+ All Categories
Home > Documents > Morphological analysis and cell viability on diamond-like carbon films containing nanocrystalline...

Morphological analysis and cell viability on diamond-like carbon films containing nanocrystalline...

Date post: 26-Nov-2023
Category:
Upload: independent
View: 0 times
Download: 0 times
Share this document with a friend
6
Applied Surface Science 275 (2013) 258–263 Contents lists available at SciVerse ScienceDirect Applied Surface Science jou rn al h omepa g e: www.elsevier.com/locate/apsusc Morphological analysis and cell viability on diamond-like carbon films containing nanocrystalline diamond particles C.N. Almeida a , B.C. Ramos a , N.S. Da-Silva b , C. Pacheco-Soares b , V.J. Trava-Airoldi c , A.O. Lobo a , F.R. Marciano a,a Laboratory of Biomedical Nanotechnology (NanoBio), Universidade do Vale do Paraíba (Univap), Av. Shishima Hifumi, 2911 São José dos Campos, 12244-000, SP, Brazil b Laboratório de Biologia Celular e Tecidual, Universidade do Vale do Paraíba (Univap), Av. Shishima Hifumi, 2911 São José dos Campos, 12244-000, SP, Brazil c Associated Laboratory of Sensors and Materials, Instituto Nacional de Pesquisas Espaciais (INPE), Av. dos Astronautas 1758, São José dos Campos, 12227-010, SP, Brazil a r t i c l e i n f o Article history: Received 1 October 2012 Received in revised form 20 December 2012 Accepted 22 December 2012 Available online 29 December 2012 Keywords: Diamond-like carbon Crystalline diamond nanoparticles Cell viability Fibroblasts a b s t r a c t The coating of orthopedic prostheses with diamond like-carbon (DLC) has been actively studied in the past years, in order to improve mechanical, tribological properties and promote the material’s biocom- patibility. Recently, the incorporation of crystalline diamond nanoparticles into the DLC film has shown effective in combating electrochemical corrosion in acidic medias. This study examines the material’s biocompatibility through testing by LDH release and MTT, on in vitro fibroblasts; using different con- centrations of diamond nanoparticles incorporated into the DLC film. Propounding its potential use in orthopedics in order to increase the corrosion resistance of prostheses and improve their relationship with the biological environment. © 2013 Elsevier B.V. All rights reserved. 1. Introduction Surfaces play a vital role in biology and medicine with most biological reactions occurring at surfaces and interfaces [1]. The successful incorporation of an implant into the body depends on tissue integration and infection resistance, which is influenced by the adherence of autologous cells and bacteria to the sur- faces [1,2]. Cell adhesion and spreading is fundamentally essential for biomaterials that are frequently used in biomedical devices [3]. In most cases, a surface modification of these biomaterials is considered to be a prerequisite for improving biocompatibil- ity, since this kind of material should also be hard, wear resistant, with a low friction coefficient and corrosion resistant for certain applications [4]. Diamond-like carbon (DLC) coatings have been actively studied over the last decade in the field of material engineering. Consist- ing of dense amorphous carbon or hydrocarbon, DLC mechanical properties fall between those of graphite and diamond [5–8]. These coatings can also impart wear resistance, hardness, and corrosion resistance to a medical device surface, and have been considered for using in a variety of cardiovascular, orthope- dic, biosensor, and implantable microelectromechanical system Corresponding author. Tel.: +55 12 3947 1100; fax: +55 12 3947 1149. E-mail address: [email protected] (F.R. Marciano). devices [6,9,10]. Recent studies have reported modified-DLC films improved biocompatibility, lubricity, stability and cell adhesion [11–14]. Nanoparticle-dispersed composite films are expected to have the potential of changing their performances according to the individual properties of nanoparticles [15]. According to Yun et al. (2008) [8], these characteristics are related to structural bonds [16,17], surface roughness [18,19] and whether the film is hydrophobic or hydrophilic [20,21]. In our previous manuscript [22], it was show for the first time the use of DLC films with nanocrystalline diamond (NCD) parti- cles incorporated in their structure. NCD particles increased DLC electrochemical corrosion resistance, reducing its nanopores and consequently preventing aggressive ions from attacking the stain- less steel surface [22,23]. Nanocomposite coatings, composed of crystalline/amorphous nanophase mixture, have recently attracted increasing interest in fundamental research and industrial applica- tions, due to the possibilities of synthesizing a surface protection layer with unique physicochemical properties that are often not attained in bulk materials [24]. However, the biological interaction of NCD–DLC films has never been studied. In the current paper, cell viability and adhesion of L929 mouse fibroblasts was investigated using two different colorimetric assays: (i) 2-(4,5-dimethyl-2- thiazolyl)-3,5-diphenyl-2H-tetrazolium bromide (MTT), and (ii) lactate dehydrogenase (LDH). It is also compared the observa- tion with Scanning Electron Microscopy (SEM) and Fluorescence Microscopy (FM). 0169-4332/$ see front matter © 2013 Elsevier B.V. All rights reserved. http://dx.doi.org/10.1016/j.apsusc.2012.12.122
Transcript

