A HighContent Screening Approach to Stem ... - Wiley Online Library

196 downloads 14778 Views 262KB Size Report
Jul 20, 2012 - Here, we review the development of HCS technique and its past application on stem cells and discuss possible ... Xiaofeng Xia, Ph.D., The Methodist Hospital Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, USA. ..... Custom- ized RNAi libraries, either in the form of virus or synthetic oligos ...
STEM CELL TECHNOLOGY: EPIGENETICS, GENOMICS, PROTEOMICS, AND METABONOMICS Concise Review: A High-Content Screening Approach to Stem Cell Research and Drug Discovery XIAOFENG XIA,a,b STEPHEN T. WONGa,b a

Department of Systems Medicine and Bioengineering, The Methodist Hospital Research Institute, Houston, TX, USA; bWeill Cornell Medical College, Cornell University, New York, NY, USA Key Words. Stem cells • High-content screening • High-throughput screening • Drug screening • Image-based screening • Cell-based assay

ABSTRACT High-throughput screening (HTS) is a technology widely used for early stages of drug discovery in pharmaceutical and biotechnology industries. Recent hardware and software improvements have enabled HTS to be used in combination with subcellular resolution microscopy, resulting in cell image-based HTS, called high-content screening (HCS). HCS allows the acquisition of deeper knowledge at a singlecell level such that more complex biological systems can be studied in a high-throughput manner. The technique is particularly well-suited for stem cell research and drug discov-

ery, which almost inevitably require single-cell resolutions for the detection of rare phenotypes in heterogeneous cultures. With growing availability of facilities, instruments, and reagent libraries, small-to-moderate scale HCS can now be carried out in regular academic labs. We envision that the HCS technique will play an increasing role in both basic mechanism study and early-stage drug discovery on stem cells. Here, we review the development of HCS technique and its past application on stem cells and discuss possible future developments. STEM CELLS 2012;30:1800–1807

Disclosure of potential conflicts of interest is found at the end of this article.

HIGH-THROUGHPUT VERSUS HIGH-CONTENT High-throughput screening (HTS) is technology developed and widely used in the pharmaceutical and biotechnology industry. Using robotics and computers, HTS achieves the full automation of biological or pharmacological experiments so that vast amounts of reagents can be rapidly evaluated to identify biologically active candidates that can potentially be used as drugs. In industry, HTS workstations are now capable of screening tens of thousands of compounds daily, providing an irreplaceable tool for lead discovery [1, 2]. Recently, with the growing availability of instruments and reagents, HTS is increasingly used in academia because of its emerging potential in basic biomedical research. HTS is made feasible by the latest technologies including robotics, liquid handling, high-sensitivity detectors, and highperformance computing. Since its establishment, its capability continues to expand with the advances in these technologies. HTS technology was initially spurred by the concept of target-based drug discovery. In the 1970s, HTS successfully demonstrated its power in identifying compounds against isolated molecular targets. From the late 1980s to the mid-1990s, great advances in combinatorial chemistry and genomics fuelled a rapid development stage of HTS: large numbers of

new compounds were synthesized for screening, and potential new drug targets were discovered at an unprecedented rate. The primary goal of HTS development during this stage was to increase its capacity to allow the screening of massive amounts of new compounds against a rapidly increasing number of new targets. To maximize throughput, assays were carried out with purified biochemical molecules in homogeneous solutions so that the reaction could be readily miniaturized and the signal quickly detected by a plate reader. This effort led to ultra-HTS capable of testing millions of compounds. However, to researchers’ and the public’s disappointment, this ‘‘quantity’’ improvement did not result in the expected increase in new drug discovery. Instead, the productivity of the whole pharmaceutical industry decreased over the past decades [3, 4]. We now realize that a ‘‘quality’’ improvement to HTS is desired, despite the higher upfront cost, to reduce the high attrition rate in downstream pipelines. Apparently, one of the causes of inapplicability of screening hits in vivo is that biochemical screening assays do not reflect the complexity of living systems. Novel higher order biological assays are needed for a more systematic and accurate evaluation of the reagent effect. To address this challenge, cell-based assays have been increasingly used in HTS since the 1990s as a more physiological alternative to biochemical assays. In vitro cultured cells provide a first-level living complexity, in which

