Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Review Functional protein micropatterning for drug design and discovery Changjiang You & Jacob Piehler† † 1. Introduction 2. Surface chemistries and surface architectures 3. Spatially resolved protein Downloaded by [Universität Osnabrueck], [Jacob Piehler] at 02:57 05 December 2015 immobilization 4. Applications for drug discovery 5. Conclusions 6. Expert opinion Department of Biology, Division of Biophysics, University of Osnabrück, Osnabrück, Germany Introduction: The past decade has witnessed tremendous progress in surface micropatterning techniques for generating arrays of various types of biomolecules. Multiplexed protein micropatterning has tremendous potential for drug discovery providing versatile means for high throughput assays required for target and lead identification as well as diagnostics and functional screening for personalized medicine. However, ensuring the functional integrity of proteins on surfaces has remained challenging, in particular in the case of membrane proteins, the most important class of drug targets. Yet, generic strategies to control functional organization of proteins into micropatterns are emerging. Areas covered: This review includes an overview introducing the most common approaches for surface modification and functional protein immobilization. The authors present the key photo and soft lithography techniques with respect to compatibility with functional protein micropatterning and multiplexing capabilities. In the second part, the authors present the key applications of protein micropatterning techniques in drug discovery with a focus on membrane protein interactions and cellular signaling. Expert opinion: With the growing importance of target discovery as well as protein-based therapeutics and personalized medicine, the application of protein arrays can play a fundamental role in drug discovery. Yet, important technical breakthroughs are still required for broad application of these approaches, which will include in vitro “copying” of proteins from cDNA arrays into micropatterns, direct protein capturing from single cells as well as protein microarrays in living cells. Keywords: cell patterning, membrane protein, protein array, protein immobilization, protein interaction, surface micropatterning Expert Opin. Drug Discov. [Early Online] 1. Introduction Modern drug discovery faces immense analytical challenges, which range from identification and validation of targets and biomarkers, selection and optimization of lead compounds to increasing diagnostic efforts for toxicological evaluation and personalized medicine. In the past decade, high-throughput assays (HTA) based on microarray technologies have dramatically reshaped this procedure in an efficient and sample-economical manner. A key requirement for HTA is to massively parallelize and miniaturize binding and activity assays in order to limit sample consumption. For this purpose, solid-phase-based assays are most suitable as they combine highly sensitive spectroscopic readout with convenient automated handling of minute sample volumes by microfluidic techniques. For generating microarrays for HTA, samples have to be immobilized on the substrate surface in a spatially resolved manner and thousands of interactions are probed simultaneously. A broad range of compound classes have been employed to generate microarrays (biochips) for HTA in modern drug discovery,[1] including small molecules, 10.1517/17460441.2016.1109625 © 2015 Taylor & Francis. ISSN 1746-0441, e-ISSN 1746-045X All rights reserved: reproduction in whole or in part not permitted 1 You & Piehler Downloaded by [Universität Osnabrueck], [Jacob Piehler] at 02:57 05 December 2015 Article highlights. ● Microarray-based technology is a cornerstone for highthroughput assays in modern drug discovery. Protein microarray techniques are gaining importance not only for target and biomarker identification and validation, but also for developing protein-based pharmaceuticals. ● The key challenge in protein microarray technology is to maintain the functional integrity of immobilized proteins, which requires the substrate surface to be engineered protein compatible as well as site-specific protein conjugation under physiological conditions. ● Multiplexed protein immobilization on biocompatible surface is very important for high-content, high-throughput screening. For this purpose, a variety of bioorthogonal conjugation methods and multiplexed micropatterning techniques have been developed. ● More sophisticated surface architectures are required for fabricating membrane protein microarrays, since the functions of membrane proteins are inextricably linked to their native lipid environment. ● Live-cell protein microarray techniques are emerging, which exploit the tremendous progress in single-cell and single-molecule analysis and promises ultrasensitive drug screening to single cell–single molecule level under native conditions. This box summarizes key points contained in the article. oligonucleotides, carbohydrates, peptides and proteins.[2,3] Due to the diversity of samples to be immobilized on different substrate materials, multifarious surface chemistries and microfabrication methods have been developed to ensure the specificity and efficiency of interaction screening. Thus, highly multiplexed oligonucleotide and peptide microarrays are routinely produced with very high quality and reproducibility. However, more than 80% of the therapeutic targets are proteins, which include membrane receptors, ion channels and enzymes.[4,5] Moreover, therapeutic proteins have tremendously gained importance during the past decade, which often require intensive further engineering efforts before being suitable for application.[6] Traditional technologies for generating microarray are often not compatible with proteins as their functional integrity is not sustained under the immobilization conditions. The metastable nature of three-dimensional protein structures, which is inextricably linked to their function, as well as the highly diverse physicochemical and functional properties of proteins demand dedicated solutions for engineering protein-compatible surface architectures. This is particularly challenging for membrane proteins which constitute a major fraction of the most relevant drug targets,[5] yet often require a specific lipid environment for maintaining their structural and functional integrity.[7] In this review, we focus on emerging strategies for spatially resolved surface functionalization suitable for 2 functional organization of proteins on surfaces. We present chemical and biochemical aspects of functional protein immobilization on solid support as well as micropatterning methodologies for spatial controlled functionalization under physiological conditions. Recent applications of using micropatterned protein arrays for drug discovery and screening are highlighted. Given the significant advances of live-cell micropatterning and single-cell analysis,[8,9] recent applications of micropatterned protein surfaces for mapping signaling pathways and protein–protein interaction in live cells are presented and their potential in drug discovery is discussed. 2. Surface chemistries and surface architectures A variety of substrate materials have been used for functional protein micropatterning, including nitrocellulose, polydimethylsiloxane (PDMS), glass, silicon wafer and gold-coated silicon/glass. Glass is widely used due to its excellent compatibility with optical detection techniques, including highly sensitive fluorescence imaging. Glass or silicon supports coated with gold or other metals are also very commonly used, as they support functionalization via self-assembled monolayers (SAMs) as well as the application of label-free detection by surface plasmon resonance. For sensitive assays, functional interfacing of proteins with these substrate materials is a major prerequisite. Except for very few robust proteins such as antibodies,[10] direct interaction of proteins with the bare substrate mostly results in loss of their functions because the interactions with the substrate surface may block active sites or even denature the native protein structure. In order to minimize such nonspecific interactions, functional protein immobilization requires coating with organic polymers for rendering the surface protein repellent (FIGURE 1A). In turn, site-specific attachment of the protein to the surface is needed in order to ensure their presentation at the surface in a functionally homogenous manner (FIGURE 1A). During the past 15 years, substantial progress in “soft” surface engineering has been made, providing a versatile spectrum of surface chemistries and surface functionalities suitable for functional protein immobilization (summarized in TABLE 1). Choosing an appropriate strategy not only depends on the substrate material, but also requires careful considerations, which will be elaborated in this section. 