8
A comprehensive biological evaluation of ceramic nanoparticles as wear debris Y.F. Zhang, MPhil a , Y.F. Zheng, PhD a, , L. Qin, PhD b,c a State Key Laboratory for Turbulence and Complex System and Department of Advanced Materials and Nanotechnology, College of Engineering, Peking University, Beijing, China b Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong c Translational Medicine Research & Development Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzheng, China Received 6 October 2010; accepted 18 April 2011 Abstract Patients are exposed internally to nanoparticles (NPs) by wear mechanisms associated with total joint arthroplasty. This tissue-specific retention implies that the biological evaluation of NPs shall be integrative and niche targeting. Here, we report that ceramic zirconia and silicon nitride NPs interfere with MG63 cells' function and remarkably stimulate the secretion of TNF-α in RAW264.7 cells. However, alumina NPs significantly promote the alkaline phosphatase (ALP) activity of MG63 cells at low concentration and do not show irritation to macrophages. In this study, we prove that ceramic materials at nanoscale are bioactive to cells. These findings also suggest that a more rational paradigm for the biosafety evaluation of NPs than is currently in place is needed before their clinical applications. From the Clinical Editor: In this study, the authors demonstrate that ceramic nano materials associated with normal wear-and-tear of joint prostheses at nanoscale are bioactive to cells. © 2011 Elsevier Inc. All rights reserved. Key words: Ceramic nanoparticles; Wear debris; Biosafety; Orthopedics Periprosthetic osteolysis is emerging as the major cause of failure in the long-term durability of total hip prosthesis. 1,2 The tissue membranes obtained from patients with aseptic loosening provided the evidence of foreign-body reaction accompanying wear particles. 3 Currently, the osteolysis has mainly been attributed to the biological responses to the repetitive exposure of wear particles at the bone-implant interface. 4 In 1972, Boutin first used ceramic material in total hip arthroplasty (THA) and not only attributed superior mechanical properties to it but also fewer wear particles, which made ceramics attractive in orthopedics. 5 Currently, the ceramic articulate components have been widely applied in THA. However, loosened ceramic prostheses retrieved from patients associated with extensive osteolysis suggests that ceramic bearings might not be as bioinert as initially perceived. 6,7 In the periprosthetic membrane of loosened ceramic prostheses, clinicians found not only abundant ceramic wear particles, including alumina and zirconia particles, but also Ti6Al4V alloy, and pure Ti particles were found with the presence of histiocytic foreign-body reaction. 8,9 As with metal and polyethylene particles, ceramic particles might be phagocytosed by macrophages. Although some authors considered the ceramics, especially zirconia particles, as the major cause of osteolysis around ceramic implants, it remains unclear whether the particle dose or composition determined the tissue's biological responses around the implant. 9 Due to less accurate techniques used in periprosthetic tissue digestion and particles' extraction, previous studies mainly focused on wear particles with sizes ranging from submicron to micron meters. 10,11 With laser capture microdissection (LCM) and transmission electron microscope (TEM), Ingham et al found more tiny alumina wear particles (590 nm) accompanied by tissue necrosis as evidenced by presence of macrophages in tissues around the alumina-on-alumina prosthesis. 12 Considering the fact that differently sized particles had discriminating effects on osteoblast function and proliferation, 13 the particle size must be considered an important parameter. Moreover, it was proven that cobaltchromium (CoCr) NPs caused more free radicals and BASIC SCIENCE Nanomedicine: Nanotechnology, Biology, and Medicine 7 (2011) 975 982 Research Article nanomedjournal.com This work is supported by National High Technology Research and Development Program of China (863 Program) under grant number 2011AA030101, National Natural Science Foundation of China (No. 51041004), and Program for New Century Excellent Talents in University (NCET-07-0033). Corresponding author. Department of Advanced Materials and Nanotechnology, College of Engineering, Peking University, No. 5 Yi-He- Yuan Road, Hai-Dian District, Beijing 100871, CHINA. E-mail address: [email protected] (Y.F. Zheng). 1549-9634/$ see front matter © 2011 Elsevier Inc. All rights reserved. doi:10.1016/j.nano.2011.04.005 Please cite this article as: Y.F. Zhang, Y.F. Zheng, L. Qin, A comprehensive biological evaluation of ceramic nanoparticles as wear debris. Nanomedicine: NBM 2011;7:975-982, doi:10.1016/j.nano.2011.04.005

A comprehensive biological evaluation of ceramic ...lbmd.coe.pku.edu.cn/PDF/2011nnbm.pdf · A comprehensive biological evaluation of ceramic nanoparticles as wear debris ... (TCP)

  • Upload
    others

  • View
    6

  • Download
    0

Embed Size (px)

Citation preview

Page 1: A comprehensive biological evaluation of ceramic ...lbmd.coe.pku.edu.cn/PDF/2011nnbm.pdf · A comprehensive biological evaluation of ceramic nanoparticles as wear debris ... (TCP)