Author contributions: X.X. and S.T.W.: wrote the manuscript. Correspondence: Xiaofeng Xia, Ph.D., The Methodist Hospital Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, USA. Telephone: 713-441-7261; Fax: 713-441-7189; e-mail: [email protected]; or Stephen T. Wong, Ph.D., The Methodist Hospital Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, USA. Telephone: 713-441-5884; Fax: 713-441-7189; e-mail: stwong@tmhs. org Received April 20, 2012; accepted for publication June 16, 2012; first published online in STEM CELLS EXPRESS July 20, 2012; C AlphaMed Press 1066-5099/2012/$30.00/0 doi: 10.1002/stem. available online without subscription through the open access option. V 1168

STEM CELLS 2012;30:1800–1807 www.StemCells.com

Xia and Wong

1801

Figure 1. High-throughput screening (HTS) and high-content screening (HCS). HTS is a method in which large amounts of reagents are arrayed and tested automatically. Screening assays can be carried out on either purified biochemical molecules or cultured cells. Various measurements can be used as readout for the effect evaluation. HCS specifically refers to the cell-based HTS that uses microscopic images as assay readout (blue branches). It requires more data analysis capability and is only recently enabled by modern computer technology. The requirement increases with the complexity of cellular image analysis.

not only the reaction with the molecular target itself is considered but also the interaction with the complete regulatory network and feedback control mechanisms [5]. This technique also provides evaluation of membrane permeability, off-target effects, and cytotoxicity, which are critical for the eventual clinical application. Moreover, in contrast to biochemical assays, cell-based assays do not require the purification of biological molecules, which is often challenging. In fact, it can even be carried out without the prior knowledge of a defined molecular target; instead, compounds can be screened with a completely unknown mechanism using phenotypic assays. These advantages have led to increasing consideration that cell-based assays are a mainstream HTS technology, and today, cell-based assays represent approximately half of all HTS performed [6]. Complete evaluation of a complex cellular network is beyond the scope of the current technique. In early applications, the complexity had to be reduced using reporter cell lines so that the target-related signal could be readily detected from the whole well; in many studies, the cells were also genetically engineered to overexpress the protein of interest to narrow the target space. This approach is compatible with a traditional plate reader. However, with such compromises, the screening clearly did not fully use the systems biology readout of cell-based assays. Signals detected are average of the www.StemCells.com

population of all the cells in the well, such that the heterogeneity of cellular response is overlooked. Also, it usually only measures the status of a single target, wasting the rich information available. Among all current techniques, microscopy provides the best solution to this problem with its spatial, temporal, and spectral resolution. Combination of microscopy and HTS has led to a new technology: high-content screening (HCS). Broadly speaking, HCS refers to any HTS that uses multiple measurements as readout. However, HCS is now often a specific name for cellular image-based HTS (Fig. 1) [7, 8]. Notably, although microscopes have been used by biologists for over a hundred years, their high-throughput application has only recently become feasible through innovations in both microscope hardware and especially automated image analysis software. The first HCS system was created by the founders of Cellomics in 1996 [7]; today, it has been widely used in all the different stages of drug development. While it retains high-throughput capacity, HCS permits the quantitative observation of comprehensive phenotypes by providing a spatial resolution at subcellular level, a temporal resolution ranging from milliseconds to days, and a spectral resolution for the observation of multiple targets. Since its introduction, HCS has undergone rapid development and its capacity has been more and more explored. In its simplest application, cells expressing different biomarkers can be counted and

Stem Cell High-Content Screening

1802

analyzed separately in a mixed culture, enabling heterogeneity studies, which are unreachable with traditional ensemble measurements [9]. Novel computer programs have been developed for the automated analysis of cell morphology, enabling phenotypic screening on many important physiological functions, such as cell migration, transformation, and neurite outgrowth [10–12]. Programs were also developed with the attempt to fully use the capacity of HCS through comprehensive phenotype profiling, encompassing the intensity and distribution of all detectable signals in the cellular image. Such techniques may enable screening with unclear biological mechanisms, and therefore greatly expanding the application of HCS [13, 14] (Fig. 1). Today, with more than a decade of development, HCS has become a mature technique that is not only used for lead discovery in industry but also for mechanism research in academia.