2.1 Protein-compatible surface modification As introduced above, the “protein compatibility” of a surface is largely equivalent to its minimized nonspecific interaction affinity toward proteins, and strategies, therefore, highly depend on the class of target proteins. Thus, soluble proteins typically require hydrophilic but largely uncharged surfaces. In contrast, membrane proteins are preferably integrated into lipid bilayers as the lipid environment often is critical for their activities. In general, molecular layers of non-charged, Expert Opin. Drug Discov. (2015) 11(1) Downloaded by [Universität Osnabrueck], [Jacob Piehler] at 02:57 05 December 2015 Functional protein micropatterning Figure 1. Surface engineering of micropatterned protein arrays. A. Generic surface architecture of functional protein micropatterning including a coating for rendering the substrate biocompatible (cyan layer) as well as spatially resolved functionalization (orange grids) for specific conjugation with biological components. Several commonly used materials for biocompatible layer are depicted. B–E. Surface modification for biocompatibility layers on different substrates. The reactive sites for introducing further functionalization are denoted by orange circles. B. Molecular scheme of PEGylated surface for glass (silica) and other metal oxide support. C. Self-assembled monolayer for ultrathin coatings of noble metal surfaces. D. Surface coating with functionalized poly-(L)-lysine graft poly(ethylene glycol) (PEG) on a negatively charged surface (material independent). E. Formation of lipid bilayer on surface provides the crucial lipid environment for keeping the activity of membrane proteins. F. Site-specific protein capturing on streptavidin-functionalized surfaces. A biotinylated antibody was used for multiplexed protein immobilization. G. Site-specific protein capturing onto tris-NTA-functionalized surfaces. H. Sitespecific protein capturing based on self-immobilizing protein tags. As an example, the HaloTag/HaloTag® ligand (HTL) reaction is shown. hydrophilic polymers such as dextran hydrogels [31] and dense layers of poly(ethylene glycol) (PEG) [32] have been proven efficient for shielding surfaces against nonspecific protein binding (FIGURE 1A). While PEGs with a molecular mass of 2000–5000 g/mol are commonly used in many cases, highly ordered SAMs formed on gold surface by alkyl thiolates with short PEG chains of 3–6 repeat units also efficiently shield the substrate surface.[33] Simple, yet highly efficient surface coating with PEG layers is achieved by a layer-by-layer assembly, in which a poly-L-lysine-graftPEG (PLL-PEG) copolymer is directly deposited on negatively charged surfaces.[14] Noncovalent assembly of lipids and lipid-like molecules into solid-supported lipid bilayers has been successfully applied for rendering surface biocompatible, which is important for functional reconstitution of membrane proteins on surfaces [16] (FIGURE 1B–E). A more sophisticated form of this surface architecture suitable for the integration of transmembrane proteins are polymer-supported membranes, where lipid bilayers are tethered above a thin polymer cushion to further minimize nonspecific interactions with the solid substrate.[18] 2.2 Surface biofunctionalization for protein immobilization On the basis of protein-repellent surfaces, further functionalization with anchoring moieties is essential for efficient protein immobilization. Amine-reactive coupling via aldehydes or activated carboxyl groups have been used for covalent tethering proteins to surfaces through lysine residues presented on protein surfaces. However, these approaches yield heterogeneously orientated attachment, which likely results in heterogeneous functionality due to blocking of relevant residues of protein.[34] Therefore, site-specific techniques for tethering proteins to surface are desired. Several chemical reactions such as Staudinger ligation, Diels–Alder reaction, azide-alkyne click reaction and thiol-ene reaction Expert Opin. Drug Discov. (2015) 11(1) 3 You & Piehler Table 1. Surface functionalization techniques for functional protein immobilization. Surface chemistry Coating of biocompatible layer Self-assembled monolayer Au–thiol interaction PEG brush Downloaded by [Universität Osnabrueck], [Jacob Piehler] at 02:57 05 December 2015 Poly lysine-graft-PEG Solid support lipid bilayer Polymer-supported, tethered lipid bilayer *Surface functionality Biotin or streptavidin Disadvantages Ref. Very efficient and stable surface coating; ultrathin layer, good coupling to detection (e.g., LSPR) Specific for applications on gold surface; Synthesis of complex compounds required Little control on composition; relatively undefined surface architecture Stability is dependent on pH and ionic strength of mediums Fragile coating, very sensitive to exposure to air [11] Covalent coupling Robust coating for versatile biofunctionalization; bottom-up synthesis from simple compounds Noncovalent, multiple One-step coating for obtaining charge–charge interaction biocompatible surface Noncovalent self-assembly One-step coating for obtaining on bare substrate biocompatible surface used for membrane proteins; self-healing Noncovalent self-assembly Functional reconstitution of on a polymer cushion transmembrane proteins is possible; lipid phase separation can be controlled Enzymatic (suicide) substrates Protein attachment Affinity capturing via biotin–streptavidin interaction Affinity capturing via Histag and Ni–NTA interaction Affinity capturing by enzymatic reaction GFP-trap (anti-GFP nanobody) Affinity capturing via GFP fusion Ni-NTA Advantages Advantages Very rapid, specific and strong protein attachment; site-specific binding is possible Small and very common affinity tag; reversibility for repetitive write–erase processes Site-specific, covalent protein attachment, feasible for fabricating multiplexed arrays Very rapid, specific and relatively stable; simple visualization/ quantification [12,13] [14,15] [16,17] Surface modification is rather error-prone; membrane assembly is not spontaneous [18– 20] Disadvantages Proteins have to be biotinylated; blocking of binding sites by endogenous biotin Reversible binding sensitive to nucleophiles, chelators, bivalent metal ions, reducing agents Relatively slow, inefficient reactions; specialized tags Ref. [21,22] Fusion of a relatively large tag required [23,24] [25– 28] [29] *The list summarizes the most common surface functionalities for protein attachment. Other specific, bioorthogonal methods for chemical coupling of proteins can be found in [30]. Further specialized approaches are given in the main text. GFP: Green fluorescence protein; LSPR: Localized surface plasmon resonance; NTA: Nitrilotriacetic acid; PEG: poly(ethylene glycol). have been exploited for site-specific protein immobilization. [30] These reactions, however, require not only couplingspecific anchoring groups on surface but also modifications of the protein, for example, by incorporation of non-natural amino acids,[35] which is not yet routinely used in most laboratories. Moreover, these chemical reactions are relatively slow, thus requiring high protein concentrations, which in turn increase the background. For these reasons, quasi-irreversible noncovalent interactions mediated via genetically encoded tags are currently favored as they allow highly efficient and specific protein capturing to surfaces. A large proportion of noncovalent protein immobilization approaches relies on either the very rapid and extremely high-affinity interaction between biotin and streptavidin (or the related proteins), or the complexation of immobilized transition metal ions by oligohistidine-tagged proteins.[21] In the first case, the surface is functionalized with streptavidin or related proteins, and the protein of interest modified with biotin by means of chemical or biochemical biotinylation are readily immobilized to the surface (FIGURE 1F). For 4 site-specific immobilization of oligohistidine-(His)-tagged proteins, functional moieties of iminodiacetic acid or nitrilotriacetic acid (NTA) are coupled to surfaces for immobilizing transition metal ions such as Zn(II), Cu(II), Ni(II) and Co (II).[36] More stable and stoichiometrically defined tethering of histidine-tagged protein was achieved by using multivalent chelators with two to four NTA moieties grafted onto different scaffolds,[24] with tris-NTA exactly matching the stoichiometry of the frequently applied hexahistidine-tag (FIGURE 1G). A broad spectrum of surface architectures functionalized with tris-NTA have been successfully implemented, yielding quasi-irreversible, site-specific protein immobilization.[24] These stable complexes, however, can be rapidly dissociated by competitors of imidazole at millimolar concentration, providing means for rapidly switching this interaction under mild conditions. Driven by the increasing demand for site-specific proteinlabeling techniques, protein immobilization via enzymatic reactions has gained much attention. In these methods, small-molecule ligands are covalently immobilized onto the Expert Opin. Drug Discov. (2015) 11(1) Downloaded by [Universität Osnabrueck], [Jacob Piehler] at 02:57 05 December 2015 Functional protein micropatterning surface. Enzymatic reactions successfully exploited for sitespecific protein immobilization include phosphopantetheinyl transfer,[25] transpeptidation [26] as well as enzymatic suicide reactions [27,37] (FIGURE 1H). For the assembly of protein arrays, highly multiplexed protein immobilization is required, and therefore orthogonal coupling approaches are desired. For this purpose, antibodies against the proteins of interest have been most frequently applied. For instance, monoclonal antibodies modified with biotin were chosen for site-specifically anchoring a variety of target proteins on the surface.