Mc

CAa

b

c

a

ARR2AA

KDCCF

1

bstbff[iiwa

oipTccd

0h

Applied Surface Science 275 (2013) 258– 263

Contents lists available at SciVerse ScienceDirect

Applied Surface Science

jou rn al h omepa g e: www.elsev ier .com/ locate /apsusc

orphological analysis and cell viability on diamond-like carbon filmsontaining nanocrystalline diamond particles

.N. Almeidaa, B.C. Ramosa, N.S. Da-Silvab, C. Pacheco-Soaresb, V.J. Trava-Airoldi c,

.O. Loboa, F.R. Marcianoa,∗

Laboratory of Biomedical Nanotechnology (NanoBio), Universidade do Vale do Paraíba (Univap), Av. Shishima Hifumi, 2911 – São José dos Campos, 12244-000, SP, BrazilLaboratório de Biologia Celular e Tecidual, Universidade do Vale do Paraíba (Univap), Av. Shishima Hifumi, 2911 – São José dos Campos, 12244-000, SP, BrazilAssociated Laboratory of Sensors and Materials, Instituto Nacional de Pesquisas Espaciais (INPE), Av. dos Astronautas 1758, São José dos Campos, 12227-010, SP, Brazil

r t i c l e i n f o

rticle history:eceived 1 October 2012eceived in revised form0 December 2012ccepted 22 December 2012

a b s t r a c t

The coating of orthopedic prostheses with diamond like-carbon (DLC) has been actively studied in thepast years, in order to improve mechanical, tribological properties and promote the material’s biocom-patibility. Recently, the incorporation of crystalline diamond nanoparticles into the DLC film has showneffective in combating electrochemical corrosion in acidic medias. This study examines the material’sbiocompatibility through testing by LDH release and MTT, on in vitro fibroblasts; using different con-

vailable online 29 December 2012

eywords:iamond-like carbonrystalline diamond nanoparticlesell viability

centrations of diamond nanoparticles incorporated into the DLC film. Propounding its potential use inorthopedics in order to increase the corrosion resistance of prostheses and improve their relationshipwith the biological environment.

© 2013 Elsevier B.V. All rights reserved.

ibroblasts

. Introduction

Surfaces play a vital role in biology and medicine with mostiological reactions occurring at surfaces and interfaces [1]. Theuccessful incorporation of an implant into the body depends onissue integration and infection resistance, which is influencedy the adherence of autologous cells and bacteria to the sur-aces [1,2]. Cell adhesion and spreading is fundamentally essentialor biomaterials that are frequently used in biomedical devices3]. In most cases, a surface modification of these biomaterialss considered to be a prerequisite for improving biocompatibil-ty, since this kind of material should also be hard, wear resistant,

ith a low friction coefficient and corrosion resistant for certainpplications [4].

Diamond-like carbon (DLC) coatings have been actively studiedver the last decade in the field of material engineering. Consist-ng of dense amorphous carbon or hydrocarbon, DLC mechanicalroperties fall between those of graphite and diamond [5–8].hese coatings can also impart wear resistance, hardness, and

orrosion resistance to a medical device surface, and have beenonsidered for using in a variety of cardiovascular, orthope-ic, biosensor, and implantable microelectromechanical system

∗ Corresponding author. Tel.: +55 12 3947 1100; fax: +55 12 3947 1149.E-mail address: [email protected] (F.R. Marciano).

