Skip to main content
Bentham Open Access logoLink to Bentham Open Access
. 2009 Nov;12(9):870–876. doi: 10.2174/138620709789383277

Applications of High Content Screening in Life Science Research

Joseph M Zock 1,*
PMCID: PMC2841426  PMID: 19938341

Abstract

Over the last decade, imaging as a detection mode for cell based assays has opened a new world of opportunities to measure “phenotypic endpoints” in both current and developing biological models. These “high content” methods combine multiple measurements of cell physiology, whether it comes from sub-cellular compartments, multicellular structures, or model organisms. The resulting multifaceted data can be used to derive new insights into complex phenomena from cell differentiation to compound pharmacology and toxicity. Exploring the major application areas through review of the growing compendium of literature provides evidence that this technology is having a tangible impact on drug discovery and the life sciences.

Keywords: High content screening, cell signaling, oncology, neurobiology, in vitro toxicology, target identification, target validation, RNAi, stem cells.

INTRODUCTION

High Content Screening (HCS) is the application of automated microscopy and image analysis to both drug discovery and cell biology. This technique has grown from an interesting proposition, to a useful technology, and onto a valuable utility over the last decade. This paper reports on observations of peer reviewed journal articles using HCS as a key component of the research and attempts to offer a glimpse at how widely adopted the technology has been in several important areas of life science research.

Predictably, early papers were focused on HCS as a novel technology. As the technology has gained wider acceptance and use, however, the focus of papers has shifted back to the biology being studied, with HCS becoming one of the tools to deliver “supportive biological context” to whatever new entity or idea is being proposed. In my mind, this trend is one of the hallmarks of true technology adoption and a good indication that HCS is here to stay.

It is interesting to note that, although most of scientific articles citing the use of HCS fall along the categories of cell signaling, oncology, neurobiology, in vitro toxicology, and target validation (i.e. RNAi), an increasing number of papers describe very novel applications. Adoption beyond the now “standard” uses for HCS shows the flexibility of the technology to expand the breadth of addressable biological processes including cardiac failure [1], gap junctions [2], immunosupression [3], osteoporosis [4], phagocytosis [5], autophagy [6], centrosome function [7], fungal pathogenesis [8], retinal repair [9], circadian rhythms [10], and screening in yeast [11] just to name a few.

As with any emerging technology, HCS is being compared to current assay methods. In many instances HCS provides significant benefits over existing approaches or at least is seen as complementary, providing additional data that can be used to make scientific conclusions. In recent papers comparing HCS approaches to a standard Enzyme - Linked ImmunoSorbent Assay (ELISA), for example, it is noted that the ability to see what is going on at individual cell resolution makes the measurements more accurate and reliable [12, 13].

Liu et al., explain how ELISA and HCS provide synergistic outputs that help determine the complex pharmacology of the compounds they are evaluating. “It is clear that the readout from the Neurofilament (NF) ELISA is a measure of neuronal survival…on the other hand, the Cellomics ArrayScan platform, as represented in this study, is meant to evaluate a compound’s ability to increase total neurite outgrowth…the utility of both assay platforms would enable the investigator to identify compounds with multiple cellular activities, such as FK506” [14].

Agler et al., cite subcellular location of protein/protein interaction as an advantage of HCS over second signal assays. The ability of HCS to identify only a defined subset of cells in the well enables transient transfection to be used for robust screening instead of the forced use of “stable clones” where over-expression can lead to aberrant physiology and toxicity. “HCS assays provide other information unavailable from fluorescence polarization (FP) and reporter assays, such as subcellular localization where protein-protein interactions occur. Within this assay triage strategy, the HCS translocation assay provides an inexpensive assay format compared to purchasing commercially available reagents for FP and reporter assays” [15].

HCS used to replace difficult assays is on the rise where the advantage might be sensitivity over existing methods, increased throughput, increased safety, and/or decreased cost. Baniecki et al., who developed HCS to identify new anti-malarial drugs, state, “we are able to detect as little as one individual parasite in our image-based DAPI P. falciparum growth assay compared to a uniform well readout of 0.25% parasites observed in the DAPI P. falciparum growth assay and [3H]hypoxanthine assay - significantly greater sensitivity and reliability” [16]. Johnson et al., at the Center for Disease Control (CDC), developed a rapid, high throughput vaccinia virus neutralization assay [17] utilizing HCS to replace assays that are “laborious, particularly for large numbers of test sample and take up to 48-72 hours for plaque formation…analysis of results takes additional time and may be subjective since the plaques are counted manually”. The HCS assay, based on detecting viral infection with GFP, “has the potential to replace plaque reduction neutralization titer (PRNT) as a lab-standard clinical sample neutralization assay due to the speed and reliability with which data is produced. In the event of an orthopoxvirus outbreak, the speed and high throughput nature of the assay may prove extremely valuable”.

Historically, HCS has its origins in drug discovery, initially providing novel secondary assay formats, selectivity screens, and cytotoxicity profiling using the multi-parameter and individual cell attributes of the approach. “High content, in context, and with correlation” describes the data coming from the current HCS platforms [18], but understanding the value and utility of what that data represents is only now becoming obvious in the literature. In a brilliant example, Young et al., elegantly show how powerful the application of multi-faceted HCS data can be in their recent paper that integrates HCS and ligand-target prediction to identify pharmacological mechanisms of action [19]. Using cell cycle permutation as the model, a series of image-based cytological features were collected. A thirty-six-feature subset was selected, defining six factors (nuclear size, replication, mitosis, nuclear morphology, EdU texture, and nuclear ellipticity). A library of 6,547 compounds was profiled. “The responses grouped active compounds into seven major categories of phenotypic effects. We then explore how phenotypic profiles of active compounds compare with chemical structure and predicted target profiles. The resulting structure-activity relationships are richer than would be possible with a single data type, and they allow us to infer mechanisms of action for some compounds.”

HCS AND CELL SIGNALING

In the drug discovery process, understanding how environmental triggers cue a particular set of biological process cascades holds the key to therapeutic control. Cell signaling, therefore, is at the root of most target-specific attempts to create drugs. In an academic setting, there is a similar need to understand how newly discovered proteins map into and across various pathways. Therefore, it is not surprising to find that many of the peer reviewed journal articles citing the use of HCS are reporting on some aspect of cell signaling. From the initial HCS paper on NFkB translocation [20] a decade ago many signaling molecule activities have been quantitated including STAT [21, 22], wnd/fzd [23], akt [24], NFAT [25], p38 [26], TGF-beta [27] and Smad2/3 [28, 29] making up signaling networks from inflammation [30] to G-Protein coupled receptors [31-33].

