Grading The Commercial Optical Biosensor Literature-Class of 2008: The Mighty Binders'
Grading The Commercial Optical Biosensor Literature-Class of 2008: The Mighty Binders'
Grading The Commercial Optical Biosensor Literature-Class of 2008: The Mighty Binders'
Received: 14 October 2009, Accepted: 19 October 2009, Published online in Wiley InterScience: 2009
(www.interscience.wiley.com) DOI:10.1002/jmr.1004
Grading the commercial optical biosensor literatureClass of 2008: The Mighty Binders
Rebecca L. Richa and David G. Myszkaa *
Optical biosensor technology continues to be the method of choice for label-free, real-time interaction analysis. But when it comes to improving the quality of the biosensor literature, education should be fundamental. Of the 1413 articles published in 2008, less than 30% would pass the requirements for high-school chemistry. To teach by example, we spotlight 10 papers that illustrate how to implement the technology properly. Then we grade every paper published in 2008 on a scale from A to F and outline what features make a biosensor article fabulous, middling or abysmal. To help improve the quality of published data, we focus on a few experimental, analysis and presentation mistakes that are alarmingly common. With the literature as a guide, we want to ensure that no user is left behind. Copyright 2009 John Wiley & Sons, Ltd. Keywords: afnity; Biacore; biomolecular interaction analysis; evanescent wave; kinetics; resonant mirror; surface plasmon resonance
Whats that old saying?oh yeah, something like If you give a man a sh, he can eat for a day. But if you teach a man to do a biosensor experiment correctly, he can buy all the damn sh he wants. For years we have taken an active approach to educating users, as well as non-users, about how to properly design, execute and analyse biosensor experiments. Unfortunately, based on our annual review of the literature we still have a lot of work to do. Only a minority of users know how to run even basic experiments correctly. And based on whats published, there is still a general perception that whatever data comes out of the biosensor must be biologically relevant and should be published. Nothing could be farther from the truth. Depressingly, and in some ways frighteningly, the biosensor user community is not the only scientic eld in the need of some reform, regulation and education. Take as an example the recent report from the National Academy of Sciences on the state of forensic science: Badly Fragmented Forensic Science System Needs Overhaul. 19 February 2009A US congressionally mandated report from the National Research Council nds serious deciencies in the nations forensic science system and calls for major reforms. Mandatory certication programs for forensic scientists are currently lacking, as are strong standards and protocols for analysing and reporting on evidence. Many professionals in the forensic science community and the medical examiner system have worked for years to achieve excellence in their elds, aiming to follow high ethical norms, develop sound professional standards and ensure accurate results in their practice. But there are great disparities among existing forensic science operations. The disparities appear in availability of skilled and well-trained personnel; and in certication, accreditation and oversight. This has left the forensic science system fragmented and the quality of practice uneven. These shortcomings pose a threat to the quality and credibility of forensic science practice and its service to the justice system.
Replace the words forensic and justice system with biosensor and biological sciences and you would have a pretty accurate depiction of what our user base suffers from as well. But until one (or both) of us is (are) elected to the National Academy of Sciences (which seems less likely now than ever after writing this review), we gure we will continue our role of educating a eld which is in desperate need of standardization. So grab your pole and put on some sunblock because youre about get schooled on the Moby Dick, 20 000 Leagues Under the Sea, Finding Nemo biosensor adventure of a lifetime. Youre gonna need a bigger boat.
ROLL CALL
First lets get everyone on the same page by summarizing the years literature. We found 1413 articles published in 2008 that describe biosensor-based experiments. In the reference list, this collection is divided into several sections: books, editorials and general reviews about biosensors (123); theory, methods development and instrument comparisons (2442) and instrument- and application-specic articles (431413). A review article by Comley is particularly worth reading because it summarized the results from recent interviews conducted as part of a biosensor market report (5). He compiled the surveys nding about interviewees familiarity with biosensor
* Correspondence to: D. G. Myszka, Center for Biomolecular Interaction Analysis, University of Utah, School of Medicine 4A417, 50 N. Medical Drive, Salt Lake City, UT 84132, USA. E-mail: [email protected] a R. L. Rich, D. G. Myszka Center for Biomolecular Interaction Analysis, University of Utah, Salt Lake City, UT, USA
R. L. RICH AND D. G. MYSZKA manufacturers and their products, user concerns about the different instruments, the biological systems examined and the type of information sought by users, and the technologys perceived challenges (i.e. factors that prevent it from becoming more widely adopted). In addition, Comley provides snapshots of the various label-free vendors and the platforms each offers. It seems like every year someone writes a general article describing surface plasmon resonance (SPR) experiments. This year it was De Crescenzo et al.s turn (6). They reviewed the physics of SPR, described Biacores dextran layer, outlined methods to reduce experimental artefacts (e.g. buffer preparation, ligand immobilization methods and interaction stoichiometry issues) and discussed the basics of kinetic analysis, including important processing steps and considerations during model tting. As illustrated in References 2937, several biosensor users continue to push the envelope in assay optimization and technology development. A number of researchers reported new approaches to tethering ligands to sensor surfaces (29,30,3235). For example, Gale and collaborators (admittedly, we are some of them) developed a continuous-ow microspotter to be used with SPR imaging instruments (3235). Unlike traditional pin-spotting techniques, this in-solution spotter does not require pure, highly concentrated ligand solutions nor does the ligand surface need to be dried after immobilization. We expect this approach will resolve many of the concerns surrounding array platforms and make these instruments useful for a wide range of applications. And, as the number and variety of optical biosensors increases, it can be daunting to understand the benets of the different platforms. Therefore, reports of comparative studies (like References 3842) are informative articles for the general biosensor community. The remainder of the reference list relates to specic instruments or applications. We found articles describing work performed using instruments offered by 30 manufacturers (Table 1). These instruments employ SPR (in either standard (431338) or array formats (13391391)) or alternative optical detection methods (13921413). The reference list is further divided by manufacturer and, since the number of Biacore references is so large, these articles are grouped by biological application. In reviewing this collection, we are encouraged by authors who put a lot of work into their experiments. For example, several research groups used multiple immobilization chemistries (e.g. References 372,947) and prepared surfaces of different ligand densities (267,452), tested their interactions in both orientations (593) and used different assay formats (101), and carefully examined the responses obtained from the reference surface (1014). Notably, Bravman et al. emphasized the care required when choosing the pH for immobilization (1341). This group nicely demonstrated that the pH which produced the highest surface density was in fact not necessarily optimal for ligand activity. Call us old school, but we think it never hurts to be reminded about the fundamentals of a good biosensor experiment. The 10 articles, summarized in the box below, describe signicant developments in biosensor applications, report wellperformed, in-depth experiments and include easy-to-interpret gures of data, and/or provide insights about where this technology is heading with respect to improved throughput and sensitivity. Consider each of these papers required reading. Advancing Applications Giannetti J Med Chem 51:574. Chavane Anal Biochem 378:158. Christensen J Aller Clin Immun 122:298. Rigorous Analyses Souphron Biochemistry 47:8961. Kalyuzhniy Proc Natl Acad Sci USA 105:5483. Wang J Virol 82:10567. Technology Validation Abdiche Prot Sci 17:1326. Abdiche Anal Biochem 377:209. Beusink Biosens Bioelectron 23:839. Rich Anal Biochem 373:112.
HONOR ROLL
The 10 articles we chose to spotlight this year exemplify the power of biosensor technology and the tenacity of some great users. These are the summa cum laudes of SPR; they give us hope that perhaps in the not-so-distant future everyone will produce high-quality biosensor data.
Giannetti et al. Surface plasmon resonance based assay for the detection and characterization of promiscuous inhibitors. J Med Chem 51: 574580. Giannetti et al.s detailed characterization of promiscuously binding compounds emphasizes how biosensors can impact both rigorous small-molecule kinetic analyses and higherthroughput screening assays (923). As these researchers noted, too often compounds that appear promising in biochemical screens are in fact false positives and biosensor analyses help weed out these non-specic binders. For example, Figure 1A shows the responses for a positivecontrol compound binding to an immobilized target. This well-behaved compound produced concentration-dependent responses that could be described by a simple interaction model. These data conrmed the target was active on the chip surface. Not all compounds, however, produce such easily interpretable responses. For example, non-stoichiometric compounds produce responses that are larger than the expected Rmax (Figure 1B top left panels) while superstoichiometric compounds display responses of more than ve times the Rmax, often without any indication of surface saturation (Figure 1B bottom left panels). The right panels of Figure 1B depict examples of compounds that are well-behaved at lower concentrations but then suddenly show unexpectedly large responses at slightly higher (concentration-dependent aggregators) and compounds that become well-behaved in the presence of detergent. But, as these researchers demonstrated, testing a compound library against only one target is not enough to determine which compounds to ag as bad binders. Some compounds behave differently depending on the target. Figure 1C shows the responses for one compound tested against seven targets and an unmodied surface. To some targets this compound displayed little or no binding while it showed intermediate but nonstoichiometric binding or superstoichiometric binding to other
2
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
Table 1. Commercial optical biosensor technologies Manufacturer Analytical m-systems Biosensing Instruments DKK-TOA EcoChemie/Autolab GE Healthcare/Biacore Instruments Website References (4352) (5359) (41,6062) (6389) (2931,3639,901230)
IBIS K-MAC Microvacuum/Articial Sensing Moritex/Nippon Laser & Electronics Nanolm Technology NanoSPR NeoSensors Ltd. Nomadics/Texas Instruments NTT-AT Optrel Plasmonic Biosensor Reichert Analytical Instruments/Xantec Resonant Probes Thermo Scientic Alphasniffer Bio-Rad GE Healthcare/Biacore GWC Technologies/Toyobo Horiba/Genoptics IBIS Plexera Corning Fareld Sensors ForteBio Maven Biotechnologies Silicon Kinetics SRU Biosystems
Standard SPR platforms Biosuplar, Biosuplar-2, micro-systems.de -3, -6 BI-2000, -2000G, -3000, biosensingusa.com -3000G SPR-20 dkktoa.co.uk Esprit, Springle ecochemie.nl biacore.com Biacore, Bialite, C, J, Q, X, 1000, 2000, 3000, S51, T100, A100, X100 IBIS II ibis-spr.n SPR LAB, SPRmicro www.kmac.to OWLS 110, OWLS 120 owls-sensors.com SPR 670 M, SPR-Cellia EP3SPR NanoSPR 310, 321, 621 IAsys, IAsys Auto SensiQ, Spreeta Handy SPR PS-0109 Multiskop Plasmonic Multichannel SR7000, SR7000DC SPTM SPR-1000 moritex.co.jp nanolm.de nanospr.com neosensors.com discoversensiq.com ntt-at.com optrel.de plasmonic.de reichertspr.com resonant-probes.de thermo.com
(1231) (12321236) (40,42,12371247,1407) (41,12481258) (1143,1259,1260) (12611264) (884,12651294) (12951308) (1139,13091311) (13121324) (1325) (13261332) (13331337) (1338) (1339) (38,39,13401357) (3235,13581362) (13631382) (13831388) (1389,1390) (1391) (40,13921395) (1273,13961408) (38,39,1409) (1410) (1411) (40,42,1412,1413)
Imaging SPR platforms BiOptix alphasniffer.com ProteOn XPR36 bio-rad.com Flexchip biacore.com SPRimager II, SPR 100, gwctechnologies.com MultiSPRinter toyobo.co.jp SPRi-Plex, SPRi-Lab genoptics-spr.com iSPR ibis-spr.n PlexArray plexera.com Non-SPR platforms Epic corning.com Analight Bio200, BioFlex, fareld-group.com CrystaLight, 4D Octet Q, QK, Red, fortebio.com Red384, QK384 LFIRE mavenbiotech.com SkiPro siliconkinetics.com BIND srubiosystems.com
targets. These data sets emphasize the need to check for binding to off targets and control/reference surfaces when screening compound libraries. Giannetti et al.s article serves two essential purposes. First, it outlines a systematic approach to identifying and classifying promiscuous binders. Second, it presents a number of data sets from poorly behaved compounds. Too often users think responses like these are good data. It is important for all users to recognize how data from promiscuous binders may look and to realize that these responses may reveal information about
stoichiometry, reversibility and changes in compound behaviour over a range of concentrations. Now while it may be frustrating to chemists to hear that their exciting leads are junk, a major advantage of the biosensor is that it can conrm real hits and identify false positives. The sooner false leads are abandoned, the better for the project overall. Work like Giannetti et al.s is particularly important as biosensor technology is increasingly used in fragment screening, where weak-afnity (but less well-characterized) compounds are screened at fairly high concentrations and in relatively high
3
www.interscience.wiley.com/journal/jmr
Figure 1. Responses for well- and poorly behaved compounds. (A) Responses of a well-behaved compound can be described by a simple interaction model (red lines). The highest concentration tested, number of replicates and binding parameters determined from the t are shown as insets. (B) Classes of poorly behaved compounds. In each panel, the expected Rmax is indicated by the red line. (In the gure of concentration-dependent aggregation responses, the insets show responses that are more like what would be expected for each compound.) (C) Protein-specic promiscuity of one compound. Adapted from Reference 923 with permission from the American Chemical Society 2008.
throughput. The ability to efciently pick out both true binders and false positives is essential when reviewing the massive amounts of data generated in these screens. And although Giannetti et al. focused on small molecules, these types of non-ideal responses are observed in all types of biological interactions. Weve added these terms for the different classes of promiscuous binders to our biosensor vocabulary list (see the end of this paper). Chavane et al. At-line quantication of bioactive antibody in bioreactor by surface plasmon resonance using epitope detection. Anal Biochem 378: 158165. Chavane et al.s report of a fully automated method to track antibody production highlights yet another unique application of biosensors (1212). This group devised a hands-off, at-line set-up (shown in Figure 2A) that tested crude medium from a bioreactor
every 10 h to quantitate the concentration of active antibody produced over 10 days. Their biosensor approach has several advantages over more traditional production-tracking methods. Standard ELISA-like assays are labour- and sample-intensive, uorescent assays require tags to engineered in and cleaved off and mass spectrometry assays measure the total production level but provide no information about activity. In contrast, the biosensor assay is label free, uses very little material and reveals both the amount and activity of the bioreactor product. But maybe best of all, its automation means the system can run unattended, even over Spring Break! It should be noted that Chavane et al. rst put a lot of effort into optimizing the assay off line. They needed to identify a suitable binding partner, appropriate immobilization chemistry and
4
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
Figure 2. At-line biosensor-based assay to track bioreactions. (A) Schematic of automated monitoring of antibody production. A pump continuously delivered ltered culture medium to s sampling vial in the biosensor. At set intervals, an aliquot of medium was diluted into running buffer using the Biacore 3000s sample transfer/mix functions and then injected across the sensor surface. The initial binding rate (measured at 90120 s) in each sensorgram was used to calculate antibody concentration. (B) Off-line assay optimization. Ten replicate antibody injections across immobilized (i) antigen peptide and (ii) full-length antigen. Corresponding responses from mock surfaces are included in each panel. (C) Standard curve generation. Left: responses for a concentration series of puried antibody. Included are replicates for antibody from crude medium (highlighted by the asterisk). Right: calibration plot derived from the responses shown. (D) Trend plot of the responses for a standard antibody sample measured throughout the analysis. Adapted from Reference 1212 with permission from Elsevier Inc. 2008.
regeneration solution and conditions that produced a mass transport-limited interaction. Figure 2B shows the responses for puried antibody binding to immobilized antigen (both fulllength and peptide), as well as to mock surfaces. The low responses from the mock surfaces indicated the antibody bound specically to the antigen surfaces. In addition, the overlay of 10 replicate antibody tests demonstrated the stability of both binding partners and the suitability of the regeneration condition. To induce mass transport-limited conditions that produce a linear association phase, Chavane et al. intentionally prepared high-density surfaces and used slow ow rates. They chose to use the peptide surface in follow-up experiments because it produced large, linear signals from which they could determine the initial binding rate, which correlates with antibody production level (Figure 2B, left panel).