169-4332/$ – see front matter © 2013 Elsevier B.V. All rights reserved.ttp://dx.doi.org/10.1016/j.apsusc.2012.12.122

devices [6,9,10]. Recent studies have reported modified-DLC filmsimproved biocompatibility, lubricity, stability and cell adhesion[11–14]. Nanoparticle-dispersed composite films are expected tohave the potential of changing their performances according tothe individual properties of nanoparticles [15]. According to Yunet al. (2008) [8], these characteristics are related to structuralbonds [16,17], surface roughness [18,19] and whether the film ishydrophobic or hydrophilic [20,21].

In our previous manuscript [22], it was show for the first timethe use of DLC films with nanocrystalline diamond (NCD) parti-cles incorporated in their structure. NCD particles increased DLCelectrochemical corrosion resistance, reducing its nanopores andconsequently preventing aggressive ions from attacking the stain-less steel surface [22,23]. Nanocomposite coatings, composed ofcrystalline/amorphous nanophase mixture, have recently attractedincreasing interest in fundamental research and industrial applica-tions, due to the possibilities of synthesizing a surface protectionlayer with unique physicochemical properties that are often notattained in bulk materials [24]. However, the biological interactionof NCD–DLC films has never been studied. In the current paper, cellviability and adhesion of L929 mouse fibroblasts was investigatedusing two different colorimetric assays: (i) 2-(4,5-dimethyl-2-

thiazolyl)-3,5-diphenyl-2H-tetrazolium bromide (MTT), and (ii)lactate dehydrogenase (LDH). It is also compared the observa-tion with Scanning Electron Microscopy (SEM) and FluorescenceMicroscopy (FM).

urface Science 275 (2013) 258– 263 259

2

msa1pva

sg−filpva6

aa

uJv

smwtaw

aclfsi

“viMcww

(po(asctsw2(cwwa

C.N. Almeida et al. / Applied S

. Experimental procedures

The F138 stainless steel (SS) substrates (1 × 1 cm2) wereechanically polished to a mirror-like finish surface, cleaned ultra-

onically in an acetone bath for 15 min and dried in nitrogentmosphere. The clean samples were mounted on a water-cooled,0 cm diameter cathode powered by a pulsed directly currentlasma enhanced chemical vapor deposition power supply, withariable pulse voltage from 0 to −1000 V, at a frequency of 20 kHznd duty-cycle of 50%.

Into the chamber (vacuum base pressure of 1.3 mPa) the sub-trates were additionally cleaned by argon discharge with 1 sccmas flow at 11.3 Pa working pressure and a discharge voltage of700 V for 10 min prior to deposition. In order to enhance the DLClm adhesion to metallic surfaces, a thin amorphous silicon inter-

ayer (thickness around 200 nm) were deposited using silane as therecursor gas (1 sccm gas flow at 11.3 Pa for 12 min and a dischargeoltage of −700 V) [25]. The DLC films were deposited using hex-ne as the feed gas to a thickness of around 2.0 �m (at 18.0 Pa for0 min and a discharge voltage of −700 V).

In order to produce NCD–DLC films, NCD particles of 500 nmverage size dispersed in hexane in different concentration (0.1, 0.3nd 0.5 g/L) replaced the pure hexane during the DLC deposition.

The dispersion of NCD nanoparticles in DLC films were analyzedsing field emission gun scanning electron microscopy (FEG–SEM),

EOL JSM-6330F, with 30.0 kV. The film morphology and roughnessalue were characterized by a Wyko NT1100 optical profiler.

The contact angle (�) of the samples was measured by using theessile drop method with a Kruss EasyDrop contact angle instru-ent (EasyDrop DSA 100). Two different test liquids (distilledater and diiodomethane) were used for surface energy calcula-

ions, according to the Owens method [26]. The liquid was droppedutomatically by a computer-controlled system. All measurementsere carried out at room temperature.

