Biotechnological applications of supersonic cluster beam-deposited nanostructured thin films:...

15
Review Article Biotechnological applications of supersonic cluster beam-deposited nanostructured thin films: Bottom-up engineering to optimize cell–protein–surface interactions Ajay Vikram Singh 1,2 1 Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180-3590 2 Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180 Received 1 November 2012; revised 3 January 2013; accepted 4 January 2013 Published online 21 March 2013 in Wiley Online Library (wileyonlinelibrary.com). DOI: 10.1002/jbm.a.34601 Abstract: Technological innovations in biomaterial sciences harness nanoparticle (NP) production, manipulation, and dep- osition with supreme precision, enabling the development of industrial processes. This review first discusses the basic components of this approach, introducing cluster sources, experimental apparatus, and growth mechanisms for NP formation. The second part of this review provides an overview of how the nanoscale bottom-up engineering can control protein adsorption, which in turn determines the fate of nanostructured coating for prokaryotic and mammalian (primary and stem) cell interactions. In addition, we briefly address the implications of the cluster beam deposition tech- nique for nanostructuration of biocompatible microdevices and its potential as a facile coating method to promote protein–surface interactions for microarray applications in biotechnology. V C 2013 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 101A: 2994–3008, 2013. Key Words: nanocluster, AFM, microarray, cell–surface inter- actions, high throughput How to cite this article: Singh AV. 2013. Biotechnological applications of supersonic cluster beam-deposited nanostructured thin films: Bottom-up engineering to optimize cell–protein–surface interactions. J Biomed Mater Res Part A 2013:101A:2994–3008. INTRODUCTION Nanoparticle (NP) applications have a long-stranding his- tory, with aerosol productions and robust steel coatings dating back 2300 and 1800 years, respectively. 1,2 In the industrial realm, NPs have been used well before the advent of nanotechnology as both fillers (carbon in rubber tires) 3 and additives (TiO 2 in dye and paints). 4,5 In the last few years, technological innovations and advances have led researchers to understand the complex interplay of nanobiointeractions which stimulated the development of medicinal devices and drug-delivery systems based on NPs. 6 However, further progress depends on fabricating new pro- tocols for superior NPs productions with controlled size/ shape and biocompatible surface functionalities. In the long term, nanoscience and technology (NST) either complement and strengthen existing technologies or substitute obsolete techniques. 7 Like other paradigms of science and technology, the commercial success of NST depends on funding opportuni- ties and minimization of cost involved in scaling up at an industrial level. The prerequisite of core manufacturing areas containing NPs and nanomaterials in device platform need capability of integrating different components in a definite hierarchy defining structure and chemical composi- tions. 7 This fact is closely associated with collaborative commons of economic production technologies such as semiconductor industry, which improve in performance per unit cost. 8 This could be achieved by synthesis of controlled dimensions with desired structure and chemical status of NPs and meticulously position transfer on suitable matrices (bottom-up engineering). 9 The care must be taken to main- tain a structure–function relationship of NPs in defined matrices or areas. Lately, a technological breakthrough is achieved by manipulating and replicating micronanoscale particle in nonwetting templates (PRINT) to fabricate shape- and size-specific microparticles and NPs. 10,11 Manipulation and controlled replications of PRINT technology put forward the ability to sort the objects in terms of a size or geometry, and positions. This in turn restores precise control over shape, size, surface stiffness, and modulus. Including PRINT, most of the controlled fabrication technologies are limited to polymeric and/or biomolecular (protein/peptide)-specific Correspondence to: A. V. Singh; e-mail: [email protected] Contract grant sponsors: Department of Biomedical Engineering (BME), Rensselaer Polytechnic Institute (RPI; AVS) 2994 V C 2013 WILEY PERIODICALS, INC.

Transcript of Biotechnological applications of supersonic cluster beam-deposited nanostructured thin films:...

Review Article

Biotechnological applications of supersonic cluster beam-depositednanostructured thin films: Bottom-up engineering to optimizecell–protein–surface interactions

Ajay Vikram Singh1,2

1Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180-35902Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180

Received 1 November 2012; revised 3 January 2013; accepted 4 January 2013

Published online 21 March 2013 in Wiley Online Library (wileyonlinelibrary.com). DOI: 10.1002/jbm.a.34601

Abstract: Technological innovations in biomaterial sciences

harness nanoparticle (NP) production, manipulation, and dep-

osition with supreme precision, enabling the development of

industrial processes. This review first discusses the basic

components of this approach, introducing cluster sources,

experimental apparatus, and growth mechanisms for NP

formation. The second part of this review provides an

overview of how the nanoscale bottom-up engineering can

control protein adsorption, which in turn determines the fate

of nanostructured coating for prokaryotic and mammalian

(primary and stem) cell interactions. In addition, we briefly

address the implications of the cluster beam deposition tech-

nique for nanostructuration of biocompatible microdevices

and its potential as a facile coating method to promote

protein–surface interactions for microarray applications in

biotechnology. VC 2013 Wiley Periodicals, Inc. J Biomed Mater Res

Part A: 101A: 2994–3008, 2013.

Key Words: nanocluster, AFM, microarray, cell–surface inter-

actions, high throughput

How to cite this article: Singh AV. 2013. Biotechnological applications of supersonic cluster beam-deposited nanostructured thinfilms: Bottom-up engineering to optimize cell–protein–surface interactions. J Biomed Mater Res Part A 2013:101A:2994–3008.

INTRODUCTION

Nanoparticle (NP) applications have a long-stranding his-tory, with aerosol productions and robust steel coatingsdating back 2300 and 1800 years, respectively.1,2 In theindustrial realm, NPs have been used well before the adventof nanotechnology as both fillers (carbon in rubber tires)3

and additives (TiO2 in dye and paints).4,5 In the last fewyears, technological innovations and advances have ledresearchers to understand the complex interplay ofnanobiointeractions which stimulated the development ofmedicinal devices and drug-delivery systems based on NPs.6

However, further progress depends on fabricating new pro-tocols for superior NPs productions with controlled size/shape and biocompatible surface functionalities. In the longterm, nanoscience and technology (NST) either complementand strengthen existing technologies or substitute obsoletetechniques.7

Like other paradigms of science and technology, thecommercial success of NST depends on funding opportuni-ties and minimization of cost involved in scaling up at anindustrial level. The prerequisite of core manufacturing

areas containing NPs and nanomaterials in device platformneed capability of integrating different components in adefinite hierarchy defining structure and chemical composi-tions.7 This fact is closely associated with collaborativecommons of economic production technologies such assemiconductor industry, which improve in performance perunit cost.8 This could be achieved by synthesis of controlleddimensions with desired structure and chemical status ofNPs and meticulously position transfer on suitable matrices(bottom-up engineering).9 The care must be taken to main-tain a structure–function relationship of NPs in definedmatrices or areas. Lately, a technological breakthrough isachieved by manipulating and replicating micronanoscaleparticle in nonwetting templates (PRINT) to fabricate shape-and size-specific microparticles and NPs.10,11 Manipulationand controlled replications of PRINT technology put forwardthe ability to sort the objects in terms of a size or geometry,and positions. This in turn restores precise control overshape, size, surface stiffness, and modulus. Including PRINT,most of the controlled fabrication technologies are limitedto polymeric and/or biomolecular (protein/peptide)-specific

Correspondence to: A. V. Singh; e-mail: [email protected]

Contract grant sponsors: Department of Biomedical Engineering (BME), Rensselaer Polytechnic Institute (RPI; AVS)

2994 VC 2013 WILEY PERIODICALS, INC.

NPs fabrication. However, inorganic metal and metal oxidesas biocompatible surface coating for implants and prostheticdevices in vivo, and lab-on-a-chip platforms for cell microar-ray/reverse transfection assays, yet to witness technologicalsuccess.12–14

Conventional route for nanostructuredmaterial productionNanostructured materials covers a vast area of materials atnanoscale as thin film metal and metal oxide coating forenergy applications and enhance mechanical stability ofsurfaces. Biocompatible transition metal oxides (e.g., gold,silver, titania, and zirconia, iron is excluded as it is toxic tocells15 are among the most popular nanostructured materi-als for creating biocompatible surfaces and functional bioen-gineered materials for promoting tissue, organs and cellularmatrix interaction.16 A fascinating prospect of engineeringbiopolymer via docking nanoscale peptides, proteins, carbo-hydrates, and lipid moieties makes them particularly suita-ble for tissue engineering and scaffold design. Anotheremerging area in nanomedicine which involves in vivoinvasive and noninvasive theranostic (therapy þ diagnosis)application17 for soft tissue is polymer thin films and versa-tile hydrogel.18 The efficient application of nanomaterialsdepends upon its synthesis routes; however, biologicalroutes are preferred over chemical routes due to cellular,immune, genotoxicity, and carcinogenesis consideration.Newly emerging techniques such as electrophoretic deposi-tion and conventional deposition of biomaterials as nano-layers via self-assembled monolayer, Langmuir–Blodgettfilms, and layer-by-layer assemblies provide precisionin coating at air–water interface.19,20 NP production ofdiverse inorganic materials using colloidal phase, wet lab,and gas phase procedure are other popular conventionalroutes.9,21,22 Among diverse protocols for the NPs produc-tion, gas-phase approaches are very popular due to massproduction via facile scaling route.23–26 These approachesprovide a high level of control on particle physicochemicalproperties such as surface charge, energy, roughness, andporosity with wettability composition.27 In situ synthesis inaerosols and postproduction thermal treatment to improveperformance are other factors attracting this route of metaland oxide NP synthesis. More interesting, surface manipula-tions with a resolution in the 100 nm range and below hasbeen demonstrated using contemporary physical vapor dep-osition (PVD) techniques.28,29

In current review, we present and discuss features ofgas-phase nanocluster production and deposition relevantfor the fabrication of nanostructured systems. In particular,we will discuss the fundamental aspects related with super-sonic cluster beam depositions (SCBDs) for gas phase NPsproduction and possibility of patterning, and coupling toplanar technologies. In last part, we present an overview ofnanostructured thin films in biological applications.