Figure 2C shows the calibration data used to quantitate the bioproduct. This gure also includes a set of replicates from a crude antibody sample (highlighted by the asterisk). The similar shapes of the responses from puried antibody and the culture medium conrmed that other components in the medium did not bind to the ligand surface. Figure 2D depicts the ageing of the ligand surface throughout the repeated binding/regeneration cycles over the 10 days of antibody culture. With this activity-monitoring information, Chavane et al. realized they needed to account for the surfaces gradual loss in activity over time when calculating the bioreactions output. Two things make this paper required reading. First, the authors did a great job describing how the biosensor can automatically track culture growth over several days. Second (and maybe even
5
www.interscience.wiley.com/journal/jmr
R. L. RICH AND D. G. MYSZKA more important), several of the elements of their assay can be applied to biosensor-based concentration determination experiments in general. Christensen et al. Several distinct properties of the IgE repertoire determine effector cell degranulation in response to allergen challenge. Allergy Clin Immun 122: 298304. Taking advantage of the biosensors exibility in assay design, Christensen et al. used both epitope mapping and kinetic assays to characterize a panel of 31 IgEs developed against major house dust mite allergen (Der p2) (358) (it might be mites). With a two-stage epitope-mapping process, they investigated the antigen/antibody binding interface. First, they tested the ability of pairs of antibodies to bind Der p2 simultaneously (assay design illustrated in the top left panel of Figure 3A). If both antibodies bound, these researchers knew that the clones bound at separate sites in Der p2 (Figure 3A, top middle panel). If the second clone did not bind (e.g. Figure 3A, top right panel), they discovered the two clones bound at an overlapping site in Der p2. Then, using a related assay design (Figure 3A, bottom left panel), Christensen et al. screened wild type or mutant rDer p2 for binding to captured clones to identify residues in Der p2 critical for the Ab/Ag interaction (Figure 3A, bottom right panel). The pair-wise mapping revealed seven distinct antibody epitopes in Der p2 (Figure 3B, left panel) and the characterization of the variants indicated nine different binding patterns (Figure 3B, middle panel). Combining the information from these two complementary mapping experiments produced a threedimensional model of the IgE epitopes in rDer p2 (Figure 3B, right panel). Following up on the mapping experiments, Christensen et al. determined kinetic parameters for the various antigen/antibody pairs. The clones displayed a range of kinetic proles (examples are shown in the top panels of Figure 3C), varied in their rDer p2 afnities by almost 30 000 fold, and were binned as high-, medium- or low-afnity binders (Figure 3C, bottom panel). Christensen et al.s coupled epitope mapping and kinetic approach illustrates the wealth of information that can be obtained using biosensors. In addition, their work incorporated features we look for in smart assay design when studying antibodies: (1) to avoid avidity effects, the antibodies were tethered to the surface rather than tested in solution and (2) by capturing the antibodies (rather than direct immobilization), the surface could be regenerated between binding cycles, thereby allowing different antibodies to be captured on the same sensor surface. Finally, this paper demonstrates the biosensors efciency. Most biosensors are automated, including the Biacore 3000 used by Christensen et al., and can test hundreds of clones in a single experiment. Souphron et al. Structural dissection of a gating mechanism preventing misactivation of ubiquitin by NEDD8s E1. Biochemistry 47: 89618969. Figure 4 depicts Souphron et al.s examination of wild type and mutant NEDDs binding to ubiquitin and related proteins (279). These data sets are excellent examples of what we look for in an equilibrium analysis. Most importantly, in each panel the responses for every analyte concentration reach a plateau during the association phase. (This plateau indicates the interaction did in fact reach equilibrium and it was therefore appropriate to perform an equilibrium analysis.) Additionally, these researchers intentionally produced ligand densities that yielded relatively low analyte binding responses (Rmax $50 RU) and tested replicate analyte samples to demonstrate the reproducibility of their experiments. And nally, the data shown in Figure 4 is easy to interpret. The data panels are scaled so we can quickly spot how well a particular NEDD protein is recognized by different ubiquitins, as well as which mutations decrease afnity or eliminate binding. Why cant all papers be this good? Kalyuzhniy et al. Adenovirus serotype 5 hexon is critical for virus infection of hepatocytes in vivo. Proc Natl Acad Sci USA 105: 54835488. Wang et al. In vitro and in vivo properties of adenovirus vectors with increased afnity to CD46. J Virol 82: 10567 10579. Work published by Shayakhmetov and co-workers exemplify well-performed kinetic analyses. In these studies of adenoviral interactions with blood coagulation factors and a receptor, the multivalent virus was immobilized (rather than tested in solution) to avoid avidity effects and surface densities were kept low to minimize other complexities. Analyte injection times were long enough for obtaining curvature in the association phase, the dissociation phases were monitored long enough to observe a decrease in response and a wide range of analyte concentrations were included in each experiment. Figure 5 demonstrates the wealth of information available from a systematic kinetic characterization of related binding pairs. In this study, Kalyuzhniy et al. determined the adenoviral specicities and afnities for a subset of blood coagulation factors, which revealed the mechanism by which these viruses bind factor X and demonstrated the importance of the hexon protein in viral infection (172). Especially noteworthy is Kalyuzhniy et al.s use of what we call the short-n-long (SNL) approach to kinetic analysis of slowly dissociating complexes (an example is shown in the upper-left panel of Figure 5). Since this interaction is so stable, the dissociation phase must be monitored for a long time (in this example, >30 min) to collect enough decay in signal to obtain a reliable kd. But these researchers recognized that there is enough decay information in the responses from the highest analyte concentration to determine kd, so they truncated the dissociation-phase data-collection times for the lower analyte concentrations. (The SNL approach improves efciency since it increases sampling throughput compared to the standard kinetic analysis format.) And, the overlay of the replicates conrmed the baseline was not drifting and the reagents were stable over the many hours required for each experiment. (We must repeat.) We were also happy to see Kalyuzhniy et al. t all the data sets to a 1:1 interaction model, even those that display some complexity (i.e. the two panels highlighted with a star in Figure 5). Rather than invent an unlikely binding scenario to account for these secondary components, the authors realized the complexity most likely arose from biologically irrelevant artefacts. With the overlay of the data and ts, we can recognize how well the reported rate constants and afnities describe these particular interactions. Figure 6 portrays the biosensor-based optimization of a transfer vector useful in gene therapies. Presuming that viral afnity for CD46, the receptor that some bre knob-containing viruses use for cell invasion, correlates with infectivity, Wang et al. constructed a panel of mutant bre knob particles and tested each against CD46 (Figure 6A). Several mutations produced particles that bound more tightly to CD46 compared to wild type, maintained their increased receptor afnity when incorporated into virions (Figure 6B), and indeed displayed greater in vivo transduction (1174).
6
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
Figure 3. Epitope mapping and kinetic characterization of Der p2-antibodies. (A) Assay designs (left) and responses (middle and right) for two complementary mapping experiments. The top panels depict the responses expected for the pair-wise analysis: a second IgE clone binding to an available site on IgE-captured rDer p2 (middle) and a second clone that would bind at the same site in rDer p2 as does the clone used to capture the antigen (right). In the analysis of antigen variants (bottom), the right panel depicts the responses for wild type and mutant rDer p2, as well as a negative control, pGAPz, binding to one IgE clone. (B) Summary of mapping experiments. Left: pair-wise analysis of 11 clones. Unlled circles: rIgE pairs that can bind rDer p2 simultaneously; lled circles, rIgE pairs that cannot. Middle: analysis of six rDer p2 variants against 11 IgE clones. **, signicantly altered; * somewhat altered; , equal; -, no binding of the rDer p2 variant compared to wild type. Right: three-dimensional model of the clones epitopes in rDer p2. (C) Top: example kinetic proles of clones binding rDer p2 with high (left), medium (middle) and low afnity (right). Bottom: summary of the afnities determined for 31 clones, with specic colours corresponding to the epitope designations in the right panel of Figure 3B. Adapted from Reference 358 with permission from the American Academy of Allergy, Asthma and Immunology 2008.
7
www.interscience.wiley.com/journal/jmr
Figure 4. Equilibrium analyses of wild type and mutant NEDDs binding to ubiquitin-like proteins and ubiquitin. In each data subset, responses for triplicate analyses of each concentration are displayed in the left panels while the right panels show the ts of the responses at equilibrium to a simple binding isotherm. Adapted from Reference 279 with permission from the American Chemical Society 2008. Wang et al.s work illustrates how kinetic information can drive a research program. The differences in afnity between wild type and various mutant knob particles were due directly to changes in the off rate, so by comparing the dissociation rate constants Wang et al. identied which mutants to incorporate into virions. From a technical standpoint, these biosensor studies are impressive for two reasons. First, Wang et al. were not intimidated by the bumpiness (due to random, short-term noise) in binding responses that may be evident when working with low-density surfaces (e.g. the upper right panel of Figure 6A). Unlike biosensor users who prefer smooth-looking, large-intensity responses, these researchers recognized that it is far better to publish data with these small deviations from ideality than to introduce artefacts that can arise at high surface densities. Second, concerned that a regeneration condition may damage the surface-tethered knob particles and/or virions, Wang et al. simply washed the surface with running buffer until the response returned to baseline (e.g. >45 min for some mutants). Sometimes doing nothing is something. Abdiche et al. Probing the binding mechanism and afnity of tanezumab, a recombinant humanized anti-NGF monoclonal antibody, using a repertoire of biosensors. Prot Sci 17: 13261335.
8
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
Figure 5. Kinetic analyses of ve blood coagulation factors binding to six adenoviruses and adenovirus capsid proteins. Red lines depict the t of a 1:1 interaction model. Each panel depicts duplicate analyses of each analyte concentration. The panels highlighted with a star are referred to specically in the text. Adapted from Reference 172 with permission from the National Academy of Sciences USA 2008.
Abdiche et al. Determining kinetics and afnities of protein interactions using a parallel real-time label-free biosensor, the Octet. Anal Biochem 377: 209217. As our options in biosensor technology multiply, it becomes increasingly important to evaluate the different instruments. Abdiche et al.s side-by-side comparison of the GE Healthcare Biacore 3000s, BioRad ProteOn XPR36s and ForteBio Octet QKs detection systems, sampling methods and throughput, as well as the precision of the obtained binding parameters, demonstrate the advantages and disadvantages of these three platforms. In one comparative study, Abdiche et al. characterized the interaction of the antibody tanezumab and human nerve growth factor (NGF) (39). This Ab/Ag pair is particularly difcult to work with using the biosensor because both binding partners are bivalent (NGF forms homodimers) and the afnity of the
interaction is tighter than 10 pM. So to obtain reliable data from, and kinetic information about, this system requires excellent experimental technique and tests the limits of these instruments capabilities. To fully examine this interaction (and to demonstrate the parameters they obtained were not inuenced by the assay orientation or immobilization method, as well as to deal with the fact that they could not nd regeneration conditions that disrupted the Ab/Ag complex without inactivating the Ab), Abdiche et al. used several assay formats, as illustrated in Figure 7A. They tried direct immobilization of both the NGF homodimer and tanezumab Fab, as well as capturing (either by streptavidin or an antibody) of NGF, the mAb and the Fab. In each experiment they worked at very low ligand densities to minimize avidity.
9
www.interscience.wiley.com/journal/jmr
Figure 6. Kinetic analyses of bre receptor CD46 binding to (A) nine adenovirus bre knob mutant particles and (B) three knob mutants incorporated into virions. Replicate responses of each analyte concentration are overlaid with the t of a 1:1 interaction model (red lines). Adapted from Reference 1174 with permission from the American Society of Microbiology 2008.
Figure 7B shows data obtained from the three biosensors for Fab binding to streptavidin-captured biotinylated NGF. The responses obtained from each instrument are easily discernable above background, concentration-dependent and reproducible. In addition, the Fab could be injected long enough to detect curvature in the association phase. Replicates overlay, indicating each instrument is stable enough to collect data even for the more than 8 h required in each binding cycle. To monitor such a slowly dissociating complex, Abdiche et al. used the SNL approach to collect the full data set from Biacore 3000; with the other two instruments, they performed one-shot kinetics to collect data for all Fab concentrations simultaneously. Although they collected dissociation-phase data for more than 8 h, the Fab/ NGF complex did not dissociate enough in each of these experiments to obtain a reliable kd. Wisely, Abdiche et al. simply reported the kd was slower than 2 106 s1. Taking the comparative study one step further, Abdiche et al. also determined the afnity of this interaction using KinExA 3000 technology. Comfortingly, they found the binding parameters obtained from the surface-based biosensors agreed well and corresponded with values from the solution-based KinExA analysis. So, heres a big raspberry to those who say surfaceand solution-based methods never match: Thpppfbt! Figure 7C and D shows the responses obtained for the interaction tested in the opposite orientation: NFG in solution tested against surface-tethered Fab or mAb. Because the ligands were captured at such low densities, similar kinetic rate constants were obtained for the NGF homodimer binding to the Fab (Figure 7C, top panel) and mAb (Figure 7C, bottom panel) surfaces. In the stepwise saturation experiment shown in Figure 7D, Abdiche et al. tracked the binding of NGF to amine-coupled Fab surface (top) and then followed how Fab in
solution bound to the now-formed NGF/FAb complexes on the surface (bottom). Together, the rate constants obtained from the experiments shown in Figure 7BD conrmed that the kinetics were not inuenced by which binding partner was tethered to the surface or by the method used to tether the ligand (either via capture or amine coupling). Thpppfbt, thpppfbt! And nally, Abdiche et al. developed a biosensor assay to serve as an alternative to functional cell-based assays. Using a solution-competition approach (Figure 7E, left panel), they quantitated the activity of tanezumab in their preparations. The method they described is widely applicable for determining active concentration. In another comparative study, Abdiche et al. took a slightly different tactic to determine the abilities of the three instruments to characterize a series of peptide/antibody interactions (38). Figure 8A and B depicts the Biacore 3000 kinetic titration (also called single-cycle) and ProteOn XPR36 one-shot approaches for measuring kinetics of peptide binding to immobilized IgG without regenerating between analyte injections. They then compared these results with those obtained from the reverse orientation: Fab in solution tested directly against immobilized peptides (Figure 8C) or in a solution-competition format (Figure 8D). The agreement between the rate constants and afnities determined using different assay designs, interaction orientations and instruments established the reliability of binding parameters reported from the Biacore 3000, ProteOn XPR36 and Octet QK. Nice job. Beusink et al. Angle-scanning SPR imaging for detection of biomolecular interactions on microarrays. Biosens Bioelectron 23: 839844. Beusink et al.s report of IBIS Technologies iSPR established the capabilities of this array platform (1389). To demonstrate iSPRs
10
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
Figure 7. Biosensor analysis of human nerve growth factor (NGF) and its neutralizing antibody, tanezumab. (A) Assay design of the different kinetic experiments performed in this study, with the symbol key at the right. (B) Kinetic analyses of Fab binding to biotinylated NGF captured on surfaces in Biacore 3000 (top), ForteBio Octet QK (middle) and BioRad ProteOn XPR36 (bottom). Left panels show zoomed-in views of the responses obtained during the association phase; right panels are the full sensorgrams, which illustrate the long dissociation-phase data collection times used in these experiments. (C) Kinetic analyses of NGF binding to captured tanezumab Fab (top) and IgG (bottom) performed using Biacore 3000. (D) Kinetic analyses of NGF binding to amine-coupled Fab (top) and Fab binding to Fab-captured NGF (bottom) performed using BioRad ProteOn XPR36. (E) Active-concentration analysis of anti-NGF IgG using Biacore 3000. Left panel: assay design; middle and right panels: responses and inhibition curve. In BD, the insets depict the assay design for each of these experiments and the black lines are the t of the replicate responses to a 1:1 interaction model. In the Octet QK experiments, binding was sinked by spiking the dissociation buffer with soluble ligand. Adapted from Reference 39 with permission from The Protein Society 2008.