L-929 mouse fibroblasts cells were provided by Cell Line Bankt Rio de Janeiro (CR019). The cells were maintained as sub-onfluent monolayer’s in minimum essential medium with 1.5 mM-glutamine adjusted to contain 2.2 g/L sodium bicarbonate 85%;etal bovine serum 10% (Gibco, BRL), 100 units/mL penicillin-treptomycin (SIGMA), and 25 �g/mL L-ascorbic acid (SIGMA). Thencubation occurred within a CO2 (5%) atmosphere at 37 ◦C.

The cytotoxicity assay was evaluated according to ISO 10993-5Biological evaluation of medical devices – test for cytotoxicity: initro methods” (or EN 30993-5), using direct contact. The cytotox-city assay was evaluated by two different colorimetric assays: (i)

TT [27] and (ii) LDH [28]. Latex fragments were used as positiveontrol [29]. Fragments of filter paper to prove the nontoxic natureere used as negative control. The dimensions of these fragmentsere the same of the substrates with DLC and NCD–DLC films.

All the samples (1 cm2) were sterilized in humid vapor121 ◦C, 1 atm) and placed in individual wells of 24-well culturelates. The cells were seeded in each well at a concentrationf 5 × 105 cells/mL, supplemented with 10% fetal bovine serumGibco, BRL). The incubation was performed under a CO2 (5%)tmosphere, at 37 ◦C during 24 h. After the incubation, the sub-trates with DLC and NCD–DLC films, and the positive and negativeontrol fragments were removed from the respective wells. Onlyhe cells adhered to the well walls were assayed with (i) MTTolution and (ii) LDH. The absorbance of the content of eachell was measured at 570 nm (MTT) and 490 nm (LDH) with a

4-well microplate reader on a spectrophotometer Spectra CountPackard). The blank reference was taken from wells without

ells, also incubated with MTT and LDH solution. The backgroundas taken from wells without cells. The optical density (OD)ere normalized by the cell culture, and expressed in percent-

ge: [ODsample − ODbackground/ODcells − ODbackground] × 100. The

Fig. 1. SEM image of NCD–DLC film produced from NCD particles of 500 nm at0.5 g/L.

experiments were carried out in quintuplicate in order to confirmthe reproducibility.

In order to analyze the surface and adhesion morphology of thecells on DLC films, the cell culture (5 × 105 cells/mL) were seededon the samples with DLC and NCD–DLC films and incubated undera CO2 (5%) atmosphere, at 37 ◦C during 24 h. After this, the mediumwas removed and the samples were fixed with a 3% glutaraldehyde(0.1 M) sodium cacodylate buffer for 1 h and dehydrated in a gradedacetone solution series (50, 70, 90, 100%) for 10 min each. The dry-ing stage used a 1:1 solution of ethanol with hexamethyldisilazane(HMDS) and the samples were dry with pure HMDS at room tem-perature. After deposition of a thin gold layer, the cell spreadingon the samples were examined by Scanning Electron Microscopy(SEM – ZEISS EVO MA10).

The actin filaments and nucleous morphology were assayed. Forthis, the cell culture (5 × 105 cells/mL) was seeded on the sampleswith DLC and NCD–DLC films and incubated under a CO2 (5%) atmo-sphere, at 37 ◦C during 24 h. After this, the medium was removedand were rinsed with PHEM buffer (MgCl2 50 mM, KCl 70 mM,EGTA 10 mM, HEPES 20 mM, PIPES 60 mM pH 6.8) fixed with a4% paraformaldehyde, 0.1% glutaraldehyde and 0.1% Triton X100in 0.1 M fosfate buffer (pH 7.2) for 30 min. Then the cells wererinsed with PHEM buffer and incubated with PHEM buffer added 1%bovine serum albumin for 10 min, treated with ammonium chlo-ride (50 mM) for 30 min. and incubated with rhodamine–phalloidin(1:100-PHEM) for 60 min and rinsed again with PHEM buffer. Thesame material were incubated with 0.03 �M of 4,6-diamidino-2-phenylindole, dihidrocloruro (DAPI) (Molecular Probes, Eugene,USA), for 10 min. Observations were made with a fluorescencemicroscope Leica DMLB and images will be captures via video cam-era digital Leica DFC 300FX.