SUPERSONIC CLUSTER BEAM DEPOSITION

Among different gas phase approaches to nanofabrication,the deposition of clusters from supersonic beams is gaining

increased attention extending the interest for this field frombasic to applied research.28 Aggregates ranging from a fewto thousands of atoms called cluster are produced and car-ried in a supersonic regime. Supersonic expansions haveseveral advantages for cluster manipulation over effusivebeams, which make this approach very powerful for thedeposition of nanostructured films. Due to the fact that clus-ter beam depositions favor the manipulation and positioningof NPs by the exploitation of NPs inertial properties, thereis great possibility to couple the SCBD with contemporarymicronanofabrication technologies.29,30

Many theoretical and experimental approaches havebeen developed to solve the problem of neutral NPs manip-ulation in the gas phase.31 The merging of solutions andmodels developed for aerosols, and for supersonic expan-sions has stimulated a novel and interdisciplinary route tosolve this problem.31 The solutions proposed and tested inthe last couple of decades have shown that the synthesis ofnanostructured materials with tailored structural and func-tional properties can be obtained by exploiting aerodynami-cally, focusing on seeded supersonic beams. In particular,the use of aerodynamic lenses32 allows an unprecedentedcontrol on NPs spatial and mass distribution, while keepingvery high fluxes and deposition rates.30 A further improve-ment in aerodynamic manipulation techniques for differentgas phase synthesis methods is opening new perspectivesfor the integration of gas phase NPs production into thelarge and well-consolidated arena of PVD technologies.33

Thanks to the use of collimated beams and stencil masks,patterned depositions with very high lateral resolution canbe obtained. For all these reasons, recently SCBD broughtforth great technological and theoretical interest consideringminimizing the cost/time involved in gas phase NPs produc-tion, manipulation and deposition of nanostructured materi-als using this technique.34,35

The SCBD apparatus houses mainly two chambers: anexpansion (source) chamber and a deposition chamber [seeFig. 1(A); for detail see Ref. 36]. The expansion chamber isequipped with a pulsed microplasma cluster source (PMCS),which represents a combination of different elements typi-cal of sputtering sources, and a laser vaporization clustersource37 as demonstrated in Figure 1(B,C). The clusterdeposition operation principle is based on the three mainevents:

1. The extraction of atoms from a solid sample2. The condensation of the atoms in clusters3. The escaping of the cluster from the source due to the

flow of an inert gas acting as the carrier of clusters.

PMCS and working principleThe group at CIMAINA (Milan, Italy) has developed thePMCS36,37 for the production of nanostructured thin films todeposit many samples in parallel minimizing both time andlabor. It consists of a ceramic cubic body [see Fig. 1(C)] inwhich a cylindrical cavity (of 1.8 cm3 volume) is drilled. Asolenoid-pulsed valve faces one side of the cavity, while theother side is closed by a removable nozzle. A channel

REVIEW ARTICLE

JOURNAL OF BIOMEDICAL MATERIALS RESEARCH A | OCT 2013 VOL 101A, ISSUE 10 2995

crosses the cavity perpendicularly to its axis and hosts atarget (generally a rod) of the material to be vaporized.Before the valve opening, the cavity has the same pressureof the vacuum chamber that hosts the source (10�6 Torr),while the pressure of the inert gas (helium or argon typi-cally) at the back of the valve is 50 bars. Once the valveopens the large pressure difference causes an adiabaticexpansion which accelerates the gas atoms to overcome thesonic speed, thus, producing a supersonic cluster beam.38

This highly collimated gas jet impacts the rod (the cathode),and after a few microseconds from the valve opening apulsed voltage is applied to the two electrodes (800 V) fora typical duration of few tens of ls. This voltage ionizes theinert gas atoms, producing plasma. Ions are then acceleratedagainst the cathode (by the negative potential) ablating thetitanium rod by sputtering. Since only the gas atoms in theMach disk of the supersonic pulse are ionized,32,39 theplasma is spatially confined. As a result, the cathode rod,which is uniformly rotated during the operation of thesource, is ablated in a well-defined region. The cluster’s con-densation begins immediately after the ablation of the tita-nium atoms in a region very close to the cathode surface.Due to the higher kinetic energy of the sputtered atoms;they expand very rapidly from the hot ablation region, thusshortening the primary aggregation process. As the gas

atoms ionized by the polarization of the electrodes repre-sent only a small fraction of the injected gas (the gas pulselasts near 300 ls, while the voltage pulse is only 60 lslong), and due to the adiabatic cooling of the gas atoms dur-ing the supersonic expansion, the mixture of gas and pri-mary clusters thermalize at a rapid pace.40 In this cold mix-ture, the secondary aggregation takes place. The blend ofcarrier gas and titanium/zirconium clusters can then gradu-ally exit the source cavity through the nozzle, and due tothe high pressure difference between the inside and the out-side of the source, a supersonic expansion of the beamoccurs [see Fig. 1(D–F)].41

In a supersonic jet, the collision rate between the atomsbecomes extremely slow, representing a transition from afluid flow dynamics to a free molecular flow dynamics. Allthe processes based on binary collisions are thereforefrozen, and thus the cluster structures are quenched. Fur-thermore, supersonic beams are much more collimated thaneffusive beams, and offer the opportunity of further decreas-ing their divergence by the nozzle. An aerodynamicallyfocusing assembly consist of five aerodynamic lenses(drilled thin plates) mounted at the end of cylindricalspacers in a sequence [Fig. 1(B)]. This configuration allowsthe clusters to concentrate on the axis of the beam. More-over; this system aerodynamically selects the size of the

FIGURE 1. Schematic of supersonic cluster beam apparatus (A); 3D view of PMCS components working (B) and its internal configuration. PMCS

working principle exhibiting microplasma formation (D), thermalization of nanoclusters (E), and expansion as a supersonic beam (F). [Color

figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

2996 SINGH BOTTOM-UP ENGINEERING TO OPTIMIZE CELL–PROTEIN–SURFACE INTERACTIONS

clusters depending on the lens geometry and on the carrier,gas used.32,40 Considering the dimension of the clustermass, there could be three outcomes:

1. If the cluster mass is too big, due to their higher inertiain comparison to the gas atoms, cluster will follow trajec-tories that cause their impact with the focus, preventingexiting from the source.

2. If the mass is too small, the clusters will substantially fol-low the gas streamlines and no focusing will happen.

3. For the proper mass range cluster with a diameter closeto a few tens of nanometers, the trajectories will be con-centrated along the beam axis. The mass range is deter-mined by the focus geometry and by the gas pressure inthe cavity.

As shown in Figure 1(D,E), at step D, ablation of a tita-nium rod takes place by an argon plasma jet, ignited by apulsed electric discharge. After the ablation, TiO2 (or anyother metal oxide) ions thermalize with argon and condenseto form clusters. The mixture of clusters and inert gas isthen extracted in the vacuum through a nozzle to form aseeded supersonic beam, which is collected on a physicalsubstrate located in the beam trajectory. The use of super-sonic beams of clusters allows an excellent control of thecluster mass distribution and kinetic energy with the possi-bility to obtain a highly collimated beam (due to aerody-namically focusing assembly) and very high depositionrates.31,42 The mass range is determined by the focalizer ge-ometry and by the gas pressure in the cavity. Exploitingsimple aerodynamic effects, one can control the divergenceof the supersonic beam and the mass distribution of theparticles.31,43 The size of the exiting nanoclusters dependson the lens geometry and on the carrier gas used. Simply,using stencil masks, patterned films with very high lateralresolutions can be obtained, and this makes SCBD compati-ble with the other planar microtechnologies.42 In a super-sonic jet, the collision rate between the atoms becomesextremely slow, representing a transition from fluid flowdynamics to free molecular flow dynamics. All the processesbased on binary collisions are therefore frozen, and thus thecluster structures are quenched. This allows a low-energyrandom stacking of NPs on the substrate: the particles donot suffer significant fragmentation at the impact, and there-fore, the resulting film keeps memory of the precursorclusters, hence this kind of material is also referred to as acluster assembly. Therefore, SCBD consists in a randomstacking of NPs that leads to materials characterized by alower density with respect to the one shown by filmsobtained from highly energetic particles.34

PMCS-SCBD as an innovative toolto produce ns-TiO2 filmsThe control of film nanotopography and coexisting capabil-ities to regulate physicochemical properties can openstimulating perspectives for applications in surface coatingin biomedicine.