BASIC SCIENCE

Nanomedicine: Nanotechnology, Biology, and Medicine7 (2011) 975–982

Research Article nanomedjournal.com

A comprehensive biological evaluation of ceramic nanoparticles as

wear debris

Y.F. Zhang, MPhila, Y.F. Zheng, PhDa,⁎, L. Qin, PhDb,c

aState Key Laboratory for Turbulence and Complex System and Department of Advanced Materials and Nanotechnology,College of Engineering, Peking University, Beijing, China

bMusculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong KongcTranslational Medicine Research & Development Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzheng, China

Received 6 October 2010; accepted 18 April 2011

Abstract

Patients are exposed internally to nanoparticles (NPs) by wear mechanisms associated with total joint arthroplasty. This tissue-specificretention implies that the biological evaluation of NPs shall be integrative and niche targeting. Here, we report that ceramic zirconia andsilicon nitride NPs interfere with MG63 cells' function and remarkably stimulate the secretion of TNF-α in RAW264.7 cells. However,alumina NPs significantly promote the alkaline phosphatase (ALP) activity of MG63 cells at low concentration and do not show irritation tomacrophages. In this study, we prove that ceramic materials at nanoscale are bioactive to cells. These findings also suggest that a morerational paradigm for the biosafety evaluation of NPs than is currently in place is needed before their clinical applications.

From the Clinical Editor: In this study, the authors demonstrate that ceramic nano materials associated with normal wear-and-tear of jointprostheses at nanoscale are bioactive to cells.© 2011 Elsevier Inc. All rights reserved.

Key words: Ceramic nanoparticles; Wear debris; Biosafety; Orthopedics

Periprosthetic osteolysis is emerging as the major cause offailure in the long-term durability of total hip prosthesis.1,2 Thetissue membranes obtained from patients with aseptic looseningprovided the evidence of foreign-body reaction accompanyingwear particles.3 Currently, the osteolysis has mainly beenattributed to the biological responses to the repetitive exposureof wear particles at the bone-implant interface.4

In 1972, Boutin first used ceramic material in total hiparthroplasty (THA) and not only attributed superior mechanicalproperties to it but also fewer wear particles, which made ceramicsattractive in orthopedics.5 Currently, the ceramic articulatecomponents have been widely applied in THA. However,loosened ceramic prostheses retrieved from patients associatedwith extensive osteolysis suggests that ceramic bearings might not

This work is supported by National High Technology Research andDevelopment Program of China (863 Program) under grant number2011AA030101, National Natural Science Foundation of China (No.51041004), and Program for New Century Excellent Talents in University(NCET-07-0033).

⁎Corresponding author. Department of Advanced Materials andNanotechnology, College of Engineering, Peking University, No. 5 Yi-He-Yuan Road, Hai-Dian District, Beijing 100871, CHINA.

E-mail address: [email protected] (Y.F. Zheng).

1549-9634/$ – see front matter © 2011 Elsevier Inc. All rights reserved.doi:10.1016/j.nano.2011.04.005

Please cite this article as: Y.F. Zhang, Y.F. Zheng, L. Qin, A comprehensive bioNBM 2011;7:975-982, doi:10.1016/j.nano.2011.04.005

be as bioinert as initially perceived.6,7 In the periprostheticmembrane of loosened ceramic prostheses, clinicians found notonly abundant ceramic wear particles, including alumina andzirconia particles, but also Ti6Al4V alloy, and pure Ti particleswere found with the presence of histiocytic foreign-bodyreaction.8,9 As with metal and polyethylene particles, ceramicparticles might be phagocytosed by macrophages. Although someauthors considered the ceramics, especially zirconia particles, asthe major cause of osteolysis around ceramic implants, it remainsunclear whether the particle dose or composition determined thetissue's biological responses around the implant.9

Due to less accurate techniques used in periprosthetic tissuedigestion and particles' extraction, previous studies mainlyfocused on wear particles with sizes ranging from submicron tomicron meters.10,11 With laser capture microdissection (LCM)and transmission electron microscope (TEM), Ingham et al foundmore tiny alumina wear particles (5–90 nm) accompanied bytissue necrosis as evidenced by presence of macrophages intissues around the alumina-on-alumina prosthesis.12 Consideringthe fact that differently sized particles had discriminating effectson osteoblast function and proliferation,13 the particle size mustbe considered an important parameter. Moreover, it was proventhat cobalt–chromium (CoCr) NPs caused more free radicals and

logical evaluation of ceramic nanoparticles as wear debris. Nanomedicine:

Page 2: A comprehensive biological evaluation of ceramic ...lbmd.coe.pku.edu.cn/PDF/2011nnbm.pdf · A comprehensive biological evaluation of ceramic nanoparticles as wear debris ... (TCP)

976 Y.F. Zhang et al / Nanomedicine: Nanotechnology, Biology, and Medicine 7 (2011) 975–982

inducedmore DNAdamage than themicron-sized particles did.14

Other studies also pointed out that NPs (b100 nm) had moreradical interference to cells in vitro than larger ones (N100 nm)from metal alloy, polyethylene and ceramics.14,15

