biomaterials.2011.01.004
[64]Yamniuk A P and Vogel H J, 2005, Calcium- and magnesium-dependent interactions between calcium- and integrin-binding protein and the integrin αIIb cytoplasmic domain. Protein Sci, 14(6): 1429–1437. http://dx.doi.org/10.1110/ps.041312805
[65]Zreiqat H, Howlett C, Zannettino A, et al., 2002, Mechanisms of magnesium-stimulated adhesion of osteoblastic cells to commonly used orthopaedic implants. J Biomed Mater Res A, 62(2): 175–184. http://dx.doi.org/10.1002/jbm.10270
[66]Bouvard D, Pouwels J, De Franceschi N, et al., 2013, Integrin inactivators: Balancing cellular functions in vitro and in vivo. Nat Rev Mol Cell Biol, 14(7): 430–442. http://dx.doi.org/10.1038/nrm3599
[67]Bourboulia D and Stetler-Stevenson W G, 2010, Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs): Positive and negative regulators in tumor cell adhesion. Semin Cancer Biol, 20(3): 161–168. http://dx.doi.org/10.1016/j.semcancer.2010.05.002

Share
Back to top
International Journal of Bioprinting, Electronic ISSN: 2424-8002 Print ISSN: 2424-7723, Published by AccScience Publishing