HCS AND ONCOLOGY

HCS found an initial foothold in oncology research, due to the early applications for measuring apoptosis [34-38] and proliferation [39-41]. Assays for cell cycle [42-44], transformation [45] and migration [46-48] followed. At one point we developed a motility assay algorithm and reagent kit that is used to access metastatic potential [49-51]. Being able to see the individual cell responses verses a “population average” has led to a better understanding of how anti-cancer compounds might differentially effect cancerous cells compared to normal cells and has been used to determine the function of cancer biomarkers [52]. Anti-cancer compounds identified utilizing HCS have started moving into clinical trials [53].

As HCS technology continued to develop, additional applications in oncology research began to emerge. The study of angiogenesis, for example, can now be routinely performed by stimulating endothelial cells to undergo angiogenic transformation in a microplate well [54, 55]. The phenotype of tube formation is striking in the sense that a multitude of cells are signaling each other and working in unison to create an extremely specific multicellular structure that has many implications on various disease states, depending on the particular focus. Stimulating neovascularization of a damaged organ or wound would be considered beneficial while inhibiting the neovascularization of a solid tumor or retina (wet form of macular degeneration) would also be therapeutic. The key here is to be able to accurately capture, measure, and report on the phenotypes by individually assessing a number of attributes. In the case of angiogenic tube formation, being able to measure tube size and shape, connectedness, number of nodes, number of cells in a tube, and even target activation in the cells in a tube allow the researcher to discriminate between compound activities. Another area of interest in oncology using HCS is the evaluation of anti-cancer compounds through the quantitation of cytoskeletal rearrangement, specifically looking at microtubule assembly/disassembly [56, 57].

A good example of HCS adoption is the NCI Institute for Chemical Genomics. They have developed and implemented HCS assays for nuclear foci formation, cell morphology changes and protein translocations. “Because such measurements are done at the cellular level rather than averaged over a well, the signal-to-noise ratio is considerably higher; each well, in essence, serves as its own set of data points” [58]. Using HCS they have been able to identify novel cell division modulators with different modes of action than the microtubule disruption of classic antimitotic compounds, secramine, an actin polymerization inhibitor reducing metastasis, and several modulators of NFAT and FOX01a nuclear translocation.

HCS AND NEUROBIOLOGY

Very early in the development of HCS we recognized the potential of imaging in the quantitation of neuronal morphology and were the first to create a product to monitor neurite outgrowth. Over the years we have evolved the algorithmic approaches a number of times, responding to the feedback from multiple users screening for stimulation of neurite outgrowth [59-62] and neuronal protection [63-65] with the resulting calculated feature set allowing the extraction of many attributes of neurons and neuronal sub-populations in both primary cells and standard cell lines.

More striking is the application of HCS to the study of a wide variety of neurological disease states, whether it is a basic understanding of the underlying biology, the creation of new models, or the screening of molecules for therapeutic intervention. Examples include Alzheimer Disease [66], Parkinson’s Disease [67, 68], Huntington Disease [69, 70], Amyotrophic Lateral Sclerosis [71] and brain cancer [72] with more articles appearing each year.

HCS AND IN VITRO TOXICOLOGY

All HCS assays can be considered “tox” assays on some level, since they measure a cell’s physiological responses to stimulus, whether it be environmental or chemical. From relatively simple measures of acute cytotoxicity, such as cell counting and cell rounding, to more specific measures of organelle health [73, 74], HCS can be applied to many situations, often as a multi-parameter assay where cross correlation of multiple endpoints can help define subtle toxic states.

It is clear that HCS has found a strong foothold in drug discovery in the area of cytotoxicity [75]. However, uses of HCS beyond straight cytotoxicity [76], where the evaluation at the cell level is predictive for downstream toxic effects in whole organisms (like us) is an important area of growth for automated imaging in drug discovery, since the increased capacity for getting critical data at the right time can mean the gain/loss of billions of dollars. Initial applications include assays for micronucleus induction [77, 78] to assess genotoxicity, phospholipidosis [79] for liver lipidosis, and developmental neurotoxicity [80]. Moving forward there is great potential in using HCS to set up new models for toxicity [81] including the use of model organisms like zebrafish and worms.

One of the most exciting results showing the potential of a multi-parameter imaging approach to in vitro toxicology is in the area of drug-induced liver injury where Xu et al., have developed a testing strategy around a panel of phenotypes that are directly linked to hepatotoxicity [82]. “When applied to over 300 drugs and chemicals including many that caused rare and idiosyncratic liver toxicity in humans, our testing strategy has a true-positive rate of 50-60% and an exceptionally low false-positive rate of 0-5%”

In another retrospective study of hepatotoxic compounds, O’Brien et al. [83]. compared the “standard 7” biochemical cytotoxicity assays used in the industry to a single, 4-dye component, multi-parameter HCS assay. The HCS assay showed a much higher sensitivity (93% vs <25%) and specificity (98% vs ~90%) than the best combination of the "standard 7" biochemical assays. These studies are being confirmed across the pharmaceutical industry [84].

HCS AND TARGET VALIDATION

The target validation area of early drug discovery, and to a large extent all basic research, is focused on identifying new components in the cell biology puzzle and validating their various functions. On the basic research side, validation adds to the understanding of the big picture. On the drug discovery side, validation provides the foundation to develop assays that reflect disease states so that molecules that perturb the disease state can be identified. Ultimate success in this area requires both relevant biological models and physiologically accurate environmental conditions. The relatively recent technological advances of using stem cells and RNAi to create cell models are a natural fit for phenotypic quantitation. Whether it is tracking the development of a differentiating population of stem cells en route to becoming muscle cells, or assessing the outcome of knocking out proliferation signals for neurite outgrowth, HCS can be applied. In a recent review of RNA interference based screening, Perrimon comments, “Perhaps the most significant advances in RNAi HTS will come from high content screening. Cell-based HCS that rely on cellular phenotypes are becoming one of the preferred methods in RNAi HTS because they generate data sets that are rich in information…the use of primary cells offers ample opportunities to carry out cell morphology screens in a biologically relevant context” [85].

Moffat et al. developed a screen based on high content imaging to identify genes required for mitotic progression and applied it to 5000 unique short hairpin RNA (shRNA) expressing lentiviruses targeting 1028 human genes [86]. The screen identified ~ 100 (new) candidate regulators of proliferation. Similar studies using HCS to monitor phenotypic endpoints have been done using short interfering RNA (siRNA) libraries [87-90].

On the stem cell front, HCS has been used to help identify components of the regulatory machinery involved in stem cell self-renewal [91, 92] and differentiation [93], primarily through the quantitation of pluripotency markers like Oct-4 in either embryonic stem cells [94, 95] or lines derived from adult tissues [96]. Downstream tracking of cell fate using various differentiation state biomarkers has also been done [97]. The unique ability of HCS to provide spatial information about cell /cell relationships is illustrated by Peerani et al., where the heterogeneous microenvironments (niches) in the organization of human embryonic stem cell (hESC) cultures influence hESC fate [98]. By evaluating niche size and cell composition against localized secretion of differentiation inducing and inhibiting factors (via siRNA knock down) they discovered, “for the first time, a role for Smad1 in the integration of spatial information and in the niche-size-dependent control of hESC self-renewal and differentiation.”