In the microenvironment between bone and prosthesis, wearparticles could directly interact with osteoblasts and suppressosteoblastic activity. Although fibroblasts stimulated by wearparticles might participate in the periprosthetic fibrogenesis andenhance the synthesis of metalloproteinases (MMPs), MMPscould affect the attachment and synthetic activity of osteoblastsand eventually cause bone loss.16 In addition, when macro-phages phagocytosed wear particles, they released many pro-inflammatory and pro-osteoclastogenic cytokines, such as tumornecrosis factor (TNF)-α, interleukin (IL)-1 and IL-6 forpromoting osteoclastogenesis.17 Several studies demonstratedthat wear particles interacted with many types of osteo-relatedcells and impaired the well-balanced coupling between boneformation and resorption.11,13,18 Accordingly, this study wasdesigned to investigate whether the nanosized ceramic particleshad any negative effect on bone-remolding processes that in turnattributed to aseptic loosening. Unlike many other studiesemphasizing the cytotoxicity of NPs, we aimed to developprotocols for systemic evaluation of NPs' biological effects in aspecial bio-microenvironment.

In this study, three kinds of ceramic NPs, i.e., alumina(Al2O3), zirconia (ZrO2), and silicon nitride (Si3N4), which arecommonly used in orthopedic fields,19 were evaluated in vitro.Human steoblast-like cell lines' (MG63) viability and prolifer-ation were evaluated by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. MG63 cells' function andmacrophage RAW264.7 cells' irritation were determinedseparately by ALP activity assay and enzyme-linked immuno-sorbent assay (ELISA) kit. In all experiments specified below,we used pure titanium (Ti) particles as control for comparison.

Methods

Materials

Alumina nanopowders (40–50nm, 99.5%, Catalog #44932;Alfa Aesar, Inc., Ward Hill, Massachusetts) and commerciallypure titanium particles (b20 μm, with average diameter of 4.50μm, 93%, Catalog #00681, Alfa Aesar, Inc.), silicon nitridenanopowders (b50 nm, N98%, Catalog #634581), and zirconia(IV) nanopowders (b50 nm, Catalog #544760) were purchasedfrom Sigma-Aldrich (St. Louis, Missouri). The experimental NPswere autoclaved at 135°C for 3 hours and then treated with 70%ethanol for 48 hours at 20°C followed by washing in phosphatebuffered saline (PBS) to remove endotoxin. The particles'endotoxin level was lower than 0.1 EU/mL, as determined by acommercial detection kit (Chromogenic endpoint TAL with aDiazo coupling kit, Xiamen Houshiji, Fujian, China).

Dispersion and characterization of NPs

All experimental NPs were dispersed in ultrapure, filtratedwater (Millipore, S185, resistance N18.2 MΩ) and deagglomer-ated by sonication (Shanghai Branson Ultrasonics Co. Ltd.,Shanghai, China). The geometric size and surface morphology of

the experimental NPs were examined with TEM microscope(TEM, FEI Tecnai F30, Eindhoven, The Netherlands), andscanning electron microscope (SEM, AMRAY 1910FE, Bed-ford, Massachusetts). The energy-dispersive x-ray spectroscope(EDX 9100, EDAX, Mahwah, New Jersey) was used to analyzethe particle composition. The crystallization of NPs was checkedby x-ray diffraction (XRD, Rigaku-D/maxrB diffractometer, FEITecnai F30) operated at 40kV and 100 mA at room temperature.The characterization showed that all experimental particlescoincided with the description of the manufacturer.

Cells and Methods

Cell culture

Human osteoblast-like cell lines MG-63 (American TypeCulture Collection, Rockville, Maryland), mouse fibroblast celllines L929 (Cell Bank of Shanghai Institutes for BiologicalSciences, Chinese Academy of Science, China) and Murinemacrophagic cell lines RAW264.7 (American Type Cell Collec-tion) were cultured in monolayer in Dulbecco modified eaglemedium (DMEM) (GIBCO-BRL, Grand Island, New York)containing 10 % fetal bovine serum (FBS) (Hyclone Laboratories,Logan, Utah) and supplemented with penicillin (50 μg/ml, Gibco-BRL) and streptomycin (50 μg/ml, Gibco-BRL) in a humidifiedatmosphere of 95% air and 5% carbon dioxide at 37°C. Cells weresubcultivated using 0.25% trypsin-(1 mML) EDTA (Gibco) afterreaching 70%–90% confluence. Cells were counted in ahemocytometer, and the cell viability was determined by theexclusion Trypan Blue (Sigma-Aldrich) method.

For the MTT assays and analysis of the total intracellularprotein (TCP) content, the cells were seeded in 24-well plates(Corning, Lowell, Massachusetts) at a density of 1.0 × 104

cells per well in 500 μL culture medium. After 12 hours, allkinds of experimental particles were freshly dispersed in thecell culture medium and diluted to appropriate concentrations(5-500 μg/mL). MG-63 and L929 cells were cultured in mediacontaining particles for 24, 48 and 72 hours. Culture mediawithout particles served as the control in each experiment.