11
www.interscience.wiley.com/journal/jmr
Figure 8. Biosensor analysis of calcitonin gene-related peptides (CGRPs) and an anti-CGRP antibody. (A) Single-cycle kinetic analyses of four CGRP/IgG interactions performed using Biacore 3000. Blue, red and green traces depict the responses from high-, medium- and low-density IgG surfaces. In the lower-right panel, the inset shows the binding isotherm obtained from an equilibrium analysis of the data in the main panel. (B) One-shot kinetic analysis of four CGRP/IgG interactions performed using BioRad ProteOn XPR36, with the binding isotherm of an equilibrium analysis shown in the lower-right panel inset. (C) Kinetic analyses of Fab binding to rat (left) and human (right) CGRPa as measured using ForteBio Octet QK (top), BioRad ProteOn XPR36 (middle) and Biacore 3000 (bottom). (D) Solution-based afnity determination of IgG (left) and Fab (right) binding to four CGRPs as measured using ForteBio Octet QK (top), BioRad ProteOn XPR36 (middle) and Biacore 3000 (bottom). In AC, smooth lines depict the t of the responses to a 1:1 interaction model. In all direct binding experiments using Octet QK, binding was sinked by spiking the dissociation buffer with soluble ligand. Adapted from Reference (38) with permission from Elsevier, Inc. 2008.
ability to simultaneously monitor interactions of ligands having widely different masses, these researchers owed a uorescently labelled biotin antibody through a ow cell containing spots of biotinylated peptide (2.4 kDa) and biotinylated IgG (150 kDa). Figure 9A shows how well the binding responses correlated with both analyte concentration and spot density. In the rst analyte injection (far left), the signal intensities correspond well to spotted peptide densities (with no signal observed for the negative-control spots). In addition, the responses decrease from left to right as the concentration of injected analyte decreases. From these data, Beusink et al. resolved the instruments limit of detection with respect to the concentrations of both analyte and ligand spotting solutions for this interaction (Figure 9B). As shown in the top panel of Figure 9C, the SPR image of the chip after the analyte injection conrms these binding results.
Antibody binding to both ligands (two rows each of biotinylated peptide and IgG) is apparent, indicating an array of ligands of different masses could be prepared and characterized. In addition, the intensities of the spots increase with increasing spot density. The correlation between spot density and uorescence intensity (Figure 9C, lower panel) supports the SPR results. Indeed, this imaging SPR instrument appears promising as one approach to increasing throughput in biosensor experiments. Rich et al. Extracting kinetic rate constants from surface plasmon resonance array systems. Anal Biochem 373: 112120. Yes, we recommend you to read our paper about extracting kinetics from Biacores Flexchip system since it demonstrates the potential of array technologies to produce reliable rate constants at spots throughout the ow cell (1362). Figure 10A shows an example of reaction spots within the Flexchip ow cell:
12
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
Figure 9. Binding studies performed using iSPR-IBIS. (A) Responses obtained for seven concentrations of an anti-biotin IgG (and a blank buffer injection) binding to ve different density spots of biotinylated peptide, a negative-control spot and a background spot. Responses from the rst injection (at far left) are for the highest IgG concentration across the seven spots; responses at the far right are from the blank buffer injection across these same spots. (B) Response intensities from panel A plotted against IgG concentration for the ve different ligand spot densities. (C) SPR (top) and uorescence microscopy (bottom) images of biotinylated ligand spots bound with uorescently labelled anti-biotin IgG. In each image, the top two rows are a peptide dilution series spotted in duplicate; the bottom two rows are an IgG dilution series in duplicate. NC negative controls (non-biotinylated peptide and IgG). Adapted from Reference 1389 with permission from Elsevier 2008.
13
www.interscience.wiley.com/journal/jmr
Figure 10. Kinetic analysis using Biacore Flexchip. (A) Screen shot of a 12 13 matrix of ligand spots in this large-format ow cell. Reaction regions of interest (ROIs) are circled in white, with one row highlighted in red. The darker circles represent reference spots centred between the reaction spots, with one row highlighted in blue. (B) Duplicate responses (obtained at matrix position D8 in panel A) for a concentration series of rat IgG binding to Protein A/ G, overlaid with the t of a 1:1 interaction model. (C) Responses and ts for rat IgG binding to the uniform-density 12 13 Protein A/G spot matrix. (D) Triplicate responses for a concentration series of human IgG binding to spots of different Protein A/G surface density. (E) Global ts to a 1:1 interaction model of triplicate responses for the human IgG concentration series binding to low-, medium- and high-density Protein A/G spots. Adapted from Reference 1362 with permission from Elsevier 2008.
throughout this 12 13 matrix of ligand (Protein A/G) spots, interstitial unmodied regions were used for referencing. Responses for rat IgG binding to one of these ligand spots were concentration-dependent, reproducible and t to a 1:1 interaction model (Figure 10B). Similar responses were obtained for the entire array of spots (Figure 10C). Across the surface, variability in the rate constants was random and less than 9%. Taking these tests one step further, we tested human IgG binding to a gradient of Protein A/G spots. As expected, binding response
intensities correlated with the concentrations of the ligand spotting solutions and, therefore, spot densities (Figure 10D). These data indicated the Flexchips lower limit of detection for this interaction was about 2 ug/ml in ligand spotting concentration and about 1 nM in analyte concentration. And nally, Figure 10E shows the global t of human IgG responses to three of these different density Protein A/G spots. Combined, these studies helped to establish array biosensors as viable platforms for kinetic analyses.
14
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
REPORT CARDS
While there were more than 1400 biosensor articles published in 2008, unfortunately the number of really good papers was rather small. Its clear that when you compare the papers en masse, too many users arent putting enough effort into assay optimization, data interpretation and data presentation. For years, we have discussed how to avoid common experimental errors and stressed the importance of showing data. In fact, in past surveys we used examples from some of the very worst articles to illustrate what is bad with the literature (18). But being the playground bully wasnt really fair since most authors got to stand around and watch us beat up on an unlucky few. This year we let no one escape. We got out our red pens and assigned each paper a grade. (Now you might ask: Who put us in charge? Well, if you want to spend six months of the year searching internet databases, downloading >4000 papers just to nd ones that actually describe biosensor experiments, and being exposed to a lot of really bad data, then next year we can send you a pen.) Our grading criteria were based on what would be expected in a high-school chemistry class. To get a good grade back in those days, we were required to do the experiment right, analyse it properly, and present it clearly. Authors whose papers got As and Bs did the experiment right. Basically, you got an A if you showed replicates of each analyte injection and included the t of a 1:1 interaction model; a B usually just lacked the t or the replicates. Cs showed potentially meaningful data that are inadequately optimized: either the experiment or analysis needed more work. Ds suffered from a variety of problems. The majority of Fs didnt show any data, although a few data-containing papers also failed because their sensorgrams couldnt possibly describe a real binding event. Now keep in mind that we were only judging the quality of the biosensor data and its presentation, not the scientic impact of the experiments. These grades are to educate users about what they are doing right and wrong, as well as to help readers identify which articles (from a biosensor standpoint) are worth checking out from the library. The bar graph in Figure 11 shows our grade distribution. Unfortunately, the majority ($67%) received Ds and Fs. If you think we were too harsh, just look through the examples in
Figure 11. Bar graph of the grade distribution for the 2008 literature.
Figures 12 through 17 and see for yourself the differences in the quality of the work we assigned each grade. A papers exemplify well-performed experiments and wellpresented results. Like all of the examples shown in Figures 12 and 13, A-quality responses are single exponentials (indicating they most likely describe actual binding events rather than depict artefacts) and are of what we call Goldilocks intensity: not too big and not too small, but just right. Also, the authors showed replicates of analyte concentrations in at least one data set to demonstrate their results are reproducible. Its frightening how few users demonstrate their data are reproducible. For quantitative analyses, the analyte concentration series spanned a wisely selected range and both data and ts were shown. As illustrated in Figure 12, A-quality kinetic papers include the overlay of a 1:1 interaction model. A-quality equilibrium papers (Figure 13) show data sets in which every response reached equilibrium during the association phase and the t to a simple binding isotherm is included. In addition, each data set in Figure 13 gets a gold star for plotting the binding isotherms with concentration on a log scale. With a log scale, we can clearly see all the data points (compared to a linear scale, which bunches the low-concentration data points all together, making it difcult to really judge the quality of the t). And note that none of these papers showed a Scatchard analysis. Scatchard plots are so old school we call them ancient school. Using a Scatchard plot was a great idea back in the days when we used an abacus to t our data. More than likely you have a computer, so just do nonlinear least-squares tting. (Are we going too fast for you?) B papers typically lack only one of the features we look for in a biosensor analysis. Its frustrating because sometimes the authors show replicates which are beautifully overlaid but then fail to show a t to the data (Figure 14A). Most often, they show a great t to a simple model and are only missing replicates (Figure 14BH). Clearly, these authors know what they are doingwith just a little more work they would get an A. And nally, the variety of experiments (e.g. screening, standard kinetics and equilibrium analyses, kinetic titrations and stability tracking) that produce at least B-quality data demonstrate that every assay format can produce data at least as good as these. C papers present data that are most likely due to an actual binding event, but the experiment and/or analysis needed to be improved before publication. For example, Figure 15A shows what we call a pearls-on-a-string analyses. These data sets have a very narrow dilution series. We love that the authors ran a lot of analyte binding tests, but the data sets would be much more informative if there were replicates of fewer analyte concentrations instead of singlets of such a narrow dilution series. Andy Warhol said Replication is the key to success, now who wants soup? Figure 15B shows two examples of complex responses that are difcult to attribute to a specic binding event. Most likely, one (or both) of the binding partners is poorly behaved and the experimental conditions could be better optimized. As weve said many times before, high ligand densities and high analyte concentrations lead to complex binding. You need to realize that there is a limit to the highest concentration you can go to with many proteins and small molecules before you introduce complicating factors like non-specic binding, crowding or aggregation. Less is more, more or less. Figure 15C shows two data sets that were t to a drifting-baseline model. Unfortunately, this model may not really
15
www.interscience.wiley.com/journal/jmr
Figure 12. Examples of A-quality kinetic analyses. Adapted from References (101,282,449,452,576,709,846) with necessary permissions granted by Cold Spring Harbor Press, the American Chemical Society, Elsevier, Inc. and Springer 2008.
account for whats going on in the reaction. Its just a crutch that many groups lean on to get a good t to poor-quality data. We call this tting to a somethingsomething model since it is unclear what portion of the response is accounted for by the drifting-baseline model (some sensorgrams drift up and some drift down). It is far better to improve the integrity of the data by
optimizing the experimental system than to model your brains out. The experiments shown in Figure 15D and E are some of the easiest to improve. To obtain reliable rate constants from a kinetic analysis, you need to have curvature in the association phase and observable decay in the dissociation phase. So to get more
16
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
Figure 13. Examples of A-quality equilibrium analyses. Adapted from References (178,185,224) with permission from Elsevier, Inc. and the National Academy of Sciences USA 2008.
curvature in the association phase of the data shown in Figure 15D, the authors just needed to inject the analyte for a longer time or test higher concentrations. (Granted, the injection time in the right panel was more than 40 min but that still wasnt long enough. Alternatively, you might try kinetic titrations if you are limited by injection volume.) And all of the examples in Figure 15D and E need longer dissociation times. Most biosensors are automated, so why not just let it run to collect more data in the dissociation phase? A well-conditioned biosensor should be able to collect reliable dissociation data for 12 h or more if you are careful and patient. D papers give biosensor technology a bad reputation, but unlike Joan Jett, we do give a damn about a bad reputation. For example, some D-quality experiments produced responses that might actually describe binding, but they are so weirdly shaped its hard to tell what is going on (Figure 16A) or display complex binding at high analyte concentrations (Figure 16B). Its apparent that most users whose papers got Ds fail to understand a sensorgram should be a single exponential. (Can you draw what a sensorgram should look like? Its not just some arbitrary curve. It has a specic shape. Knowing how your data should look is Step 1 in improving biosensor analysis. Now you only have 11 steps to go.). There are two things that made us so mad these papers automatically got Ds. If you used an equilibrium analysis to determine equilibrium binding constants from responses that did not come to equilibrium (like those in Figure 16C), you got a D. (Actually, you should have gotton an F for False reporting of binding constants.) Its also common to see people plot equilibrium responses against concentration but not include a t, as in
Figure 16D. They do this because they either dont understand how to t the data or they t it to a simple binding isotherm and it didnt t well so they choose not to show it. Also, just connecting the data points with a line is not tting the data. And if you t your kinetic data using a conformational-change model (e.g. Figure 16E), you also got a D. We will talk more about this later but whats funny is that most people who used a conformational-change model said something like we used a conformational-change model because it t the data better than a simple model. Nice going, Einstein. You do know that you cant prove a model is correct by simply tting it to the data? (If that sounds new to you, then go ride in an elevator at constant velocity and tell us if you are moving.) Whats even funnier is that most users of the conformational-change model dont even report the correct units for the conformational-change step. And the same can be said for those who use a bivalent analyte model. Nothing says I dont know what Im doing more than not knowing what youre doing. And another thing about model tting. We were sad and disappointed to see how many authors went model surng. They t their data sets to just about every available model, showed all these ts, and reported the rate constants obtained from each t. It shouldnt be left up to the reader to gure out what is going on in your reaction. Do they expect us to do their homework for them? Most F papers show no data whatsoever but still report binding constants (my dog ate my homework). Without seeing the data, we have to wonder if the authors of F papers did the experiment right or, in fact, did it at all. With todays opportunities to include data in on-line supplements, the excuse that theres no room in
17
www.interscience.wiley.com/journal/jmr
Figure 14. Examples of B-quality data. Adapted from References (384,411,470,505,697,728,731,852,997) with necessary permissions granted by
Macmillan Publishers Ltd: [Nature Immunology], Elsevier, Inc., the American Chemical Society, the National Academy of Sciences USA and Wiley 2008.
18
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
Figure 15. Examples of C-quality data. Adapted from References (93,192,261,377,472,653,686,881,918,1030) with necessary permissions obtained from
the American Chemical Society, Elsevier, Inc., Wiley Interscience and BioMed Central Ltd. 2008.
19
www.interscience.wiley.com/journal/jmr
Figure 16. Examples of D-quality data. Adapted from References (216,225,344,523,529,762,819,951,1074,1097) with necessary permissions granted by
Elsevier, Inc., the American Chemical Society, the National Academy of Sciences, USA, Macmillan Publishers Ltd: The EMBO Journal and the American Society for Cell Biology 2008.
the paper doesnt y. Its unforgivable 01to report binding constants without a gure of the data. Come on, its show-and-tell. You spent all that time collecting data. Why not share it with the rest of the class? We know the answer. Its like Michael Jackson said: Because its Bad, its Badcome on (Bad Badreally, really Bad) You know its Bad, its Badyou know it
(Bad Badreally, really Bad) You know its Bad, its Badyou know it, you know (Bad Badreally, really Bad) And the whole world has the answer right now Just to tell you once again... whos Bad Whos bad? Your data are bad. Other F papers include some data, but they are absurdly awful. Figure 17 shows examples of
20
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
Figure 17. Examples of F-quality data. Adapted from References (276,916,1075,1134) with permission from Elsevier, Ltd. 2008. published data that are in desperate need of improvement. We wish Michael was still around to help us write a catchy song about that. Who can generate poor-quality data? (we can, and we do) Who publishes whatever comes out of the machine Often without even showing the data? (we can, and we do) All the time! Okay, we will stick to writing literature reviews rather than songs if you promise to put a little more effort into your experiments and publications. modelling a step function of equal intensity throughout the association phase. Figure 18A is a good example of a data set that contains this small jump in the association phase and Mattu et al. were justied in including the bulk-shift correction when tting their data. (Excellent t to the data. This paper would have received an A if this group had run some replicates and not so many pearls on a string.) In contrast, Figure 18BD shows several data sets in which the bulk-shift correction should not have been used. We call these data sets bulk-shift INcorrection because this correction factor was used to mask complexity in the responses. Notice how the bulk shift improves the t but it is not really describing the complex kinetics in the data set itself. You often nd users like these stating the data t well to a simple 1:1 interaction model when in reality they are not using a simple model at all. The two examples in Figure 18D take this to an extreme level and use the bulk shift to account for over 80% of their binding signal. Dont let these groups fool you. Do you see what I see? People often ask us why we insist on seeing ts to the data. A t instantly shows the reader you know what you were doing (and showing the data overlaid with the t does not take up any more space in the article so we dont want to hear any excuses). To illustrate the importance of showing ts to your data, we simulated what the t would have looked like in several data sets
DETENTION
Although the bell may have sounded, schools not over for some of you. Sit back down. We need to talk with you about a few problems that are showing up in the literature with increasing frequency. Bulk-shift INcorrection The bulk refractive index correction was introduced to account for small residual jumps in signal that may be apparent when working with low-intensity responses. The correction involves
21
www.interscience.wiley.com/journal/jmr
Figure 18. Examples of using the bulk-shift correction (A) appropriately and (BD) inappropriately. Adapted from References
(136,403,778,746,839,947,1055) with permission from Elsevier, Inc., Springer and the American Chemical Society 2008.