FUTURE DIRECTIONS AND OUTLOOK

It is clear from the published literature cited above that HCS has moved beyond the “proving the technology” phase and is now entering the “broad adoption” phase where the needs of an ever increasing user base will drive the technology to be more cost effective, easy to use, and robust. Development of new reagents [99] and micro-environments, combined with continued application of autofluorescent proteins [100] will open the door to even more ways to quantify cellular behavior. A focus on the productivity metrics of HCS, both in creating the information (i.e. assay prep automation, reagent kits, algorithms) and understanding it (i.e. data management, visualization, mining) will supplement the detection platforms, allowing more routine generation of HCS data as part of decision support in the life sciences. While the core applications of HCS will evolve to mass use, technology development will progress toward expanding biological data relevance through use of multicellular assemblies, tissues, organs, and organisms. An additional trend to incorporate “cellomics” with genomics and proteomics technologies will provide an unprecedented picture of cell functions.

The outlook for HCS is very bright, considering that many countries are proposing moratoriums to reduce animal testing [101, 102] in the “not-to-distant” future. The development of in vitro cell based assay tools like HCS could not be more timely. As someone who has participated in developing this technology from its inception, I can honestly say, due to the passion and dedication of “highly productive users”, HCS is taking its place as a powerful tool to help weave the “fabric of scientific knowledge” for years to come.

Table 1.

Key Application Areas of HCS and Example References

Application Area Example References
Cell Signaling NFkB 20
STAT 21,22
Wnd/fzd 23
akt 24
NFAT 25,58
P38 26
TGF-beta 27,29
Smad2/3 28,29
GPCR 31-33,100
Glucocorticoid receptor 15
FOXO1a 58
yeast 11
Cell Physiology Proliferation 12,39-41,90
Phosphorylation 13
Phagocytosis 5
Autophagy 6
Gap junction induction 2
Mitochondrial health 73,74,82-84
Nuclear morphology 19,83,84,88
Apoptosis 34-38,65
Membrane permeability 82-84
Cell cycle 19,42-44,58,86,99
Motility/migration 46-51
Cytoskeletal rearrangement 7,33,56,57
Neurite outgrowth 9,14,59-65,72,80
Transformation 45
In Vitro Toxicology Micronucleus induction 77,78
Phospholipidosis 79
Neurotoxicity 80
Organelle health 73,74,82,84
Hepatotoxicity 82-84
Organism Physiology Cardiac failure 1
Circadian rhythms 10
Immunosupression 3
Osteoporosis 4
Fungal pathogenesis 8
Virus neutralization 17
Parasite infection 16
Angiogenesis 54,55
Alzheimer’s Disease 66
Parkinson’s Disease 67,68
Huntington’s Disease 69,70
Amyotrophic Lateral Sclerosis 71
Target Validation shRNA interference library 86
siRNA interference library 87-90
Stem cell self-renewal 91,92,98
Stem cell differentiation 94-96,98