For ALP activity analysis, the MG63 cells were plated at50,000 cells per well in 24-well plates (Corning) in 500 μLculture medium per well (medium supplemented with 50 mg/mLascorbic acid (Sigma) and 10 mM β-glycerophosphate). At 24hours the media were replaced with experimental mediacontaining dilutions of the particle stock solutions, and thecells were cultured for an additional 48 hours.

For the measurement of cytokine levels by ELISA, RAW264.7 cells (2 × 106 cells per well in a 6-well plate) were incubatedfor 24 hours to reach confluence, then removed the media andreplaced with 2 ml experimental media containing differentconcentrations of experimental particles for another 24 hours.Separate wells were challenged with lipopolysaccharide (LPS)5EU/ml (Xiamen Houshiji, Fujian, China) as positive control.

Cytotoxicity

MTT assays were used to evaluate the proliferation of MG63and L929 cells cultured in media containing different

Page 3: A comprehensive biological evaluation of ceramic ...lbmd.coe.pku.edu.cn/PDF/2011nnbm.pdf · A comprehensive biological evaluation of ceramic nanoparticles as wear debris ... (TCP)

977Y.F. Zhang et al / Nanomedicine: Nanotechnology, Biology, and Medicine 7 (2011) 975–982

concentrations (5–500 μg/mL) of the evaluated experimentalparticles. Mitochondrial enzymes in metabolically active cellsdecomposed the tetrazolium salt into a colored formazanproduct. Cells in the 24-well plates were treated with culturemedia containing different concentrations of particles for 24, 48and 72 hours. After each time point, the medium was aspirated.The cells were gently rinsed with phosphate-buffered salinesolution (1000 μL). Fresh serum-free medium (450 μL) wasadded to each well as well as a MTT stock solution (50 μL, 5 mg/mL). Cells were incubated for 2 hours at 37°C. The medium wasremoved, 500 μL of 5% SDS was added to each well and theplates incubated for a further 24 hours at 37°C. Followingincubation, 200 μL of culture medium was aspirated from eachwell and the optical density was measured at 570 nm (correctionwavelength 630 nm). At least three independent experimentswere carried out for statistical analysis.

Total intracellular protein content

At the end of 72 hours' incubation, MG63 and L929 cells werelysed using 0.5% (v/v) Triton X-100 and twice freeze-thawedcycles. Total protein content in the cell lysates was determinedspectrophotometrically using a commercially available kit (Sigma-Aldrich) and following manufacturer's instructions. For thispurpose, aliquots of each protein-containing supernatant wereincubated with a solution of copper sulfate and bicinchoninic acidat 37°C for 30 minutes. Light absorbance of these samples wasmeasured at 570 nm (correction wavelength 630 nm).

ALP activity

ALP activity in the osteoblast cell layer was measured usingthe assay by measuring the release of p-nitrophenol from p-nitrophenylphosphate at pH 10.2 as described by Barbara et al.20

When the cells were cultured for 48 hours with mediacontaining various dilutions of the experimental particle solutions,the media was aspirated, the cells were washed three times withPBS, and 500 μl of 0.5% (v/v) Triton X-100 was added to eachwell. The 24-well culture plate was freeze-thawed twice, the celllysateswere sonicated for 1minute on ice and eachwellmixedwitha Titre-tek pipettor. The cell lysatewas centrifuged and a volume of100μl supernatant cell lysate was tested, 50% of the cell lysate wasused for determination of cell total protein content and another 50%cell lysate was used for detection of ALP activity.

In addition, p-Nitrophenol was produced in the presence ofALP and the absorbance was measured with a microplate readerat 405 nm (correction wavelength 630 nm). The change in therate of absorbance was directly proportional to the ALP activity.Data were normalized by the total cell protein measured with acommercial kit (Sigma-Aldrich).

Cytokine levels of macrophages

The level of tumor necrosis factor-α (TNF-α) was determinedby a commercially available kit (R&D Systems, Abingdon,United Kingdom). The assay was performed according to themanufacturers' instructions. Briefly, cell culture supernatantswere collected immediately after the treatment and spun at13,000g for 10 minutes to remove any particulates. The mediumwas added to a 96-well plate precoated with affinity-purified

polyclonal antibodies specific for the mouse TNF-α. An enzyme-linked polyclonal antibody specific for the mouse TNF-α wasadded to the wells and left to react for 2 hours, followed by a finalwash to remove any unbound antibody-enzyme reagent. Theintensity of the color detected at 450 nm (correction wavelength570 nm) was measured after addition of a substrate solution andwas proportional to the amount of TNF-α produced.

Uptake of NPs by MG63 and their subcellular localization

MG63 cells were plated in 100 mm culture dish (1.0 × 107

cells per well) and incubated at 37°C for 24 hours. At the end ofthe incubation period, the cells were treated with experimentalNPs (300 μg/mL) for 6 hours. Cells were observed by TEM.

Statistical analysis

The data were expressed as the mean ± standard deviation(SD). Student's t test was used for comparison, with P values ofless than 0.01 being considered significant.