(shown in Figure 19). Remember, each of these papers showed a gure of data (shown in the left panels of Figure 19) and included rate constants (listed on the right) but did not include the kinetic model t overlaid with the data. Our simulated responses are shown in red. In Figure 19A, our simulation closely mimics the authors data, indicating their reported rate constants are believable. This data
set even included replicates; if these authors had just shown the t, their paper would have received an A. The data in Figure 19BD are a different matter altogether. In most cases, the authors data do not look anything like our simulations, leaving us to wonder where they got their binding parameters from. We call these hate constants because we hate to see numbers published that are so misleading.
22
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
Figure 19. Data sets (left panels) for which we simulated responses (right panels) based on the rate constants reported by the original authors.
Adapted from References (198,220,355,651) with permission from Elsevier, Inc. and Informa UK Ltd. 2008.
Even by just glancing at the data in Figure 19BD, you should be able to spot some problems. For example, you should know that a kd of 102 s1 is relatively fast, unlike the authors responses shown Figure 19C. We dont think you should be allowed to step up to the biosensor unless you can do a few simple things, like recognize some ballpark rate constants (at the very least, you should know what the proles of different off rates look like). Then you can tell if the parameters you report are at all close to what youd expect.
And then there is the data in Figure 19D. Fortunately, we werent the only ones who were surprised by the rate constants reported from this data set. In their Letter to the Editor, Dolimbek et al. commented (8), The Biacore experiments (in Reference 651) dont make a lot of sense. The methods do not seem to t the experimental data. There is no dose-dependent response. . .. . ..a much greater response differential would be expected. But the results are not what would be expected, and it is uncertain what they were actually owing over the cell. Hence it is questionable
23
www.interscience.wiley.com/journal/jmr
R. L. RICH AND D. G. MYSZKA how the afnities were calculated. Its so gratifying to see members of the community taking a more active role in teaching one another (maybe next year we should send Dolimbek a red pen). We also want to point out that the authors of Figure 19D reported an association rate constant of 3.88 104. Really, you have an association rate constant that is 104? That seems a tad slow to us. Cha-cha-cha-changes Why do people persist in clinging to a conformational change model simply because it ts their data? As we pointed out earlier, you cannot prove a model is correct by tting it to a data set (jump on that elevator). The reason is that multiple models may t the data equally well. Of course, the only way to prove a model is correct is through experimentation. To prove the conformation change model is applicable to your interaction, you need to test analyte binding for different lengths of contact time. Most users forget, however, the key is that you have to saturate the surface for each contact time. Otherwise the method cannot be used to distinguish between a conformational change and something else like surface heterogeneity. Murat et al. demonstrate the correct way of testing for a conformation change (Figure 20A). This group collected binding data with different association times, but notice how the response for even the shortest contact time reaches the same magnitude as that of the longest contact time. And notice that the units for the response are still in RU, which is correct (they didnt rescale or normalize). They appropriately showed that contact time did not inuence the dissociation phase in their system. They get to go home early today. The data in Figure 20B show how NOT to test a conformational-change model. Sure, these authors varied the association phase time and saw differences in the dissociation rates, but look carefully at the units of the Y-axis. They scaled the responses at the end of the association phase but tried to hide this rescaling by naming the Y-axis Response (RU) when its clearly not. It should be labelled Normalized Response and it would not have units. Its also clear to see, in the shapes of the response proles collected for different times, that the reactions have not all saturated the surface, which is required for this test. So while the dissociation phases for the different contact times of analyte look different, this does not prove its a result of a conformational change. How can we be so sure? Take a look at the responses in Figure 20C. These look strikingly similar to the ones in Figure 20B and they also clearly show differences in dissociation rate that correlate with contact time. Raise your hand if you think this proves that this data set is a result of a conformation change reaction. You can put your hands down now. Youre wrong. We simulated this data set with a surface heterogeneity model. We could have simulated similar looking data using a solution heterogeneity, a bivalent analyte and/or a aggregation model. Dont be fooled by people claiming to prove they have a conformational change when in fact they dont know what they are doing. And how relevant do you think a so-called conformational change is when it occurs on a time scale of 1015 min? You know Occums Razor: the simplest model is correct. Well we have a new razor for you, Myszkas Razor: Stop using a conformational change model to t your poor-quality data.
Figure 20. Data sets in which the injection time was varied. Panels A
and B were adapted from References (798,953) with permission from Elsevier, Inc. and Wiley-VCH Verlag GmbH & Co. 2008.
Whats in a name? My mother [DGM] always said If you cant say anything nice, dont say anything at all. And never drink wine out of a bottle bigger than your head. Well, weve got something to say to those of you who either dont proofread your publications or dont know how to report binding constants in a paper. Heres the deal. The IUPAC IUBMB, which is the international governing body responsible for standardizing scientic language, denes the association event for a biomolecular interaction as ka
FINISHING SCHOOL
In order to standardize the biosensor literature, consider these recommendations as your new school uniform.
24
www.interscience.wiley.com/journal/jmr
COMMERCIAL OPTICAL BIOSENSOR LITERATURE and the dissociation event as kd. Note its a small letter k that is in italics and a subscript little a or d that is not in italics. Period. They are not Ka, KA, kfor, k1, ka, kass, ka, kassoc, kon, ka app and Kd, KD, krev, kd, kd, kdiss, kdissoc, k1, koff, kd app. It is truly amazing the variety of notation you can nd in the literature. If you dont believe us, look back at the binding constants reported in Figure 19. These were taken directly from the authors papers without editing. Notice that none of these four examples conform to the IUPAC standard for reporting rate constants. And the same can be said for the equilibrium dissociation constant, which should be presented as KD. Never report your afnities as an equilibrium association constant (KA); youre not doing isothermal titration calorimetry here (yes, yes we know KD 1/KA and so does everyone else so you dont need to provide both in your paper). Reporting the correct units for parameters is also important. The standard units for the association rate constant are M1 s1 and the dissociation rate constant is s1. Weve seen the association rate constant reported with units of M s, M/s, s/M, mole1 s1, M and just s. About 15% of the time there are no units at all. It may seem like nitpicking, but is it really that hard to report binding constants with the correct units? Plus it gives the reader of your paper a sense that you know what you are doing. Of course, statistical errors tell us only how well a parameter is dened for a model within a particular data set. To dene the experimental error you would need to repeat the entire study. This may sound crazy but hear us out. To be a scientist means you often have to run an entire experiment more than once to see how unknown variables affect the results. You would then take the average of the measurements for the parameters and use their standard deviation to report the experimental standard error. We could nd only a handful of papers which provide convincing evidence that they replicated the entire study. Unfortunately, it seems that fully automated instruments have made us complacent and spawned an attitude of If it came out of this fancy and expensive machine with a computer, I dont need to repeat my experiment. (Hal! Open the pod bay doors!).
Show me the error of my ways Now if you really want to impress your colleagues, you should present the binding constants with the correct standard errors. (Gosh, at this point we would be happy if you reported any errors at all). An association rate constant like 5.25 104 M1 s1 is actually meaningless without standard errors associated with it. There are two types of standard errors that we are concerned about, statistical and experimental. Statistical errors are presented by the tting software, typically to what is referred to as one standard deviation which has a condence interval of 68% (see Wikipedia for details on condence intervals; were limited in space here). Basically, it denes how well we know a parameter in the model for a given data set. Now many users are shocked to see that the statistical error may be really tiny, like only a tenth of a percentage of the parameter value itself. For example, in an information-rich (lots of curvature) data set, the computer may tell you the association rate constant is (5.2578 0.0015) 104. The fact that the standard error is small compared to the parameter value tells us in this case that the association rate constant is well dened. If the computer said the association rate constant was (5.2578 1.5245) 104, this would say the parameter is not as well dened. However, it doesnt mean the value is wrong. Now it is just as important to report the correct number of signicant digits when publishing binding constants. You cant simply publish whatever numbers the computer provides you. (Open the pod bay doors, Hal.) The rule is to round up the value of the error to its rst signicant digit. This then denes the number of signicant digits to report in the binding constant. So, the correct way of reporting the rst example in the paragraph above would be (5.258 0.002) 104. You see how we rounded up the value of the error as well as the binding constant to four signicant digits? The second example would be reported at (5 2) 104. Here, you try one. How would you report the dissociation rate constant if the computer gave you 1.8309 103 5.7251 105 to you? If you said (1.83 0.06) 103, you would be correct. If you got it wrong, read this paragraph again.
Figure 21. Figures scaled to emphasize (A) the spikes and (B) the
baseline. Adapted from References (194,916,919,940) with permission from Elsevier, Inc. and the American Chemical Society 2008.
25
www.interscience.wiley.com/journal/jmr
R. L. RICH AND D. G. MYSZKA Tuck in your shirt For years we have been encouraging users to show gures of their data in publications, and we are happy to see that a majority of papers (>70%) now do present some data. The next issue that we would like to address is just how the data are presented. One of the common and comical ways we see data presented is illustrated in Figure 21A. Note how these gures have been scaled so that the spikes (which often occur at the transitions between running buffer and sample) are visible. This forces the responses (the curvy parts) that we really care about seeing to be really small. We call this kind of plotting scaling-to-the-spikes syndrome or SSS plotting for short. The data shown back in Figure 19D are another good example of this. Another common ailment in plotting is to show too much baseline before or after the injection. While the binding data presented in Figure 21B are in fact pretty good, it sure would have been nice if the authors focused on the response curves so we could see them better (and the overlay of the t if they had included one) rather than consuming half the space in the gure with a at line. Compare the sloppy plots of Figure 21 with those shown in Figures 12 through 14. You see how easy it is to interpret well-presented data? Another way we want to further standardize the biosensor community is to encourage everyone to publish good-looking gures. Too often authors simply dump low-resolution screenshots from the biosensor software directly into their paper. Oftentimes, this means the gure is scaled to include those irrelevant data spikes and/or the lines are too thin or too faint and the axis labels are too small to read. Making a good-quality gure is an essential part of becoming a professional biosensor scientist. bulk-shift INcorrection concentrationdependent aggregator detergent-sensitive binder double referencing Goldilocks responses Hal-ing your results hate constants it might be mites ka kd KD using the bulk-shift correction to mask complex data. analyte that displays inexplicably large responses at high concentrations. analyte that becomes well-behaved upon the addition of detergent. subtracting the responses from a reference surface and buffer blanks. responses that are neither too big nor too small, but just right. publishing whatever the computer tells you. reported rate constants that do not describe the binding responses. the idea that mites are the cause of all diseases. association rate constant, with units dened as M1 s1 by IUPAC IUBMB. dissociation rate constant, with units dened as s1 by IUPAC IUBMB. equilibrium dissociation constant, with units dened as M by IUPAC IUBMB. testing several analyte concentrations in one binding cycle. data that are bad, bad really bad, you know it. Stop using a conformational-change model to t your poor-quality data. analytes that bind more than the predicted Rmax. slang term for the dissociation rate constant. anything before 1990, the year the rst Biacore biosensor was released. slang term for the association rate constant. testing several analyte concentrations in parallel. testing a ne dilution series of analyte concentrations. poorly behaved analytes that produce unrealistic responses. overlaid responses from repeated tests of the same analyte concentration. short-n-long kinetic analysis of stable complexes, in which the dissociation phase of only the highest analyte concentration is monitored for a long time. scaling-to-the-spikes syndrome. sound signifying derision directed towards anyone claiming biosensordetermined binding parameters never match those from solution-based experiments.
kinetic titration Michael Jackson data Myszkas Razor non-stoichiometric binder off rate old school on rate one-shot kinetics pearls-on-a-string analysis promiscuous binder replicates
BIOSENSOR PLEDGE
Now before we nish with todays exercises, we have homework for you. Cut out the Biosensor Pledge and tape it to your instrument. Each morning, before beginning your experiment, read it out loud so your labmates can hear. See you again next year.
Biosensor Pledge
1. I pledge to do my experiment right. 2. I pledge to report how I did my experiment. 3. I pledge to publish easily interpretable gures of my data that include replicates and an overlay of the t to a simple model. 4. I pledge to report the binding constants and standard errors correctly.
SNL approach
26
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
REFERENCES
Books, review articles and editorials
1. Abdulhalim I, Zourob M, Lakhtakia A. 2008. Surface plasmon resonance for biosensing: a mini-review. Electromagnetics 28: 214242. 2. Ahmed FE. 2008. Mining the oncoproteome and studying molecular interactions for biomarker development by 2DE, ChIP and SPR technologies. Expert Rev. Proteomics 5: 469496. 3. Borisov SM, Wolfbeis OS. 2008. Optical biosensors. Chem. Rev. 108: 423461. 4. Chen H, Yuan L, Song W, Wu Z, Li D. 2008. Biocompatible polymer materials: role of protein-surface interactions. Prog. Polym. Sci. 33: 10591087. 5. Comley J. 2008. Progress in the implementation of label-free detection. Drug Discov. World Fall: 2849. 6. De Crescenzo G, Boucher C, Durocher Y, Jolicoeur M. 2008. Kinetic characterization by surface plasmon resonance-based biosensors: principle and emerging trends. Cell. Mol. Bioeng. 1: 204215. 7. Derelov M, Detterfelt J, Bjorkman M, Mandenius C-F. 2008. Engineering design methodology for bio-mechatronic products. Biotechnol. Prog. 24: 232244. 8. Dolimbek BZ, Steward LE, Aoki KR, Atassi MZ. 2008. The binding sites on botulinum neurotoxin B for synaptotagmin and for blocking antibodies. Biochem. Biophys. Res. Commun. 376: 631632. 9. Fan X, White IM, Shopova SI, Zhu H, Suter JD, Sun Y. 2008. Sensitive optical biosensors for unlabeled targets: a review. Anal. Chim. Acta 620: 826. 10. Homola J. 2008. Surface plasmon resonance sensors for detection of chemical and biological species. Chem. Rev. 108: 462493. 11. Homola J, Yee SS, Myszka D. 2008. Surface plasmon resonance biosensors. In Optical Biosensors: Today and Tomorrow, Ligler FS, Taitt CR (eds). Elsevier B.V.: Amsterdam, The Netherlands; 683688. 12. Horing NJM, Cui HL. 2008. Surface-plasmon-resonance based optical biosensing. In Spectral Sensing Research for Water Monitoring Applications and Science and Technology for Chemical, Biological and Radiological Defense, Woolard D, Jensen J (eds). World Scientic Publishing Co. Pte. Ltd.: Singapore; 329336. 13. Karlinsey JM, Landers JP. 2008. Optical detection systems for microchips. In Handbook of Capillary and Microchip Electrophoresis and Associated Microtechniques, Landers JP (ed.). CRC Press: Boca Raton, FL; 12531276. 14. Luong JHT, Male KB, Glennon JD. 2008. Biosensor technology: technology push versus market pull. Biotechnol. Adv. 26: 492500. 15. Monch W, Zappe H. 2008. Biophotonics. In Comprehensive Microsystems, Yogesh G, Osamu T, Hans ZP (eds). Elsevier B.V.: Amsterdam, The Netherlands; 293321. 16. Orellana G, Haigh D. 2008. New trends in ber-optic chemical and biological sensors. Curr. Anal. Chem. 4: 273295. 17. Pace TCS, Bohne C. 2008. Dynamics of guest binding to supramolecular systems: techniques and selected examples. Adv. Phys. Org. Chem. 42: 167223. 18. Rich RL, Myszka DG. 2008. Survey of the year 2007 commercial optical biosensor literature. J. Mol. Recognit. 21: 355400. 19. Sadana A, Sadana N. 2008. Fractal Analysis of the Binding and Dissociation Kinetics for Different Analytes on Biosensor Surfaces. Elsevier B.V.: Amsterdam, The Netherlands. 20. Sheikh S, Blaszykowski C, Thompson M. 2008. Acoustic wave-based detection in bioanalytical chemistry: competition for surface plasmon resonance? Anal. Lett. 41: 25252538. 21. Tanious FA, Nguyen B, Wilson WD. 2008. Biosensor-surface plasmon resonance methods for quantitative analysis of biomolecular interactions. Methods Cell Biol. 84: 5377. 22. Tsapikouni TS, Missirlis YF. 2008. Protein-material interactions: from micro-to-nano scale. Mater. Sci. Eng. B 152: 27. 23. Wolfbeis OS. 2008. Fiber-optic chemical sensors and biosensors. Anal. Chem. 80: 42694283. 25. Friedrich D, Please C, Melvin T. 2008. Optimisation of analyte transport in integrated microuidic afnity sensors for the quantication of low levels of analyte. Sens. Actuat. B 131: 323332. 26. Gorshkova II, Svitel J, Razjouyan F, Schuck P. 2008. Bayesian analysis of heterogeneity in the distribution of binding properties of immobilized surface sites. Langmuir 24: 1157711586. 27. Squires T, Messinger R, Manalis S. 2008. Making it stick: convection, reaction and diffusion in surface-based biosensors. Nat. Biotechnol. 26: 417426. 28. Sytchkova AK. 2008. Measurement of optical constants of thin lms by non conventional ellipsometry, photothermal deection spectroscopy and plasmon resonance spectroscopy. Proc. SPIE 7101: 71010R-1.