REFERENCES

  • 1.Harrison BC, Kim MS, van Rooji E, Plato CF, Papst PJ, Vega RB, McAnally JA, Richardson JA, Bassel-Duby R, Olson EN, McKinsey TA. Regulation of cardiac stress signaling by protein kinase D1. Mol Cell. Biol. 2006;26(10):3875–3888. doi: 10.1128/MCB.26.10.3875-3888.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Li Z, Yan Y, Powers EA, Ying X, Janjua K, Garyantes T, Baron B. Identification of gap junction blockers using automated fluorescence microscopy imaging. J. Biomol. Screen. 2003;8:489–499. doi: 10.1177/1087057103257309. [DOI] [PubMed] [Google Scholar]
  • 3.Yan P, Nanamori M, Sun M, Zhou C, Cheng N, Li N, Zheng W, Xiao L, Xie X, Ye RD, Wang MW. The immunosuppressant cyclosporin A antagonizes human formyl peptide receptor through inhibition of cognate ligand binding. J. Immunol. 2006;177:7050–7058. doi: 10.4049/jimmunol.177.10.7050. [DOI] [PubMed] [Google Scholar]
  • 4.Buckbinder L, Crawford DT, Qi H, Ke HZ, Olson LM, Long KR, Bonnette PC, Baumann AP, Hambor JE, Grasser WA, Pan LC, Owen TA, Luzzio MJ, Hulford CA, Gebhard DF, Paralkar VM, Simmons HA, Kath JC, Roberts WG, Smock SL, Guzman-Perez A, Brown TA, Li M. Proline-rich tyrosine kinase 2 regulates osteoprogenitor cells and bone formation and offers an anabolic treatment approach for osteoporosis. Proc. Natl. Acad. Sci. USA. 2007;104(25):10619–10624. doi: 10.1073/pnas.0701421104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Steinberg BE, Scott CC, Grinstein S. High-throughput assays of phagocytosis, phagosome maturation, and bacterial invasion. Am. J. Physiol. Cell. Physiol. 2007;292:945–952. doi: 10.1152/ajpcell.00358.2006. [DOI] [PubMed] [Google Scholar]
  • 6.Zhang L, Yu J, Pan H, Hu P, Hao Y, Cai W, Zhu H, Yu AD, Xie X, Ma D, Yuan J. Small molecule regulators of autophagy identified by an image-based high-throughput screen. Proc. Natl. Acad. Sci. USA. 2007;104(48):19023–19028. doi: 10.1073/pnas.0709695104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Rogers GC, Rusan NM, Peifer M, Rogers SL. A Multicomponent assembly pathway contributes to the formation of acentrosomal microtubule arrays in interphase drosophila cells. Mol. Biol. Cell. 2008;19:3163–3178. doi: 10.1091/mbc.E07-10-1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Wheeler RT, Fink GR. A drug-sensitive genetic network masks fungi from the immune system. PLoS Pathog. 2006;2(4):e35–328-339. doi: 10.1371/journal.ppat.0020035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Kerrison JB, Lewis RN, Otteson DC, Zack DJ. Bone morphogenic proteins promote neurite outgrowth in retinal ganglion cells. Mol. Vision. 2005;11:208–215. [PubMed] [Google Scholar]
  • 10.Badura L, Swanson T, Adamowicz W, Adams J, Cianfrogna J, Fisher K, Holland J, Kleiman R, Nelson F, Reynolds L, St.; Germain K, Schaeffer E, Tate B, Sprouse J. An inhibitor of casein kinase I induces phase delays in circadian rhythms under free-running and entrained conditions. J. Pharmacol. Exp. Ther. 2007;322:730–738. doi: 10.1124/jpet.107.122846. [DOI] [PubMed] [Google Scholar]
  • 11.Benanti JA, Cheung SK, Brady MC, Toczyski DP. A proteomic screen reveals SCF[Grr1] targets that regulate the glycolytic-gluconeogenic switch. Nat. Cell Biol. 2007;9(10):1184–1191. doi: 10.1038/ncb1639. [DOI] [PubMed] [Google Scholar]
  • 12.Gasparri F, Mariani M, Sola F, Galvani A. Quantification of the proliferation index of human dermal fibroblast cultures with the ArrayScan high content screening reader. J. Biomol. Screen. 2004;9:232–243. doi: 10.1177/1087057103262836. [DOI] [PubMed] [Google Scholar]
  • 13.Vogt A, Cooley KA, Brisson M, Tarpley MG, Wipf P, Lazo JS. Cell-active dual specificity phosphatase inhibitors identified by high content screening. Chem. Biol. 2003;10:733–742. doi: 10.1016/s1074-5521(03)00170-4. [DOI] [PubMed] [Google Scholar]
  • 14.Liu D, McIlvain HB, Fennell M, Dunlop J, Wood A, Zaleska MM, Graziani EI, Pong K. Screening of immunophilin ligands by quantitative analysis of neurofilament expression and neurite outgrowth in cultured neurons and cells. J. Neurosci. Methods. 2007;163:310–320. doi: 10.1016/j.jneumeth.2007.03.018. [DOI] [PubMed] [Google Scholar]
  • 15.Agler M, Prack M, Zhu Y, Kolb J, Nowak K, Ryseck R, Shen D, Cvijic ME, Somerville J, Nadler S, Chen T. A high content glucocorticoid receptor translocation assay for compound mechanism-of-action evaluation. J. Biomol. Screen. 2007;12:1029–1041. doi: 10.1177/1087057107309353. [DOI] [PubMed] [Google Scholar]
  • 16.Baniecki ML, Wirth DF, Clardy J. High-Throughput Plasmodium falciparum growth assay for malaria drug discovery. Antimicrob. Agents Chemother. 2007;51(2):716–723. doi: 10.1128/AAC.01144-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Johnson MC, Damon IK, Karem KL. A Rapid, high-throughput vaccinia virus neutralization assay for testing smallpox vaccine efficacy based on detection of green fluorescent protein. J. Virol. Methods. 2008;150:14–20. doi: 10.1016/j.jviromet.2008.02.009. [DOI] [PubMed] [Google Scholar]
  • 18.Keefer S, Zock J. Approaching High Content Screening and Analysis: Practical Advice for Users . In: Haney SA, editor. High Content Screening: Science, Techniques and Applications. Wiley-Interscience; 2008. pp. 3–24. [Google Scholar]
  • 19.Young DW, Bender A, Hoyt J, McWhinnie E, Chirn GW, Tao CY, Tallarico JA, Labow M, Jenkins JL, Mitchison TJ, Feng Y. Integrating high content screening and ligand-target prediction to identify mechanism of action. Nat. Chem. Biol. 2008;4:59–68. doi: 10.1038/nchembio.2007.53. [DOI] [PubMed] [Google Scholar]
  • 20.Ding GJF, Fischer PA, Boltz RC, Schmidt JA, Colaianne JJ, Gough A, Rubin RA, Miller DK. Characterization and quantitation of NF-kB nuclear translocation induced by interleukin-1 and tumor necrosis factor-a. J. Biol. Chem. 1998;273:28897–28905. doi: 10.1074/jbc.273.44.28897. [DOI] [PubMed] [Google Scholar]
  • 21.Vakkila J, DeMarco RA, Lotze MT. Imaging analysis of STAT1 and NF-kB translocation in dendritic cells at the single cell level. J. Immunol. Methods. 2004;294:123–134. doi: 10.1016/j.jim.2004.09.007. [DOI] [PubMed] [Google Scholar]