3. Results and discussion

The surface morphology of NCD–DLC film shown in Fig. 1 con-firmed that NCD particles were really incorporated in DLC films.SEM image also shows some nanoparticles completely immersedand others partially immersed in DLC surface. Despite the nanopar-ticles had a tendency to form aggregates on the surface duringthe deposition process [30], it was possible to reach a satisfatorydensity of NCD particles with the adopted methodology.

The surface roughness, maximum roughness height (Rt), was

measured over an area of 736 �m × 480 �m and can be seen onFig. 2. The as-deposited DLC films presented roughness valuesaround 1.46 �m. The films became rougher with the increase ofNCD concentration in hexane.

260 C.N. Almeida et al. / Applied Surface Science 275 (2013) 258– 263

Fig. 2. Maximum roughness height (Rt) of DLC films according to the NCD concen-tration in hexane. The error bars represent the standard deviation of 5 differentsamples.

Table 1Contact angle of DLC and NCD–DLC films with different NCD concentrations. Eachmean value corresponds to the average value on five different areas.

NCD Concentration (g/L) Contact angle, � (◦)

Water Diiodomethane

0.0 50.8 ± 7.6 35.0 ± 3.30.1 66.7 ± 2.1 37.7 ± 1.1

aoicsspt

sbs

Ooonwwcft

TSca

Fig. 3. L929 mouse fibroblast mitochondrial activity (cell viability) curve measured

0.3 83.8 ± 3.4 43.4 ± 1.60.5 89.8 ± 2.5 38.3 ± 0.3

The contact angles of the samples formed with distilled waternd diiodomethane are shown in Table 1. As the concentrationf NCD particles in DLC films increased, the water contact anglencreased from 50.8 to ∼80◦, and in the case of diiodomethane, theontact angle remained almost constant. Usually, a hydrophobicurface has a contact angle higher than 70◦, while a hydrophilicurface has a contact angle lower than 70◦ [31]. The hydrophobicroperty of NCD–DLC films could be explained by the presence ofhe NCD particles on the surface [32].

Another important property directly related to the cell cultureubstrate is the surface free energy of the substrate, which haseen shown to be related to the function of tissue cultured on theubstrate, including attachment, spreading, and growth [32].

The surface energy components obtained according to thewens method [33] are listed in Table 2. The total surface energy (�)f 53.0 mN/m for the as-deposited DLC films is estimated as the sumf a dispersive component (�d = 39.9 mN/m) and a polar compo-ent (�p = 14.1 mN/m). The interfacial free energy determines theetting characteristics, and hence, the wall shear stress generated

hen the liquid comes into contact with the surface [34]. As the

oncentration of NCD particles in DLC films increased, the total sur-ace energy decreased from 53.0 to 41.5 mN/m, which is attributedo the reduction in the polar component that is controlled by

able 2urface energy components of DLC and TiO2–DLC films with different TiO2 con-entrations. Each mean value correspond to the average value on five differentreas.

TiO2 Concentration(g/L)

Surface free energy (mN/m) �p/(�d + �p)

Dispersive (�d) Polar (�p) Total

0 36.0 4.0 40.0 0.100.1 29.8 20.9 50.7 0.410.5 27.4 22.5 49.9 0.451.0 24.7 29.0 53.7 0.54

by MTT assay. The percentage of cell viability was calculated by normalization ofoptical density to the negative control.

different intermolecular forces, permanent and induced dipolesand hydrogen bonds [34–36]. Furthermore, fibroblast spreadinghas been correlated with surface free energy, with greatest spread-ing on substrates with surface free energy greater than 45 (mN/m)[37], which does not occur in NCD–DLC films in the concentrationof 0.3 and 0.5 g/L.

The MTT assay offers a quantitative, convenient method for eval-uating a cell population’s response to external factors, whether itbe an increase in cell growth, no effect, or a decrease in growth.The MTT assay is a colorimetric assay system which measuresthe reduction of a tetrazolium component (MTT) into an insol-uble formazan product by NAD(P)H-dependent oxidoreductaseenzymes largely in the cytosolic compartment of the cell, specif-ically in mitochondrial compartment. The mitochondrial enzymesuccinate-dehydrogenase within viable cells is able to cleave thetetrazolium salt MTT into a blue colored product (formazan). Theamount of color produced is directly proportional to the number ofviable cells [38]. Among the applications for the method are drugsensitivity, cytotoxicity, response to growth factors, and cell acti-vation. For this, the increasing of concentration of NCD in DLC filmsis in direct relation with cell viability and mitochondrial activity.