STEM CELLS AS AN ASSAY VEHICLE FOR HCS In order for HCS results to be able to be faithfully transferred to downstream applications in vivo, screenings should ideally be carried out on cells from the final tissue of interest. However, under most circumstances, such cells cannot be obtained and alternatives have to be used. Several advantages make immortalized cell lines currently the most commonly used alternative: first, they are cheap and can be amplified in virtually unlimited quantities, providing a consistent and homogeneous vehicle source; second, they can be easily modified genetically to generate pure clones of reporter lines, allowing straightforward readout that can be conveniently automated. Despite these advantages, the genetic backgrounds of cell lines differ considerably from those of native cells. The marked difference often makes the screening result inapplicable in vivo [15]. Based on this concern, some HCS applications have used primary mammalian cells, which can be derived from the tissue of interest and therefore more closely resemble the endogenous state. However, a major limitation that prevents use of primary cells as a standard for HCS is their relative scarcity—it is often difficult to obtain large amount of cells reproducibly. Moreover, while several specialized primary cultures, such as hepatocytes, umbilical endothelial cells, and keratinocytes, can be established from adult human, many primary cells need to be cultured from embryonic tissues so that they have to be obtained from non-human vertebrates. The biochemical differences between species certainly pose an obstacle for the final application of the screening results [16]. In addition, transfection of primary cells is challenging, often preventing incorporation of reporter genes. Limitations to immortalized cell lines and primary cells can be addressed, at least to a certain extent, using stem cells [16]. Stem cells are nontransformed cells derived from primary tissue. Certain stem cell lines, such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), can be amplified through self-renewal so that unlimited supplies may be obtained. As we will show in the later section, these cell sources have been successfully used either directly for HCS or to produce large amount of definitive stem cells for HCS. Various techniques have been developed for the highly efficient genetic modification of stem cells, with many reporter lines established that can be used for high-throughput applications [17]. Stem cells can be generated from human tissue and, more importantly, they can be subsequently differentiated in vitro into any desired tissue type, providing a unique cell source for personalized drug discovery [18–21]. Numerous studies have

confirmed that stem cell derivatives highly resemble cells from the native tissue, both phenotypically and transcriptionally [22, 23]. All these properties make stem cells ideal vehicles for high-throughput drug screening. Conversely, stem cells can potentially be used as regenerative medicine directly, raising great interest in screening for reagents that can regulate their self-renewal and differentiation. HCS is particularly well-suited for such screening, which almost inevitably requires single-cell resolution for the analysis of morphology and biomarker expression in highly heterogeneous cultures. Application of HCS on stem cells is also propelled by the lack of understanding of stem cell biology. The capability of phenotypic screening enables HCS to be carried out when a defined molecular target is unavailable, and in return the identified hits may be used as probes for dissecting the underlying mechanism. With such positive feedback, HCS provides a powerful tool for stem cell research and is expected to eventually lead to more novel medicine discovery.

PROGRESS OF HCS ON STEM CELLS While the potential of stem cells for high-throughput drug screening, especially for toxicity assessment and personalized medicine discovery [24, 25], is well-recognized and major pharmaceutical companies are already investing in it, it may still take years for the value to be fully realized due to the lack of understanding on stem cell biology, especially on ESCs and iPSCs. Nevertheless, pioneering work on HCS with stem cells has already been reported in the literature, and its power for identifying stem cell management reagents as well as dissecting the underlying mechanism has already emerged [26]. One of the obstacles to the clinical application of human ESCs (hESCs) is that their maintenance requires feeder cells as well as medium containing serum derivatives. Moreover, hESCs survive poorly under single-cell enzymatic dissociation, adding to the complexity of in vitro amplification and manipulation of the cells. In order to develop techniques for highly reproducible large-scale amplification of hESCs under chemically defined conditions, several HCSs have been carried out for compounds that maintain the survival and selfrenewal of ESCs, especially hESCs, in feeder-free conditions [27–32]. These screens used fluorescence images generated from Oct4 expression to identify and count the pluripotent cells and evaluate the compound effect on stem cell selfrenewal. Along with a number of marketed drugs and natural compounds, several kinase inhibitors were identified through these studies to control hESC self-renewal. These include rhoassociated protein kinase (ROCK) inhibitors and RasGAP/ ERK1 dual inhibitor. While ROCK inhibition is previously known to be important for ESC survival, dual inhibition of RasGAP and ERK1 revealed novel mechanism regulating self-renewal. More recently, the mechanism on ERK1 was confirmed and identified to be through interaction with Klf4 [33]. These screening studies not only provided a novel ESC culture technique but also deepened our understanding of the mechanism of ESC self-renewal. Using a library of purified recombinant proteins, a similar HCS effort also identified pigment epithelium-derived factor to promote long-term pluripotent growth of hESCs without basic fibroblast growth factor or TGFbeta/Activin/Nodal ligand supplementation [34], providing direct evidence for the involvement of the protein in hESC self-renewal. HCS was used to identify reagents that direct stem cells to differentiate into various lineages. One such application is the endodermal differentiation of ESCs, with particular