[38] However, antibody binding to proteins often blocks active sites and thus considerably bias functional assays. In other attempts, the huge multiplexing capability of oligonucleotide pairing has been exploited. Single-stranded DNA with different oligonucleotide sequences were coupled to the surface in a spatially resolved manner and proteins modified with the complementary strands were captured to the surface via sequence-specific DNA hybridization, thus enabling in situ assembly of protein arrays.[39] 3. Spatially resolved protein immobilization The repertoire of techniques for spatial organization of proteins on surfaces in micrometer and nanometer dimensions is rapidly expanding in line with the rapidly growing application of protein arrays. Micro- and nanopatterned surface modification can be achieved by self-assembly as exploited for block copolymer micelle nanolithography.[40] While such bottom-up techniques are powerful for reaching nanometer resolution, the ability to control and vary geometries is rather limited. Therefore, top-down approaches are desired, which can be achieved either by local deposition of materials on the surface (soft lithography techniques) or by photochemical manipulation (photolithography techniques). Most frequently used techniques are briefly presented here as comprehensive in-depth background has been reviewed recently.[21,41] 3.1 Local deposition by soft lithographic techniques Piezoelectric dispensing by exploiting ink-jet technology has been a very straightforward approach to the assembly of spatially resolved surface modification, which has been exploited for generating the first-generation protein arrays. [2] While this technology is low-cost, robust and mature, features below 10 µm can only be obtained with relatively high technological efforts.[42] In the late 1990s, the concept of soft lithography for spatially resolved deposition of (bio) molecule onto surfaces with submicrometer resolution was introduced by George M Whitesides and coworkers to cover a set of microfabrication methods using molds, stamps or channels made by elastomeric materials.[43] Soft lithography is highly versatile as very different compounds can be directly transferred onto surfaces. The most commonly used elastomeric material is PDMS. The currently most broadly applied soft lithographic technique is microcontact printing (µCP), where the patterns of a master PDMS stamp coated with ink are transferred to the substrate surface through face-to-face contact (FIGURE 2A). By using stamps with different featured sizes, patterning resolution down to tens of nanometers has been obtained by µCP [43] (FIGURE 2B). µCP has been very broadly applied for patterning proteins and other biomolecules in very different formats.[44,45] Applications include direct deposition of proteins, for example, antibodies or streptavidin onto reactive or adhesive surfaces. However, this approach is not well compatible with most proteins as the printing process is prone to protein denaturation. Therefore, µCP of functional groups for site-specific protein capturing is preferred, which has been extensively applied to alkylthiolates on gold surfaces to form micropatterned SAM. [43] Likewise, µCP of biotin to an activated PEG polymer brush on glass surface has been implemented to prepare protein microarrays.[46] Approaches for Ni-NTA-based protein patterning by µCP of reactive alkyl thiols followed by chemical coupling of NTA [23], as well as direct µCP or by backfilling of NTA derivatives have been described.[11] Traditional µCP is designed to generate binary micropatterns. Substantially higher multiplexing has been achieved by using microscopic or nanoscopic stamps (“pens”) for locally depositing compounds (“inks”). This has been initially implemented by dip-pen nanolithography (DPN) using the tip of an atomic force microscope (AFM) as a pen to directly create patterns with inks on the substrate [48] (FIGURE 2C). With the options of using different proteins as inks, this method offers a great flexibility for making multiplexed protein arrays with length scale down to 50 nm on gold surface. Since different types of tips and substrates are implemented lately, it is also named as scanning probe lithography. DPN had been used for protein nanopatterning, for example, depositing antibodies on hydrophilic surface by DPN has been successfully demonstrated [51] as well as specific capturing of His-tagged proteins into nanopatterns.[49] By the combination of block copolymer micelle nanolithography and DPN, single-molecule protein arrays were obtained. [52] Several further techniques based on nano-manipulation via the AFM tip have been devised, including native protein nanolithography.[53] However, preparation of protein nanoarrays by this method is slow since writing proteins with nanometer profiles has to be carried out one by one. To overcome this limitation, functional immobilization of proteins on PEGmodified SAM surfaces by parallel DPN was developed, which helps improving the throughput.[54] More recently, Si-based nanostencil was introduced to make protein nanoarrays as “nanostencil lithography.”[55] This method can be deemed as a variation of parallel DPN. Instead of using tips to dip the ink, a nanostencil with ink reservoir was applied on the surface for allowing protein inks being patterned on substrate through apertures. Multiplexed DPN of lipid bilayer was realized on glass substrates with subcellular Expert Opin. Drug Discov. (2015) 11(1) 5 Downloaded by [Universität Osnabrueck], [Jacob Piehler] at 02:57 05 December 2015 You & Piehler Figure 2. Examples of soft lithography and dip-pen nanolithography (DPN) for preparing protein microarrys. A. Scheme of microcontact printing. Polydimethylsiloxane (PDMS) stamp soaked with poly-(L)-lysine-PEG-HaloTag® ligand (PLL-PEG-HTL) contacts a plasma-cleaned glass substrate (i), thus transfer the PLL-PEG-HTL patterning to the surface (ii). Backfilling with PLLPEG-OMe blocks the uncoated regions (iii). The obtained micropatterned surface is used for affinity-capturing HaloTag-fused proteins (iv). B. Scheme of nanocontact printing. A planar PDMS stamp is incubated with a protein solution, blow-dried (i), and quickly brought into contact with a photopolymer (Norland Optical Adhesive 63) replica that was plasma activated, and separated to lift-off proteins in the contact areas (ii). The flat PDMS stamp is then printed onto a glass slide (iii) and separated, producing protein nanopatterns on the slide (iv). (Adapted with permission from John Wiley and Sons.[47]) C. Surface nanopatterning by DPN. Top: principle of surface deposition of chemical “ink” adsorbed onto an atomic force microscope tip. (Reprinted with permission from the American Association for the Advancement of Science.[48]) Bottom: functional protein nanoarray fabricated by DPN and protein capturing. (Reprinted with permission from the American Chemical Society.[49]) D. Multiplexed protein immobilization by polymer pen lithography (PPL). Top: scheme depicting the deposition of different biomolecules adsorbed onto a polymer pen array. Bottom: multicolor micropatterns generated by PPL with fluorescent lipids. (Reprinted with permission from John Wiley and Sons.[50]) dimensions (down to 200 nm), which was functionalized with proteins for T-cell activation.[56] An even more rigorous development into parallel deposition has been achieved by polymer pen lithography (PPL).[57] Rather than AFM tips, this technique employs elastomeric polymer molds comprising an array of pyramid-shaped “pens” (FIGURE 2D). After inking these pens with the desired compounds, these are transferred onto surfaces by means of a piezoelectric system, which allows control of lateral position as well as the size of deposited features via the axial force. Thus, huge throughput of the fabrication process was achieved with 150,000 features per second for sub-100-nm structures.[57] In combination of multiplexed inking of polymer pen arrays, highly multiplexed deposition of relatively complex features has been demonstrated.[50,58] 3.2 Photolithographic techniques Photolitographic protein micropatterning is typically based on a photosensitive molecular layer directly grafted on a biocompatible surface coating. Typical components are 6 nitroveratryl group, azide, tetrazine, C=C bond containing compounds and maleimide. Photolithographic modification by means of illumination through a photomask or spatially controlled focused light is employed for generating micropatterns. Due to the diffraction limitation of light, conventional photolithography obtains patterns down to a length scale in the range of hundreds of nanometers. By exploiting the concept of stimulated emission depletion, resolution beyond the diffraction limit down to ~40 nm was achieved, [59] which has been used for fabricating protein nanoarrays.[60] The simplest approaches for photolithographic micropatterning are based on photodestruction of functional groups on the surface. This can be done by directly destroying capturing groups such as NTA, which can be photodegraded by a light-induced Fenton reaction.