  • 22.Grace MJ, Lee S, Bradshaw S, Chapman J, Spond J, Cox S, DeLorenzo M, Brassardi D, Wylie D, Cannon-Carlson S, Cullen C, Indelicato S, Voloch M, Bordens R. Site of pegylation and polyethylene glycol molecule size attenuate inferferon-alpha antiviral and antiproliferative activities through the JAK/STAT signaling pathway. J. Biol. Chem. 2004;280(8):6327–6336. doi: 10.1074/jbc.M412134200. [DOI] [PubMed] [Google Scholar]
  • 23.Borchert KM, Galvin RJ, Frolik CA, Hale LV, Halladay DL, Gonyier RJ, Trask OJ, Nickischer DR, Houck KA. High content screening assay for activators of the wnt/fzd pathway in primary human cells. Assay Drug Dev. Technol. 2005;3(2):133–141. doi: 10.1089/adt.2005.3.133. [DOI] [PubMed] [Google Scholar]
  • 24.Lundholt BK, Linde V, Loechel F, Pedersen HC, Moller S, Praestegaard M, Mikkelsen I, Scudder K, Bjorn SP, Heide M, Arkhammar PO, Terry R, Nielsen SJ. Identification of Akt pathway inhibitors using Redistribution® screening on the FLIPR and the InCell 3000 Analyzer. J. Biomol. Screen. 2005;10:20–29. doi: 10.1177/1087057104269989. [DOI] [PubMed] [Google Scholar]
  • 25.Van Sant C, Wang G, Anderson MG, Trask OJ, Lesniewski R, Semizarov A. Endothelin signaling in osteoblasts: global genome view and implication of the calcineurin/NFAT pathway. Mol. Cancer Ther. 2007;6(1):253–261. doi: 10.1158/1535-7163.MCT-06-0574. [DOI] [PubMed] [Google Scholar]
  • 26.Ross S, Chen T, Yu V, Tudor Y, Zhang D, Liu L, Tamayo N, Dominguez C, Powers D. High content screening analysis of the p38 pathway: profiling of structurally related p38alpha kinase inhibitors using cell-based assays. Assay Drug. Dev. Technol. 2006;4(4):397–409. doi: 10.1089/adt.2006.4.397. [DOI] [PubMed] [Google Scholar]
  • 27.Sarker KP, Kataoka H, Chan A, Netherton SJ, Pot I, Huynh MA, Feng X, Bonni A, Riabowol K, Bonni S. ING2 as a novel mediator of transforming growth factor-β-dependent responses in epithelial cells. J. Biol. Chem. 2008;283:13269–13279. doi: 10.1074/jbc.M708834200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Dawes LJ, Angell H, Sleeman M, Reddan JR, Wormstone IM. TGFb isoform dependant Smad2/3 kinetics in human lens epithelial cells: a cellomics analysis. Exp. Eye Res. 2007;84:1009–1012. doi: 10.1016/j.exer.2006.12.017. [DOI] [PubMed] [Google Scholar]
  • 29.Wormstone IM, Dawes LJ, Sleeman M, Anderson IK, Reddan JR. FGF promotes TGF-beta-induced matrix contraction and suppresses expression of the transdifferentiation marker alphaSMA. Invest. Ophthalmol. Vis. Sci. 2008;49:3726. doi: 10.1167/iovs.07-0586. [DOI] [PubMed] [Google Scholar]
  • 30.Bertelsen M, Sanfridson A. Inflammatory pathway analysis using a high content screening platform. Assay Drug Dev. Technol. 2005;3:261–271. doi: 10.1089/adt.2005.3.261. [DOI] [PubMed] [Google Scholar]
  • 31.Schlag BD, Lou Z, Fennell M, Dunlop J. Ligand dependency of 5-HT2C receptor internalization. J. Pharmacol. Exp. Ther. 2004;310:865–870. doi: 10.1124/jpet.104.067306. [DOI] [PubMed] [Google Scholar]
  • 32.Fowler A, Seifert N, Acker V, Woehrle T, Kilpert C, de Saizieu A. A nonradioactive high-throughput / high content assay for measurement of the human serotonin reuptake transporter function in vitro. J. Biomol. Screen. 2006;11:1027–1034. doi: 10.1177/1087057106294698. [DOI] [PubMed] [Google Scholar]
  • 33.Barnard R, Barnard A, Salmon G, Liu W, Sreckovic S. Histamine-induced actin polymerization in human eosinophils: an imaging approach for histamine H4 receptor. Cytometry A. 2008;73(4):299–304. doi: 10.1002/cyto.a.20514. [DOI] [PubMed] [Google Scholar]
  • 34.Inglefield JR, Larson CJ, Gibson SJ, Lebrec H, Miller RL. Apoptotic responses in squamous carcinoma and epithelial cells to small-molecule toll-like receptor agonists evaluated with automated cytometry. J. Biomol. Screen. 2006;11(6):575–585. doi: 10.1177/1087057106288051. [DOI] [PubMed] [Google Scholar]
  • 35.Lovbor H, Gullbo J, Larsson R. Screening for apoptosis-classical and emerging techniques. Anticancer Drugs. 2005;16:593–599. doi: 10.1097/00001813-200507000-00001. [DOI] [PubMed] [Google Scholar]
  • 36.To K, Zhao Y, Jiang H, Hu K, Wang M, Wu J, Lee C, Yokom W, Stratford AL, Klinge U, Mertens PR, Chen CS, Bally M, Yapp D, Dunn SE. The phosphoinositide-dependent kinase-1 inhibitor 2-amino-N-[4-[5-(2-phenanthrenyl0-3-(trifluoromethyl)-1hH-pyrazol-1-yl]phenyl]-acetamide (OSU-03012) prevents Y-box binding protein-1 from inducing epidermal growth factor receptor. Mol. Pharmacol. 2007;72(3):641–652. doi: 10.1124/mol.107.036111. [DOI] [PubMed] [Google Scholar]
  • 37.Vogt A, McDonald PR, Tamewitz A, Sikorski RP, Wipf P, Skoko III JJ, Lazo JS. A cell-active inhibitor of mitogen-activated protein kinase phosphatases restores paclitaxel-induced apoptosis in dexamethasone-protected cancer cells. Mol. Cancer Ther. 2008;7:330–340. doi: 10.1158/1535-7163.MCT-07-2165. [DOI] [PubMed] [Google Scholar]
  • 38.Cummings J, Hodgkinson C, Odedra R, Sini P, Heaton SP, Mundt KE, Ward TH, Wilkinson RW, Growcott J, Hughes A, Dive C. Preclinical evaluation of M30 and M65 ELISAs as biomarkers of drug induced tumor cell death and antitumor activity. Mol. Cancer Ther. 2008;7:455–463. doi: 10.1158/1535-7163.MCT-07-2136. [DOI] [PubMed] [Google Scholar]
  • 39.Soncini C, Carpinelli P, Gianellini L, Fancelli D, Vianello P, Rusconi L, Storici P, Zugnoni P, Pesenti E, Croci V, Ceruti R, Giorgini ML, Cappella P, Ballinari D, Sola F, Varasi M, Bravo R, Moll J. PHA-680632, a novel Aurora kinase inhibitor with potent antitumoral activity. Clin. Cancer Res. 2006;12(13):4080–4089. doi: 10.1158/1078-0432.CCR-05-1964. [DOI] [PubMed] [Google Scholar]
  • 40.Gasparri F, Mariani M, Sola F, Galvani A. Quantification of the proliferation index of human dermal fibroblast cultures with the ArrayScan high content screening reader. J. Biomol. Screen. 2004;9:232–243. doi: 10.1177/1087057103262836. [DOI] [PubMed] [Google Scholar]
  • 41.Breier JM, Radio NM, Mundy WR, Shafer TJ. Development of a high-throughput screening assay for chemical effects on proliferation and viability of immortalized human neural progenitor cells. Toxicol. Sci. 2008;105:119–133. doi: 10.1093/toxsci/kfn115. [DOI] [PubMed] [Google Scholar]