Recently, we have demonstrated that bacterial coloniza-tion on biomaterial surface can be successfully controlled

by tuning the surface nanotopography using SCBD tech-nique.44 Moreover, facile operation and ambient depositionconditions in SCBD make it technology of choice not onlyfor thin film coating [Fig. 2(A–H), left panel] but also inte-grate contemporary micronanofabrication approaches, suchas colloidal lithography [Fig. 2(I–Q), right panel].45 Albeitcomplex, objects characterized at different scales anddimensions are hierarchically organized in biological sys-tems.44 The study of nanostructured titanium oxide (ns-TiO2) thin films production using argon as carrier gas beganapproximately 10 years ago. Cluster-assembled ns-TiO2 filmshave a semiconducting behavior that was initially studiedfor application in catalysis and sensors.30,45–47 Recently, webegan to study and to use ns-TiO2 thin films as a substratefor biological applications because it is possible to use themwith optical instruments, like phase contrast microscopy orconfocal microscopy,48,49 since ns-TiO2 argon thin films areoptically transparent with an estimated refraction index of1.7.50 Also ns-TiO2 thin films have structure constituted bynanocrystals embedded in an amorphous material whichshow good adhesion on various substrates (e.g., glass, mica,and polymer). It is an important parameter for the evalua-tion of the successful performance of cluster-assembledcoatings for various technical and technological applica-tions.51,52 Through the use of a device manipulator and ras-tering, it is possible to produce films whose thickness andcharacteristics are uniform over an extended area, by slidingthe substrate in the plane perpendicular to the beam. Weobtain, on micro- to nanometer scale uniform layer ofns-TiO2 in which grain features are the result of growthdynamics, which follow a ballistic aggregation process, typi-cal of low mobility deposition regimes, provided by lowenergy deposition on cold substrates.30,53

Physical and chemical properties of the clusterassembled filmsAtomic force microscopic (AFM) images of a cluster-assembled TiO2 (left panel) films with nanoscale roughnessare shown in Figure 2(A–H). At the nanoscale, both thefilms expose a granular surface with a grain diameter rang-ing from few up to 20 nm mimicking to that of extracellularmatrix (ECM). This is the result of ballistic deposition ofsize-dispersed clusters, which impinge on the surface andstick together. The cluster growth mechanism leads to ahighly porous material, with a large specific area and asmall average surface slope despite the large surfaceroughness.54

The nanostructure is characterized by nanocrystallineregions embedded in an amorphous matrix. The nanograinsare randomly assembled to constitute a porous structuresuch as those typical of the ballistic aggregation regime, thedensity of the films being roughly half of the correspondingbulk phase (2.5–2.7 g/cm3 against 3.9–4.3 g/cm3 for bulkTiO2). As reported in previous works49 the surface of anas-deposited cluster assembled TiOx is characterized by aremarkable abundance of Ti-3d defect states in the bandgap related to surfacing oxygen vacancies and under coordi-nated titanium atoms.55 These states are known to favor the

REVIEW ARTICLE

JOURNAL OF BIOMEDICAL MATERIALS RESEARCH A | OCT 2013 VOL 101A, ISSUE 10 2997

dissociative adsorption of water molecules and to play arole in the chemisorptions of a variety of organic mole-cules.56–58 Despite the mean clusters’ diameter, nanostruc-tured films are actually comparable with the thickness ofthe native oxide film grown on pure titanium,55,58 as the de-posited cluster assembled titania exhibit a surface muchmore under-stoichiometric (TiOx). However, post depositionannealing in a dry air reveals surface stoichiometric strictlyclose to MO2 for inorganic materials such as zirconium andtitanium (M: Ti/Zr).59,60

Biotechnological application of nanostructuredthin filmsBiocompatible substrate for primary, cancerous cell cul-ture, and stem cell expansion. Referring to biocompatibil-ity of inorganic and polymeric materials, over the years,many definitions have been offered, nevertheless, updated

manifesto and biocompatibility definition for the 21st cen-tury smart biomaterials, as stated by Prof. BD Ratner ‘‘Bio-compatibility refers to the ability of a material to locallytrigger and guide non-fibrotic wound healing, reconstructionand tissue integration’’.61 In true clinical in vivo set up, it fol-lows that biocompatibility is not just a property of the ma-terial, but it implies the interaction between the materialand the host relatively to a precise function.61,62 This inter-action is strictly connected to the protein-adsorption pro-cess, as cells or any other biomolecules never interactdirectly with the surface. Indeed, as soon as a biomaterialsurface comes into contact with blood, serum or any otherbiological fluid, it is immediately covered by proteins thatform an adsorbed layer. In other words, the interactionbetween a material and the biological environment is medi-ated by the protein adsorption process.58,63,64 More relevantliterature explaining the role of nanostructured surfaces in

FIGURE 2. Cluster beam deposition as a tool for controlled synthesis of nanostructured thin films deposition (left panel) and integrating it with

contemporary planar technologies (right panel). (A–D) Representative height maps in three-dimensional view of ns-TiO2 films produced by

SCBD with increasing thickness (50, 100, 200, and 300 nm)44; (E–H) representative surface profiles exhibiting variations in root mean square

roughness (Rq), specific area (Aspec), correlation length, skewness, and kurtosis, as well as in pore width and depth distributions, as discussed

in the main text. (I–Q) Schematic presentation of the different steps of the NSL-SCBD-based micro- and nanopatterning.45 (J) Glass cover slip as

the starting substrate. (J) Drop-coated FITC-BSA (green) and spin-coated polystyrene microspheres (light blue); the microspheres form a closely

packed hexagonal lattice. (K) ns-TiO2 coating over microspheres obtained by SCBD (yellow). (L) ns-TiO2 micro/nanopatterns generated by soni-

cation in DI. (M) Plating of bacterial cells limits the bacterial interaction with nanotriangles. Right panel (N): AFM image of micro/nanopatterns

of ns-TiO2 þ BSA obtained with 3 lm microspheres (60 lm � 60 lm). (O) Magnified topographic image of (P) showing hexagonal distribution of

triangular nanopatterns (ns-TiO2; 15 lm � 15 lm; z scale: 50 nm). (Q) Magnified image of the ns-TiO2 triangle inside the encircled area of image

(G) showing granular morphology of ns-TiO2 (500 nm � 500 nm; z scale: 50 nm). (D) Overlay of reflectance and fluorescence images showing

both the reflective ns-TiO2 islands and the precoated FITC-BSA underneath, visible as the green fluorescent signal (scale bar: 10 lm). [Color fig-

ure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

2998 SINGH BOTTOM-UP ENGINEERING TO OPTIMIZE CELL–PROTEIN–SURFACE INTERACTIONS

proteins and biointerfacial interactions which are beyondthe scope of this review can be found elsewhere.65,66

A recent work characterized the biocompatibility of ns-TiOx films produced by the deposition of a supersonic beamof TiOx clusters via analyzing different interactions (van derWaals, electrostatic, and chemical bonding) between AFMtip and surface.53 The findings corroborate past andcurrent cellular studies showing the new material is highlybiocompatible, and it supports normal growth and adhesionof immortalized, tumor and primary cells, and stemcells without coating with ECM proteins (Fig. 3).41,67,68

Therefore, ns-TiOx is proposed as an optimal substrate fordifferent applications in cell-based assays (cell microarray),biosensors, or microfabricated devices for medical applica-tions69–72 (Table I). The size of the clusters composing thens-TiOx film as well the size of many ECM proteins is of theorder of a few nanometers. As an immediate implication, ona sufficiently small scale (i.e., few hundreds of nanometers)the surface morphology of cluster-assembled ns-TiOx canmimic the natural ECM environment. This topography pro-vides a synthetic cue for ECM protein adsorption from cellculture media, thus, generating biochemical cue with correctorientation and clustering of integrin proteins, promotingcell–ECM interaction.

Given that the topographic features of the substrate playan essential role, the morphology of the cluster assembledns-TiOx may favor the interaction of cells with its surface.48

Therefore, it is of extreme interest to quantitatively charac-terize the interaction of this new material with proteins, asthis is the basis for controlling cell–surface interactions.67

Considering our recent study on nanostructured zirconiumoxide, to the best of our knowledge, bottom-up approach ofzirconia film synthesis in form of nanoclusters, using PMCStechnique is truly novel with defined nanoscale physical andchemical properties.9 The nanoscale morphology and sur-face topography of zirconia shares much similarity with thetitania except surface chemistry. Titanium oxides films haveversatile chemistry as described in above sections, whereaszirconium oxide behaves like almost bioinert surfaces.Human osteosarcoma cell line (MG-63) and Pheochromocy-toma (PC12) cell lines seeded on the cubic ns-ZrOx filmsmade by the bottom-up approach demonstrated precisesensing of nanoscale roughness of different zirconia filmswith increasing focal adhesion complex formation (Fig. 4).9

Application of nanostructured film in designing cell andprotein microarray. Living-cell microarrays are powerfultools for functional genomics and drug discovery.70–72 Inthis perspective, new approaches have been proposed tophenotype screening through the creation of living-cellmicroarrays, which exploit the ‘‘reverse transfection’’ of com-plementary DNAs or small interfering RNAs or the ‘‘reverseinfection’’ by lentiviral vectors to allow the simultaneoushigh-throughput analysis of the function of many differentgenes on a glass slide.71,73,74 However, despite severalattempts to improve this technology, it is still a challenge toobtain microarrays of cells with competence to overexpress-ing or downregulating specific genes to address complex

phenotypes.75–77 The bottleneck of technology relies in effi-cient immobilization of molecule of interest on a putativesurface, exhibiting correct functional state to be expressedin living cells.78–80 Other than this, there is a need of experi-mental protocols to fabricate substrate topography andchemistry on an artificial substrates which ‘‘mimic’’ theextracellular environment surrounding cells in vivo, likely topresent more appropriate physiological cues to culturedcells.81,82 In this regard, researchers immobilized viral vec-tors on a ns-TiO2 film obtained by depositing a supersonicbeam of titania clusters on a glass substrate. The SCBD-based technique demonstrated the validation of the retrovi-ral cell array by overexpression of green fluorescent protein(GFP) reporter genes in primary and cancer cells, and byRNA interference of p53 in primary cells by analyzingeffects in cell growth (Fig. 5). It has been demonstrated thatns-TiO2 retroviral arrays are an enabling tool for the studyof gene function of families of genes for complex pheno-types and for the identification of novel drug target.83