[61] Alternatively, chemical groups for coupling of functional groups can be eliminated by light. Thus, photolithographic patterning of maleimide-modified surface has been introduced [13], which are used for surface functionalization via thiols Expert Opin. Drug Discov. (2015) 11(1) Downloaded by [Universität Osnabrueck], [Jacob Piehler] at 02:57 05 December 2015 Functional protein micropatterning (FIGURE 3A). A strikingly high signal to background was achieved, which facilitated single-molecule studies in microarrays requiring high contrast (FIGURE 3B). Maleimide patterning also obtained very homogenous patterning of polymer-supported membranes that allowed assessing diffusion and interaction of reconstituted transmembrane proteins on the single molecule level without biasing their diffusion and interaction properties as resolved by single molecule imaging [62] (FIGURE 3C, D). Photolithographic patterning based on azide functionalization has gained considerable attention nowadays as the azide is a small, photosensitive moiety and widely used in azide-alkyne click reaction. Extracellular matrix (ECM) proteins containing RGD or REDV peptides as well as the unnatural amino acid azidophenylalanine (pN3-Phe) were spreaded on glass coverslides and crosslinked by UV-illumination through a photomask. By washing with 6 M guanidine hydrochloride, the protein was selectively removed in the non-illuminated regions, yielding guided cell adhesion these micropatterned surfaces.[63] Direct photopatterning of biomolecules via azide groups, that is, positive photolithography, were successfully demonstrated.[64] With the ability to site-specifically introduce amino acids with clickable side chains into recombinant proteins, exciting applications of these approaches for protein patterning can be envisaged. More versatile and sophisticated surface modification can be achieved by photocaging of functional groups. o-Nitroveratryloxycarbonyl (NVOC) is a very common photosensitive moiety applied for caging amine groups on the surface [65] (FIGURE 3E). A general photopatterning strategy for binary surface functionalization using NVOC contains two steps: uncaging the NVOC-protected amino groups with a photomask under UV irradiation, followed by chemical or biochemical functionalization of the amine groups in the uncaged areas for capturing proteins (FIGURE 3F).[66] This approach has been successfully applied for site-specific noncovalent and covalent protein immobilization [25,66] (FIGURE 3G). Likewise, photolithographic writing of SNAP-tag fusion proteins was achieved using caged benzylguanine.[28] Figure 3. Micropatterned surface functionalization by photolithography. A–D. Surface micropatterning based on maleimide photodestruction. Spatially resolved surface functionalization was used for single-molecule protein-binding assays. (A, B: Adapted with permission from the American Chemical Society.[13] C, D: Adapted with permission from John Wiley and Sons. [62]) E–G. Binary surface patterning based on caging of surface amines. E. The structure of the photoprotection group onitroveratryloxycarbonyl (NVOC). F. Scheme of photolithographic patterning of lipid anchors on PEGylated surface. Top to bottom: UV light decages NVOC-protected amine groups via a photomask, followed by coupling of functional groups type A. A second UV exposure without mask decages the remaining NOVCs for sequential coupling of functional groups type B. G. Binding experiment with binary micropatterned surface comprising biotin and tris-NTA functionalities. Reversible protein– protein interaction with a soluble receptor domain is demonstrated by fluorescence imaging. (Adapted with permission from the American Chemical Society.[25]) Expert Opin. Drug Discov. (2015) 11(1) 7 Downloaded by [Universität Osnabrueck], [Jacob Piehler] at 02:57 05 December 2015 You & Piehler While these photolithographic techniques are powerful for spatially organizing one or two different proteins on surfaces, higher multiplexing requires techniques for directly targeting proteins into microscopic structures. Photolithographic “writing” of proteins has been achieved by different approaches. Nitroveratryl groups have also been applied for photocaging biomolecules. By local uncaging of surface NiNTA complexed with photocleavable oligohistidine peptides, in situ micropatterning of His-tagged proteins on surfaces by laser lithography was achieved with diffraction-limited resolution.[67] Likewise, caged glutathione was employed for micropatterning of proteins fused to glutathione S-transferase,[68] a common protein used for affinity purification. These techniques very elegantly allow multiplexed writing and erasing of proteins on surfaces, yet suffer from instabilities and relatively high background due to the noncovalent nature of the interactions. More robust laser lithography is achieved by photocatalysed formation of covalent bonds. Thus, protein micropatterning by means of photoinitiated thiol-ene click reactions has been implemented.[69] Another click reaction based on tetrazine-norbornene chemistry was recently reported for forming cell-laden hydrogels. Duplexed protein patterning in this “click gel” was realized by taking advantage of orthogonal thiol-ene photoreaction and Diels–Alder reaction of tetrazine-norbornene.[12] Related to photolithographic techniques, also electrons provide versatile means for spatially resolved chemical manipulation. Electron beams (e-beams) can be focused to spot sizes of a few nanometers, making e-beam lithography a powerful way for fabricating nanoarrays. By e-beam-controlled reduction and cross-linking of aromatic SAMs, nanopatterned amino groups on gold surfaces were obtained for further biofunctionalization and nanopatterning of biomolecules at the submicron scales.[70] Multiplexed protein patterning with nanometer scale by e-beam lithography was achieved by depositing functionalized PEG dendrimers, yielding a line width of ~100 nm.[71] Alternatively, electron-beam-induced deposition has been used for depositing carbon nanostructures onto a PEG polymer brush for subsequent adsorption of proteins with very high contrast.[72] It should be noted, however, that application of these methods is limited to conductive substrates. 4. Applications for drug discovery 4.1 Profiling protein–protein interaction networks Early applications of large-scale protein arrays have been focused on protein interaction analyses, which have had substantial impact in proteomic research for drug discovery and, in particular, for personalized medicine.[73] Compared to affinity-capturing techniques coupled with mass spectrometry (AC/MS) and two-hybrid technologies, the advantage of protein microarrays is that they contain nearly all proteins coded by whole genome, and they can be assayed 8 simultaneously in vitro. The first proteome chip contained 94% (~5800 of 6200) of the yeast proteins with N-terminal GST-His-tag tandem affinity purification tag.[34] Since then, this technology has become an important tool in several fields of drug discovery [74] and protein microarrays of kinases, phosphatases and proteases have become available. [75] High-throughput global identification of protein kinase substrates and protein kinase–phosphatase interaction networks have emerged.[76,77] Such global network mapping is very important for clinical diagnosis because the anomaly regulation of protein kinase activity is strongly related to diseases such as cancer, and the inhibitor or activators for these interactions are important drug candidates. Fluorescence detection techniques are currently the preferred readout methods as they provide unrivaled signal-to-background ratios with a sensitivity down to the level of single molecules.[78] However, recent development of ultrasensitive label-free detection by LSPR offers interesting options, [15,79] as often the labeling is a critical and time-consuming step in assay development. A key shortcoming of state-of-the-art protein array technology is the necessity to produce and purify proteins prior to deposition on the surface, a very demanding and timeconsuming process. Owing to the fragile folding, storage of proteins and protein arrays often leads to loss of function and therefore in situ assembly of protein arrays is highly desired. For this reason, current developments in the field aim for closer linking protein production and assembly into arrays, for example, by local in vitro translation on micropatterned support [80,81] or by in situ protein capturing from cells.[29] For plasma membrane receptors, which are key drug targets, yet are often very difficult to isolate, livecell protein capturing has been implemented [22] (also see Section 4.3). Recently, this technology has been adapted to allow multiplexed protein interaction analysis inside a living cell.[82] To this end, an antibody array fabricated by DPN was used to micropattern cytosolic proteins through an engineered transmembrane linker, allowing for monitoring activation of protein kinase A upon G-protein-coupled receptor (GPCR) stimulation. 4.2 Screening GPCR ligand binding GPCRs represent the largest family of therapeutic targets.[5] These receptors are the targets of more than 50% of the current pharmaceutical agents on the market, with a global market reaching ~100 billion USD in 2013.[83] Over a long time, GPCR drug discovery by high-throughput screening (HTS) for hit identification has remained the major focus of drug discovery worldwide.