Methods development
29. Bolivar JG, Soper SA, McCarley RL. 2008. Nitroavidin as a ligand for the surface capture and release of biotinylated proteins. Anal. Chem. 80: 93369342. 30. Clow F, Fraser JD, Proft T. 2008. Immobilization of proteins to biacore sensor chips using Staphylococcus aureus sortase A. Biotechnol. Lett. 30: 16031607. 31. De Crescenzo G, Woodward L, Srinivasan B. 2008. Online optimization of surface plasmon resonance-based biosensor experiments for improved throughput and condence. J. Mol. Recognit. 21: 256266. 32. Eddings MA, Miles AR, Eckman JW, Kim J, Rich RL, Gale BK, Myszka DG. 2008. Improved continuous-ow print head for micro-array deposition. Anal. Biochem. 382: 5559. 33. Natarajan S, Chang-Yen DA, Gale BK. 2008. Large-area, highaspect-ratio SU-8 molds for the fabrication of PDMS microuidic devices. J. Micromech. Microeng. 18: 045021. 34. Natarajan S, Hatch A, Myszka DG, Gale BK. 2008. Optimal conditions for protein array deposition using continuous ow. Anal. Chem. 80: 85618567. 35. Natarajan S, Katsamba PS, Miles A, Eckman J, Papalia GA, Rich RL, Gale BK, Myszka DG. 2008. Continuous-ow microuidic printing of proteins for array-based applications including surface plasmon resonance imaging. Anal. Biochem. 373: 141146. 36. Sexton BA, Feltis BN, Davis TJ. 2008. Characterisation of gold surface plasmon resonance sensor substrates. Sens. Actuators A 141: 471475. 37. Van Vlierberghe S, De Wael K, Buschop H, Adriaens A, Schacht E, Dubruel P. 2008. Ozonization and cyclic voltammetry as efcient methods for the regeneration of gelatin-coated SPR chips. Macromol. Biosci. 8: 10901097.
Instrument comparisons
38. Abdiche Y, Malashock D, Pinkerton A, Pons J. 2008. Determining kinetics and afnities of protein interactions using a parallel real-time label-free biosensor, the Octet. Anal. Biochem. 377: 209217. 39. Abdiche YN, Malashock DS, Pons J. 2008. Probing the binding mechanism and afnity of tanezumab, a recombinant humanized anti-NGF monoclonal antibody, using a repertoire of biosensors. Prot. Sci. 17: 13261335. 40. Cottier K, Horvath R. 2008. Imageless microscopy of surface patterns using optical waveguides. Appl. Phys. B 91: 319327. 41. Miura N, Shankaran D, Gobi K, Kawaguchi T, Kim S. 2008. An overview of the development and application of surface plasmon resonance based immunosensors for detection of small molecules. Sensor Lett. 6: 891902. 42. Xi B, Yu N, Wang X, Xu X, Abassi Y. 2008. The application of cell-based label-free technology in drug discovery. Biotechnol. J. 3: 484495.
27
www.interscience.wiley.com/journal/jmr
EcoChemie/Autolab
D 63. Atkins KL, Burman JD, Chamberlain ES, Cooper JE, Poutrel B, Bagby S, Jenkins ATA, Feil EJ, van den Elsen JMH. 2008. S. aureus IgG-binding proteins SpA and Sbi: host specicity and mechanisms of immune complex formation. Mol. Immunol. 45: 16001611. D 64. Aung KMM, Ho X, Su X. 2008. DNA assembly on streptavidin modied surface: a study using quartz crystal microbalance with dissipation or resistance measurements. Sens. Actuators B 131: 371378. D 65. Dong H, Cao X, Li CM, Hu W. 2008. An in situ electrochemical surface plasmon resonance immunosensor with polypyrrole propylic acid lm: comparison between SPR and electrochemical responses from polymer formation to protein immunosensing. Biosens. Bioelectron. 23: 10551062. F 66. Du Y, Li B, Wei H, Wang Y, Wang E. 2008. Multifunctional label-free electrochemical biosensor based on an integrated aptamer. Anal. Chem. 80: 51105117. D 67. Garca-Aljaro C, Munoz-Berbel X, Jenkins ATA, Blanch AR, Munoz FX. 2008. Surface plasmon resonance assay for real-time monitoring of somatic coliphages in wastewaters. Appl. Environ. Microbiol. 74: 40544058. D 68. Gehlot R, Sharma K, Mathew M, Kumbhat S. 2008. Surface plasmon resonance based biosensor for label free detection of cholesterol. Indian J. Chem. 47A: 18041808. D 69. Gondran C, Dubois M-P, Fort S, Cosnier S, Szunerits S. 2008. Detection of carbohydrate-binding proteins by oligosaccharidemodied polypyrrole interfaces using electrochemical surface plasmon resonance. Analyst 133: 206212. D 70. Hu W, Li CM, Dong H. 2008. Poly(pyrrole-co-pyrrole propylic acid) lm and its application in label-free surface plasmon resonance immunosensors. Anal. Chim. Acta 630: 6774. C 71. Jiang X, Cao Z, Tang H, Tan L, Xie Q, Yao S. 2008. Electrochemical surface plasmon resonance studies on the deposition of the charge-transfer complex from electrooxidation of o-tolidine and effects of dermatan sulfate. Electrochem. Commun. 10: 12351237. D 72. Kim SW, Kim M-G, Kim J, Lee H-S, Ro H-S. 2008. Detection of the mycovirus OMSV in the edible mushroom, Pleurotus ostreatus, using an SPR biosensor chip. J. Virol. Methods 148: 120124. D 73. Lee SJ, Youn B-S, Park JW, Niazi JH, Kim YS, Gu MB. 2008. ssDNA aptamer-based surface plasmon resonance biosensor for the detection of retinol binding protein 4 for the early diagnosis of type 2 diabetes. Anal. Chem. 80: 28672873. D 74. Manesse M, Stambouli V, Boukherroub R, Szunerits S. 2008. Electrochemical impedance spectroscopy and surface plasmon resonance studies of DNA hybridization on gold/SiOx interfaces. Analyst 133: 10971103. D 75. Matharu Z, Arya SK, Sumana G, Gupta V, Malhotra BD. 2008. Self-assembled monolayer for low density lipoprotein detection. J. Mol. Recognit. 21: 419424. D 76. McIlwee HA, Schauer CL, Praig VG, Boukherroub R, Szunerits S. 2008. Thin chitosan lms as a platform for SPR sensing of ferric ions. Analyst 133: 673677. D 77. Mendes RK, Carvalhal RF, Kubota LT. 2008. Effects of different self-assembled monolayers on enzyme immobilization procedures in peroxidase-based biosensor development. J. Electroanal. Chem. 612: 164172. F 78. Mir M, Jenkins ATA, Katakis I. 2008. Ultrasensitive detection based on an aptamer beacon electron transfer chain. Electrochem. Commun. 10: 15331536. F 79. Mir M, Katakis I. 2008. Target label-free, reagentless electrochemical DNA biosensor based on sub-optimum displacement. Talanta 75: 432441. F 80. Munoz-Berbel X, Vigues N, Jenkins ATA, Mas J, Munoz FJ. 2008. Impedimetric approach for quantifying low bacteria concentrations based on the changes produced in the electrode-solution interface during the pre-attachment stage. Biosens. Bioelectron. 23: 15401546. D 81. Prabhakar N, Arora K, Arya SK, Solanki PR, Iwamoto M, Singh H, Malhotra BD. 2008. Nucleic acid sensor for M. tuberculosis detection based on surface plasmon resonance. Analyst 133: 15871592. D 82. Ryu J, Joung HA, Kim MG, Park CB. 2008. Surface plasmon resonance analysis of Alzheimers b-amyloid aggregation on a solid surface: from monomers to fully-grown brils. Anal. Chem. 80: 24002407.
Biosensing Instruments
D 53. Choi S, Yang Y, Chae J. 2008. Surface plasmon resonance protein sensor using Vroman effect. Biosens. Bioelectron. 24: 893899. D 54. Du M, Zhou F. 2008. Postcolumn renewal of sensor surfaces for high-performance liquid chromatography-surface plasmon resonance detection. Anal. Chem. 80: 42254230. D 55. He J, Lin L, Zhang P, Spadola Q, Xi Z, Fu Q, Lindsay S. 2008. Transverse tunneling through DNA hydrogen bonded to an electrode. Nanoletters 8: 25302534. F 56. Hebeisen P, Kuhn B, Kohler P, Gubler M, Huber W, Kitas E, Schott B, Benz J, Joseph C, Ruf A. 2008. Allosteric FBPase inhibitors gain 105 times in potency when simultaneously binding two neighboring AMP sites. Bioorg. Med. Chem. Lett. 18: 47084712. D 57. Qu N, Wan B, Guo L-H. 2008. Label-free electrochemical differentiation of phosphorylated and non-phosphorylated peptide by electro-catalyzed tyrosine oxidation. Analyst 133: 12461249. D 58. Wain AJ, Do HNL, Mandal HS, Kraatz H-B, Zhou F. 2008. The inuence of molecular dipole moment on the redox-induced reorganization of a-helical peptide self-assembled monolayers: an electrochemical SPR investigation. J. Phys. Chem. C 112: 1451314519. D 59. Xin Y, Gao Y, Guo J, Chen Q, Xiang J, Zhou F. 2008. Real-time detection of Cu2 sequestration and release by immobilized apometallothioneins using SECM combined with SPR. Biosens. Bioelectron. 24: 369375.
DKK-TOA
D 60. Li Y, Ren J, Nakajima H, Kim BK, Soh N, Nakano K, Imato T. 2008. Flow sandwich immunoassay for specic anti-OVA IgG antibody by use of surface plasmon resonance sensor. Talanta 77: 473478. D 61. Masadome T, Yano Y, Imato T. 2008. Surface plasmon resonance immunosensor for anionic surfactants based on an indirect competitive immunoreaction. Anal. Lett. 41: 640648. C 62. Tanaka M, Li Y, Nakajima H, Soh N, Nakano K, Sakamoto K, Imato T. 2008. Sequential injection ow immunoassay based on surface plasmon resonance sensors. J. Flow Inject. Anal. 25: 172182.
28
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
D 105.
D 106.
D 107.
GE Healthcare/Biacore Proteins
D 90. Acebron SP, Fernandez-Saiz V, Taneva SG, Moro F, Muga A. 2008. DnaJ recruits DnaK to protein aggregates. J. Biol. Chem. 283: 13811390. D 91. Ambrosio AL, Taelman VF, Lee HX, Metzinger CA, Cofnier C, De Robertis EM. 2008. Crossveinless-2 is a BMP feedback inhibitor that binds chordin/BMP to regulate Xenopus embryonic patterning. Dev. Cell 15: 248260. F 92. Amemiya Y, Azmi P, Seth A. 2008. Autoubiquitination of BCA2 RING E3 ligase regulates its own stability and affects cell migration. Mol. Cancer Res. 6: 13851396. C 93. Amen M, Espinoza HM, Cox C, Liang X, Wang J, Link TME, Brennan RG, Martin JF, Amendt BA. 2008. Chromatin-associated HMG-17 is a major regulator of homeodomain transcription factor activity modulated by Wnt/b-catenin signaling. Nucleic Acids Res. 36: 462476. C 94. Amsili S, Zer H, Hinderlich S, Krause S, Becker-Cohen M, MacArthur DG, North KN, Mitrani-Rosenbaum S. 2008. UDPN-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE) binds to alpha-actinin 1: novel pathways in skeletal muscle? PLoS ONE 3: e2477. C 95. Anderka O, Boyken J, Aschenbach U, Batzer A, Boscheinen O, Schmoll D. 2008. Biophysical characterization of the interaction between hepatic glucokinase and its regulatory protein. J. Biol. Chem. 283: 3133331340. B 96. Aoki N, Sakiyama A, Kuroki K, Maenaka K, Kohda D, Deshimaru M, Terada S. 2008. Serotriin, a CRISP family protein with binding afnity for small serum protein-2 in snake serum. Biochim. Biophys. Acta 1784: 621628. F 97. Arrigoni G, Pagano MA, Sarno S, Cesaro L, James P, Pinna LA. 2008. Mass spectrometry analysis of a protein kinase CK2b subunit interactome isolated from mouse brain by afnity chromatography. J. Proteome Res. 7: 9901000. D 98. Attali C, Frolet C, Durmort C, Offant J, Vernet T, Di Guilmi AM. 2008. Streptococcus pneumoniae choline-binding protein E interaction with plasminogen/plasmin stimulates migration across the extracellular matrix. Infect. Immun. 76: 466476. C 99. Austermann J, Nazmi AR, Muller-Tidow C, Gerke V. 2008. Charac terization of the Ca2-regulated ezrin-S100P interaction and its role in tumor cell migration. J. Biol. Chem. 283: 2933129340. D 100. Bahar MW, Kenyon JC, Putz MM, Abrescia NGA, Pease JE, Wise EL, Stuart DI, Smith GL, Grimes JM. 2008. Structure and function of A41, a vaccinia virus chemokine binding protein. PLoS Pathog. 4: e5. A 101. Beattie J, Phillips K, Shand JH, Szymanowska M, Flint DJ, Allan GJ. 2008. Molecular interactions in the insulin-like growth factor (IGF)
D 111.
D 115.
C 116. B 117.
B 118.
B 119.
D 120.
29
www.interscience.wiley.com/journal/jmr
F 121.
D 122.
F 123.
D 124. D 125.
B 126.
D 130.
C 131. F 132.
B 133.
B 134.
D 135.
C 136.
B 137. F 138.
30
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
31
www.interscience.wiley.com/journal/jmr
D 221. D 222.
D 223.
F 227. C 228.
D 229.
D 236.
D 237.
D 238.
32
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
33
www.interscience.wiley.com/journal/jmr
D 300.
D 301.
F 302. D 303.
B 304. F 305.
B 306. C 307.
C 308.
C 309.
F 310. C 311.
F 312.
B 313.
D 314.
34
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
C 335. C 336.
D 337.
F 338. F 339.
F 340. F 341.
F 342.
B 343.
D 344. F 345.
F 346.
F 347.
B 348.
F 349.
F 350.
Antibodies
F 332. Adekar SP, Jones RM, Elias MD, Al-Saleem FH, Root MJ, Simpson LL, Dessain SK. 2008. Hybridoma populations enriched for afnitymatured human IgGs yield high-afnity antibodies specic for botulinum neurotoxins. J. Immunol. Methods 333: 156166. B 333. Akimoto T, Wada S, Karube I. 2008. A surface plasmon resonance probe without optical bers as a portable sensing device. Anal. Chim. Acta 610: 119124. C 334. Alam SM, Scearce RM, Parks RJ, Plonk K, Plonk SG, Sutherland LL, Gorny MK, Zolla-Pazner S, VanLeeuwen S, Moody MA, Xia S-M, Monteori DC, Tomaras GD, Weinhold KJ, Karim SA, Hicks CB, Liao H-X, Robinson J, Shaw GM, Haynes BF. 2008. Human immunodeD 351. C 352.