After the successful completion of the Human GenomeProject, the researchers involved in Human Proteome Orga-nization are looking forward in exploring a global HumanProteome Project, which is designed to map the entirehuman protein set (proteome).84 However, analysis of pro-tein expression and functions of human proteome data baseavailable has revealed there is tremendous complexity andvariability involved in the individual amino acids in proteinsequences.85,86 This inherent complexity of the human pro-teome has encouraged the development of sophisticatedmultiplexed technologies for more appropriate methods ofanalysis. As a consequence, the trend of detection technolo-gies has moved from low-throughput analytical methodsuch as enzyme-linked immunosorbent assay, Western blotto ‘‘high-content/high-throughput’’ approaches.87 In this con-text, the development of novel high throughput miniaturizeddevices, such as biochips and microarrays, can offer a validapproach to interrogate the high content proteome by multi-plexing the information at a reasonable cost.87,88 Due to thisfact, other than living cell microarray, protein microarraytechnologies are rapidly expanding to fulfill current needsof proteome discovery for disease management.87,89,90

As explained in the previous sections, ns-TiOx surface ischaracterized by morphology at the nanoscale that can betuned to modulate specific biomolecule–material interactions.58

It is important to note that topographical features withdimensions similar to the surface bound biomolecule (�10nm) can significantly affect their morphology and activityfor cell–surface interactions.56,91 Carbone et al.50 presenteda systematic characterization of ns-TiOx coatings as proteinbinding surfaces for antibody microarray using SCBD pro-duced nanostructured film. Comparing the performances ofthe coating with those of the most common commercialsubstrates in protein and antibody microarray assays, thismethod demonstrated equivalent efficiency. Through a ro-bust statistical evaluation of repeatability in terms of coeffi-cient of variation analysis, they demonstrate that ns-TiOxcan be used as a reliable substrate for biochips in analyticalprotein microarray application.48

REVIEW ARTICLE

JOURNAL OF BIOMEDICAL MATERIALS RESEARCH A | OCT 2013 VOL 101A, ISSUE 10 2999

FIGURE 3. Immunofluorescence analysis of cytoskeleton and cellular adhesion markers on gelatin- and cluster-assembled nanostructured (ns)

TiO2-coated coverslips at short and long-term points. (A) MEFs, (B) Tig3-hTert, (C) U2OS, and (D) human primary melanocytes (small arrows

identify ‘‘long’’ and big arrows identify ‘‘short’’ focal adhesions). (E–G) In vitro endothelial differentiation of human circulating CD133Þ cells after

expansion on cluster-assembled ns-TiO2 layers and control condition, performed in coculture with a line of human umbilical vein endothelial

cells (HUVEC), used also as a positive control. 2 h after deposition, CD133Þ cells still can be distinguished from HUVEC cells used as feeder (A);

after 14 days of coculture, CD133Þ cells appear as endothelial structures and adherent mature endothelial cells CD31Þ (red) (B). Endothelial

marker expression has been verified through reverse transcriptase polymerase chain reaction analysis (C), both for CD133Þ cells previously

expanded on cluster-assembled ns-TiO2 layers (1, as-deposited; 2, annealed at 400�C) and in control conditions. (Reproduced with permission

from Refs. 51, 68.) [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

3000 SINGH BOTTOM-UP ENGINEERING TO OPTIMIZE CELL–PROTEIN–SURFACE INTERACTIONS

TA

BLE

I.A

pp

licati

on

of

Nan

ost

ruct

ure

dFilm

sin

Bio

med

icin

eP

rod

uced

by

Clu

ste

rB

eam

Dep

osit

ion

Bio

tech

no

log

yA

pp

lica

tio

ns

SC

BDþ

PM

CS

Inte

gra

ted

wit

hC

on

tem

po

rary

Tech

niq

ue

Co

mp

ati

bilit

yw

ith

Pla

nar

Mic

roN

an

ofa

bri

cati

on

Tech

niq

ue

Un

derl

yin

gM

ech

an

ism

Rem

ark

Refe

ren

ce

Nan

ost

ruct

ura

tio

no

fm

icro

devic

eC

ollo

idal

lith

og

rap

hy

(NS

L)

an

dS

CB

DY

es

Mic

rosp

here

mo

no

layer

use

das

mask

for

nan

op

att

ern

ing

of

TiO

xS

trate

gy

cou

ldb

eu

sefu

lto

pro

mo

tem

am

malian

cells

an

din

hib

its

bact

eri

al

colo

niz

ati

on

inin

viv

op

rost

heti

cd

evic

es

45

SC

BDþ

e-b

eam

lith

og

rap

hy

an

dlift

off

Yes

Faci

lep

roto

col

of

mic

ron

an

ofa

bri

cati

on

via

lift

off

pro

ced

ure

ino

rgan

icso

lven

tS

pati

al

con

tro

lo

fce

llad

hesi

on

(PC

12)

over

mic

rod

evic

e63

SC

BDþ

collo

idal

lith

og

rap

hy

Yes

Mic

ron

an

op

att

ern

ing

of

top

og

rap

hic

(TiO

x)

an

dch

em

ical

cues

(BS

A)

Pro

toco

lp

erm

its

toco

ntr

ol

com

ple

xce

llfu

nct

ion

such

as

dif

fere

nti

ati

on

sca

nb

ein

du

ced

64

Invit

rom

od

el

for

pro

kary

oti

cce

ll-

nan

ost

ruct

ure

dti

tan

iaco

mm

un

icati

on

SC

BDþ

PM

CS

NA

Bact

eri

al

ad

hesi

on

isco

ntr

olled

by

ns-

TiO

xfi

lmro

ug

hn

ess

Stu

dy

dem

on

stra

tes

qu

an

tifi

cati

on

of

bact

eri

al–

cell

surf

ace

inte

ract

ion

s44

Bio

com

pati

ble

coati

ng

for

stem

cell

bio

log

yS

CB

PM

CS

NA

Hu

man

circ

ula

tin

gm

yo

gen

icp

rog

en

ito

rssh

ow

invit

roca

paci

tyto

dif

fere

nti

ate

into

myo

gen

icce

lls

Po

ten

tial

for

stem

cell

exp

an

sio

nan

dd

iffe

ren

tiati

on

inn

clin

ical

set

up

68

Bio

com

pati

ble

coati

ng

for

pri

mary

an

dca

nce

rce

llcu

ltu

re

SC

BDþ

PM

CS

NA

Exce

llen

tp

latf

orm

for

cell

cult

ure

surf

ace

coati

ng

w/o

mo

difi

cati

on

wit

hE

CM

pro

tein

s

clu

ster-

ass

em

ble

dT

iOx

pro

mo

tes

ad

sorp

tio

nan

dst

ab

ilit

yo

fE

CM

pro

tein

41

Pro

tein

–su

rface

inte

ract

ion

mic

roarr

ay

(PS

IM)

SC

BDþ

PM

CSþ

inkj

et

pri

nti

ng

Yes

Nan

om

ete

r-sc

ale

mo

rph

olo

gy

on

reg

ula

tes

pro

tein

ad

sorp

tio

no

nm

ate

rial

oxid

esu

rface

s

qu

an

tita

tive

hig

h-t

hro

ug

hp

ut

chara

cteri

zati

on

of

pro

tein

–su

rface

inte

ract

ion

67

Pro

tein

an

dan

tib

od

ym

icro

arr

ay

Nan

ocl

ust

er

of

TiO

2Y

es

An

tib

od

yad

sorp

tio

nw

ith

corr

ect

fold

ing

an

dep

ito

pe

exp

ress

ion

Co

mm

erc

ial

bio

chip

sin

an

aly

tica

lp

rote

inm

icro

arr

ay

50

Flu

ore

scen

cein

situ

hyb

rid

izati

on

(FIS

H)

mic

roch

ip

Mic

rofl

uid

icin

teg

rate

dw

ith

nan

ocl

ust

ers

of

TiO

2

Yes

Nan

om

ate

rial

pro

mo

tes

hem

ato

po

ieti

cce

llim

mo

biliz

ati

on

inco

nd

itio

ns

of

shear

stre

ss

Mic

rofl

uid

ic-b

ase

dsc

reen

ing

chro

mo

som

al

ab

err

ati

on

sin

can

cer

114

Retr

ovir

al

mic

roarr

ay-

base

dp

latf

orm

Mic

rosp

ott

ing

inte

gra

ted

wit

hS

CB

DY

es

Retr

ovir

al

arr

ay

over

exp

ress

ion

ind

iffe

ren

tce

lllin

es

over

TiO

2-c

oate

dst

rep

tavid

in

Cell-b

ase

dm

icro

arr

ay

for

ph

en

oty

pe

scre

en

ing

84

Bo

tto

m-u

pen

gin

eeri

ng

of

zirc

on

ium

oxid

eth

infi

lms

Clu

ster

beam

-dep

osi

ted

zirc

on

iaY

es

Ns-

ZrO

x-c

oate

dfi

lms

dem

on

stra

teexce

llen

tb

ioco

mp

ati

bilit

yag

ain

std

iffe

ren

tce

lllin

es

Po

ten

tial

for

ad

van

ceap

plica

tio

ns

ind

en

tist

ry9

Cell

ad

hesi

on

an

dp

rolife

rati

on

Nan

ost

ruct

ure

dca

rbo

nfi

lms

Yes

Su

rface

mo

rph

olo

gy

pla

ys

vit

al

role

inca

rbo

n–c

ell

inte

ract

ion

Nan

ost

ruct

ure

dca

rbo

nco

uld

be

use

das

next

gen

era

tio

nce

llcu

ltu

resu

bst

rate

115

An

tib

od

yp

uri

fica

tio

n-

ind

ep

en

den

tm

icro

arr

ays

(PIM

)