Fig. 3 shows the mitochondrial activity found for DLC andNCD–DLC films after 24 h. A high index of mitochondrial activityis shown for DLC and NCD–DLC films. The increasing concentra-tion of NCD nanoparticles in DLC films enhanced the mitochondrialactivity on these samples.

Cell morphology was assessed by SEM and is shown in Fig. 4. L-929 mouse fibroblast cells are often elongated, much flattened withmicrovilli and/or cell prolongations on their surface. The presenceof a pseudoconfluent layer of elongated and adherent cells demon-strated very good adhesion and good spreading of the fibroblastson the DLC and NCD–DLC coatings. Cellular processes linking thesecells were also observed in all of the samples.

The Lactate dehydrogenase (LDH), which is a soluble cytosolicenzyme present in most eukaryotic cells, releases into culturemedium upon cell death due to damage of plasma membrane. Theincrease of the LDH activity in culture supernatant is proportionalto the number of lysed cells. If the cells are lysed prior to assayingthe medium, an increase or decrease in cell numbers results in aconcomitant change in the amount of substrate converted. Thisindicates the degree of inhibition of cell growth (cytotoxicity)caused by the test material. If cell-free aliquots of the medium from

cultures given different treatments are assayed, then the amount ofLDH activity can be used as an indicator of relative cell viability aswell as a function of membrane integrity. This technique has been

C.N. Almeida et al. / Applied Surface Science 275 (2013) 258– 263 261

F (b) DLc

uts

fiiIoi

FDd

ig. 4. SEM images of L929 mouse fibroblast spreading on (a) F318 stainless steel,

ontrol).

tilized as conventional cytotoxicity resulting from interaction of aest material with the cell. In this case, the decrease of LDH activityhow the plasma membrane integrity [39,40].

Fig. 5 shows the results of cell proliferation on DLC and NCD–DLClms obtained by LDH total assay. The increase of the LDH activity

n culture supernatant is proportional to the number of lysed cells.

n total LDH assay all cells of each well were quantified. A indexf LDH activity is shown for DLC and NCD–DLC films. The increas-ng concentration of NCD nanoparticles in DLC films decreases the

ig. 5. Integrity of L-929 cells measured by LDH released in the interaction withLC and NCD–DLC films. The percentage was calculated by normalization of opticalensity to the negative control.

C, NCD–DLC films in (c) 0.1, (d) 0.3 and (e) 0.5 g/L, and (f) latex fragments (positive

LDH activity on these samples and shown the integrity of plasmamembrane. These results (enhance the mitochondrial activity anddecrease of LDH activity) confirm that the increasing concentrationof NCD nanoparticles in DLC films allowed the biocompatibility (nocytotoxicity) in this cell lineage (L-929 cells).

The cytoskeleton distribution analyzed with a fluorescentmarker rhodamine–phalloidin (Fig. 6). DAPI was used to marknucleolus.

In Fig. 6, cells on the samples were examined by therhodamine–phalloidin (red), and DAPI (blue) at 24 h. A high cellattachment was obtained on all the samples (DLC and NCD–DLCfilms). It was not possible to see any significative difference onthe number of cell attachment and the cell morphology betweenDLC and NCD–DLC films. As observed in MTT and LDH assay, therhodamine–phalloidin and DAPI test show a strong compatibilityof the DLC films. The increased of cells observed in 0.5 g/L TiO2-DLC films (with DAPI marker) and no alterations in cytoskeletoncomponent (rhodamine–phalloidin marker) confirm these results.