[84] Commonly used HTS assays for GPCR ligand binding and functional assays include the key detection points in GPCR signaling pathway, such as detection of ligand binding, GTPγS binding, cAMP, IP3/IP1, Ca2+ flux, β-arrestin recruitment, receptor internalization and receptor dimerization.[85] Expert Opin. Drug Discov. (2015) 11(1) Downloaded by [Universität Osnabrueck], [Jacob Piehler] at 02:57 05 December 2015 Functional protein micropatterning Over the past decade, versatile techniques for functional micropatterning of membrane proteins have been established, which facilitate in vitro HTS of GPCR ligand binding. [16,86] GPCR arrays of the β1-adrenergic receptor, the neurotensin receptor (NTR1) and the dopamine receptor based on microspotted lipid membrane have been successfully fabricated. Specific ligand binding to these GPCR microarrays was confirmed and IC50 value of neurotensin to NTR1 down to 2 nM was measured by competitive ligand chasing.[87] In the following work reported by Pfizer Inc. (New York, NY, USA) and Corning Inc. (New York, NY, USA), multiplexed GPCR microarrays of α-adrenergic receptors were developed for ligand-binding assays based on 125Ilabeled agonist clonidine. A 200- to 400-fold decreased sample preparation was sufficient for analysis, which is very useful for HTS.[88] Another type of GPCR microarray was achieved by embedding membrane proteins into liposomes prepared from Sf9 cellular membrane extracts. Site-directed immobilization of the liposomes on protein repelling surface was achieved by hybridization of cholesterol-modified oligonucleotides between the liposomes and the surfaces. The functionalities of GPCR models in liposomes, for example, H1-histamine receptor and the M2-muscarinic receptor, were confirmed by ligand-binding assays and G-protein signaling assays under a microarray reader format.[19,20] This liposome-based strategy yielded impressive performance and can be very efficient for HTS of potential GPCR ligands since the proof of concept of singleliposome arrays has been established by using µCP.[89] To further simplify the sample preparation of GPCR microarray and effectively screen GPCR drugs in native cellular environments, live-cell microarrays in the format of well-plate, micropatterned surface, and microfluidic systems have been developed.[90] 4.3 Probing signal transduction in live cells Large-scale monitoring of signal transduction pathways stands as the largest challenge in drug discovery, as almost all known diseases exhibit dysfunctional aspects in signal transduction networks,[91] and therefore the vast majority of drug target are part of these.[5] Many signaling pathways have been reasonably mapped at molecular level, uncovering a highly complex network of subtle and interdependent cross talk. The integration of this complexity into cellular decision making is still largely unclear. Therefore, the overall potency of a drug has to be verified under physiological conditions. Monitoring signaling pathways and screening the perturbations of drugs to these pathways with multiplexed assays thus has unique potential to bridge the gap between molecularlevel assays and the animal tests. 4.3.1 Cell adhesion and integrin signal transduction The interaction and migration of cells within tissues play a key role in stem cell differentiation and organogenesis, but also in tumor development and metastasis. Promising new drug targets such as the Wnt/β-catenin pathway involved in controlling these processes are currently emerging.[92] Micro- and nanopatterned surface functionalization offers unique means to mimic and study these extremely complex processes in vitro and thus to identify key molecular and cellular determinants and their potential manipulation by drugs.[93] Cell adhesion and apoptosis on micropatterned RGD peptides [94] and ECM proteins [95] have been examined to elucidate the mechanism of integrin signal transduction, which have shown determinant functions for regulating cell survival, differentiation and growth. By using copolymer micelle nanolithography, induced cell polarization and migration were examined in detail by spacing adhesive ligands in a gradient in nanoscale [94] (FIGURE 4A). Interestingly, these experiments revealed that inducing cell adhesion on ordered RGD nanopattern requires the RGD peptide being spaced below 70 nm. This critical distance confirms that the formation of stable focal adhesion and actin fiber networks have a distance threshold of 70 nm. Hierarchical RGD micro-/nanopattern designed using a similar strategy was recently used for investigation of stem cell differentiation.[96] Strikingly, growth of mammalian cells on micropatterned surface showed preserved left–right patterning, a phenomenon termed as “chirality”.[97] Chirality has also been detected for other single-cell types and multicellular structures, which highlights the possibility of chiral morphogenesis during stem cell differentiation. 4.3.2 Receptor tyrosine kinase signal transduction Receptor tyrosine kinases (RTKs) are major regulatory hubs controlling cell growth and differentiation, and therefore play a prominent role in tumor development. For this reason, several RTKs have become key drug targets.[100] Closer studies revealed an enormous complexity of RTK activation,[101] demanding new tools for testing the molecular mechanisms responsible for inhibition. Live-cell micropatterning (cf. Section 4.1) has opened unique avenues to study RTK signaling in situ. For this purpose, cells are cultured on micropatterned monoclonal antibodies against the target receptor, which will be captured and thus spatially rearranged within the plasma membrane. [22] Thus, interactions with other receptor subunits and/ or cytosolic effector proteins can be readily monitored. [22,98,102] By using tag-specific surface capturing rather than antibodies, generic and robust surface micropatterning for probing assembly and signaling of RTK and related receptors in living cells was recently implemented.[98] These surfaces allow control of cell attachment via RGD peptides and assembly of signaling complexes via receptors fused to the HaloTag (FIGURE 4B, C). Thus, protein– protein interactions involved in receptor dimerization and recruitment of cytosolic effector proteins could be quantified.[98] By using micropatterned lipid bilayers, cell–cell Expert Opin. Drug Discov. (2015) 11(1) 9 Downloaded by [Universität Osnabrueck], [Jacob Piehler] at 02:57 05 December 2015 You & Piehler Figure 4. Live-cell micropatterning for mapping signaling pathways. A. Scheme of probing cell polarization and migration on micropatterns prepared by copolymer micelle nanolithography. The scanning electron microscopy image shows an ultrasmall cellular protrusion. Inset highlights the cRGDfK-functionalized gold nanoparticles which are adhesion patches for single integrin receptors.[94] (Reprinted with permission from the American Chemical Society.[94]) B, C. Micropatterning of signaling complexes in living cells. B. Scheme depicting a binary RGD- and HTL-micropatterned surface used for probing type I interferon signaling in live cells. Cells are attached to the RGD patterns via focal adhesion. Interferon receptor subunit IFNAR2 fused with HaloTag was captured into the HTL-functionalized regions. C. Fluorescence images depicting processes involved in the assembly of active signaling complexes in micropatterns: ligand-induced receptor dimerization on the plasma membrane (left image); interaction with the cytosolic Janus kinases (center) and recruitment of cytosolic effector proteins. (Adapted with permission from The Rockefeller University Press.[98]) D. Reorganization of ligand-stimulated EphA2 by micropatterned ephrin-A1–functionalized supported membranes. Bright field images (left) and epifluorescence images of labeled EphA2 (right). Scale bar: 5 µm. (Reprinted with permission from the American Association for the Advancement of Science.[99]) E. Single-cell pull down for probing the kinetics and the stoichiometry of protein complexes. (Adapted with permission from the American Chemical Society.[29]) interactions can be mimicked to identify the determinants of cellular recognition processes. Thus, immune synapse formation on surfaces was achieved on micropatterned surface which mimics the antigen-presenting cells and signaling pathways involved in T-cell activation were probed.[17] The spatial control of EphA2 RTK signal activation during cell–cell contact formation was quantitatively explored by a similar approach [99], revealing a striking correlation between spatial receptor organization and invasion potential of different mammary epithelial cell lines (FIGURE 4D). Further exciting possibilities are opened by combination of these approaches with spatial control of cell adhesion using RGD nanopatterns.