B 353.
35
www.interscience.wiley.com/journal/jmr
F 375.
C 376.
C 377.
F 378.
B 379. C 380.
F 381.
F 382.
F 383.
B 384.
F 385.
F 386.
F 387.
F 388. F 389.
36
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
C 390. D 391.
F 392.
F 393.
C 394.
F 395.
F 396.
F 397.
C 398. B 399.
C 400.
F 401.
C 402.
C 403. B 404.
D 405. F 406.
37
www.interscience.wiley.com/journal/jmr
F 425.
D 426.
D 427. D 428.
D 429.
D 430.
F 431.
F 432.
D 433.
F 434.
F 435. D 436.
F 437.
F 438.
38
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
39
www.interscience.wiley.com/journal/jmr
F 496.
D 512.
D 497.
B 513. B 514.
C 498.
F 515.
D 499. F 500.
C 516.
A 501.
D 517. F 518.
D 502.
D 519.
F 503. F 504.
D 520.
Receptors
B 505. Adams E, Strop P, Shin S, Chien YH, Garcia KC. 2008. An autonomous CDR3d is sufcient for recognition of the nonclassical MHC class I molecules T10 and T22 by gd T cells. Nat. Immunol. 9: 777784. F 506. Ahlgren S, Orlova A, Rosik D, Sandstrom M, Sjoberg A, Baastrup B, Widmark O, Fant G, Feldwisch J, Tolmachev V. 2008. Evaluation of maleimide derivative of DOTA for site-specic labeling of recombinant afbody molecules. Bioconjug. Chem. 19: 235243. D 507. Ambjrn M, Asmussen JW, Lindstam M, Gotfryd K, Jacobsen C, Kiselyov VV, Moestrup SK, Penkowa M, Bock E, Berezin V. 2008. Metallothionein and a peptide modeled after metallothionein, EmtinB, induce neuronal differentiation and survival through binding to receptors of the low-density lipoprotein receptor family. J. Neurochem. 104: 2137. D 508. An X, Gauthier E, Zhang X, Guo X, Anstee DJ, Mohandas N, Chasis JA. 2008. Adhesive activity of Lu glycoproteins is regulated by interaction with spectrin. Blood 112: 52125218. C 509. Ananyeva NM, Makogonenko YM, Sarafanov AG, Pechik IV, Gorlatova N, Radtke KP, Shima M, Saenko EL. 2008. Interaction of coagulation factor VIII with members of the low-density lipoprotein receptor family follows common mechanism and involves consensus residues within the A2 binding site 484509. Blood Coagul. Fibrinolysis 19: 543555. C 510. Ananyeva NM, Makogonenko YM, Kouiavskaia DV, Ruiz J, Limburg V, Meijer AB, Khrenov AV, Shima M, Strickland DK, Saenko EL. 2008. The binding sites for the very low density lipoprotein receptor and low-density lipoprotein receptor-related protein are shared within coagulation factor VIII. Blood Coagul. Fibrinolysis 19: 166177. D 511. Andersen J, Justesen S, Fleckenstein B, Michaelsen T, Berntzen G, Kenanova VE, Daba MB, Lauvrak V, Buus S, Sandlie I. 2008. Ligand
D 521.
D 522.
D 523.
D 524.
D 528.
40
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
41
www.interscience.wiley.com/journal/jmr
D 566. F 567.
C 586. C 587.
C 588.
F 568. C 569.
B 589.
D 570.
F 590.
C 571. F 572.
D 591.
C 573.
D 592.
C 574.
B 593. D 594.
D 575.
A 576. C 577.
B 595.
F 596.
F 578.
B 597. D 598.
F 579. B 580.
D 599.
B 581.
F 600.
B 601.
C 602.
D 603.
42
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
C 604. D 605.
F 606.
D 607.
B 608. D 609.
D 610.
C 617.
D 618.
F 619.
C 620.
D 621.
43
www.interscience.wiley.com/journal/jmr
F 639.
D 640.
C 641.
C 642.
D 643.
B 644.
F 645.
B 646. F 647.
F 652.
C 653.
C 654.
44
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
B 673.
D 674.
C 675.
Peptides
C 692. Ampapathi RS, Creath AL, Lou DI, Craft JW Jr, Blanke SR, Legge GB. 2008. Order-disorder-order transitions mediate the activation of cholera toxin. J. Mol. Biol. 377: 748760. D 693. Andersen L, Wind T, Hansen H, Andreasen P. 2008. A cyclic peptidylic inhibitor of murine urokinase-type plasminogen activator: changing species specicity by substitution of a single residue. Biochem. J. 412: 447457. F 694. Andjelic CD, Planelles V, Barrows LR. 2008. Characterizing the anti-HIV activity of papuamide A. Mar. Drugs 6: 528549. B 695. Austin RJ, Ja WW, Roberts RW. 2008. Evolution of class-specic peptides targeting a hot spot of the Gas subunit. J. Mol. Biol. 377: 14061418. F 696. Auvynet C, El Amri C, Lacombe C, Bruston F, Bourdais J, Nicolas P, Rosenstein Y. 2008. Structural requirements for antimicrobial versus chemoattractant activities for dermaseptin S9. FEBS J. 275: 41344151. B 697. Benard SA, Smith TM, Cunningham K, Jacob J, DeSilva T, Lin L, Shaw GD, Kriz R, Kelleher KS. 2008. Identication of peptide antagonists to glycoprotein Iba that selectively inhibit von Willebrand factor dependent platelet aggregation. Biochemistry 47: 46744682. C 698. Beverly KN, Sawaya MR, Schmid E, Koehler CM. 2008. The Tim8Tim13 complex has multiple substrate binding sites and binds cooperatively to Tim23. J. Mol. Biol. 382: 11441156. F 699. Bookwalter JE, Jurcisek JA, Gray-Owen SD, Fernandez S, McGillivary G, Bakaletz LO. 2008. A carcinoembryonic antigen-related cell adhesion molecule 1 homologue plays a pivotal role in nontypeable Haemophilus inuenzae colonization of the chinchilla nasopharynx via the outer membrane protein P5-homologous adhesin. Infect. Immun. 76: 4855. D 700. Brewster LP, Washington C, Brey EM, Gassman A, Subramanian A, Calceterra J, Wolf W, Hall CL, Velander WH, Burgess WH, Greisler HP. 2008. Construction and characterization of a thrombinresistant designer FGF-based collagen binding domain angiogen. Biomaterials 29: 327336. D 701. Buxbaum JN, Ye Z, Reixach N, Friske L, Levy C, Das P, Golde T, Masliah E, Roberts AR, Bartfai T. 2008. Transthyretin protects Alzheimers mice from the behavioral and biochemical effects of Ab toxicity. Proc. Natl Acad. Sci. USA 105: 26812686. D 702. Cabaleiro-Lago C, Quinlan-Pluck F, Lynch I, Lindman S, Minogue AM, Thulin E, Walsh DM, Dawson KA, Linse S. 2008. Inhibition of amyloid b protein brillation by polymeric nanoparticles. J. Am. Chem. Soc. 130: 1543715443. D 703. Cao W, Bao C, Padalko E, Lowenstein CJ. 2008. Acetylation of mitogen-activated protein kinase phosphatase-1 inhibits Toll-like receptor signaling. J. Exp. Med. 205: 14911503. D 704. Cendron AC, Wines BD, Brownlee RTC, Ramsland PA, Pietersz GA, Hogarth PM. 2008. An FcgRIIa-binding peptide that mimics the interaction between FcgRIIa and IgG. Mol. Immunol. 45: 307319. D 705. Champion EA, Lane BH, Jackrel ME, Regan L, Baserga SJ. 2008. A direct interaction between the Utp6 half-a-tetratricopeptide repeat domain and a specic peptide in Utp21 is essential for efcient pre-rRNA processing. Mol. Cell. Biol. 28: 65476556. F 706. Cortajarena AL, Yi F, Regan L. 2008. Designed TPR modules as novel anticancer agents. ACS Chem. Biol. 3: 161166. B 707. Cushing PR, Fellows A, Villone D, Boisguerin P, Madden DR. 2008. The relative binding afnities of PDZ partners for CFTR: a biochemical basis for efcient endocytic recycling. Biochemistry 47: 1008410098. F 708. Deroo S, Fischer A, Beaupain N, Counson M, Boutonnet N, Pletinckx J, Loverix S, Beirnaert E, De Haard H, Schmit J-C, Lasters I. 2008. Non-immunized natural human heavy chain CDR3 repertoires allow the isolation of high afnity peptides mimicking a human inuenza hemagglutinin epitope. Mol. Immunol. 45: 13661373.
D 676.
B 677.
D 678.
F 679.
F 680. F 681.
b 682.
D 683.
F 684.
F 685. C 686.
C 687.
D 688.
F 689.
45
www.interscience.wiley.com/journal/jmr
B 728. F 729.
C 737.
D 738. D 739.
F 740.
C 741.
F 742.
D 743.
B 744.
D 745.
46
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
47
www.interscience.wiley.com/journal/jmr
Oligonucleotides
D 812. Adhikari S, Kennel SJ, Roy G, Mitra PS, Mitra S, Roy R. 2008. Discrimination of lesion removal of N-methylpurine-DNA glycosylase revealed by a potent neutralizing monoclonal antibody. DNA Repair 7: 3139. C 813. Adhikari S, Manthena PV, Uren A, Roy R. 2008. Expression, purication and characterization of codon-optimized human N-methylpurine-DNA glycosylase from Escherichia coli. Protein Expr. Purif. 58: 257262. C 814. Adhikari S, Uren A, Roy R. 2008. Dipole-dipole interaction stabilizes the transition state of apurinic/apyrimidinic endonucleaseabasic site interaction. J. Biol. Chem. 283: 13341339. D 815. Ahel I, Ahel D, Matsusaka T, Clark AJ, Pines J, Boulton SJ, West SC. 2008. Poly(ADP-ribose)-binding zinc nger motifs in DNA repair/ checkpoint proteins. Nature 451: 8185. F 816. Arora A, Kaur H, Wengel J, Maiti S. 2008. Effect of locked nucleic acid (LNA) modication on hybridization kinetics of DNA duplex. Nucleic Acids Symp. 52: 417418. D 817. Arora A, Maiti S. 2008. Effect of loop orientation on quadruplex TMPyP4 interaction. J. Phys. Chem. B 112: 81518159. C 818. Arraiano CM, Barbas A, Amblar M. 2008. Characterizing ribonucleases in vitro: examples of synergies between biochemical and structural analysis. Meth. Enzymol. 447: 131160. D 819. Balamurugan S, Obubuafo A, McCarley RL, Soper SA, Spivak DA. 2008. Effect of linker structure on surface density of aptamer monolayers and their corresponding protein binding efciency. Anal. Chem. 80: 96309634. D 820. Barbas A, Matos RG, Amblar M, Lopez-Vinas E, Gomez-Puertas P, Arraiano CM. 2008. New insights into the mechanism of RNA degradation by ribonuclease II. J. Biol. Chem. 283: 1307013076. ` B 821. Bessiere D, Lacroix C, Campagne S, Ecochard V, Guillet V, Mourey L, Lopez F, Czaplicki J, Demange P, Milon A, Girard J-P, Gervais V. 2008. Structure-function analysis of the THAP zinc nger of THAP1, a large C2CH DNA-binding module linked to Rb/E2F pathways. J. Biol. Chem. 283: 43524363. D 822. Cohen C, Forzan M, Sproat B, Pantophlet R, McGowan I, Burton D, James W. 2008. An aptamer that neutralizes R5 strains of HIV-1 binds to core residues of gp120 in the CCR5 binding site. Virology 381: 4654. D 823. Cyr JL, Heinen CD. 2008. Hereditary cancer-associated missense mutations in hMSH6 Duncouple ATP hydrolysis from DNA mismatch binding. J. Biol. Chem. 283: 3164161648.
48
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
C 844.
F 851. B 852.
D 856. B 857.
F 858.
D 859. C 860.
D 861.
F 862.
F 863. B 864.
49
www.interscience.wiley.com/journal/jmr
C 898. F 899.
Small molecules
D 904. Albert JS, Edwards PD. 2008. Identication of high-afnity b-secretase inhibitors using fragment-based lead generation. In Fragment-Based Drug Discovery: A Practical Approach, Zartler EA, Shapiro MJ (eds). John Wiley & Sons, Ltd: Malden, MA; 261279. D 905. Angell RM, Angell TD, Bamborough P, Bamford MJ, Chung C-w, Cockerill SG, Flack SS, Jones KL, Laine DI, Longstaff T, Ludbrook S, Pearson R, Smith KJ, Smee PA, Somers DO, Walker AL. 2008. Biphenyl amide p38 kinase inhibitors 4: DFG-in and DFG-out binding modes. Bioorg. Med. Chem. Lett. 18: 44334437. F 906. Antonysamy SS, Aubol B, Blaney J, Browner MF, Giannetti AM, Harris SF, Hebert N, Hendle J, Hopkins S, Jefferson E, Kissinger C, Leveque V, Marciano D, McGee E, Najera I, Nolan B, Tomimoto M, Torres E, Wright T. 2008. Fragment-based discovery of hepatitis C virus NS5b RNA polymerase inhibitors. Bioorg. Med. Chem. Lett. 18: 29902995.
50
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
51
www.interscience.wiley.com/journal/jmr
52
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
Carbohydrates
F 984. Adams EL, Rice PJ, Graves B, Ensley HE, Yu H, Brown GD, Gordon S, Monteiro MA, Papp-Szabo E, Lowman DW, Power TD, Wempe MF, Williams DL. 2008. Differential high-afnity interaction of dectin-1 with natural or synthetic glucans is dependent upon primary structure and is inuenced by polymer chain length and sidechain branching. J. Pharmacol. Exp. Ther. 325: 115123. D 985. Ai J, Xin X, Zheng M, Wang S, Peng S, Li J, Wang L, Jiang H, Geng M. 2008. A triad of Lys12, Lys41, Arg78 spatial domain, a novel identied heparin binding site on Tat protein, facilitates Tat-driven cell adhesion. PLos ONE 3: 2662. C 986. Becerra SP, Perez-Mediavilla LA, Weldon JE, Locatelli-Hoops S, Senanayake P, Notari L, Notario V, Hollyeld JG. 2008. Pigment epithelium-derived factor binds to hyaluronan. J. Biol. Chem. 283: 3331033320. B 987. Brooks CL, Muller-Loennies S, Brade L, Kosma P, Hirama T, MacK enzie CR, Brade H, Evans SV. 2008. Exploration of specicity in germline monoclonal antibody recognition of a range of natural and synthetic epitopes. J. Mol. Biol. 377: 450468. D 988. Byres E, Paton AW, Paton JC, Loing JC, Smith DF, Wilce MCJ, Talbot UM, Chong DC, Yu H, Huang S, Chen X, Varki NM, Varki A, Rossjohn J, Beddoe T. 2008. Incorporation of a non-human glycan mediates human susceptibility to a bacterial toxin. Nature 456: 648652. D 989. Cain SA, Baldwin AK, Mahalingam Y, Raynal B, Jowitt TA, Shuttleworth CA, Couchman JR, Kielty CM. 2008. Heparan sulfate regulates brillin-1 N- and C-terminal interactions. J. Biol. Chem. 283: 2701727027. D 990. Chatterjee U, Bose PP, Dey S, Singh TP, Chatterjee BP. 2008. Antiproliferative effect of T/Tn specic Artocarpus lakoocha agglutinin (ALA) on human leukemic cells (Jurkat, U937, K562) and their imaging by QD-ALA nanoconjugate. Glycoconj. J. 25: 741752. F 991. Clarke AJ, Hurtado-Guerrero R, Pathak S, Schuttelkopf AW, Bor odkin V, Shepherd SM, Ibrahim AFM, van Aalten DMF. 2008. Structural insights into mechanism and specicity of O-GlcNAc transferase. EMBO J. 27: 27802788. C 992. Cle C, Gunning AP, Syson K, Bowater L, Field RA, Bornemann S. 2008. Detection of transglucosidase-catalyzed polysaccharide synthesis on a surface in real time using surface plasmon resonance spectroscopy. J. Am. Chem. Soc. 130: 1523415235. D 993. Collot M, Sendid B, Fievez Al, Savaux C, Standaert-Vitse A, Tabouret M, Drucbert AS, Marie Danze P, Poulain D, Mallet J-M. 2008. Biotin sulfone as a new tool for synthetic oligosaccharide immobilization: application to multiple analysis proling and surface plasmonic analysis of anti-Candida albicans antibody reactivity against a and b (1 ! 2) oligomannosides. J. Med. Chem. 51: 62016210. C 994. Coombe DR, Stevenson SM, Kinnear BF, Gandhi NS, Mancera RL, Osmond RI, Kett WC. 2008. Platelet endothelial cell adhesion molecule 1 (PECAM-1) and its interactions with glycosaminoglycans: 2. biochemical analyses. Biochemistry 47: 48634875. D 995. Demolliens A, Boucher C, Durocher Y, Jolicoeur M, Buschmann MD, De Crescenzo G. 2008. Tyrosinase-catalyzed synthesis of a
53
www.interscience.wiley.com/journal/jmr
D 1015.