  • 42.Barabasz A, Foley B, Otto JC, Scott A, Rice J. The use of high content screening for the discovery and characterization of compounds that modulate mitotic index and cell cycle progression by differing mechanisms of action. Assay Drug Dev. Technol. 2006;4(2):153–163. doi: 10.1089/adt.2006.4.153. [DOI] [PubMed] [Google Scholar]
  • 43.Gasparri F, Ciavolella A, Galvani A. Cell cycle inhibitor profiling by high content analysis. Adv. Exp. Med. Biol. 2007;604:137–148. doi: 10.1007/978-0-387-69116-9_13. [DOI] [PubMed] [Google Scholar]
  • 44.Gasparri F, Cappella P, Galvani A. Multiparametric cell cycle analysis by automated microscopy. J. Biomol. Screen. 2006;11:586–598. doi: 10.1177/1087057106289406. [DOI] [PubMed] [Google Scholar]
  • 45.Li Y, Pan J, Li JL, Lee JH, Tunkey C, Saraf K, Garbe JC, Whitley MZ, Jelinsky SA, Stampfer MR, Haney SA. Transcriptional changes associate with breast cancer occur as normal human mammary epithelial cells overcome senescence barrier and become immortalized. Mol. Cancer. 2007;6(7):1–17. doi: 10.1186/1476-4598-6-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Mastyugin V, McWhinnie E, Labow M, Buxton F. A quantitative high-throughput endothelial cell migration assay. J. Biomol. Screen. 2004;9:712–718. doi: 10.1177/1087057104269495. [DOI] [PubMed] [Google Scholar]
  • 47.Richards GR, Millard RM, Leveridge M, Kerby J, Simpson PB. Quantitative assays of chemotaxis and chemokinesis for human neural cells. Assay Drug Dev. Technol. 2004;2:465–472. doi: 10.1089/adt.2004.2.465. [DOI] [PubMed] [Google Scholar]
  • 48.Nam J-S, Ino Y, Kanai Y, Sakamoto M, Hirohashi S. 5-aza-2'-deoxycytidine restores the E-cadherin system in E-cadherin-silenced cancer cells and reduces cancer metastasis. Clin. Exp. Metastasis. 2004;21:49–56. doi: 10.1023/b:clin.0000017180.19881.c1. [DOI] [PubMed] [Google Scholar]
  • 49.Bhawe KM, Blake RA, Clary DO, Flanagan PM. An automated image capture and quantitation approach to identify proteins affecting tumor cell proliferation. J. Biomol. Screen. 2004;9:216–222. doi: 10.1177/1087057103262842. [DOI] [PubMed] [Google Scholar]
  • 50.Shimamra T, Yasuda J, Ino Y, Gotoh M, Tsuchiya A, Nakajima A, Sakamoto M, Kanai Y, Hirohashi S. Dysadherin expression facilitates cell motility and metastatic potential of human pancreatic cancer cells. Cancer Res. 2004;64:6989–6995. doi: 10.1158/0008-5472.CAN-04-1166. [DOI] [PubMed] [Google Scholar]
  • 51.Chuma M, Sakamoto M, Yasuda J, Fujii G, Nakanishi K, Tsuchiya A, Ohta T, Asaka M, Hirohashi S. Overexpression of cortactin is involved in motility and metastasis of hepatocellular carcinoma. J. Hepatol. 2004;41:629–636. doi: 10.1016/j.jhep.2004.06.018. [DOI] [PubMed] [Google Scholar]
  • 52.O'Brien C, Cavet G, Pandita A, Hu X, Haydu L, Mohan S, Toy K, Rivers CS, Modrusan Z, Amler LC, Lackner MR. Functional genomics identifies ABCC3 as a mediator of taxane resistance in HER2-amplified breast cancer. Cancer Res. 2008;68:5380–5389. doi: 10.1158/0008-5472.CAN-08-0234. [DOI] [PubMed] [Google Scholar]
  • 53.Sartore-Bianchi A, Gasparri F, Galvani A, Nici L, Darnowski JW, Barbone D, Fennell DA, Gaudino G, Porta C, Mutti L. Bortezomib inhibits nuclear factor-kappa B dependent survival and has potent in vivo activity in mesothelioma. Clin. Cancer Res. 2007;13(19):5942–5951. doi: 10.1158/1078-0432.CCR-07-0536. [DOI] [PubMed] [Google Scholar]
  • 54.Liu L, Cao Y, Chen C, Zhang X, McNabola A, Wilkie D, Wilhelm S, Lynch M, Carter C. Sorafenib blocks the RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and induces tumor cell apoptosis in hepatocellular carcinoma model PLC/PRF/5. Cancer Res. 2006;66(24):11851–11858. doi: 10.1158/0008-5472.CAN-06-1377. [DOI] [PubMed] [Google Scholar]
  • 55.Basu P, Ghosh R, Grove L, Klei L, Barchowsky A. Angiogenic potential of 3-nitro-4-hydroxy benzene arsonic acid (Roxarsone) Environ. Health Perspect. 2008;114(4):520–523. doi: 10.1289/ehp.10885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Wei SY, Li M, Tang SA, Sun W, Xu B, Cui JR, Lin WH. Induction of apoptosis accompanying with G1 phase arrest and microtubule disasembly in human hepatoma cells by jaspolide B, a new isomalabaricane-type triterpene. Cancer Lett. 2008;262:114–122. doi: 10.1016/j.canlet.2007.11.039. [DOI] [PubMed] [Google Scholar]
  • 57.Minguez JM, Giuliano KA, Balachandran R, Madiraju C, Curran DP, Day BW. Synthesis and high content cell-based profiling of simplified analogues of the microtubule stabilizer (+)-discodermolide. Mol. Cancer Ther. 2002;1:1305–1313. [PubMed] [Google Scholar]
  • 58.Tolliday N, Clemens PA, Ferraiolo P, Koehler AN, Lewis TA, Li X, Schreiber SL, Gerhard DS, Eliasof S. Small molecules, big players: the National Cancer Institute's initiative for chemical genetics. Cancer Res. 2006;66(18):8935–8942. doi: 10.1158/0008-5472.CAN-06-2552. [DOI] [PubMed] [Google Scholar]
  • 59.Richards GR, Smith AJ, Parry F, Platts A, Chan GK, Leveridge M, Kerby JE, Simpson PB. A morphology- and kinetics-based cascade for human neural cell high content screening. Assay Drug Dev. Technol. 2006;4(2):143–152. doi: 10.1089/adt.2006.4.143. [DOI] [PubMed] [Google Scholar]
  • 60.McIlvain HB, Baudy A, Sullivan K, Liu D, Pong K, Fennell M, Dunlop J. Pituitary adenylate cyclase-activating peptide (PACAP) induces differentiation in the neuronal F11 cell line through a PKA-dependent pathway. Brain Res. 2006;1077(1):16–23. doi: 10.1016/j.brainres.2005.12.130. [DOI] [PubMed] [Google Scholar]
  • 61.Loh SH, Francescut L, Lingor P, Bahr M, Nicotera P. Identification of new kinase clusters required for neurite outgrowth and retraction by loss-of-function RNA interference screen. Cell Death Differ. 2008;15(2):283–298. doi: 10.1038/sj.cdd.4402258. [DOI] [PubMed] [Google Scholar]