Tit

an

iafi

lms

Yes

Dir

ect

bact

eri

al

cell

spo

ttin

go

nT

iO2

coate

dsl

ides

Vari

ab

led

om

ain

so

fsi

ng

leh

eavy-c

hain

an

tib

od

ies

ag

ain

stfi

bro

bla

stg

row

thfa

cto

r28

rece

pto

r1

(FG

FR

1)

were

use

dto

cap

ture

the

an

tig

en

106

Inviv

osi

mu

lati

on

of

bact

eri

a–

nan

ost

ruct

ure

dsu

rface

inte

ract

ion

PM

CS

-dep

osi

ted

thin

film

Yes

Bact

eri

ase

nse

nan

osc

ale

rou

gh

ness

wit

hu

ltra

pre

cisi

on

Invit

rost

ud

yd

em

on

stra

tes

nan

osc

ale

rou

gh

ness

cou

ldb

eu

sed

toco

ntr

ol

bact

eri

al

cell

colo

niz

ati

on

on

imp

lan

tsu

rface

116

REVIEW ARTICLE

JOURNAL OF BIOMEDICAL MATERIALS RESEARCH A | OCT 2013 VOL 101A, ISSUE 10 3001

Immunocell arrays technology based on antibody–nanostructured surface interaction. The knowledge of sig-naling pathways, which are triggered by physiological andpathological conditions or drug treatment, is essential for thecomprehension of the biological events that regulate cellularresponses.92,93 Recently novel platforms based on ‘‘Reverse-Phase Protein Arrays or Reverse Phase Protein Microarray(RPMA)’’ have proven to be useful in the study of differentpathways.94 However, it still lack the possibility to detectevents in the complexity of a cellular context.95,96 RPMA istechnically efficient in analyzing any fluid from cellularlysates or body fluids (serum, cerebrospinal fluid, urine, vit-reous, saliva, etc.) printed with micro- or nanospotter anddetected by secondary labeled antibody via chemilumines-cent, fluorescent or colorimetric assays. Unlike conventionalprotein array which are preferably designed with microscaleddot-blot platforms, RPMA utilizes either of the micro- ornanoscaled dot-blot platform which allows measurement ofprotein expression.97 The advantage of RPMA technologyover its contemporary counterparts is that it provides high

dimensional proteomic data in a high throughput, sensitiveand quantitative manner. An efficient modified version ofRPMA is immune cell array for multiplexed analysis of signal-ing pathways in drug response screening. An ‘‘immunocell-array’’ of cells on chip was developed using ns-TiOx as novelcoating substrate, where upon cell plating, culture, drugtreatment, and fixation, by spotting specific antibodies (Fig.6).98 On the chip, one can detect the localization and state ofhundreds of proteins involved in specific signaling pathways.By applying this technology to mammalian cells, the group atTethis s.r.l. analyzed signaling proteins involved in theresponse to DNA damage and identified a chromatin remod-eling pathway following bleomycin treatment.99 The technol-ogy developed manifests a new tool for the array-based mul-tiplexed analysis of signaling pathways in drug responsescreening.100 It also applies to the proteomics of profilingpatient cells, and ultimately for the high-throughput screen-ing of antibodies for immunofluorescence application.101,102

The conational microarray platforms utilizes surfacecoating to immobilize protein/peptide/antibody in chip

FIGURE 4. Zirconium oxide as novel coating for cell culture substrate. (A–F) Fluorescent immune labeling of adhesion complexes and stress

fiber organization of the Pheochromocytoma (PC12) cell line on nanostructured ZrOx films. PC12 cultured on ns-ZrOx (50–100–200 nm) sub-

strates formed well-organized stress fiber and numerous premature (large arrows) and mature adhesions (small arrows). (G–L) MG-63 cells.

Adhesions plaques were numerous and located predominantly at the cell periphery. (A, C, and E) cell clusters and an individual [squared cell

from cluster, (B, D, and F)] magnified (4�) view of PC12 cell. Similarly, (G, I, and K) MG-63 cell cluster and magnified (H, J, and L) view.9 [Color

figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

3002 SINGH BOTTOM-UP ENGINEERING TO OPTIMIZE CELL–PROTEIN–SURFACE INTERACTIONS

format with correct accessibility of the antibody active moi-eties or functional groups involved in complementary reac-tion. On the other hand, immobilizing whole cells on bio-compatible metal/metal oxide coated substrate expressingantibodies to capture antigen could be more productive inpreparing sensitive and specific microarrays for antigendetection. In a similar approach, nanostructured thin filmswere used for spotting bacteria that expose recombinantantibodies on their external surface directly on nanostruc-tured-TiO2 or epoxy slides [purification-independent micro-array (PIM)]. This is a simple and reliable alternative forpreparing sensitive and specific microarrays for antigendetection. Variable domains of single heavy-chain antibodiesagainst fibroblast growth factor receptor 1 (FGFR1) wereused to capture the antigen diluted in serum or bovine se-rum albumin (BSA) solution.103–106 The FGFR1 detectionwas performed by either direct antigen labeling or using asandwich system in which FGFR1 was first bound to itsantibody and successively identified using a labeled FGF. Inboth cases, the signal distribution within each spot was uni-

form and spot morphology regular. The signal-to-noise(S/N) ratio was extremely elevated, and the specificity ofthe system was proven statistically.104 The level of detectionof the system for the antigen was calculated being 0.4ng/mL and the dynamic range between 0.4 ng/mL and 10lg/mL. The microarrays prepared with bacteria exposingantibodies remain fully functional for at least 31 days afterspotting on the ns-TiO2 films. The current method is suita-ble for other antigens–antibody pairs and expects that itcould be easily adapted to further applications such as thedisplay of scFv and IgG antibodies or the autoantibodydetection using protein PIMs.106

Protein–surface interaction microarrays. Protein surfaceinteractions is the first of complex cascade of events regu-lates many phenomena at the nanobiointerface such asin vivo inflammatory responses, cell adhesion and differen-tiation on synthetic surfaces in vitro. Protein surface interac-tion microarray (PSIM) is a powerful method for studyingin details the interaction between proteins and

FIGURE 5. Retroviral microarrays for the overexpression of Enhanced Green Fluorescent Protein-EGFP-fused proteins; (A) 10� immunofluores-

cence panel of a 10 � 24 arrays of MCF10A cells infected by PINCO and PINCO GFP-NPM vectors in an alternate fashion. Colors as in panel (A)

blue, cell nuclei (DAPI staining); green, GFP fluorescence. (B) 20� magnification of fields from spots in a selected area of the array; GFP fluores-

cence only is shown. (C) 10� immunofluorescence panel of a 12 � 24 arrays of U2OS cells infected by the same vectors in an alternate fashion.

(D) 20� magnification of fields from spots in a selected area of the U2OS array; GFP fluorescence only is shown. (E) Scatter plots of GFP inten-

sity versus DAPI intensity in populations infected by PINCO (blue) and PINCO GFP-NPM (red) in MCF10A cells. The percentage of infected cells

is reported. (F) Same as the panel (E) for U2OS cells (Printed with permission Ref. 84). [Color figure can be viewed in the online issue, which is

available at wileyonlinelibrary.com.]

REVIEW ARTICLE

JOURNAL OF BIOMEDICAL MATERIALS RESEARCH A | OCT 2013 VOL 101A, ISSUE 10 3003

nanostructured surfaces considering relevance of the media-tion adsorbed protein in biomaterial–cell interac-tions.12,67,106,107 PSIM is based on protein array technology,and it allows to study in one single experiment hundreds ofdifferent protein surface interactions. PSIM protocol consistsin spotting small-volume droplets (30 nL) of fluorescent-labeled proteins on the surface under investigation[Fig. 7(A)]. After incubation, blocking, washing, and drying,the amount of adsorbed proteins is evaluated by readingthe fluorescent signal with a commercial microarray scan-ner108 [Fig. 7(B)]. Using PSIM it is possible to compare, onthe same biomaterial sample, the amount of adsorbed pro-teins for a panel of proteins under various conditions suchas protein concentration and pH.109 Since the experimentcan be performed in parallel on several biomaterial samples,PSIM allows characterizing the role of surface synthesis pa-rameters in protein immobilization. PSIM is applicable tomany nanostructured surfaces, and it is a very flexiblemethod, in each of the 300 drops that can be spotted on aglass slide (25 mm � 75 mm), it is possible to change pro-

tein concentration, pH, buffer, salt concentration or proteintype. New spotting technologies give the possibility to study1200 different protein surface interactions in a singleexperiment. The high number of spots can also be used tomake replicates, and to produce very good statistics foreach interaction.110,111

The recent application of a high-throughput characteriza-tion approach to study the effect of surface nanoscale mor-phology on protein adsorption shed more light on this phe-nomenon. PSIM has been applied to ns-TiOx for carrying outsystematic characterization of the adsorption of proteins onnanostructured surfaces.64,67 With PSIM, it was possible tostudy the adsorption of a panel of three proteins (BSA, fibri-nogen, and streptavidin) on a ns-TiOx library composed byfive families of samples each with different surface morphol-ogy110 [Fig. 7(C–E)]. Thanks to PSIM high-throughput power,it was possible to design an experiment in which 1200 pro-tein–surface interaction were studied in order to reproduceprotein adsorption isotherms for the different proteins [Fig.7(C–E)]. Results confirm that nanoscale morphology signifi-cantly enhances the adsorption of proteins. Varying surfaceroughness from 15 nm to 30 nm, amount of adsorbed pro-teins increases up to 600%, well beyond the correspondingincrease of specific area.111 The application of PSIM, in com-bination with other quantitative methods, demonstrated thatthe increase is related to the formation of protein clusters incorrespondence of surface nanometric pores. The nanometricpores inducing protein clusterization112 are characterized byan aspect ratio higher than a threshold value (depends onthe protein characteristic).67,113