Results

Cytotoxic assay analysis and decreased viability

The cytotoxic assay (MTT) on Al2O3, ZrO2 and Si3N4 NPs withMG63 osteoblast-like cells at 24, 48 and 72 hours' incubation timepoints were performed, with Ti microparticles as control (Figure 1).A significant dose-dependent increase was shown in the absorbanceof Si3N4 NPs, and the ZrO2 NPs group revealed a dose-dependentdecrease from 15 ppm to 150 ppm at 24 hours. When incubationtime extended to 48 hours and 72 hours, the absorbance at allconcentrations was close to or exceeded that of the control level. Atthe highest dose (500 ppm) of ZrO2 NPs, the absorbancesignificantly exceeded the control. In comparison with Si3N4 andZrO2 NPs, Al2O3 NPs did not show significant effect on cellularviability. In contrast, pure Ti microparticles showed disciplinarydose-dependent decrease in the absorbance, and the suppression ofcellular viability eased over the entire incubation time.

Because the increasing absorbance obtained in the presenceof Si3N4 and ZrO2 NPs could be related to the increase in cellularviability or the possible increase in the cell number, the totalintracellular proteins of MG63 at 72 hours were also measured asan index of cell proliferation (Figure 2). A slight decrease inintracellular protein content was found in both Ti microparticlesand ZrO2 and Al2O3 NP groups (statistically significant at 150and 500 ppm), whereas it remained stable in both the Si3N4 NPgroup and the control group. Interestingly, to L929 cells (FigureS1in the Supplementary Materials), Si3N4 NPs could obviouslydecrease the TCP of L929 cells in a dose-dependent manner at 72hours, but it has no effect on the TCP of MG63 cells.

Both in MTT and TCP assay, Al2O3 NPs had no detachableadverse effect on MG63 cells and L929 cells in variousconcentrations at any time point.

ALP activity

The increase in ALP activity reflects maturation of osteoblastdifferentiation. As shown in Figure 3, ZrO2 and Si3N4 NPscaused notable increases (P b 0.01) in the intrinsic ALP activity

Page 4: A comprehensive biological evaluation of ceramic ...lbmd.coe.pku.edu.cn/PDF/2011nnbm.pdf · A comprehensive biological evaluation of ceramic nanoparticles as wear debris ... (TCP)

Figure 1. MTT assay showing the cytotoxicity of (A) Al2O3 NPs; (B) ZrO2 NPs; (C) Si3N4 NPs and (D) pure Ti microparticles to MG63 cells at differentconcentrations. Experiments were carried out with 24, 48, 72 h incubation time with MG63 cells. *P b 0.01 in comparison with controls (n = 4).

978 Y.F. Zhang et al / Nanomedicine: Nanotechnology, Biology, and Medicine 7 (2011) 975–982

of MG63 cells at high concentrations (especially at 500 ppm). Incontrast, Al2O3 NPs obviously increased ALP activity of MG63cells at low doses (15 ppm) and decreased at high doses.

We also tested the effect of differently sizedAl2O3 particles onthe ALP activity of MG63 cells (Figure S2 in the SupplementaryMaterials). The results showed that Al2O3 (b1 μm) increased theALP activity of MG63 cells in a dose-dependent manner andAl2O3 (b10 μm but larger N1 μm) did not have such property.This indicated that the sizes of the particles might have differentcontributions to osteoblast function and the smallest Al2O3

particles weremore potent to increase ALP activity ofMG63 cellsat low concentrations (15 ppm).

In comparison with Ti microparticles, ceramic NPs signifi-cantly promoted the ALP activity of MG63 cells at either low orhigh doses.

Tumor necrosis factor-α (TNF-α) production in RAW264.7 cells

The production levels of TNF-α released in the supernatant ofthe cultures at 24 hours for the various particles and twoconcentrations are shown in Figure 4. The RAW264.7 cellsincubated with 500 ppm ZrO2, Si3N4 NPs had almost three timesmore production of TNF-α than the control. Even at a lowerconcentration (100 ppm), cells challenged by ZrO2 and Si3N4

NPs significantly elevated TNF-α production.

We also tested the TNF-α expression of RAW264.7 cellsstimulated by different sized Al2O3 particles. All sizes of Al2O3

particles were found to have negative effect on this assay (FigureS3, Supplementary Materials).

Cellular vesicles formation and change observed by TEM

TEM image of MG63 cells treated with 300 μg/mL Al2O3,ZrO2 and Si3N4 NPs for 6 hours showed the intracellularvesicles containing phagocyted NP agglomerates (Figure 5).The cell membrane began phagocyting and NPs wereobserved in Figure 5, A. Most of the particles phagocytedby the cell were agglomerating and included into a vesicle orlysosome; only a few NPs were free in the different sites ofthe cell. It could be clearly observed that the individual and/oragglomerates of NPs were internalizing and particles wrappedin the endosome and/or lysosomal vesicles ultimately weretranslocated to the perinuclear region of the cell (Figure 5, Aand 5, G).