D 1033.
D 1016. C 1017.
F 1034.
D 1035.
C 1018.
C 1036.
D 1019.
C 1037.
D 1020.
F 1038.
D 1021.
C 1039.
D 1022.
D 1040.
C 1023.
D 1041. C 1042.
D 1024.
D 1043.
F 1044.
Extracellular matrix
D 1045. Adams JC, Bentley AA, Kvansakul M, Hatherley D, Hohenester E. 2008. Extracellular matrix retention of thrombospondin 1 is controlled by its conserved C-terminal region. J. Cell Sci. 121: 784795. B 1046. Esgleas M, Li Y, Hancock MA, Harel J, Dubreuil JD, Gottschalk M. 2008. Isolation and characterization of a-enolase, a novel bronectin-binding protein from Streptococcus suis. Microbiology 154: 26682679. C 1047. Fenton KA, Mjelle JE, Jakobsen S, Olsen R, Rekvig OP. 2008. Renal expression of polyomavirus large T antigen is associated with nephritis in human systemic lupus erythematosus. Mol. Immunol. 45: 31173124. C 1048. Gronwald W, Bomke J, Maurer T, Domogalla B, Huber F, Schumann F, Kremer W, Fink F, Rysiok T, Frech M, Kalbitzer HR. 2008. Structure of the leech protein Saratin and characterization of its binding to collagen. J. Mol. Biol. 381: 913927. D 1049. Hussain M, Haggar A, Peters G, Chhatwal GS, Herrmann M, Flock J-I, Sinha B. 2008. More than one tandem repeat domain of
C 1029.
C 1030.
C 1031.
C 1032.
54
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
D 1053.
D 1054.
C 1059.
B 1060.
D 1061. D 1062.
55
www.interscience.wiley.com/journal/jmr
56
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
B 1129. D 1130.
C 1131.
B 1137.
F 1138.
F 1139.
D 1140.
D 1144.
D 1148.
57
www.interscience.wiley.com/journal/jmr
Clinical support
B 1177. Boghaert ER, Khandke KM, Sridharan L, Dougher M, DiJoseph JF, Kunz A, Hamann PR, Moran J, Chaudhary I, Damle NK. 2008. Determination of pharmacokinetic values of calicheamicin-antibody conjugates in mice by plasmon resonance analysis of small (5 ml) blood samples. Cancer Chemother. Pharmacol. 61: 10271035. F 1178. Bright RA, Carter DM, Crevar CJ, Toapanta FR, Steckbeck JD, Cole KS, Kumar NM, Pushko P, Smith G, Tumpey TM, Ross TM. 2008. Cross-clade protective immune responses to inuenza viruses with H5N1 HA and NA elicited by an inuenza virus-like particle. PLos ONE 3: 1501. F 1179. Evans PA, Hawkins K, Lawrence M, Williams RL, Barrow MS, Thirumalai N, Williams PR. 2008. Rheometry and associated techniques for blood coagulation studies. Med. Eng. Phys. 30: 6716679. C 1180. Gibbs E, Oger J. 2008. A biosensor-based characterization of the afnity maturation of the immune response against interferon-b and correlations with neutralizing antibodies in treated multiple sclerosis patients. J. Interferon Cytokine Res. 28: 713724. F 1181. Landsberger M, Staudt A, Choudhury S, Trimpert C, Herda LR, Klingel K, Kandolf R, Schultheiss H-P, Kroemer HK, Volker U, Felix SB. 2008. Potential role of antibodies against cardiac Kv channel-interacting protein 2 in dilated cardiomyopathy. Am. Heart J. 156: 9299.e2. F 1182. Latrofa F, Ricci D, Grasso L, Vitti P, Masserini L, Basolo F, Ugolini C, Mascia G, Lucacchini A, Pinchera A. 2008. Characterization of thyroglobulin epitopes in patients with autoimmune and nonautoimmune thyroid diseases using recombinant human monoclonal thyroglobulin autoantibodies. J. Clin. Endocrinol. Metab. 93: 591596. F 1183. Mahmood K, Bright RA, Mytle N, Carter DM, Crevar CJ, Achenbach JE, Heaton PM, Tumpey TM, Ross TM. 2008. H5N1 VLP vaccine induced protection in ferrets against lethal challenge with highly pathogenic H5N1 inuenza viruses. Vaccine 26: 53935399. F 1184. Mao W, Iwai C, Liu J, Sheu S-S, Fu M, Liang C-s. 2008. Darbepoetin alfa exerts a cardioprotective effect in autoimmune cardiomyopathy via reduction of ER stress and activation of the PI3K/Akt and STAT3 pathways. J. Mol. Cell. Cardiol. 45: 250260.
58
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
F 1204.
Other applications
C 1225. Bingel-Erlenmeyer R, Kohler R, Kramer G, Sandikci A, Antolic S, Maier T, Schaftzel C, Wiedmann B, Bukau B, Ban N. 2008. A peptide deformylase-ribosome complex reveals mechanism of nascent chain processing. Nature 452: 108112. C 1226. Cain SA, Raynal B, Hodson N, Shuttleworth A, Kielty CM. 2008. Biomolecular analysis of elastic bre molecules. Methods 45: 4252. D 1227. Chiou J-C, Li X-P, Remacha M, Ballesta JPG, Tumer NE. 2008. The ribosomal stalk is required for ribosome binding, depurination of the rRNA and cytotoxicity of ricin A chain in Saccharomyces cerevisiae. Mol. Microbiol. 70: 14411452. D 1228. Lunder M, Bratkovic T, Anderluh G, Strukelj B, Kreft S. 2008. Afnity ranking of phage-displayed peptides: enzyme-linked immunosorbent assay versus surface plasmon resonance. Acta Chim. Slov. 55: 233235. C 1229. Miller J, Le Coq J, Hodes A, Barbalat R, Miller J, Ghosh P. 2008. Selective ligand recognition by a diversity-generating retroelement variable protein. PLoS Biol. 6: e131. C 1230. Tchesnokova V, Aprikian P, Yakovenko O, LaRock C, Kidd B, Vogel V, Thomas W, Sokurenko E. 2008. Integrin-like allosteric properties of the catch bond-forming FimH adhesin of Escherichia coli. J. Biol. Chem. 283: 78237833.
Crude analytes
A 1212. Chavane N, Jacquemart R, Hoemann CD, Jolicoeur M, De Crescenzo G. 2008. At-line quantication of bioactive antibody in bioreactor by surface plasmon resonance using epitope detection. Anal. Biochem. 378: 158165. B 1213. Jacquemart R, Chavane N, Durocher Y, Hoemann C, De Crescenzo G, Jolicoeur M. 2008. At-line monitoring of bioreactor protein production by surface plasmon resonance. Biotechnol. Bioeng. 100: 184188. D 1214. Piggott A, Karuso P. 2008. Rapid identication of a protein binding partner for the marine natural product kahalalide F by using reverse chemical proteomics. ChemBioChem 9: 524530. F 1215. Piletska EV, Villoslada FN, Chianella I, Bossi A, Karim K, Whitcombe MJ, Piletsky SA, Doucette GJ, Ramsdell JS. 2008. Extraction of domoic acid from seawater and urine using a resin based on 2-(triuoromethyl)acrylic acid. Anal. Chim. Acta 610: 3543. D 1216. Shafqat J, Ishrat M, Jagerbrink T, Sillard R, Maeorg U, Efendic S, Berggren P-O, Zaitsev SV, Jornvall H. 2008. Proteins in the insulin-secreting cell line MIN6 bind the imidazoline compound BL11282. FEBS Lett. 582: 16131617. D 1217. Takashina K, Bessho T, Mori R, Kawai K, Eguchi J, Saito K-I. 2008. MKC-231, a choline uptake enhancer: (3) mode of action of MKC-231 in the enhancement of high-afnity choline uptake. J. Neural Trasm. 115: 10371046. D 1218. Thongborisute J, Takeuchi H. 2008. Evaluation of mucoadhesiveness of polymers by BIACORE method and mucin-particle method. Int. J. Pharm. 354: 204209.
IBIS
D 1231. Kuil J, van Wandelen LTM, de Mol NJ, Liskamp RMJ. 2008. A photoswitchable ITAM peptidomimetic: synthesis and real time surface plasmon resonance (SPR) analysis of the effects of cis-trans isomerization on binding. Bioorg. Med. Chem. 16: 13931399.
K-MAC
D 1232. Chen H, Cheng H, Lee J, Kim J-H, Hyun MH, Koh K. 2008. Surface plasmon resonance spectroscopic chiral discrimination using self-assembled leucine derivative monolayer. Talanta 76: 4953. F 1233. Cho H, Jang D, Lee S, Ku S, Kim H, Yu K, Lee J. 2008. Characterization of novel self assembled materials (SAM) for surface modication of sensor chip. Biochip J. 2: 206211. D 1234. Kim H-C, Lee S-K, Jeon WB, Lyu H-K, Lee SW, Jeong SW. 2008. Detection of C-reactive protein on a functional poly(thiophene) self-assembled monolayer using surface plasmon resonance. Ultramicroscopy 108: 13791383. D 1235. Kim S-W, Kim M-G, Kim J, Lee H-S, Ro H-S. 2008. Detection of the mycovirus OMSV in the edible mushroom, Pleurotus ostreatus, using an SPR biosensor chip. J. Virol. Methods 148: 120124. F 1236. Lee S-K, Kim H-C, Cho S-J, Jeong SW, Jeon WB. 2008. Binding behavior of CRP and anti-CRP antibody analyzed with SPR and AFM measurement. Ultramicroscopy 108: 13741378.
59
www.interscience.wiley.com/journal/jmr
NanoSPR
D 1261. Akkilic N, Mustafaev M, Chegel V. 2008. Conformational dynamics of poly(acrylic acid)-bovine serum albumin polycomplexes at different pH conditions. Macromol. Symp. 269: 138144. D 1262. Benilova I, Chegel VI, Ushenin YV, Vidic J, Soldatkin AP, Martelet C, Pajot E, Jaffrezic-Renault N. 2008. Stimulation of human olfactory receptor 1740 with odorants probed by surface plasmon resonance. Eur. Biophys. J. 37: 807814. D 1263. Chegel V, Chegel Y, Guiver MD, Lopatynskyi A, Lopatynska O, Lozovski V. 2008. 3D-quantication of biomolecular covers using surface plasmon-polariton resonance experiment. Sens. Actuators B 134: 6671. D 1264. Uematsu T, Kuwabata S. 2008. In situ surface plasmon resonance measurements of self-assembled monolayers of ferrocenylalkylthiols under constant potentials. Anal. Sci. 24: 307312.
NeoSensors
C 1265. Alev C, Urschel S, Sonntag S, Zoidl G, Fort AG, Hoher T, Matsubara M, Willecke K, Spray DC, Dermietzel R. 2008. The neuronal connexin36 interacts with and is phosphorylated by CaMKII in a way similar to CaMKII interaction with glutamate receptors. Proc. Natl Acad. Sci. USA 105: 2096420969. D 1266. Atsumi S, Inoue Y, Ishizaka T, Mizuno E, Yoshizawa Y, Kitami M, Sato R. 2008. Location of the Bombyx mori 175kDa cadherin-like protein-binding site on Bacillus thuringiensis Cry1Aa toxin. FEBS J. 275: 49134926. D 1267. Benadie Y, Deysel M, Siko DGR, Roberts VV, Van Wyngaardt S, Thanyani ST, Sekanka G, Ten Bokum AMC, Collett LA, Grooten J, Baird MS, Verschoor JA. 2008. Cholesteroid nature of free mycolic acids from M. tuberculosis. Chem. Phys. Lipids 152: 95103. D 1268. Catlow KR, Deakin JA, Wei Z, Delehedde M, Fernig DG, Gherardi E, Gallagher JT, Pavao MSG, Lyon M. 2008. Interactions of hepatocyte growth factor/scatter factor with various glycosaminoglycans reveal an important interplay between the presence of iduronate and sulfate density. J. Biol. Chem. 283: 52355248. D 1269. Chen H, He X, Wang Z, Wu D, Zhang H, Xu C, He H, Cui L, Ba D, He W. 2008. Identication of human T cell receptor gd-recognized epitopes/proteins via CDR3d peptide-based immunobiochemical strategy. J. Biol. Chem. 283: 1252812537. C 1270. Cuccioloni M, Mozzicafreddo M, Barocci S, Ciuti F, Pecorelli I, Eleuteri AM, Spina M, Fioretti E, Angeletti M. 2008. Biosensorbased screening method for the detection of aatoxins B1G1. Anal. Chem. 80: 92509256. D 1271. Hamma-Kourbali Y, Bernard-Pierrot I, Heroult M, Dalle S, Caruelle D, Milhiet PE, Fernig DG, Delbe J, Courty J. 2008. Inhibition of the mitogenic, angiogenic and tumorigenic activities of pleiotrophin by a synthetic peptide corresponding to its C-thrombospondin repeat-I domain. J. Cell. Physiol. 214: 250259. D 1272. Huang J, Lin Q, Yu J, Ge S, Li J, Yu M, Zhao Z, Wang X, Zhang X, He X, Yuan L, Yin H, Osa T, Chen K, Chen Q. 2008. Comparison of a resonant mirror biosensor (IAsys) and a quartz crystal microbalance (QCM) for the study on interaction between Paeoniae Radix 801 and endothelin-1. Sensors 8: 82758290. D 1273. Ismail TM, Fernig DG, Rudland PS, Terry CJ, Wang G, Barraclough R. 2008. The basic C-terminal amino acids of calcium-binding protein S100A4 promote metastasis. Carcinogenesis 29: 22592266. F 1274. Ivanov YD, Ivanov AV, Petushkova NA, Gara OG, Kuznetsov VY, Podoplelov AV, Archakov AI. 2008. The optical biosensor study of the redox partner interaction with the cytochrome P450 2B4-containing monooxygenase system under hydroxylation conditions. Biochemistry (Moscow) 2: 367372. D 1275. Kabir-Salmani M, Fukuda MN, Kanai-Azuma M, Ahmed N, Shiokawa S, Akimoto Y, Sakai K, Nagamori S, Kanai Y, Sugihara K, Iwashita M. 2008. The membrane-spanning domain of CD98 heavy chain promotes avb3 integrin signals in human extravillous trophoblasts. Mol. Endocrin. 22: 707715. C 1276. Kaur KJ, Sarkar P, Nagpal S, Khan T, Salunke DM. 2008. Structure-function analyses involving palindromic analogs of tritrypticin suggest autonomy of anti-endotoxin and antibacterial activities. Prot. Sci. 17: 545554. F 1277. Li B, Zhang R, Li J, Zhang L, Ding G, Luo P, He S, Dong Y, Jiang W, Lu Y, Cao H, Zheng J, Zhou H. 2008. Antimalarial artesunate protects sepsis model mice against heat-killed Escherichia coli challenge
Nanolm Technology
B 1259. Klenkar G, Liedberg B. 2008. A microarray chip for label-free detection of narcotics. Anal. Bioanal. Chem. 391: 1679 1688. C 1260. Klenkar G, Brian B, Ederth T, Stengel G, Hook F, Piehler J, Liedberg B. 2008. Addressable adsorption of lipid vesicles and subsequent protein interaction studies. Biointerphases 3: 2937.