Fibroblasts play a particularly important role in the woundrepair process as one of the first tissues involved with the repairof damaged or diseased tissue. Substrate properties including thewater contact angle [41], surface free energy [28], and roughness[41,42] can influence cellular processes of attachment, spreadingand growth. According to Chai et al. [4], since DLC is not a specificmaterial but a group of materials with a wide range of atomic bondstructures and properties depending on the preparation method, it

must be noted that many previous works have reported contradic-tory results about the effect of DLC coating on the improvementof biomedical performance of materials. However, one impor-tant aspect should be carefully addressed: DLC coating exhibits a

262 C.N. Almeida et al. / Applied Surface Science 275 (2013) 258– 263

F d) 0.r

wts

4

dfiNrcidatuN

A

mà2

[

[

[

[[

[[

ig. 6. Cells on (a) stainless steel, (b) DLC, (c) 0.1 g/L, NCD–DLC, (hodamine–phalloidinrhodamine–phalloidin and DAPI at 24 h.

ide range of atomic bond structures, depending on the deposi-ion conditions, and consequently bulk materials properties andurfaces.

. Conclusion

In this paper, L929 viability and spreading was studied oniamond-like carbon films containing NCD nanoparticles. Thebroblast mitochondrial activity increases with the presence ofCD particles. In addition, the percentage of cell death (LDH

eleased) decreased. A high cell attachment was obtained on mostoncentrated NCD–DLC films. Microscopy images evidenced thencreasing number of cell projections (philopodia and lamellipo-ia). These factors are indicatives of a higher biocompatibillity anddhesion between L929 cells and the films. It was not possibleo see statistical difference between the different concentrationsed in this study. These results show the potential use of DLC andCD–DLC films in biomedical applications.

cknowledgements

This study was supported by Conselho Nacional de Desenvolvi-ento Científico e Tecnológico (CNPq) and Fundac ão de Amparo

Pesquisa do Estado de São Paulo (FAPESP 2011/17877-7 and011/20345-7).

[

[

3 g/L NCD–DLC films and (e) 0.5 g/L NCD–DLC films marked by the

References

[1] D.G. Castner, B.D. Ratner, Surface Science 500 (2002) 28.[2] L.K. Randeniya, A. Bendavid, P.J. Martin, M.S. Amin, R. Rohanizadeh, F. Tang, J.M.

Cairney, Diamond and Related Materials 19 (2010) 329.[3] A.G. Karakec ili, M. Gümüsderelioglu, Colloids and Surfaces B 61 (2008) 216.[4] F. Chai, N. Mathis, N. Blanchemain, C. Meunier, H.F. Hildebrand, Acta Biomate-

rialia 4 (2008) 1369.[5] J. Robertson, Materials Science and Engineering: R 37 (2002) 129.[6] C. Donnet, J. Fontaine, T. Le Mogne, M. Belin, C. Héau, J.P. Terrat, F. Vaux, G. Pont,

Surface and Coatings Technology 120 (1999) 548.[7] C. Donnet, A. Grill, Surface and Coatings Technology 94 (1997) 456.[8] D.Y. Yun, W.S. Choi, Y.S. Park, B. Hong, Applied Surface Science 254 (2008)

7925.[9] A. Shirakura, M. Nakaya, Y. Koga, H. Kodama, T. Hasebe, T. Suzuki, Thin Solid

Films 84 (2006) 494.10] M.L. Morrison, R.A. Buchanan, P.K. Liaw, C.J. Berry, R.L. Brigmon, L. Riester, H.

Abernathy, C. Jin, R.J. Narayan, Diamond and Related Materials 15 (2006) 138.11] E.T. Uzumaki, C.S. Lambert, A.R. Santos Jr., C.A.C. Zavaglia, Thin Solid Films 515

(2006) 293.12] S. Zhang, H. Du, S.E. Ong, K.N. Aung, H.C. Too, X. Miao, Thin Solid Films 515

(2006) 66.13] R. Hauert, Diamond and Related Materials 12 (2003) 583.14] T. Nakamura, T. Ohna, M. Suzuki, M. Ishihara, A. Tanaka, Y. Koga, Surface Science

580 (2005) 101.15] M. Ban, N. Hasegawa, Diamond and Related Materials 25 (2012) 92.16] J.Y. Chen, L.P. Wang, K.Y. Fu, N. Huang, Y. Leng, Y.X. Leng, P. Yang, J. Wang, G.J.

Wan, H. Sun, X.B. Tian, P.K. Chu, Surface and Coatings Technology 156 (2002)

289.

17] Q. Zhao, Y. Liu, C. Wang, S. Wang, Diamond and Related Materials 16 (2007)1862.