[103] 10 5. Conclusions In the past decade, spatially controlled protein immobilization has become a versatile and reliable technology with numerous existing and potential applications in drug discovery. Key challenges for combining biocompatible surface architectures and bioorthogonal coupling reactions with functional micropatterning have been resolved. To this achievement, development of affinity tags and bioconjugation techniques for site-specific protein modification has been instrumental. Thus, maximized protein functionality has been achieved in artificial environments for reliably probing the interactions between proteins and protein substrates. Expert Opin. Drug Discov. (2015) 11(1) Functional protein micropatterning Downloaded by [Universität Osnabrueck], [Jacob Piehler] at 02:57 05 December 2015 The sensitivity of surface-based detection techniques down to the single-molecule level by both fluorescence-based and label-free imaging techniques nowadays allows enormous miniaturization and parallelization for quantitative binding and activity assays. With these tools, the scope of drug discovery is profoundly expanded to reach whole proteomes. Yet, manufacturing of high-density multiplexed protein arrays remains challenging because of the limited stability of proteins. Moreover, membrane proteins as a key class of drug targets are typically not well covered by whole proteome analysis techniques as they require dedicated approaches toward functional immobilization. 6. Expert opinion During the past decade, drug discovery has become a much more demanding and multifaceted process. Key challenges include more efficient target identification, development and testing macromolecular drugs as well as coping with the complex demands of personalized medicine. Robust and user-friendly protein-based assay technologies providing high level of multiplexing and flexible design will play a key role in addressing these challenges. Conventional protein arrays are, meanwhile, commercially available from various suppliers. These are typically designed for proteome-wide screens with the aim to cover a large proportion of a given proteome. For drug discovery, however, more complex, dedicated and adaptable assay formats will be requested for functional analysis of a subset of interesting protein candidates. Implementation of such assays will profit from several major biotechnological innovations, which are currently emerging. One major current limitation in multiplexed protein assay development results from the limited availability of specific binders with well-characterized protein recognition properties. While in the past monoclonal antibodies has been the solution of choice, alternative approaches for generating high-affinity binders are now available, which have the advantage of being much more readily accessible. Next to recombinant antibody fragments, which are directly obtained by in vitro selection, simpler scaffolds such as nanobodies or DARPins have been designed,[104] which can be readily produced by recombinant protein expression or by in vitro translation. As a fully synthetic alternative, aptamer technology has the potential to play an important part in this field.[105] However, considerable and concerted efforts will be required to provide a large and validated repertoire of recombinant or synthetic binders to the scientific community.[106] As a second important technology for site-specific protein attachment, incorporation of non-natural amino acids into recombinant protein is coming of age, being available for different cell types and providing a large variety of possible modifications. After further optimization of the efficiency and selectivity of coupling reactions, this tool will be the key for generic site-specific immobilization and labeling of proteins on very small scale. Functional reconstitution and micropatterning of membrane proteins – the most important class of drug targets – on surfaces has been achieved by membrane architectures such as vesicles and solid-supported membranes, as well as nanodisk technology.[107] These approaches depend on isolation of membrane proteins by detergent solubilization, which requires time-consuming identification of a suitable detergent for a specific membrane protein. As the functions of membrane proteins are inextricably linked to lipid environment [7], appropriate selection of lipids for functional reconstitution is required. Currently, techniques for more direct transfer of membrane proteins to surfaces are emerging, which do not require the extraction from their lipid environment. Promising strategies include budding of proteoliposomes from plasma membranes [89] as well as extraction of membrane proteins within their lipid environment by amphipathic polymers (lipodisq).[108] Successful extraction of GPCRs along with endogenous lipids in lipodisqs was recently demonstrated in a detergent-free manner,[109] highlighting the potentials for a wide application in drug discovery. As a promising alternative strategy, cell-free production of membrane proteins by in vitro translation opens exciting possibility to in situ reconstitute GPCR into suitable membrane architectures.[110] In general, more closely interfacing protein production and array fabrication will be an important trend in this field. Next to generating protein arrays by local in vitro translation of cDNA libraries (cf. Section 4.1), strategies for capturing proteins directly from cells offer promising opportunities. In particular for diagnostic screening required for personalized drug selection, assays need to be performed from minute sample quantities, ideally from a single cell. Single-cell analysis, moreover, opens new avenues to deconvolute the cellular heterogeneity.[111] Cell-dependent variations of stem cell signaling and differentiation responses were examined by large scale (~2000 cells) single-cell western blotting (scWestern) using an open-microwell array.[112] This approach has already been applied to demonstrate the possibility for quantifying cell-to-cell variance to pharmaceutical stimuli.[113] Alternative strategies aim for performing protein assays directly on the cell culture substrate. In case of plasma membrane protein such as receptors and transporters, capturing to surfaces within living cells opens exciting possibilities (cf. Section 4.3.2). In-cell micropatterning of soluble proteins is possible [82], yet requires genetic manipulation. For exploiting the growing availability of high-affinity binders and the very high sensitivity of surface-sensitive detection, single-cell pulldown assays are highly promising, which have been recently demonstrated for recombinant proteins [29] (FIGURE 4E). By taking advantage of efficient cell capturing to surface in combination with local high-density arrays of binders in close proximity to captured cell, this approach allows efficient isolation of proteins from primary cells. Thus, a broad application of ultrahigh-sensitive HTS in the format of single cell– single molecule can be envisioned, which ultimately will fuel drug development for personalized medicine. Expert Opin. Drug Discov. (2015) 11(1) 11 You & Piehler Financial & Competing Interests Disclosure Part of this work was funded by the Deutsche Forschungsgemeinschaft (DFG). J Piehler is the co-inventor of tris-NTA patent EP1778651 entitled “Multivalent chelators for modifying and organizing of target molecules” (WO2005EP08124/DE20041038134/US20040614950P). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. Bibliography Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers. Downloaded by [Universität Osnabrueck], [Jacob Piehler] at 02:57 05 December 2015 1. 2. Mannhold R, Kubinyi H, Folkers G. High-throughput screening in drug discovery. Weinheim: Wiley-VCH Verlag GmbH & Co. KGaA; 2006. MacBeath G, Schreiber SL. Printing proteins as microarrays for high-throughput function determination. Science. 2000;289 (5485):1760–1763. 3. Phizicky E, Bastiaens PIH, Zhu H, et al. Protein analysis on a proteomic scale. Nature. 2003;422(6928):208–215. 4. Bleicher KH, Bohm H-J, Muller K, et al. Hit and lead generation: beyond highthroughput screening. Nat Rev Drug Discov. 2003;2(5):369–378. 5. Overington JP, Al-Lazikani B, Hopkins AL. How many drug targets are there? Nat Rev Drug Discov. 2006;5(12):993– 996. 6. Kimchi-Sarfaty C, Schiller T, HamasakiKatagiri N, et al. Building better drugs: developing and regulating engineered therapeutic proteins. Trends Pharmacol Sci. 2013;34(10):534–548. 7. 8. • Laganowsky A, Reading E, Allison TM, et al. Membrane proteins bind lipids selectively to modulate their structure and function. Nature. 2014;510 (7503):172–175. Kam LC, Shen K, Dustin ML. Micro- and nanoscale engineering of cell signaling. Annu Rev Biomed Eng. 2013;15(1):305– 326. A review highlighting micro- and nanofabrication techniques employed for unraveling spatial control of cellular signaling 9. Valiokas R. Nanobiochips. Cell Mol Life Sci. 2012;69(3):347–356. 10. Mooney JF, Hunt AJ, McIntosh JR, et al. Patterning of functional antibodies and other proteins by photolithography of silane monolayers. Proc Natl Acad Sci USA. 1996;93(22):12287–12291. 12 11. Klenkar G, Valiokas R, Lundström I, et al. Piezo dispensed microarray of multivalent chelating thiols for dissecting complex protein-protein interactions. Anal Chem. 2006;78:3643–3650. 12. Alge DL, Azagarsamy MA, Donohue DF, et al. Synthetically tractable click hydrogels for three-dimensional cell culture formed using tetrazine–norbornene chemistry. Biomacromolecules. 2013;14(4):949–953. • Bio-orthogonal thiol-ene photoreaction and Diels–Alder reaction of tetrazinenorbornene were used for fabricating three-dimensional cell culture hydrogels, a powerful new tool for the development of cell-instructive scaffold for tissue engineering applications. 13. 14. 15. 16. 17. •• Waichman S, You C, Beutel O, et al. Maleimide photolithography for singlemolecule protein−protein interaction analysis in micropatterns. Anal Chem. 2011;83(2):501–508. VandeVondele S, Voros J, Hubbell JA. RGD-grafted poly-L-lysine-graft-(polyethylene glycol) copolymers block nonspecific protein adsorption while promoting cell adhesion. Biotechnol Bioeng. 