CONCLUDING REMARKS

The SCBD technique provides a cutting-edge solution tocontrol surface texture without changing chemistry (biomo-lecular information) at the biomaterial–tissue interface in aspatially controlled manner, which is greatly compromisedin both contemporary gas and chemical vapor depositiontechnologies. In the present review, we demonstrated thatthe cluster beam depositions technique is a vital tool fornanostructured film production with controlled surface to-pography, without changing surface chemical compositions.The most important property of the clusters produced atnanoscale via SCBD is the ECM mimicking topography,which opens a plethora of applications in biotechnology: anefficient surface coating to the cell culture substrate formammalian as well as prokaryotic (bacterial) cell adhesion.Many cell culture-based biological assay need surface coat-ing by ECM proteins or poly-L-lysine for cell adhesion whichoften alters surface chemistry. This in turn might affect thegenomic and proteomic profile of the cells. In addition, bio-compatibility of ns-TiOx/ZrOx films makes it future technol-ogy for the coating implants surface to promote interactionsof the prosthesis with surrounding tissue/organ in vivo. Theprotein surface interactions make it a valuable tool fordesigning HTP protein microarrays for functional genomicsand drug screening; immunocell array and ECM-based cellarrays. Moreover, SCBD compatibility with the planar tech-nologies makes it the technique of choice for the

FIGURE 6. (A) Mouse IgG spot morphology and background on the

ns-TiOx slide. The excellent morphology is demonstrated by surface

plot (B) and plot profile (C) of a selected 3 � 3 arrays of mouse IgG

spots detected by anti-mouse Cy3 antibody. (D) ns-TiOx slide back-

ground in comparison with epoxy and NC slides. (E) Schematic depic-

tion of the immunocell-array technology. Cells are cultured directly on

slides and processed at the appropriate time for immunostaining, and

images are acquired with an automated fluorescence microscope and

analyzed by imaging software (adopted with permission from Refs.

50,101. [Color figure can be viewed in the online issue, which is avail-

able at wileyonlinelibrary.com.]

3004 SINGH BOTTOM-UP ENGINEERING TO OPTIMIZE CELL–PROTEIN–SURFACE INTERACTIONS

future Bio-MEMS and Bio-NEMS device productions(Table I).114–116 Further exploration should be focused onintegrating SCBD with silicon-based microtechnologies toreach the next level of technological innovations.

ACKNOWLEDGMENTS

We thank for the support from Ahmad Abu-Hakmeh in revisingthe manuscript for syntax, grammatical errors, and specific re-vision to biological part.

REFERENCES

1. Santamaria A. Historical overview of nanotechnology and nano-

toxicology. Methods Mol Biol 2012;926:1–12.

2. Nano-express from New Delhi? Nat Mater 2005;4(8):575.

3. Zhou J, Wu S, Yu T, Xie Z. 2011 International Conference on

Modified Pyrolytic Carbon Black from Scrap Tires and Its Rein-

forcement Performance in Natural Rubber. Computer Distributed

Control and Intelligent Environmental Monitoring (CDCIEM);

2011. p 472–475.

4. Braun JH, Baidins A, Marganski RE. TiO2 pigment technology: A

review. Prog Org Coat 1992;20(2):105–138.

5. Ulrike D. The surface science of titanium dioxide. Surf Sci Rep

2003;48(5–8):53–229.

6. Eran B. In: Eitan S,Shimon U, editors. Combining Top-Down

and Bottom-Up Segmentation; 2004. p 46.

7. Eaton MAW. How do we develop nanopharmaceuticals under

open innovation? Nanomed Nanotechnol Biol Med 2011;7(4):

371–375

8. Neher E. Molecular biology meets microelectronics. Nat Biotech-

nol 2001;19(2):114

9. Singh AV, Ferri M, Tamplenizza M, Borghi F, Divitini G, Ducati C,

Lenardi C, Piazzoni C, Merlini M, Podest�a A, Milani P. Bottom-up

engineering of the surface roughness of nanostructured cubic

zirconia to control cell adhesion. Nanotechnology 2012;23:

475101.

10. Perry JL, Herlihy KP, Napier ME, DeSimone JM. PRINT: A novel

platform toward shape and size specific nanoparticle theranos-

tics. Acc Chem Res 2011;44(10):990–998

11. Mihail CR. Nanotechnology: Convergence with modern biology

and medicine. Curr Opin Biotechnol 2003;14(3):337–346

12. Wilson CJ, Clegg RE, Leavesley DI, Pearcy MJ. Mediation of bio-

material–cell interactions by adsorbed proteins: A review. Tissue

Eng 2005;11(1–2):1–18.

13. Yarmush ML, King KR. Living-cell microarrays. Annu Rev

Biomed Eng 2009;11(1):235–257

14. Galli C, Collaud Coen M, Hauert R, Katanaev VL, Wymann MP,

Gr€oning P, Schlapbach L. Protein adsorption on topographically

nanostructured titanium. Surf Sci 2001;474(1–3):L180–L184.

15. Singh AV, Vyas V, Montani E, Cartelli D, Parazzoli D, Oldani A,

Zeri G, Orioli E, Gemmati D, Zamboni P. Investigation of in vitro

cytotoxicity of the redox state of ionic iron in neuroblastoma

cells. J Neurosci Rural Pract 2012;3(3):301–310

16. Li J. Nanostructured Biomaterials.New York, NY:Springer; 2010.

p 222.

17. Singh AV, Khare M, Gade WN, Zamboni P. Theranostic implica-

tions of nanotechnology in multiple sclerosis: A future perspec-

tive. Autoimmune Dis 2012;2012:1–12.

18. Vendra VK, Lin Wu, Krishnan S. Polymer Thin Films for Biomedical

Applications.Weinheim, Germany:Wiley-VCH Verlag; 2010. p 54.

19. Boccaccini AR, Keim S, Ma R, Li Y, Zhitomirsky I. Electrophoretic

deposition of biomaterials. J R Soc Interface 2010;7(Suppl 5):

S581–S613.

20. Ariga K, Nakanishi T, Michinobu T. Immobilization of biomateri-

als to nano-assembled films (self-assembled monolayers, Lang-

muir–Blodgett films, and layer-by-layer assemblies) and their

related functions. J Nanosci Nanotechnol 2006;6(8):2278–2301

21. Singh AV, Patil R, Kasture MB, Gade WN, Prasad BL. Synthesis

of Ag–Pt alloy nanoparticles in aqueous bovine serum albumin

foam and their cytocompatibility against human gingival fibro-

blasts. Colloids Surf B: Biointerfaces 2009;69(2):239–245

22. Singh AV, Bandgar BM, Kasture M, Prasad BLV, Sastry M. Syn-

thesis of gold, silver and their alloy nanoparticles using bovine

serum albumin as foaming and stabilizing. J Mater Chem 2005;

15(48):5115–5121

FIGURE 7. Sketch of the six steps PSIM protocol. The proposed method verified the feasibility of this approach with specific assays and demon-

strated that the fluorescent signal is proportional to the amount of adsorbed proteins and that the fluorescent marker used in the experiment

does not influence the protein–surface interaction process. Finally, using fluorescence recovery after photobleaching (FRAP), it is also verified

that the adsorbed proteins remain immobilized on the surface after adsorption. (B) Raw data obtained by reading the slides of the PSIM experi-

ment in which eight concentrations of BSA, fibrinogen and streptavidin were spotted in 10 replicates on the five nanostructured titania samples,

with different surface morphology. (C) adsorbed fibrinogen, (D) BSA, and (e) streptavidin as a function of protein concentration (adsorption iso-

therms) for five ns-TiOx samples with different surface roughness. PSIM allowed obtaining adsorption isotherms on nanostructured surfaces for

the first time. Data are fitted with Langmuir isotherm in order to calculate saturation uptake and binding affinity. Error bars correspond to stand-

ard deviation of the 10 replicates (Ref. 67). [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

REVIEW ARTICLE

JOURNAL OF BIOMEDICAL MATERIALS RESEARCH A | OCT 2013 VOL 101A, ISSUE 10 3005

23. Stark WJ, Pratsinis SE. Aerosol flame reactors for manufacture

of nanoparticles. Powder Technol 2002;126(2):103–108

24. Wegner K, Pratsinis SE. Scale-up of nanoparticle synthesis in dif-

fusion flame reactors. Chem Eng Sci 2003;58(20):4581–4589

25. Kammler HK, M€adler L, Pratsinis SE. Flame synthesis of nano-

particles. Chem Eng Technol 2001;24(6):583–596

26. Singh Y, Javier RN, Ehrman SH, Magnusson MH, Deppert K.

Approaches to increasing yield in evaporation/condensation

nanoparticle generation. J Aerosol Sci 2002;33(9):1309–1325

27. Wegner K, Pratsinis SE. Nozzle-quenching process for controlled

flame synthesis of titania nanoparticles. AIChE J 2003;49(7)

1667–1675

28. Milani P, Barborini E, Piseri P, Bottani CE, Ferrari AC, Li Bassi A.

Nanostructured carbon films from supersonic cluster beam dep-

osition: Structure and morphology. Eur Phy J D 1999;9(1):63–68

29. Milani P, Piseri P, Barborini E, Podesta A, Lenardi C. Cluster

beam synthesis of nanostructured thin films. J Vac Sci Technol

2001;19(4):2025–2033

30. Mazza T, Barborini E, Kholmanov IN, Piseri P, Bongiorno G,

Vinati S, Milani P, Ducati C, Cattaneo D, Li Bassi A, Bottani CE,

Taurino AM, Siciliano P. Libraries of cluster-assembled titania

films for chemical sensing. Appl Phys Lett 2005;87:103108.

31. Piseri P, Podesta A, Barborini E, Milani P. Production and charac-

terization of highly intense and collimated cluster beams by iner-

tial focusing in supersonic expansions. Rev Sci Instrum 2001;

72(5):2261–2267

32. Tafreshi HV, Piseri P, Benedek G, Milani P. The role of gas dy-

namics in operation conditions of a pulsed microplasma cluster

source for nanostructured thin films deposition. J Nanosci Nano-

technol 2006;6:1140–1149.