  • 62.Qian MD, Zhang J, Tan XY, Wood A, Gill D, Cho S. Novel agonist monoclonal antibodies activate TrkB receptors and demonstrate potent neurotrophic activities. J. Neurosci. 2006;26:9394–9403. doi: 10.1523/JNEUROSCI.1118-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Zhou W, Zhu X, Zhu L, Cui YY, Wang H, Qi H, Ren QS, Chen HZ. Neuroprotection of muscarinic receptor agonist pilocarpine against glutamate-induced apoptosis in retinal neurons. Cell. Mol. Neurobiol. 2008;28:263–275. doi: 10.1007/s10571-007-9251-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Ruan B, Pong K, Jow F, Bowlby M, Crozier RA, Liu D, Liang S, Chen Y, Mercado ML, Feng X, Bennett F, von Schack D, McDonald L, Zaleska MM, Wood A, Reinhart PH, Magola RL, Skotnicki J, Pangalos MN, Koehn FE, Carter GT, Abou-Gharbia M, Graziani EI. Binding of rapamycin analogs to calcium channels and FKBP52 contributes to their neuroprotective activities. Proc. Natl. Acad. Sci. USA. 2008;105(1):33–38. doi: 10.1073/pnas.0710424105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Fennell M, Chan H, Wood A. Multiparameter measurement of caspase 3 activation and apoptotic cell death in NT2 neuronal precursor cells using high content analysis. J. Biomol. Screen. 2006;11(3):296–302. doi: 10.1177/1087057105284618. [DOI] [PubMed] [Google Scholar]
  • 66.Culbert AA, Skaper SD, Howlett DR, Evans NA, Facci L, Soden PE, Seymour ZM, Guillot F, Gaestel M, Richardson JC. MAPK-activated protein kinase 2 deficiency in micoglia inhibits pro-inflammatory mediator release and resultant neurotoxicity: relevance to neuroinflammation in a transgenic mouse model of Alzheimer Disease. J. Biol. Chem. 2006;281(33):23658–23667. doi: 10.1074/jbc.M513646200. [DOI] [PubMed] [Google Scholar]
  • 67.Lotharius J, Falsig J, van Beek J, Payne S, Dringen R, Brundin P, Leist M. Progressive degenration of human mesencephalic neuron-derived cells triggered by dopamine-dependent oxidative stress is dependent on a mixed-lineage kinase pathway. J. Neurosci. 2005;25(27):6329–6342. doi: 10.1523/JNEUROSCI.1746-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Plun-Favreau H, Klupsch K, Moisoi N, Gandhi S, Kjaer S, Frith D, Harvey K, Deas E, Harvey RJ, McDonald N, Wood NW, Martins LM, Downward J. The mitochondrial protease HtrA2 is regulated by Parkinson's Disease-associated linase PINK1. Nat. Cell Biol. 2007;9(11):1227–1229. doi: 10.1038/ncb1644. [DOI] [PubMed] [Google Scholar]
  • 69.Doi H, Okamura K, Bauer PO, Furukawa Y, Shimizu H, Kurosawa M, Machida Y, Miyazaki H, Mitsui K, Kuriowa Y, Nukina N. RNA-binding protein TLS is a major nuclear aggregate-interacting proein in Huntingtin exon 1 with expanded polyglutamine-expressing cells. J. Biol. Chem. 2007;283(10):6489–6500. doi: 10.1074/jbc.M705306200. [DOI] [PubMed] [Google Scholar]
  • 70.Rigamonti D, Bolognini D, Mutti C, Zuccato C, Tartari M, Sola F, Valenza M, Kazantsev AG, Cattaneo E. Loss of Huntingtin function complemented by small molecules acting as repressor element 1/neuron restrictive silencer element silencer modulators. J. Biol. Chem. 2007;282:24554–24562. doi: 10.1074/jbc.M609885200. [DOI] [PubMed] [Google Scholar]
  • 71.Furukawa Y, Kaneko K, Yamanaka K, O'Halloran TV, Nukina N. Complete loss of post-translational modifications triggers fibrillar aggregation of sod1 in familial form of ALS. JBC manuscript M802083200. J. Biol. Chem. 2008;283(35):24167–24176. doi: 10.1074/jbc.M802083200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Wickstrom M, Johnsen JI, Ponthan F, Segerstrom L, Sveinbjornsson B, Lindskog M, Lovborg H, Viktorsson K, Lewensohn R, Kogner P, Larsson R, Gullbo J. The novel melphalan prodrug J1 inhibits neuroblastoma growth in vitro and in vivo. Mol. Cancer Ther. 2007;6:2409–2417. doi: 10.1158/1535-7163.MCT-07-0156. [DOI] [PubMed] [Google Scholar]
  • 73.Dykens JA, Jamieson JD, Marroquin LD, Nadanaciva S, Xu JJ, Dunn MC, Smith AR, Will Y. In vitro assessment of mitochondrial dysfunction and cytotoxicity of nefazodone, trazodone, and buspirone. Toxicol. Sci. 2008;103(2):335–45. doi: 10.1093/toxsci/kfn056. [DOI] [PubMed] [Google Scholar]
  • 74.Bova MP, Tam D, McMahon G, Mattson MN. Troglitaone induces a rapid drop of mitochodrial membrane potential in liver HepG2 cells. Tox. Lett. 2005;155:41–50. doi: 10.1016/j.toxlet.2004.08.009. [DOI] [PubMed] [Google Scholar]
  • 75.Xu JJ, Dunn MC, Smith A. Applications of cell based imaging technologies in toxicity screening in drug discovery and development. American Drug Discov. 2006;1(1):20–26. [Google Scholar]
  • 76.Jeffrey KD, Alejandro EU, Luciani DS, Kalynyak TB, Hu X, Li H, Lin Y, Townsend RR, Polonsky KS, Johnson JD. Carboxypeptidase E mediates palmitate-induced γ-cell ER stress and apoptosis. Proc. Natl. Acad. Sci. USA. 2008;105:8452–8457. doi: 10.1073/pnas.0711232105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Diaz D, Scott A, Carmichael P, Shi W, Costales C. Evaluation of an automated in vitro micronucleus assay in CHO-K1 cells. Mutat. Res. 2007;630(1-2):1–13. doi: 10.1016/j.mrgentox.2007.02.006. [DOI] [PubMed] [Google Scholar]
  • 78.Scott A, Malcomber S, Maskell S, Moore C, Windebank S, Diaz D, Carmichael P. An assessment of the performance of an automated scoring system (Cellomics) for the in vitro micronucleus assa in CHO-K1 cells. Toxicology. 2007;231(2-3):111–112. [Google Scholar]
  • 79.Morelli JK, Buehrle M, Pognan F, Barone LR, Fieles W, Ciaccio PJ. Validation of an in vitro screen for phospholipidosis using a high content biology platform. Cell. Biol. Toxicol. 2006;22(1):15–27. doi: 10.1007/s10565-006-0176-z. [DOI] [PubMed] [Google Scholar]
  • 80.Radio NM, Breier JM, Shafer TJ, Mundy WR. Assessment of chemical effects on neurite outgrowth in PC12 cells using high content screening. Toxicol. Sci. 2008;105:106–118. doi: 10.1093/toxsci/kfn114. [DOI] [PubMed] [Google Scholar]
  • 81.Yu X, Sidhu JS, Hong S, Faustman EM. Essential role of extracellular matrix overlay in establishing the functional integrity of primary neonatal rat sertoli cell/gonocyte cocultures: an improved in vitro model for assessment of male reproductive toxicity. Toxicol. Sci. 2005;84:378–393. doi: 10.1093/toxsci/kfi085. [DOI] [PubMed] [Google Scholar]
  • 82.Xu JJ, Henstock PV, Dunn MC, Smith AR, Chabot JR, de Graaf D. Cellular imaging predictions of clinical drug-induced liver injury. Toxicolog. Sci. 2008;105(1):97–105. doi: 10.1093/toxsci/kfn109. [DOI] [PubMed] [Google Scholar]