Cells that phagocyted Si3N4 and ZrO2 NPs (especially Si3N4)showed rich mitochondria and ectatic endoplasmic reticulum(ER) in cytoplasm (Figure 5, C-5, D). In the cells withphagocyted Si3N4 NPs, the mitochondria were swollen, partialcrista fragmentation and endoplasmic reticulum dilation werealso found (Figure 5, E_5, F).

Page 5: A comprehensive biological evaluation of ceramic ...lbmd.coe.pku.edu.cn/PDF/2011nnbm.pdf · A comprehensive biological evaluation of ceramic nanoparticles as wear debris ... (TCP)

Figure 2. Total intercellular protein content curve of MG63 cells treated withAl2O3, Si3N4, ZrO2 NPs and pure Ti particles at different concentrationsduring 72 h. *P b 0.01 in comparison with controls.

Figure 3. ALP activity of MG63 cells treated with Al2O3, Si3N4, ZrO2 NPsand pure Ti particles at three different concentrations during 48 h. *P b 0.01in comparison with controls.

Figure 4. TNF-α amount of RAW264.7 cells treated with Al2O3, Si3N4, ZrO2

NPs and pure Ti particles at low and high concentrations during 24 h. *P b0.05, **P b 0.01 in comparison with negative controls.

979Y.F. Zhang et al / Nanomedicine: Nanotechnology, Biology, and Medicine 7 (2011) 975–982

Discussion

This study demonstrated that ceramic NPs were bioactive tocells. With MG63 cells, all ceramic NPs showed no significanttoxicity as tested up to 72 hours. The increases in absorbance(MTT assay) observed in Si3N4 and ZrO2 NPs were not due tothe increased proliferation of cells. These results were inagreement with that reported by Di Virgilio et al21 Furthermore,our TEM observation showed the increasing number ofmitochondria in cells that phagocyted Si3N4 and ZrO2 NPs.This explained the increase in absorbance observed in Si3N4 andZrO2 NPs as the MTT salt was mainly reduced by dehydroge-nases in mitochondria.22 The cell respiratory burst caused byNPs could increase the reduction of MTT salt. From the TEMimages of cells that phagocyted Si3N4 NPs, we found theswelling mitochondria, partial crista fragmentation and endo-plasmic reticulum dilation. This implied dysfunction in cellularenergy production and protein synthesis.

Referring to the report by others that CoCr NPs induced morecell DNA damage than the larger CoCr particles,14 our findingssuggest that ceramic NPs were less toxic than alloy NPs were.Notably, Si3N4 NPs decreased the TCP of L929 cells in a dose-dependent manner but not to MG63 cells (refer to theSupplementary Materials, Figure S1). This phenomenon indi-cated that the biological effect of these NPs was stronglydependent on the cell types, and the discriminating effects mightinhibit the formation of fibrous tissue around the prosthesis.

With increase in concentrations of Si3N4 and ZrO2 NPs, theALP activity of MG63 cells increased. However, Al2O3 NPsalready significantly increased the ALP activity of MG63 cellsat low concentration (15 ppm). We also observed that the ALPactivity of MG63 cells treated with Ti microparticles was lowerthan that treated by ceramic NPs. This observation mightindicate the superiority of ceramics in osteogenesis incomparison with metals.

Ingham et al found that the particles with different sizes(nanometers to several hundred micrometers) appeared in allkinds of implant bearings in vivo.12,23 The smaller ultra-highmolecular weight polyethylene (UHMWPE) particles (0.1–1.0 μm) were more reactive than particles above the 1 μm. Of

the cytokines secreted by macrophages, TNF-α was consideredto be the most direct and important marker for bone resorption.17

In our study, Al2O3 particles of all three sizes (b100 nm, b1 μm,and b10 μm) (see Supplementary Materials Figure S3) did notirritate RAW264.7 cells. However, noticeable irritation ofRAW264.7 cells by Si3N4 and ZrO2 NPs was observed, andthese activities were found stronger than that of Ti microparti-cles. This result indicated that ZrO2 and Si3N4 NPs might bemore irritating to macrophages than Ti microparticles. Thechemical composition of NPs might play an important role in celland tissue response.

We proved that ceramic NPs, especially ZrO2 and Si3N4 NPs,were bioactive to osteoblast and macrophage-like cells. Thisconcerned the application of these two kinds of ceramic materialsin orthopedics. As Al2O3 NPs did not show significant irritatingeffect on osteoblast-like cells and macrophage-like cells, thebiological reaction observed in the ceramic periprostheticmembrane might be induced by particles of other compositions,such as polyethylene and metal alloy.