2003;82(7):784–790. Bhagawati M, You C, Piehler J. Quantitative real-time imaging of protein–protein interactions by LSPR detection with micropatterned gold nanoparticles. Anal Chem. 2013;85 (20):9564–9571. Groves JT, Boxer SG. Micropattern formation in supported lipid membranes. Acc Chem Res. 2002;35(3):149–157. Mossman KD, Campi G, Groves JT, et al. Altered TCR signaling from geometrically repatterned immunological synapses. Science. 2005;310(5751):1191–1193. Formation of artificial immune synapses was explored on functionalized solidsupported membranes mimicking antigen-presenting cells. Using micropatterning, the role of interface geometry was correlated with signaling involved in T-cell activation. Expert Opin. Drug Discov. (2015) 11(1) 18. Tanaka M, Sackmann E. Polymer-supported membranes as models of the cell surface. Nature. 2005;437(7059):656–663. 19. Bailey K, Bally M, Leifert W, et al. Gprotein coupled receptor array technologies: site directed immobilisation of liposomes containing the H1-histamine or M2-muscarinic receptors. Proteomics. 2009;9(8):2052–2063. 20. Bally M, Bailey K, Sugihara K, et al. Liposome and lipid bilayer arrays towards biosensing applications. Small. 2010;6 (22):2481–2497. 21. You C, Bhagawati M, Brecht A, et al. Affinity capturing for targeting proteins into micro and nanostructures. Anal Bioanal Chem. 2009;393(6–7):1563– 1570. 22. Schwarzenbacher M, Kaltenbrunner M, Brameshuber M, et al. Micropatterning for quantitative analysis of protein-protein interactions in living cells. Nat Meth. 2008;5(12):1053–1060. •• Surface micropatterning for capturing plasma membrane bait proteins in living cells was established. Interactions with fluorescence-labeled prey protein with the bait were probed by ensemble and singlemolecule techniques. 23. Wegner GJ, Lee HJ, Marriott G, et al. Fabrication of histidine-tagged fusion protein arrays for surface plasmon resonance imaging studies of protein-protein and protein-DNA interactions. Anal Chem. 2003;75(18):4740–4746. 24. You C, Piehler J. Multivalent chelators for spatially and temporally controlled protein functionalization. Anal Bioanal Chem. 2014;406(14):3345–3357. 25. Waichman S, Bhagawati M, Podoplelova Y, et al. Functional immobilization and patterning of proteins by an enzymatic transfer reaction. Anal Chem. 2010;82 (4):1478–1485. 26. Ito T, Sadamoto R, Naruchi K, et al. Highly oriented recombinant glycosyltransferases: site-specific immobilization of unstable membrane proteins by using Functional protein micropatterning Staphylococcus aureus Sortase A. Biochemistry. 2010;49(11):2604–2614. 27. Downloaded by [Universität Osnabrueck], [Jacob Piehler] at 02:57 05 December 2015 28. Taniguchi Y, Kawakami M. Application of halotag protein to covalent immobilization of recombinant proteins for single molecule force spectroscopy. Langmuir. 2010;26(13):10433–10436. 36. Yoshitani N, Saito K, Saikawa W, et al. NTA-mediated protein capturing strategy in screening experiments for small organic molecules by surface plasmon resonance. Proteomics. 2007;7(4):494–499. 37. Engin S, Trouillet V, Franz CM, et al. Benzylguanine thiol self-assembled monolayers for the immobilization of snap-tag proteins on microcontact-printed surface structures. Langmuir. 2010;26(9):6097– 6101. Banala S, Arnold A, Johnsson K. Caged substrates for protein labeling and immobilization. Chembiochem. 2008;9(1):38– 41. 38. 29. Wedeking T, Lochte S, Richter CP, et al. Single cell GFP-trap reveals stoichiometry and dynamics of cytosolic protein complexes. Nano Lett. 2015;15(5):3610–3615. • Micropatterned surface functionalization with an anti-GFP nanobody enabled for probing individual protein complexes captured protein complexes from single cells. 39. Chen Y-X, Triola G, Waldmann H. Bioorthogonal chemistry for site-specific labeling and surface immobilization of proteins. Acc Chem Res. 2011;44(9):762– 773. 40. 30. 31. 32. 33. 34. • 35. Lofas S, Johnsson B. A novel hydrogel matrix on gold surfaces in surface-plasmon resonance sensors for fast and efficient covalent immobilization of ligands. J Chem Soc-Chem Commun. 1990;21:1526–1528. Piehler J, Brecht A, Valiokas R, et al. A high-density poly(ethylene glycol) polymer brush for immobilization on glass-type surfaces. Biosens Bioelectron. 2000;15(9– 10):473–481. Mrksich M, Whitesides GM. Using selfassembled monolayers to understand the interactions of man-made surfaces with proteins and cells. Annu Rev Biophys Biomol Struct. 1996;25:55–78. Zhu H, Bilgin M, Bangham R, et al. Global analysis of protein activities using proteome chips. Science. 2001;293 (5537):2101–2105. The first report of protein activity analysis on genome-wide scale, in which site-specific protein immobilization was demonstrated to be superior over uncontrolled protein attachment. Kim CH, Axup JY, Schultz PG. Protein conjugation with genetically encoded unnatural amino acids. Curr Opin Chem Biol. 2013;17(3):412–419. Ludden MJ, Li X, Greve J, et al. Assembly of bionanostructures onto beta-cyclodextrin molecular printboards for antibody recognition and lymphocyte cell counting. J Am Chem Soc. 2008;130(22):6964– 6973. Arrabito G, Reisewitz S, Dehmelt L, et al. Biochips for cell biology by combined dippen nanolithography and DNA-directed protein immobilization. Small. 2013;9 (24):4243–4249. Lohmüller T, Aydin D, Schwieder M, et al. Nanopatterning by block copolymer micelle nanolithography and bioinspired applications. Biointerphases. 2011;6(1): MR1–MR12. 41. Zhou X, Boey F, Huo F, et al. Chemically functionalized surface patterning. Small. 2011;7(16):2273–2289. 42. Park JU, Hardy M, Kang SJ, et al. Highresolution electrohydrodynamic jet printing. Nat Mater. 2007;6(10):782–789. 43. Xia Y, Whitesides GM. Soft lithography. Angew Chem Int Ed. 1998;37(5):550–575. 44. Whitesides GM, Ostuni E, Takayama S, et al. Soft lithography in biology and biochemistry. Annu Rev Biomed Eng. 2001;3:335–373. 45. Voskuhl J, Brinkmann J, Jonkheijm P. Advances in contact printing technologies of carbohydrate, peptide and protein arrays. Curr Opin Chem Biol. 2014;18:1–7. 46. Park JP, Lee SJ, Park TJ, et al. Microcontact printing of biotin for selective immobilization of streptavidin-fused proteins and SPR analysis. Biotechnol Bioprocess Eng. 2004;9(2):137–142. 47. 48. Ricoult SG, Pla-Roca M, Safavieh R, et al. Large dynamic range digital nanodot gradients of biomolecules made by low-cost nanocontact printing for cell haptotaxis. Small. 2013;9(19):3308–3313. Piner RD, Zhu J, Xu F, et al. “Dip-Pen” nanolithography. Science. 1999;283 (5402):661–663. Expert Opin. Drug Discov. (2015) 11(1) 49. Rakickas T, Gavutis M, Reichel A, et al. Protein-protein interactions in reversibly assembled nanopatterns. Nano Lett. 2008;8(10):3369–3375. 50. Brinkmann F, Hirtz M, Greiner AM, et al. Interdigitated multicolored bioink micropatterns by multiplexed polymer pen lithography. Small. 2013;9 (19):3266–3275. • Polymer pen lithography for multiplexed protein deposition of complex features was employed for studying the interaction of fibroblasts and neurons with micropatterned functionalized surfaces. 51. Lee K-B, Park S-J, Mirkin CA, et al. Protein nanoarrays generated by dip-pen nanolithography. Science. 2002;295 (5560):1702–1705. •• Dip-pen nanolithography was used to fabricate protein arrays with down to 100 nm features. These nanoarrays exhibited almost no detectable nonspecific binding of proteins in complex mixtures of proteins. 52. Chai J, Wong LS, Giam L, et al. Singlemolecule protein arrays enabled by scanning probe block copolymer lithography. Proc Natl Acad Sci. 2011;108(49):19521– 19525. 53. Tinazli A, Piehler J, Beuttler M, et al. Native protein nanolithography that can write, read and erase. Nat Nanotechnol. 2007;2(4):220–225. 54. Lee SW, Oh BK, Sanedrin RG, et al. Biologically active protein nanoarrays generated using parallel dip-pen nanolithography. Advanced Mater. 2006;18(9):1133– 1136. 55. Huang M, Galarreta BC, Artar A, et al. Reusable nanostencils for creating multiple biofunctional molecular nanopatterns on polymer substrate. Nano Lett. 2012;12 (9):4817–4822. 56. Sekula S, Fuchs J, Weg-Remers S, et al. Multiplexed lipid dip-pen nanolithography on subcellular scales for the templating of functional proteins and cell culture. Small. 2008;4(10):1785–1793. 57. Huo F, Zheng Z, Zheng G, et al. Polymer pen lithography. Science. 2008;321 (5896):1658–1660. • Polymer pen lithography for large-scale functional surface micropatterning is described, combining the advantages of dip-pen lithography and microcontact printing. 13 Downloaded by [Universität Osnabrueck], [Jacob Piehler] at 02:57 05 December 2015 You & Piehler 58. Zheng Z, Daniel WL, Giam LR, et al. Multiplexed protein arrays enabled by polymer pen lithography: addressing the inking challenge. Angew Chem Int Ed. 2009;48(41):7626–7629. 59. Li L, Gattass RR, Gershgoren E, et al. Achieving λ/20 resolution by one-color initiation and deactivation of polymerization. Science. 2009;324(5929):910–913. 60. Wiesbauer M, Wollhofen R, Vasic B, et al. Nano-anchors with single protein capacity produced with STED lithography. Nano Lett. 2013;13(11):5672–5678. 61. Bhagawati M, Ghosh S, Reichel A, et al. Organization of motor proteins into functional micropatterns fabricated by a photoinduced Fenton reaction. Angew Chem Int Ed Engl. 2009;48(48):9188–9191. 