60
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
D 1278.
C 1279. D 1280.
D 1281.
D 1282.
D 1283.
D 1284.
F 1285. C 1286.
D 1287.
D 1288. D 1289.
NTT-AT
C 1309. Kurita R, Hirata Y, Yabuki S, Yokota Y, Kato D, Sato Y, Mizutani F, Niwa O. 2008. Surface modication of thin polyion complex lm for surface plasmon resonance immunosensor. Sens. Actuators B 130: 320325. D 1310. Kurita R, Nakamoto K, Ueda A, Niwa O. 2008. Comparison of electrochemical and surface plasmon resonance immunosensor responses on single thin lm. Electroanalysis 20: 22412246. F 1311. Nakamoto K, Kurita R, Sekioka N, Niwa O. 2008. Simultaneous on-chip surface plasmon resonance measurement of disease marker protein and small metabolite combined with immunoand enzymatic reactions. Chem. Lett. 37: 698699.
F 1290.
D 1291.
F 1292. F 1293.
Optrel
D 1312. Choi J-W, Kim YJ, Kim S-U, Min J, Oh B-K. 2008. The fabrication of functional biosurface composed of iron storage protein, ferritin. Ultramicroscopy 108: 13561359. F 1313. Chung SG, Kim JC, Park C-H, Ahn W-S, Kim Y-W, Choi J-W. 2008. Volatile organic compound specic detection by electrochemical signals using a cell-based sensor. J. Microbiol. Biotechnol. 18: 145152. D 1314. Hook F, Stengel G, Dahlin AB, Gunnarsson A, Jonsson MP, Jonsson P, Reimhult E, Simonsson L, Svedhem S. 2008. Supported lipid bilayers, tethered lipid vesicles, and vesicle fusion investigated using gravimetric, plasmonic, and microscopy techniques. Biointerphases 3: FA108FA116. D 1315. Huang C, Jiang G, Advincula R. 2008. Electrochemical crosslinking and patterning of nanostructured polyelectrolytecarbazole precursor ultrathin lms. Macromolecules 41: 46614670. F 1316. Kaewtong C, Jiang G, Park Y, Fulghum T, Baba A, Pulpoka B, Advincula R. 2008. Azacalix[3]arene-carbazole conjugated poly-
F 1294.
Nomadics/Texas Instruments
F 1295. Atkinson KR, Lo KR, Payne SR, Mitchell JS, Ingram JR. 2008. Rapid saliva processing techniques for near real-time analysis of salivary steroids and protein. J. Clin. Lab. Anal. 22: 395402.
61
www.interscience.wiley.com/journal/jmr
D 1320.
F 1321.
F 1322. F 1323.
Thermo Scientic
D 1338. Boddohi S, Killingsworth CE, Kipper MJ. 2008. Polyelectrolyte multilayer assembly as a function of pH and ionic strength using the polysaccharides chitosan and heparin. Biomacromolecules 9: 20212028.
F 1324.
Plasmonic Biosensor
C 1325. Mazumdar SD, Barlen B, Kramer T, Keusgen M. 2008. A rapid serological assay for prediction of Salmonella infection status in slaughter pigs using surface plasmon resonance. J. Microbiol. Methods 75: 545550.
Bio-Rad
D 1340. Artzy-Schnirman A, Brod E, Epel M, Dines M, Hammer T, Benhar I, Reiter Y, Sivan U. 2008. A two-state electronic antigen and an antibody selected to discriminate between these states. Nanoletters 8: 33983403. B 1341. Bravman T, Bronner V, Nahshol O, Schreiber G. 2008. The ProteOn XPR36TM Array System high throughput kinetic binding analysis of biomolecular interactions. Cell. Mol. Bioeng. 1: 216228. D 1342. Brehin A-C, Rubrecht L, Navarro-Sanchez ME, Marechal V, Frenkiel ` M-P, Lapalud P, Laune D, Sall AA, Despres P. 2008. Production and characterization of mouse monoclonal antibodies reactive to Chikungunya envelope E2 glycoprotein. Virology 371: 185195. D 1343. Brod E, Nimri S, Turner B, Sivan U. 2008. Electrical control over antibody-antigen binding. Sens. Actuators B 128: 560565. B 1344. Bronner V, Nahshol O, Bravman T. 2008. Evaluating candidate lead compounds by rapid analysis of drug interactions with human serum albumin. Am. Biotechnol. Lab. 26: 1416. C 1345. Cohen-Ben-Lulu GN, Francis NR, Shimoni E, Noy D, Davidov Y, Prasad K, Sagi Y, Cecchini G, Johnstone RM, Eisenbach M. 2008. The bacterial agellar switch complex is getting more complex. EMBO J. 27: 11341144. C 1346. Dubin-Bar D, Bitan A, Bakhrat A, Kaiden-Hasson R, Etzion S, Shaanan B, Abdu U. 2008. The Drosophila IKK-related kinase (Ik2) and Spindle-F proteins are part of a complex that regulates cytoskeleton organization during oogenesis. BMC Cell Biol. 9: 51. D 1347. Goldin N, Arzoine L, Heyfets A, Israelson A, Zaslavsky Z, Bravman T, Bronner V, Notcovich A, Shoshan-Barmatz V, Flescher E. 2008. Methyl jasmonate binds to and detaches mitochondria-bound hexokinase. Oncogene 27: 46364643. B 1348. Kalie E, Jaitin DA, Podoplelova Y, Piehler J, Schreiber G. 2008. The stability of the ternary interferon-receptor complex rather than the afnity to the individual subunits dictates differential biological activities. J. Biol. Chem. 283: 3292532936. D 1349. Katz C, Benyamini H, Rotem S, Lebendiker M, Danieli T, Iosub A, Refaely H, Dines M, Bronner V, Bravman T, Shalev DE, Rudiger S, Friedler A. 2008. Molecular basis of the interaction between the
Reichert Analytical/Xantex
D 1326. Chelmowski R, Koster SD, Kerstan A, Prekelt A, Grunwald C, Winkler T, Metzler-Nolte N, Terfort A, Woll C. 2008. Peptide-based SAMs that resist the adsorption of proteins. J. Am. Chem. Soc. 130: 1495214953. B 1327. Lehmann D, Seneviratne A, Smrcka A. 2008. Small molecule disruption of G protein beta gamma subunit signaling inhibits neutrophil chemotaxis and inammation. Mol. Pharmacol. 73: 410418. F 1328. Melzak KA, Bender F, Tsortos A, Gizeli E. 2008. Probing mechanical properties of liposomes using acoustic sensors. Langmuir 24: 91729180. F 1329. Saitakis M, Dellaporta A, Gizeli E. 2008. Measurement of twodimensional binding constants between cell-bound major histocompatibility complex and immobilized antibodies with an acoustic biosensor. Biophys. J. 95: 49634971. F 1330. Saitakis M, Dellaporta A, Gizeli E. 2008. A surface acoustic wave sensor for the study of membrane-protein/ligand interactions using whole cells. Int. Frequency Control Symp. Proc. 1: 356359. D 1331. Shekhah O, Roques N, Mugnaini V, Munuera C, Ocal C, Veciana J, Woll C. 2008. Grafting of monocarboxylic substituted polychlorotriphenylmethyl radicals onto a COOH-functionalized selfassembled monolayer through copper (II) metal ions. Langmuir 24: 66406648. F 1332. Tsortos A, Papadakis G, Mitsakakis K, Melzak KA, Gizeli E. 2008. Quantitative determination of size and shape of surface-bound DNA using an acoustic wave sensor. Biophys. J. 94: 27062715.
Resonant Probes
F 1333. Feller L, Bearinger JP, Wu L, Hubbell JA, Textor M, Tosatti S. 2008. Micropatterning of gold substrates based on poly(propylene sulde-bl-ethylene glycol), (PPS-PEG) background passivation and the molecular-assembly patterning by lift-off (MAPL) technique. Surface Sci. 602: 23052310.
62
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164
B 1350.
F 1351.
C 1352.
F 1353. D 1354.
C 1355.
D 1356. D 1357.
GE Healthcare/Biacore
F 1358. Chorley BN, Wang X, Campbell MR, Pittman GS, Noureddine MA, Bell DA. 2008. Discovery and verication of functional single nucleotide polymorphisms in regulatory genomic regions: current and developing technologies. Mutat. Res. 659: 147157. C 1359. de Boer AR, Hokke CH, Deelder AM, Wuhrer M. 2008. Serum antibody screening by surface plasmon resonance using a natural glycan microarray. Glycoconj. J. 25: 7584. B 1360. Karamanska R, Clarke J, Blixt O, Macrae JI, Zhang JQ, Crocker PR, Laurent N, Wright A, Flitsch SL, Russell DA, Field RA. 2008. Surface plasmon resonance imaging for real-time, label-free analysis of protein interactions with carbohydrate microarrays. Glycoconj. J. 25: 6974. D 1361. Laurent N, Voglmeir J, Flitsch SL. 2008. Glycoarraystools for determining protein-carbohydrate interactions and glycoenzyme specicity. ChemComm; 37: 44004412. A 1362. Rich RL, Cannon MJ, Jenkins J, Pandian P, Sundaram S, Magyar R, Brockman J, Lambert J, Myszka DG. 2008. Extracting kinetic rate constants from surface plasmon resonance array systems. Anal. Biochem. 373: 112120.
Horiba/Genoptics
C 1383. Corne C, Fiche J-B, Gasparutto D, Cunin V, Suraniti E, Buhot A, Fuchs J, Calemczuk R, Livache T, Favier A. 2008. SPR imaging for label-free multiplexed analyses of DNA N-glycosylase interactions with damaged DNA duplexes. Analyst 133: 1036 1045. D 1384. Corne C, Fiche J-B, Cunin V, Buhot A, Fuchs J, Calemczuk R, Favier A, Livache T, Gasparutto D. 2008. SPR-imaging based assays on an oligonucleotide-array to analyze DNA lesions recognition and excision by repair proteins. Nucleic Acids Symp. Ser. 52: 249250. F 1385. Diltemiz SE, Denizli A, Ersoz A, Say R. 2008. Molecularly imprinted ligand-exchange recognition assay of DNA by SPR system using guanosine and guanine recognition sites of DNA. Sens. Actuators B 133: 484488. D 1386. Fiche JB, Fuchs J, Buhot A, Calemczuk R, Livache T. 2008. Point mutation detection by surface plasmon resonance imaging coupled with a temperature scan method in a model system. Anal. Chem. 80: 10491057. C 1387. Mercey E, Sadir R, Maillart E, Roget A, Baleux F, Lortat-Jacob H, Livache T. 2008. Polypyrrole oligosaccharide array and surface plasmon resonance imaging for the measurement of glycosaminoglycan binding interactions. Anal. Chem. 80: 34763482. F 1388. Ruiz A, Buzanska L, Gilliland D, Rauscher H, Sirghi L, Sobanski T, Zychowicz M, Ceriotti L, Bretagnol F, Coecke S, Colpo P, Rossi F.
GWC Technologies/Toyobo
C 1363. DAgata R, Grasso G, Spoto G. 2008. Real-time binding kinetics monitored with surface plasmon resonance imaging in a diffusion-free environment. Open Spectrosc. J. 2: 19. D 1364. Feng W-Y, Chiu N-F, Lu H-H, Shih H-C, Yang D, Lin CW. 2008. Surface plasmon resonance biochip based on ZnO thin lm for nitric oxide sensing. Conf. Proc. IEEE Eng. Med. Biol. Sci. 2008: 57575760. D 1365. Garcia BH 2nd, Goodman RM. 2008. Use of surface plasmon resonance imaging to study viral RNA:protein interactions. J. Virol. Methods 147: 1825. D 1366. Grant CF, Kanda V, Yu H, Bundle DR, McDermott MT. 2008. Optimization of immobilized bacterial disaccharides for surface plasmon resonance imaging measurements of antibody binding. Langmuir 24: 1412514132. D 1367. Grasso G, Rizzarelli E, Spoto G. 2008. How the binding and degrading capabilities of insulin degrading enzyme are affected by ubiquitin. Biochim. Biophys. Acta 1784: 11221126.
63
www.interscience.wiley.com/journal/jmr
IBIS
A 1389. Beusink JB, Lokate AM, Besselink GA, Pruijn GJ, Schasfoort RB. 2008. Angle-scanning SPR imaging for detection of biomolecular interactions on microarrays. Biosens. Bioelectron. 23: 839844. C 1390. Raz SR, Bremer MGEG, Giesbers M, Norde W. 2008. Development of a biosensor microarray towards food screening, using imaging surface plasmon resonance. Biosens. Bioelectron. 24: 552557.
Plexera
C 1391. Lausted CG, Hu Z, Hood LE. 2008. Quantitative serum proteomics from surface plasmon resonance imaging. Mol. Cell. Proteomics 7: 24642474.
ForteBio
C 1409. Do T, Ho F, Heidecker B, Witte K, Chang L, Lerner L. 2008. A rapid method for determining dynamic binding capacity of resins for the purication of proteins. Protein Expr. Purif. 60: 147150.
Fareld Sensors
C 1396. Aulin C, Varga I, Claesson PM, Wagberg L, Lindstrom T. 2008. Buildup of polyelectrolyte multilayers of polyethyleneimine and microbrillated cellulose studied by in situ dual-polarization interferometry and quartz crystal microbalance with dissipation. Langmuir 24: 25092518. F 1397. Azemi E, Stauffer WR, Gostock MS, Lagenaur CF, Cui XT. 2008. Surface immobilization of neural adhesion molecule L1 for improving the biocompatibility of chronic neural probes: in vitro characterization. Acta Biomater. 4: 12081217. F 1398. Boudjemline A, Clarke DT, Freeman NJ, Nicholson JM, Jones GR. 2008. Early stages of protein crystallization as revealed by emerging optical waveguide technology. J. Appl. Cryst. 41: 523530. D 1399. Edmondson S, Vo C-D, Armes S, Unali G-F, Weir MP. 2008. Layer-by-layer deposition of polyelectrolyte macroinitiators for enhanced initiator density in surface-initiated ATRP. Langmuir 24: 72087215. D 1400. Johnson S, Evans D, Laurenson S, Paul D, Davies AG, Ferrigno PK, Walti C. 2008. Surface-immobilized peptide aptamers as probe molecules for protein detection. Anal. Chem. 80: 978983. F 1401. Khan TR, Grandin HM, Mashaghi A, Textor M, Reimhult E, Raviakine I. 2008. Lipid redistribution in phosphatidylserine-containing vesicles adsorbing on titania. Biointerphases 3: FA90FA95.
Maven Biotechnologies
C 1410. Ralin D, Dultz S, Silver J, Travis J, Kullolli M, Hancock W, Hincapie M. 2008. Kinetic analysis of glycoproteinlectin interactions by label-free internal reection ellipsometry. Clin. Proteomics 4: 3746.
Silicon Kinetics
C 1411. Latterich MCJ. 2008. Label-free detection of biomolecular interactions in real time with a nano-porous silicon-based detection method. Proteome Sci. 6: 31.
SRU Biosystems
C 1412. Chan LL, Gosangari SL, Watkin KL, Cunningham BT. 2008. Labelfree imaging of cancer cells using photonic crystal biosensors and application to cytotoxicity screening of a natural compound library. Sens. Actuators B 132: 418425. C 1413. Chan LL, Pineda M, Heeres JT, Hergenrother PJ, Cunningham BT. 2008. A general method for discovering inhibitors of protein-DNA interactions using photonic crystal biosensors. ACS Chem. Biol. 3: 437448.
64
www.interscience.wiley.com/journal/jmr
Copyright 2009 John Wiley & Sons, Ltd. J. Mol. Recognit. 2010; 23: 164