The internalization of the NPs was an energy-dependentprocess and controlled by multiple mechanisms, including

Page 6: A comprehensive biological evaluation of ceramic ...lbmd.coe.pku.edu.cn/PDF/2011nnbm.pdf · A comprehensive biological evaluation of ceramic nanoparticles as wear debris ... (TCP)

980 Y.F. Zhang et al / Nanomedicine: Nanotechnology, Biology, and Medicine 7 (2011) 975–982

Page 7: A comprehensive biological evaluation of ceramic ...lbmd.coe.pku.edu.cn/PDF/2011nnbm.pdf · A comprehensive biological evaluation of ceramic nanoparticles as wear debris ... (TCP)

981Y.F. Zhang et al / Nanomedicine: Nanotechnology, Biology, and Medicine 7 (2011) 975–982

clathrin-caveolae-mediated endocytosis and macropinocytosis.24

A large number of nanoparticles' internalization into cell neededsufficient energy and proteins and it would facilitate the cells toenhance the function of mitochondria and ribosome. Theswelling mitochondria, partial crista fragmentation and endo-plasmic reticulum dilation observed during this study (see Figure5, C-D) might be explained by a mechanism known as“supercompensation.” Considering that NPs themselves did nothave biomolecule surface structures, we would speculate thatNPs might promote the ALP synthesis in MG63 cells in anindirect way. In a recent work by Bhabra et al, CoCr NPs werefound to be able to cause DNA damage without significant celldeath across a cellular barrier, and the damage was mediated by anovel mechanism involving adenosine triphosphate (ATP)transmission and intercellular signaling through junctions and/or channels between the cells.25 Moreover, one most recent studyfocused on the “corona” of a select group of proteins on thesurface of NP,26 and the authors strongly suggested theimportance of surface proteins on NPs in the particle-cellinteractions. Nevertheless, the effect of NPs on organisms mightinvolve complex mechanisms and their cytobiological effectsneed further investigations.

Wear particles generated from the prosthesis may enterinto the periprosthetic tissue and interact directly withosteoblasts and macrophages. The activated macrophagesand suppressed osteoblasts might reduce bone formation andosteoclastic bone resorption.17 Wear particles were also foundin liver, spleen, and abdominal para-acortic lymph nodes,27

which suggested the possibility of hematogenous dissemina-tion and systemic effects of wear particles. In this study, weevaluated three kinds of ceramic NPs with emphasis on theirinteraction with osteoblast and macrophage-like cells. Theseresults provided new and useful information of ceramicmaterials that might influence osteogenesis at the bone-implant interface. Based on our findings, we suggested thatalumina might be still the most suitable ceramic material inorthopedics. However, the use of silicon nitride and zirconiananoceramics in biomaterial and tissue engineering fieldsshould be prudent. Considering the osteolysis in associationwith alumina bearings, we speculated that alumina wearparticles might not be the main cause resulting in implantfailure. Nanomaterials were increasingly applied in thebiomedical field, especially as targeting therapies for boneor brain applications. Their retention would be more tissue-specific and privileged. This implied that the evaluation ofNPs' biosafety needed a more rational paradigm in the futurethan had been used in the past.

In summary, our in vitro findings implied that ceramic NPswere biologically radical. Our study suggested that their role inosteogenesis at implant-bone bed needed to be redefined, i.e.,

Figure 5. (A) TEM images of MG63 cells at 37°C (incubation with Al2O3 NPs forwith actin rearrangement near the plasma membrane and extension into the extracNPs for 6 h); (C) TEM images of MG63 cells at 37°C (incubation with Si3N4 NPsfor 6 h), bold arrow pointing to NPs, thin arrows pointing to an increasing numbercells at 37°C (incubation with Si3N4 NPs for 6 h), Arrows pointing to swelling mdilation; (F) TEM images of MG63 cells at 37°C (incubation with Si3N4 NPs forimages of MG63 cells at 37°C (incubation with NPs for 6 h), arrows pointing to thnear the plasma membrane and extension into the extracellular space.

because alumina might remain as one of good options fororthopedic applications, the potential concerns regarding in vivoapplications of silicon nitride and zirconia nanomaterials shouldbe addressed experimentally.

Appendix A. Supplementary data

Supplementary data associated with this article can be found,in the online version, at doi:10.1016/j.nano.2011.04.005.

References

1. Goldring SR, Clark CR, Wright TM. The problem in total jointarthroplasty: Aseptic loosening. J Bone Joint Surg Am 1993;75:799-801.

2. Clohisy JC, Calvert G, Tull F, McDonald D, Maloney WJ. Reasons forrevision hip surgery: A retrospective review. Clin Orthop Relat Res2004:188-92.

3. Willert HG, Bertram H, Buchhorn GH. Osteolysis in alloarthroplasty ofthe hip. The role of bone cement fragmentation. Clin Orthop Relat Res1990:108-21.

4. Amstutz HC, Campbell P, Kossovsky N, Clarke IC. Mechanism andclinical significance of wear debris-induced osteolysis. Clin Orthop RelatRes 1992:7-18.

5. Zichner LP, Willert HG. Comparison of alumina-polyethylene andmetal-polyethylene in clinical trials. Clin Orthop Relat Res 1992:86-94.

6. Wirganowicz PZ, Thomas BJ. Massive osteolysis after ceramic onceramic total hip arthroplasty. A case report. Clin Orthop Relat Res 1997:100-4.