62. • 63. 64. 65. Waichman S, Roder F, Richter CP, et al. Diffusion and interaction dynamics of individual membrane protein complexes confined in micropatterned polymer-supported membranes. Small. 2013;9(4):570– 577. Photolithography via maleimide-vinylether polymerization was used to construct biocompatible surface micropatterning with ultralow fluorescence background. This method was successfully employed for reconstituting transmembrane receptors into micropatterned polymer-supported membranes for single-molecule studies. Carrico IS, Maskarinec SA, Heilshorn SC, et al. Lithographic patterning of photoreactive cell-adhesive proteins. J Am Chem Soc. 2007;129(16):4874–4875. El Muslemany KM, Twite AA, ElSohly AM, et al. Photoactivated bioconjugation between ortho-azidophenols and anilines: a facile approach to biomolecular photopatterning. J Am Chem Soc. 2014;136 (36):12600–12606. Ryan D, Parviz BA, Linder V, et al. Patterning multiple aligned self-assembled monolayers using light. Langmuir. 2004;20(21):9080–9088. 66. Alonso JM, Reichel A, Piehler J, et al. Photopatterned surfaces for site-specific and functional immobilization of proteins. Langmuir. 2008;24(2):448–457. 67. Bhagawati M, Lata S, Tampé R, et al. Native laser lithography of His-tagged proteins by uncaging of multivalent chelators. J Am Chem Soc. 2010;132 (17):5932–5933. 14 68. Gatterdam V, Stoess T, Menge C, et al. Caged glutathione - triggering protein interaction by light. Angew Chem Int Ed Engl. 2012;51(16):3960–3963. 69. Jonkheijm P, Weinrich D, Kohn M, et al. Photochemical surface patterning by the thiol-ene reaction. Angew Chem Int Ed Engl. 2008;47(23):4421–4424. 70. Turchanin A, Tinazli A, El-Desawy M, et al. Molecular self-assembly, chemical lithography, and biochemical tweezers: A path for the fabrication of functional nanometer-scale protein arrays. Adv Mater. 2008;20(3):471–477. 71. Christman KL, Schopf E, Broyer RM, et al. Positioning multiple proteins at the nanoscale with electron beam cross-linked functional polymers. J Am Chem Soc. 2009;131(2):521–527. 72. Schlapak R, Danzberger J, Haselgrubler T, et al. Painting with biomolecules at the nanoscale: biofunctionalization with tunable surface densities. Nano Lett. 2012;12 (4):1983–1989. 81. Ramachandran N, Raphael JV, Hainsworth E, et al. Next-generation high-density self-assembling functional protein arrays. Nat Methods. 2008;5 (6):535–538. 82. Gandor S, Reisewitz S, Venkatachalapathy M, et al. A protein-interaction array inside a living cell. Angew Chem Int Ed. 2013;52(18):4790–4794. • Multiplexed antibody arrays fabricated by dip-pen nanolithography were employed for monitoring protein interactions during signal activation in living cells. 83. Ezer U. G-protein coupled receptor (GPCR) targeting: technologies and global markets [Internet]. 2014. [cited 2014 Aug]. Available from: http://www.bccre search.com/market-research/biotechnol ogy/GPCR-targeting-technology-bio136a. html 84. Kenakin T. Efficacy at g-protein-coupled receptors. Nat Rev Drug Discov. 2002;1 (2):103–110. 73. Yu X, Schneiderhan-Marra N, Joos TO. Protein microarrays for personalized medicine. Clin Chem. 2010;56(3):376–387. 85. Zhang R, Xie X. Tools for GPCR drug discovery. Acta Pharmacol Sin. 2012;33 (3):372–384. 74. Tu S, Jiang HW, Liu CX, et al. Protein microarrays for studies of drug mechanisms and biomarker discovery in the era of systems biology. Curr Pharm Des. 2014;20(1):49–55. 86. Kelly C, Craighead H. Nanofabrication for the analysis and manipulation of membranes. Ann Biomed Eng. 2012;40 (6):1356–1366. 87. 75. Sutandy FX, Qian J, Chen CS, et al. Overview of protein microarrays. Curr Protoc Protein Sci. 2013;Chapter 27: Unit 27 1. Fang Y, Frutos AG, Lahiri J. Membrane protein microarrays. J Am Chem Soc. 2002;124(11):2394–2395. 88. Posner B, Hong Y, Benvenuti E, et al. Multiplexing G protein-coupled receptors in microarrays: a radioligand-binding assay. Anal Biochem. 2007;365 (2):266–273. 76. Mok J, Im H, Snyder M. Global identification of protein kinase substrates by protein microarray analysis. Nat Protoc. 2009;4(12):1820–1827. 89. 77. Breitkreutz A, Choi H, Sharom JR, et al. A global protein kinase and phosphatase interaction network in yeast. Science. 2010;328(5981):1043–1046. Stamou D, Duschl C, Delamarche E, et al. Self-assembled microarrays of attoliter molecular vessels. Angew Chem Int Ed. 2003;42(45):5580–5583. 90. 78. Howorka S, Hesse J. Microarrays and single molecules: an exciting combination. Soft Matter. 2014;10(7):931–941. Martins SAM, Trabuco JRC, Monteiro GA, et al. Towards the miniaturization of GPCR-based live-cell screening assays. Trends Biotechnol. 2012;30(11):566–574. 79. Li T, Liu X, Liu D, et al. Sensitive detection of protein kinase A activity in cell lysates by peptide microarray-based assay. Anal Chem. 2013;85(15):7033–7037. 91. Persidis A. Signal transduction as a drugdiscovery platform. Nat Biotech. 1998;16 (11):1082–1083. 92. 80. Bhide M, Natarajan S, Hresko S, et al. Rapid in vitro protein synthesis pipeline: a promising tool for cost-effective protein array design. Mol Biosyst. 2014;10 (6):1236–1245. Yao H, Ashihara E, Maekawa T. Targeting the Wnt/beta-catenin signaling pathway in human cancers. Expert Opin Ther Targets. 2011;15(7):873–887. 93. Quist AP, Oscarsson S. Micropatterned surfaces: techniques and applications in Expert Opin. Drug Discov. (2015) 11(1) Functional protein micropatterning cell biology. Expert Opin Drug Discov. 2010;5(6):569–581. Downloaded by [Universität Osnabrueck], [Jacob Piehler] at 02:57 05 December 2015 94. Arnold M, Hirschfeld-Warneken VC, Lohmüller T, et al. Induction of cell polarization and migration by a gradient of nanoscale variations in adhesive ligand spacing. Nano Lett. 2008;8(7):2063– 2069. 95. Chen CS, Mrksich M, Huang S, et al. Geometric control of cell life and death. Science. 1997;276(5317):1425–1428. 96. Wang X, Li S, Yan C, et al. Fabrication of RGD micro/nanopattern and corresponding study of stem cell differentiation. Nano Lett. 2015;15(3):1457–1467. 97. 98. • 99. 100. Wan L, Ronaldson K, Park M, et al. From the cover: micropatterned mammalian cells exhibit phenotype-specific left-right asymmetry. Proc Natl Acad Sci USA. 2011;108:12295–12300. Lochte S, Waichman S, Beutel O, et al. Live cell micropatterning reveals the dynamics of signaling complexes at the plasma membrane. J Cell Biol. 2014;207 (3):407–418. Tag-specific live-cell micropatterning for quantitative analyses of protein interactions involved in receptor assembly and effector recruitment was established. Salaita K, Nair PM, Petit RS, et al. Restriction of receptor movement alters cellular response: physical force sensing by EphA2. Science. 2010;327 (5971):1380–1385. Takeuchi K, Ito F. Receptor tyrosine kinases and targeted cancer therapeutics. Biol Pharm Bull. 2011;34(12):1774–1780. 101. Casaletto JB, McClatchey AI. Spatial regulation of receptor tyrosine kinases in development and cancer. Nat Rev Cancer. 2012;12(6):387–400. 102. Lanzerstorfer P, Borgmann D, Schutz G, et al. Quantification and kinetic analysis of Grb2-EGFR interaction on micro-patterned surfaces for the characterization of EGFR-modulating substances. PLoS One. 2014;9(3):e92151. 103. 104. 105. 106. 107. 108. Lohmuller T, Triffo S, O’Donoghue GP, et al. Supported membranes embedded with fixed arrays of gold nanoparticles. Nano Lett. 2011;11(11):4912–4918. Helma J, Cardoso MC, Muyldermans S, et al. Nanobodies and recombinant binders in cell biology. J Cell Biol. 2015;209 (5):633–644. Thiviyanathan V, Gorenstein DG. Aptamers and the next generation of diagnostic reagents. Proteomics Clin Appl. 2012;6(11–12):563–573. Bradbury A, Pluckthun A. Reproducibility: standardize antibodies used in research. Nature. 2015;518 (7537):27–29. Nath A, Atkins WM, Sligar SG. Applications of phospholipid bilayer nanodiscs in the study of membranes and membrane proteins. Biochemistry. 2007;46(8):2059–2069. Orwick-Rydmark M, Lovett JE, Graziadei A, et al. Detergent-free incorporation of a seven-transmembrane receptor protein into nanosized bilayer lipodisq particles for functional and biophysical studies. Nano Lett. 2012;12(9):4687–4692. Expert Opin. Drug Discov. (2015) 11(1) • Styrene-maleic anhydride lipodisq nanoparticles with an average of one bacteriorhodopsin per particle were prepared without the use of detergents, demonstrating the ability to directly extract membrane proteins together with their native lipid environments. 109. Jamshad M, Charlton J, Lin YP, et al. Gprotein coupled receptor solubilization and purification for biophysical analysis and functional studies, in the total absence of detergent. Biosci Rep. 2015;35(2):e00188. 110. Orban E, Proverbio D, Haberstock S, et al. Cell-free expression of G-protein-coupled receptors. Methods Mol Biol. 2015;1261:171–195. 111. Tsioris K, Torres AJ, Douce TB, et al. A new toolbox for assessing single cells. Annu Rev Chem Biomol Eng. 2014;5 (1):455–477. 112. Hughes AJ, Spelke DP, Xu Z, et al. Singlecell western blotting. Nat Methods. 2014;11(7):749–755. 113. Kang CC, Lin JM, Xu Z, et al. Single-cell Western blotting after whole-cell imaging to assess cancer chemotherapeutic response. Anal Chem. 2014;86 (20):10429–10436. Affiliation Changjiang You & Jacob Piehler† † Author for correspondence Department of Biology, Division of Biophysics, University of Osnabrück, Osnabrück 49076, Germany E-mail: [email protected] 15