  • 83.O'brien PJ, Irwin W, Diaz D, Howard-Cofield E, Krejsa CM, Slaughter MR, Gao B, Kaludercic N, Angeline A, Bernardi P, Brain P, Hougham C. High concordance of drug-induced human hepatotoxicity with in vitro cytotoxicity measured in a novel cell-based model using high content screening. Arch. Toxicol. 2006;80(9):580–604. doi: 10.1007/s00204-006-0091-3. [DOI] [PubMed] [Google Scholar]
  • 84.Abraham VC, Towne DL, Waring JF, Warrior U, Burns DJ. Application of a high content multiparameter cytotoxicity assay to prioritize compounds based on toxicity potential in humans. J. Biomol. Screen. 2008;13:527–537. doi: 10.1177/1087057108318428. [DOI] [PubMed] [Google Scholar]
  • 85.Perrimon N, Mathey-Prevot B. Applications of high-throughput RNA interference screens to problems in cell and developmental biology. Genetics. 2007;175:7–16. doi: 10.1534/genetics.106.069963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Moffat J, Grueneberg DA, Yang X, Kim SY, Kloepfer AM, Hinkle G, Piqani B, Eisenhaure TM, Luo B, Grenier JK, Carpenter AE, Foo SY, Stewart SA, Stockwell BR, Hacohen N, Hahn WC, Lander ES, Sabatini DM, Root DE. A lentiviral RNAi library for human and mouse genes applied to an arrayed viral high content screen. Cell. 2006;24:1283–1298. doi: 10.1016/j.cell.2006.01.040. [DOI] [PubMed] [Google Scholar]
  • 87.Björklund M, Taipale M, Varjosalo M, Saharinen J, Lahdenperä J, Taipale J. Identification of pathways regulating cell size and cell-cycle progression by RNAi. Nature. 2006;439:1009–1013. doi: 10.1038/nature04469. [DOI] [PubMed] [Google Scholar]
  • 88.Low J, Huang S, Dowless M, Blosser W, Vincent T, Davis S, Hodson J, Koller E, Marcusson E, Blanchard K, Stancato L. High content imaging analysis of the knockdown effects of validated siRNAs and antisense oligonucleotides. J. Biomol. Screen. 2007;12(6):775–788. doi: 10.1177/1087057107302675. [DOI] [PubMed] [Google Scholar]
  • 89.Xin H, Bernal A, Amato FA, Pinhasov A, Kauffman J, Brenneman DE, Derian CK, Andrade-Gordon P, Plata-Salamán CR, Ilyin SE. High-throughput siRNA-based functional target validation. J. Biomol. Screen. 2004;9(4 ):286–293. doi: 10.1177/1087057104263533. [DOI] [PubMed] [Google Scholar]
  • 90.Trammell MA, Mahoney NM, Agard DA, Vale RD. Mob4 plays a role in spindle focusing in Drosophila S2 cells. J. Cell Sci. 2008;121:1284–1292. doi: 10.1242/jcs.017210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Walker E, Ohishi M, Davey RE, Zhang W, Cassar PA, Tanaka TS, Der SD, Morris Q, Hughes TR, Zandstra PW, Stanford WL. Prediction and testing of novel transcription networks regulating embryonic stem cell self-renewal and commitment. Stem Cell. 2007;1:71–86. doi: 10.1016/j.stem.2007.04.002. [DOI] [PubMed] [Google Scholar]
  • 92.Davey RE, Onishi K, Mahdavi A, Zandstra PW. LIF-mediated control of embryonic stem cell self-renewal emerges due to an autoregulatory loop. FASEB J. 2007;21(9):2020–2032. doi: 10.1096/fj.06-7852com. [DOI] [PubMed] [Google Scholar]
  • 93.Bauwens CL, Peerani R, Niebruegge S, Woodhouse KA, Kumacheva E, Husain M, Zandstra PW. Control of human embryonic stem cell colony and aggregate size heterogeneity influences differentiation trajectories. Stem Cells. 2008;26(9):2300–2310. doi: 10.1634/stemcells.2008-0183. [DOI] [PubMed] [Google Scholar]
  • 94.Cole MF, Johnstone SE, Newman JJ, Kagey MH, Young RA. Tcf3 is an integral component of the core regulatory circuitry of embryonic stem cells. Genes Dev. 2008;22:746–755. doi: 10.1101/gad.1642408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Tay YM, Tam WL, Ang YS, Gaughwin PM, Yang HH, Wang W, Liu R, George J, Ng HH, Perera RJ, Lufkin T, Rigoutsos I, Thomson AM, Lim B. MicroRNA-134 modulates the differentiation of mouse embryonic stem cells where it cuses post-transcriptional attenuation of naog and LRH1. Stem Cells. 2007;26(1):17–29. doi: 10.1634/stemcells.2007-0295. [DOI] [PubMed] [Google Scholar]
  • 96.Lawrence JM, Singhal S, Bhatia B, Keegan DJ, Reh TA, Luthert PJ, Khaw PT, Limb GA. MIO-M1 cells aand similar Muller glial cell lines derived from adult human retina exhibit neural stem cell characteristics. Stem Cells. 2007;25:2033–2043. doi: 10.1634/stemcells.2006-0724. [DOI] [PubMed] [Google Scholar]
  • 97.Arrell DK, Niederlander NJ, Faustino RS, Behfar A, Terzic A. Cardioinductive network guiding stem cell differentiation revealed by proteomic cartography of TNF α -primed endodermal secretome. Stem Cells. 2007;26:387–400. doi: 10.1634/stemcells.2007-0599. [DOI] [PubMed] [Google Scholar]
  • 98.Peerani R, Rao BM, Bauwens C, Yin T, Wood GA, Nagy A, Kumacheva E, Zandstra PW. Niche-mediated control of human embryonic stem cell self-renewal and diferentiation. EMBO J. 2007;26:4744–4755. doi: 10.1038/sj.emboj.7601896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Cappella P, Gasparri F, Pulici M, Moll J. A novel method based on click chemistry which overcomes limitations of cell cycle analysis by classical determination of BrdU incorporation, allowing multiplex antibody staining. Cytometry A. 2008;73(7):626–636. doi: 10.1002/cyto.a.20582. [DOI] [PubMed] [Google Scholar]
  • 100.Lamian V, Rich A, Ma Z, Li J, Seethala R, Gordon D, Dubaquie Y. Characterization of agonist-induced motilin receptor trafficking and its implications for tachyphylaxis. Mol. Pharmacol. 2006;69:109 – 118. doi: 10.1124/mol.105.017111. [DOI] [PubMed] [Google Scholar]
  • 101.The Committee on Toxicity Testing and Assessment of Environmental Agents Toxicity testing in the 21 century: a vision and a strategy. Natl. Res. Council . 2007:216. ISBN: 0-3-9-10993-0. [Google Scholar]
  • 102.NICEATM and ICCVAM The NICEATM-ICCVAM five year plan: a plan to advance alternative test methods of high scientific quality to protect and advance the health of people, animals, and the environment. NIH Publication No. 08-6410, p. 59.

Articles from Combinatorial Chemistry & High Throughput Screening are provided here courtesy of Bentham Science Publishers

RESOURCES