33. Mazza T, Barborini E, Kholmanov IN, Piseri P, Bongiorno G,

Vinati S, MIlani P, Ducati C, Li Bassi A, Bottani CE, Taurino AM,

Siciliano P. Libraries of cluster-assembled titania films for chemi-

cal sensing. Appl Phys Lett 2005;87(10).

34. Wegner K, Piseri P, Tafreshi V, Milani P. Cluster beam deposi-

tion: A tool for nanoscale science and technology. J Phys D:

Appl Phys 2006;39(22):R439–R459.

35. Chiappini C, Piseri P, Vinati S, Milani P. Supersonic cluster beam

deposition of nanostructured thin films with uniform thickness

via continuously graded exposure control. Rev Sci Instrum 2007;

78(6).

36. Barborini E, Piseri P, Milani P. A pulsed microplasma source of

high intensity supersonic carbon cluster beams. J Phys D: Appl

Phys 1999;32:L105.

37. Barborini E, Piseri P, Milani P. A pulsed microplasma source of

high intensity supersonic carbon cluster beams. J Phys D: Appl

Phys 1999;32:L105–L109.

38. Andersen N. In: Scoles G, editor. Atomic and Molecular Beam

Methods, Vol. 1. New York, NY and Oxford, UK: Oxford Univer-

sity Press; 1988. p 206–207.

39. Barborini E, Piseri P, Li Bassi A, Ferrari AC, Bottani CE, Milani P.

Synthesis of carbon films with controlled nanostructure by sepa-

ration of neutral clusters in supersonic beams. Chem Phys Lett

1999;300(5):633–638

40. Tafreshi HV, Benedek G, Piseri P, Vinati S, Barborini E, Milani P.

A simple nozzle configuration for the production of low diver-

gence supersonic cluster beam by aerodynamic focusing. Aero-

sol Sci Technol 2002;36(5):593–606

41. Milani P, Piseri P, Barborini E, Podesta A, Lenardi C. Cluster

beam synthesis of nanostructured thin films. J Vac Sci Technol

A: Vac Surf Films 2001;19:2025.

42. Barborini E, Piseri P, Podesta A, Milani P. Cluster beam micro-

fabrication of patterns of three-dimensional nanostructured

objects. Appl Phys Lett 2000;77(7):1059–1061

43. Piseri P, Tafreshi HV, Milani P. Manipulation of nanoparticles in

supersonic beams for the production of nanostructured materi-

als. Curr Opin Solid State Mater Sci8:195–202.

44. Singh AV, Vyas V, Patil R, Sharma V, Scopelliti PE, Bongiorno G,

Podest�a A, Lenardi C, Gade WN, Milani P. Quantitative character-

ization of the influence of the nanoscale morphology of nano-

structured surfaces on bacterial adhesion and biofilm formation.

PLoS One 2011;6(9):e25029.

45. Singh AV, Salve TS, Cortelli D, Dellasega D, Podesta A, Milani P,

Gade WN. Biofilm formation on nanostructured titanium oxide

surfaces and a micro/nanofabrication-based preventive strategy

using colloidal lithography. Biofabrication 2012;4(3).

46. Fioretti F, Mendoza-Palomares C, Helms M, Al Alam D, Richert

L, Arntz Y, Rinckenbach S, Garnier F, Haıkel Y, Gangloff SC,

Benkirane-Jessel N. Nanostructured assemblies for dental

application. ACS Nano 2010;4(6):3277–3287

47. Barborini E, Vinati S, Leccardi M, Repetto P, Bertolini G, Rorato

O, Lorenzelli L, Decarli M, Guarnieri V, Ducati C, Milani P. Batch

fabrication of metal oxide sensors on micro-hotplates. J Micro-

mech Microeng 2008;18:7.

48. Cassina V, Gerosa L, Podest�a A, Ferrari G, Sampietro M, Fioren-

tini F, Mazza T, Lenardi C, Milani P. Nanoscale electrical proper-

ties of cluster-assembled palladium oxide thin films. Phys Rev B

2009;79(11):115422

49. Della Foglia F, Losco T, Piseri P, Milani P, Selli E. Photocatalytic

activity of nanostructured TiO2 films produced by supersonic

cluster beam deposition. J Nanopart Res 2009;11(6):1339–1348

50. Carbone R, De Marni M, Zanardi A, Vinati S, Barborini E, Forna-

sari L, Milani P. Characterization of cluster-assembled nanostruc-

tured titanium oxide coatings as substrates for protein arrays.

Anal Biochem 2009;394(1):7–12

51. Carbone R, Marangi I, Zanardi A, Giorgetti L, Chierici E, Berlanda

G, Podest�a A, Fiorentini F, Bongiorno G, Piseri P, Pelicci PG, Milani

P. Biocompatibility of cluster-assembled nanostructured TiO2 with

primary and cancer cells. Biomaterials 2006;27(17) 3221–3329

52. Barborini E, Conti AM, Kholmanov I, Piseri P, Podest�a A, Milani

P, Cepek C, Sakho O, Macovez R, Sancrotti M. Nanostructured

TiO2 Films with 2 eV optical gap. Adv Mater 2005;17(15):

1842–1846

53. Vyas V, Podest�a A, Milani P. Probing nanoscale interactions on

biocompatible cluster-assembled titanium oxide surfaces by

atomic force microscopy. J Nanosci Nanotechnol 2011;11(6):

4739–4748

54. Caruso T, Lenardi C, Mazza T, Policicchio A, Bongiorno G, Agos-

tino RG, Chiarello G, Colavita E, Finetti P, Prince KC, Ducati C,

Piseri P, Milani P. Photoemission investigations on nanostruc-

tured TiO2 grown by cluster assembling. Surf Sci 2007;601(13):

2688–2691

55. Vandamme N, Verschoren G, Depuydt A, Cannaerts M, Bouwen

W, Lievens P, Silverans CE, Van Haesendonck C. Deposition of

gold clusters on atomically flat gold films. Appl Phys A: Mater

Sci Proc 2001;72(8):S177–S180.

56. Barabasi A, Stanley H. Fractal Concepts in Surface Growth—Cam-

bridge Books Online. New York: Cambridge University Press;

1995.

57. Olson CL, Nelson J, Islam MS. Defect chemistry, surface struc-

tures, and lithium insertion in anatase TiO2. J Phys Chem B

2006;110(20):9995–10001

58. Giorgetti L, Bongiorno G, Podest�a A, Berlanda G, Scopelliti PE,

Carbone R, Milani P. Adsorption and stability of streptavidin on

cluster-assembled nanostructured TiOx films. Langmuir 2008;

24(20):11637–11644

59. Fabre RM, Talham DR. Stable supported lipid bilayers on zirco-

nium phosphonate surfaces. Langmuir 2009;25(21):12644–12652

60. Denry I, Kelly JR. State of the art of zirconia for dental applica-

tions. Dent Mater 2008;24(3):299–307

61. Ratner B. The biocompatibility manifesto: biocompatibility for

the twenty-first century. J CardiovasTransl Res 2011:1–5.

62. Oliver RG. In: Brantley WA,Eliades T, editors. Orthodontic Mate-

rials. Scientific and Clinical Aspects.Stuttgart, Germany:Thieme;

2001. p 310.

63. Singh AV, Lenardi C, Gailite L, Gianfelice A, Milani P. A simple

lift-off-based patterning method for micro- and nanostructuring

of functional substrates for cell culture. J Micromech Microeng

2009;19(11):115028

64. Singh AV, Gailite L, Vyas V, Lenardi C, Forti S, Matteoli M,

Milani P. Rapid prototyping of nano- and micro-patterned sub-

strates for the control of cell neuritogenesis by topographic and

chemical cues. Mater Sci Eng C 2011;31(5):892–899

65. Schmidt SR, Waldeck H, Kao WJ. Protein Adsorption to Biomate-

rials. New York, NY: Springer; 2009. p 1–18.

3006 SINGH BOTTOM-UP ENGINEERING TO OPTIMIZE CELL–PROTEIN–SURFACE INTERACTIONS

66. Koegler P, Clayton A, Thissen H, Santos GN, Kingshott P. The

influence of nanostructured materials on biointerfacial interac-

tions. Adv Drug Deliv Rev 2012;64(15):1820–1839

67. Scopelliti PE, Borgonovo A, Indrieri M, Giorgetti L, Bongiorno G,

Carbone R, Podest�a A, Milani P. The effect of surface nano-

metre-scale morphology on protein adsorption. PLoS One 2010;

5(7):e11862.

68. Belicchi M, Erratico S, Razini P, Meregalli M, Cattaneo A, Jac-

chetti E, Farini A, Villa C, Bresolin N, Porretti L, Lenardi C, Milani

P, Torrente Y. Ex vivo expansion of human circulating myogenic

progenitors on cluster-assembled nanostructured TiO2. Biomate-

rials 2010;31(20):5385–5396

69. Scopelliti PE, Giorgetti L, Bongiorno G, Podest�a A, Berlanda G,

Carbone R, Milani P. Adsorption and stability of streptavidin on

cluster-assembled nanostructured TiOx films. Biophys J 2009;

96(3, Suppl 1):50a-a.

70. Giorgetti L, Zanardi A, Venturini S, Carbone R. Immunocell-

array: A novel technology for pathway discovery and cell profil-

ing. Expert Rev Proteomics 2007;4(5):609–616

71. Wheeler DB, Carpenter AE, Sabatini DM. Cell microarrays and

RNA interference chip away at gene function. Nat Genet 2005;

37(6 Suppl):S25–S30.