18] W.J. Ma, A.J. Ruys, R.S. Mason, P.J. Martin, A. Bendavid, Z. Liu, M. Ionescu, H.Zreiqat, Biomaterials 28 (2007) 1620.

urface

[

[

[[

[

[

[

[[[

[

[

[

[

[

[

[

[

[

[

[

[

C.N. Almeida et al. / Applied S

19] J.A. McLaughlin, B. Meenan, P. Maguire, N. Jamieson, Diamond and RelatedMaterials 5 (1996) 486.

20] T. Yokota, T. Terai, T. Kobayashi, T. Meguro, M. Iwaki, Surface and CoatingsTechnology 201 (2007) 8048.

21] A. Rich, A.K. Harris, Journal of Cell Science 50 (1981) 1.22] F.R. Marciano, E.C. Almeida, L.F. Bonetti, E.J. Corat, V.J. Trava-Airoldi, Journal of

Colloid and Interface Science 342 (2010) 636.23] F.R. Marciano, E.C. Almeida, D.A. Lima-Oliveira, E.J. Corat, V.J. Trava-Airoldi,

Surface and Coatings Technology 204 (2010) 2600.24] Y.T. Pei, D. Galvan, J.T.M. De Hosson, A. Cavaleiro, Surface and Coatings Tech-

nology 198 (2005) 44.25] F.R. Marciano, L.F. Bonetti, N.S. Da-Silva, E.J. Corat, V.J. Trava-Airoldi, Applied

Surface Science 255 (2009) 8377.26] R.P. Schneider, Journal of Colloid and Interface Science 182 (1996) 204.27] T. Mosmann, Journal of Immunological Methods 65 (1983) 55.28] T. Decker, M.L. Lohmann-Matthes, Journal of Immunological Methods 115

(1988) 61.29] A.O. Lobo, E.F. Antunes, M.B.S. Palma, C. Pacheco-Soares, V.J. Trava-Airoldi, E.J.

Corat, Materials Science and Engineering C 28 (2008) 532.30] A. Sileikate, I. Prosycevas, J. Puiso, A. Juratis, A. Guobiene, Materials Science 12

(2006) 287.31] H.W. Choi, R.H. Dauskardt, S.C. Lee, K.R. Lee, K.H. Oh, Diamond and Related

Materials 17 (2008) 252.

[

[

Science 275 (2013) 258– 263 263

32] F.R. Marciano, L.F. Bonetti, D.A. Lima-Oliveira, C.B. Mello, M. Ueda, E.J. Corat, V.J.Trava-Airoldi, Diamond and Related Materials 19 (2010) 1139.

33] D.K. Owens, R.C. Wendt, Journal of Applied Polymer Science 13 (1969)1741.

34] C.J. Van Oss, R.J. Good, M.K. Chaundhury, Journal of Colloid and Interface Science111 (1986) 378.

35] P.K. Roy, H.W. Choi, J.W. Yi, M.W. Moon, K.R. Lee, D.K. Han, J.H. Shin, A. Kamijo,T. Hasebe, Acta Biomaterialia 5 (2009) 249.

36] A. Bendavid, P.J. Martin, L. Randeniya, M.S. Amin, Diamond and Related Mate-rials 18 (2009) 66.

37] N. Hallab, K. Bundy, K. O’Connor, R.L. Moses, J.J. Jacobs, Tissue Engineering 7(2001) 55.

38] H. Wan, R. Williams, P. Doherty, D.F. Williams, Journal of Materials Science 5(1994) 154.

39] T. Decker, M.L. Lohmann-Matthes, Journal of Immunological Methods 15 (1988)61.

40] C. Legrand, J.M. Bour, C. Jacob, J. Capiaumont, A. Martial, A. Marc, M. Wudtke, G.Kretzmer, C. Demangel, D. Duval, J. Hache, Journal of Biotechnology 25 (1992)

231.

41] Y. Tamada, Y. Ikada, Journal of Biomedical Materials Research 28 (1994)783.

42] P. van der Valk, A.W.J. van Pelt, H.J. Busscher, H.P. de Jong, C.R.H. Wildevuur, J.Arends, Journal of Biomedical Materials Research 17 (1983) 807.


Recommended