7. Yoon TR, Rowe SM, Jung ST, Seon KJ, Maloney WJ. Osteolysis inassociation with a total hip arthroplasty with ceramic bearing surfaces.J Bone Joint Surg Am 1998;80:1459-68.

8. Lerouge S, Huk O, Yahia L, Witvoet J, Sedel L. Ceramic-ceramic andmetal-polyethylene total hip replacements: comparison of pseudomem-branes after loosening. J Bone Joint Surg Br 1997;79:135-9.

9. Mochida Y, Boehler M, Salzer M, Bauer TW. Debris from failedceramic-on-ceramic and ceramic-on-polyethylene hip prostheses. ClinOrthop Relat Res 2001:113-25.

10. Maloney WJ, Jasty M, Harris WH, Galante JO, Callaghan JJ. Endostealerosion in association with stable uncemented femoral components.J Bone Joint Surg Am 1990;72:1025-34.

11. Goodman SB, Ma T, Chiu R, Ramachandran R, Smith RL. Effects oforthopaedic wear particles on osteoprogenitor cells. Biomaterials2006;27:6096-101.

12. Hatton A, et al. Alumina-alumina artificial hip joints. Part I: a histologicalanalysis and characterisation of wear debris by laser capture microdis-section of tissues retrieved at revision. Biomaterials 2002;23:3429-40.

13. ChoiMG, et al. Effects of titanium particle size on osteoblast functions invitro and in vivo. Proc Natl Acad Sci USA 2005;102:4578-83.

14. Papageorgiou I, et al. The effect of nano- and micron-sized particles ofcobalt-chromium alloy on human fibroblasts in vitro. Biomaterials2007;28:2946-58.

6 h), Arrows pointing to the process of internalization at the surface associatedellular space; (B) TEM images of MG63 cells at 37°C (incubation with ZrO2

for 6 h); (D) TEM images of MG63 cells at 37°C (incubation with ZrO2 NPsof mitochondria and endoplasmic reticulum (ER); (E) TEM images of MG63itochondria with partial crista fragmentation and endoplasmic reticulum (ER)6 h), thin arrows pointing to endoplasmic reticulum (ER) dilation; (G) TEMe process of internalization at the surface associated with actin rearrangement

Page 8: A comprehensive biological evaluation of ceramic ...lbmd.coe.pku.edu.cn/PDF/2011nnbm.pdf · A comprehensive biological evaluation of ceramic nanoparticles as wear debris ... (TCP)

982 Y.F. Zhang et al / Nanomedicine: Nanotechnology, Biology, and Medicine 7 (2011) 975–982

15. Germain MA, et al. Comparison of the cytotoxicity of clinically relevantcobalt-chromium and alumina ceramic wear particles in vitro. Bio-materials 2003;24:469-79.

16. Koreny T, et al. The role of fibroblasts and fibroblast-derived factors inperiprosthetic osteolysis. Arthritis Rheum 2006;54:3221-32.

17. Ingham E, Fisher J. The role of macrophages in osteolysis of totaljoint replacement. Biomaterials 2005;26:1271-86.

18. Goodman S, Aspenberg P, Song Y, Regula D, Lidgren L. Polyethyleneand titanium alloy particles reduce bone formation. Dose-dependence inbone harvest chamber experiments in rabbits. Acta Orthop Scand 1996;67:599-605.

19. Bal BS, et al. Fabrication and testing of silicon nitride bearings in totalhip arthroplasty: winner of the 2007 qHAPq PAUL Award. JArthroplasty 2009;24:110-6.

20. Boyan BD, Schwartz Z, Bonewald LF, Swain LD. Localization of 1,25-(OH)2D3-responsive alkaline phosphatase in osteoblast-like cells (ROS17/2.8, MG 63, and MC 3T3) and growth cartilage cells in culture. J BiolChem 1989;264:11879-86.

21. Di Virgilio AL, Reigosa M, de Mele MF. Response of UMR 106 cellsexposed to titanium oxide and aluminum oxide nanoparticles. J BiomedMater Res A 2010;92:80-6.

22. Liu Y, Peterson DA, Kimura H, Schubert D. Mechanism of cellular 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) re-duction. J Neurochem 1997;69:581-93.

23. Richards L, et al. Identification of nanometre-sized ultra-high molecularweight polyethylene wear particles in samples retrieved in vivo. J BoneJoint Surg Br 2008;90B:1106-13.

24. Gratton SE, et al. The effect of particle design on cellular internalizationpathways. Proc Natl Acad Sci USA 2008;105:11613-8.

25. Bhabra G, et al. Nanoparticles can cause DNA damage across a cellularbarrier. Nat Nanotechnol 2009;4:876-83.

26. Walczyk D, Bombelli FB, Monopoli MP, Lynch I, Dawson KA. Whatthe cell qseesq in bionanoscience. J Am Chem Soc 2010;132:5761-8.

27. Urban RM, et al. Dissemination of wear particles to the liver, spleen, andabdominal lymph nodes of patients with hip or knee replacement. J BoneJoint Surg Am 2000;82:457-76.