72. Bailey SN, Ali SM, Carpenter AE, Higgins CO, Sabatini DM.

Microarrays of lentiviruses for gene function screens in immor-

talized and primary cells. Nat Methods 2006;3(2):117–122

73. Wu RZ, Bailey SN, Sabatini DM. Cell-biological applications of

transfected-cell microarrays. Trends Cell Biol 2002;12(10):

485–488

74. Wheeler DB, Bailey SN, Guertin DA, Carpenter AE, Higgins CO, Saba-

tini DM. RNAi living-cell microarrays for loss-of-function screens in

Drosophila melanogaster cells. Nat Methods 2004;1(2):127–132

75. Ziauddin J, Sabatini DM. Microarrays of cells expressing defined

cDNAs. Nature 2001;411(6833):107–110

76. Hubbell JA. Materials as morphogenetic guides in tissue engi-

neering. Curr Opin Biotechnol 2003;14(5):551–558

77. Stevens MM, George JH. Exploring and engineering the cell sur-

face interface. Science 2005;310(5751):1135–1138

78. Haouzi D, Baghdiguian S, Granier G, Travo P, Mangeat P, Hibner

U. Three-dimensional polarization sensitizes hapatocytes to Fas/

CD95 apoptotic signalling. J Cell Sci 2005;118(12):2763–2773

79. Hook AL, Thissen H, Voelcker NH. Surface manipulation of bio-

molecules for cell microarray applications. Trends Biotechnol

2006;24(10):471–477

80. Falconnet D, Csucs G, Michelle Grandin H, Textor M. Surface en-

gineering approaches to micropattern surfaces for cell-based

assays. Biomaterials 2006;27(16):3044–3063

81. Echeverri CJ, Perrimon N. High-throughput RNAi screening in

cultured cells: A user’s guide. Nat Rev Genet 2006;7(5):373–384

82. Jeffrey AH. Materials as morphogenetic guides in tissue engi-

neering. Curr Opin Biotechnol 2003;14(5):551–558

83. Haouzi D, Baghdiguian S, Granier G, Travo P, Mangeat P, Hibner

U. Three-dimensional polarization sensitizes hepatocytes to Fas/

CD95 apoptotic signalling. J Cell Sci 2005;118(12):2763–2773

84. Carbone R, Giorgetti L, Zanardi A, Marangi I, Chierici E, Bon-

giorno G, Fiorentini F, Faretta M, Piseri P, Pelicci PG, Milani P.

Retroviral microarray-based platform on nanostructured TiO2 for

functional genomics and drug discovery. Biomaterials 2007;

28(13):2244–2253

85. Legrain P, Aebersold R, Archakov A, Bairoch A, Bala K, Beretta

L, Bergeron J, Borchers CH, Corthals GL, Costello CE, Deutsch

EW, Domon B, Hancock W, He F, Hochstrasser D, Marko-Varga

G, Salekdeh GH, Sechi S, Snyder M, Srivastava S, Uhl�en M, Wu

CH, Yamamoto T, Paik YK, Omenn GS. The human proteome

project: current state and future direction. Mol Cell Proteomics

2011;10(7).

86. Hanash SM, Pitteri SJ, Faca VM. Mining the plasma proteome

for cancer biomarkers. Nature 2008;452(7187):571–579

87. Harrison PM, Kumar A, Lang N, Snyder M, Gerstein M. A ques-

tion of size: The eukaryotic proteome and the problems in defin-

ing it. Nucleic Acids Res 2002;30(5):1083–1090

88. Bertone P, Snyder M. Advances in functional protein microarray

technology. FEBS J 2005;272(21):5400–5411

89. Saerens D, Ghassabeh GH, Muyldermans S. Antibody technol-

ogy in proteomics. Brief Funct Genomics Proteomics 2008;7(4):

275–282

90. Ling MM, Ricks C, Lea P. Multiplexing molecular diagnostics and

immunoassays using emerging microarray technologies. Expert

Rev Mol Diagn 2007;7(1):87–98

91. Saerens D, Ghassabeh GH, Muyldermans S. Antibody technol-

ogy in proteomics. Brief Funct Genomics Proteomics 2008;7(4):

275–282

92. Kasemo B, Gold J. Implant surfaces and interface processes.

Adv Dent Res 1999;13(1):8–20

93. Zanardi A, Giorgetti L, Botrugno OA, Minucci S, Milani P, Pelicci

PG, Carbone R. Immunocell-array for molecular dissection of

multiple signaling pathways in mammalian cells. Mol Cell Pro-

teomics 2007;6(5):939–947

94. Sevecka M, Wolf-Yadlin A, MacBeath G. Lysate microarrays ena-

ble high-throughput, quantitative investigations of cellular sig-

naling. Mol Cell Proteomics 2011;10(4):M110.005363.

95. Liotta LA, Kohn EC, Petricoin EF. Clinical Proteomics. JAMA

2001;286(18):2211–2214

96. Petricoin EF 3rd, Bichsel VE, Calvert VS, Espina V, Winters M,

Young L, Belluco C, Trock BJ, Lippman M, Fishman DA, Sgroi

DC, Munson PJ, Esserman LJ, Liotta LA. Mapping molecular net-

works using proteomics: A vision for patient-tailored combina-

tion therapy. J Clin Oncol 2005;23(15):3614–3621

97. Spurrier B, Ramalingam S, Nishizuka S. Reverse-phase protein

lysate microarrays for cell signaling analysis. Nat Protoc 2008;

3(11):1796–1808

98. Chan SM, Ermann J, Su L, Fathman CG, Utz PJ. Protein microar-

rays for multiplex analysis of signal transduction pathways. Nat

Med 2004;10(12):1390–1396

99. Cadigan KM, Liu YI. Wnt signaling: Complexity at the surface. J

Cell Sci 2006;119(3):395–402

100. Tomilin NV, Solovjeva LV, Svetlova MP, Pleskach NM, Zalen-

skaya IA, Yau PM, Bradbury EM. Visualization of focal nuclear

sites of DNA repair synthesis induced by bleomycin in human

cells. Radiat Res 2001;156(4):347–354

101. Giorgetti L, Zanardi A, Venturini S, Carbone R. Immunocell-

array: A novel technology for pathways discovery and cell profil-

ing 2007;4(5):609–616

102. Ronzoni S, Faretta M, Ballarini M, Pelicci P, Minucci S. New

method to detect histone acetylation levels by flow cytometry.

Cytometry Part A 2005;66A(1):52–61.

103. Perlman ZE, Slack MD, Feng Y, Mitchison TJ, Wu LF, Altschuler

SJ. Multidimensional drug profiling by automated microscopy.

Science 2004;306(5699):1194–1198

104. Francavilla C, Cattaneo P, Berezin V, Bock E, Ami D, de Marco A,

Christofori G, Cavallaro U. The binding of NCAM to FGFR1 indu-

ces a specific cellular response mediated by receptor trafficking.

J Cell Biol 2009;187(7):1101–1116

105. Guilleaume B, Buneß A, Schmidt C, Klimek F, Moldenhauer G,

Huber W, Arlt D, Korf U, Wiemann S, Poustka A. Systematic

comparison of surface coatings for protein microarrays. Proteo-

mics 2005;5(18):4705–4712

106. De Marni ML, Monegal A, Venturini S, Vinati S, Carbone R, de Marco

A. Antibody purification-independent microarrays (PIM) by direct

bacteria spotting on TiO2-treated slides. Methods 2012;56:317–325.

107. Bengt K. Biological surface science. Curr Opin Solid State Mater

Sci 1998;3(5):451–459

108. Lipski AM, Jaquiery C, Choi H, Eberli D, Stevens M, Martin I,

Chen IW, Sastry PV. Nanoscale engineering of biomaterial surfa-

ces. Adv Mater 2007;19(4):553–557

109. Rocker C, Potzl M, Zhang F, Parak WJ, Nienhaus GU. A quantita-

tive fluorescence study of protein monolayer formation on col-

loidal nanoparticles. Nat Nano 2009;4(9):577–580

110. Nel AE, M€adler L, Velegol D, Xia T, Hoek EMV, Somasundaran P,

Klaessig F, Castranova V, Thompson M. Understanding biophy-

sicochemical interactions at the nano–bio interface. Nat Mater

2009;8(7):543–557

111. Roach P, Farrar D, Perry CC. Interpretation of protein adsorption:

Surface-induced conformational changes. J Am Chem Soc 2005;

127(22):8168–8173

REVIEW ARTICLE

JOURNAL OF BIOMEDICAL MATERIALS RESEARCH A | OCT 2013 VOL 101A, ISSUE 10 3007

112. Roach P, Farrar D, Perry CC. Surface tailoring for controlled pro-

tein adsorption: Effect of topography at the nanometer scale and

chemistry. J Am Chem Soc 2006;128(12):3939–3945

113. Chayen NE, Saridakis E, Sear RP. Experiment and theory for het-

erogeneous nucleation of protein crystals in a porous medium.

Proc Natl Acad Sci USA 2006;103(3):597–601

114. Zanardi A, Bandiera B, Bertolini F, Corsini CA, Gregato G, Milani P,

Barborini E, Carbone R. Miniaturized FISH for screening of onco-

hematological malignancies. Biotechniques 2010;49:497–504.

115. Lenardi C, Perego C, Cassina V, Podest�a A, D’Amico A, Gualand-

ris D, Vinati S, Fiorentini F, Bongiorno G, Piserj P, Vellea Sacchi

F, Milani P. Adhesion and proliferation of fibroblasts on cluster-

assembled nanostructured carbon films: The role of surface mor-

phology. J Nanosci Nanotechnol 2006;6(12):3718–3730

116. Singh AV, Galuzzi M, Borghi F, Vyas V, Podesta A, Indrieri M,

Gade WN. Interaction of bacterial cells with cluster-assembled

nanostructured titania surfaces: An atomic force microscopy

study. J Nanosci Nanotechnol 2013;13:77–85.

3008 SINGH BOTTOM-UP ENGINEERING TO OPTIMIZE CELL–PROTEIN–SURFACE INTERACTIONS