Xia and Wong

interest in pancreatic differentiation for diabetes treatment. Using the expression of endoderm marker Sox17 as an indicator, HCSs identified compounds that can induce more than 80% of ESCs to an endodermal lineage [35, 36]. A subsequent study using the compound stauprimide as probe identified NME2 as the protein target, to regulate ESC self-renewal and endodermal differentiation through nuclear localization [36]. This study showed for the first time a role of NME2 in ESC pluripotency, demonstrating the power of HCS in studying stem cell biology. To further induce ESC-derived endodermal cells to a pancreatic lineage, HCS was also carried out using PdX1 fluorescence images as readout. The screening identified a small molecule, ()-indolactam V, that induces the differentiation of a substantial number of pancreatic progenitors from hESCs [37]. Further study indicated that the biological effect is achieved at least in part through the activation of protein kinase C signaling. HCS was also used in search for reagents that induce stem cells into neuronal cells that can potentially be used as transplantation medicine for neurodegenerative diseases. In one study, a screening of a combinatorial heterocycle library of 50,000 compounds identified a new compound, neuropathiazol, able to significantly drive neural differentiation from rat neural progenitor cells, as indicated by the bIII tubulin fluorescence images [38]. In a more basic mechanism-oriented study, a genome-wide screening of approximately 6,500 genes identified eight known and seven new genes that can specify mouse ESCs into an ectodermal neural lineage, as indicated by the number of Ta1-eGFP expressing cells [39]. Mesodermal differentiation of ESCs was also explored with the HCS technique. Using a transgenic cell line that expresses mCherry under a MYH6 promoter, compound IWR-1 was identified that increases the number of cardiomyocytes differentiated from hESC mesodermal derivatives [40]. Transcriptional profiling of the inhibitor-treated samples confirmed Wnt inhibition to be critical for cardiogenesis. More recently, iPSC has become attractive for its potential to overcome the ethical concerns of ESCs as well as the immunorejection problem [41]. However, the current technique to generate iPSCs uses a transgenic method, raising concerns regarding its future clinical utility. In order to identify small molecules that can replace the transgenes, several HCSs have been carried out, using either Oct4-GFP [42] expression or alkaline phosphatase (ALP) staining combined with morphology recognition [43] as indicators for pluripotent clones. These efforts have led to the identification of several compounds that can successfully replace some of the transgenes (e.g., Sox2), and it is expected that the effort may eventually lead to a transgene-free approach for iPSC generation. In addition to regeneration medicine study, HCS has also attracted interest in cancer research. Increasing evidence suggests that the capability of a tumor to grow and propagate resides in a very small population of cancer stem cells (CSC) [44, 45]. In order to achieve the effective treatment of cancer, it is the CSC subpopulation that needs to be eliminated instead of the bulk tumor cells. Traditional HTS is very limited in such drug discovery as it only measures the ensemble effect. Besides, therapeutic agents may work through induction of differentiation rather than apoptosis, making them indistinguishable by viability assays. HCS is ideal for such application. By subpopulation analysis and morphological examination, a recent work screened the family of cancer testis antigens, on their effect on glioblastoma tumor stem cell differentiation, and differential effects of gene knockdown were observed [46]. Using dye-efflux as an indicator, compounds that can specifically eliminate the stem-like cancer cells were also identified through HCS [9]. The resolution that HCS brings to a screen at the subpopulation level makes it an excellent tool for CSC drug discovery. www.StemCells.com

1803

Applications of HCS on stem cells have just emerged; while its power has already been demonstrated, its capability has not been fully explored. Most current studies have only applied the simplest image analysis function of HCS, that is, nuclei counting. More sophisticated functions of HCS will soon be used and expectedly will promote significant advance in stem cell research considering the past success on other cells [11, 47, 48]. It is worth noting that using cellular images as readout, HCS may also help to reduce cost and allow regular academic labs to carry out screening studies that used to be unaffordable. In a recent report, bright-field images were used for result evaluation in a screening of more than 1,000 compounds. Through image analysis, the cell confluence was automatically calculated to assess the growth rate of stem cells, leading to the discovery of compounds that can specifically inhibit glioma stem cells but not normal neural stem cells [49]. In this approach, only minimal reagent is required and the cost of screening is greatly reduced, facilitating its application in academia. A summary of the reviewed HCSs with stem cells is listed in Table 1.

GROWING AVAILABILITY OF HCS FOR ACADEMIA HTS is a technology traditionally used in industry. However, there is a trend for HTS, and especially HCS, to be increasingly used in academia. Many universities and research institutes have set up HCS facilities that used to be found only in industry. Microscopes and associated image analysis software specifically designed for HCS are commercially available from several vendors (for reviews, please read [8, 50]). More and more academic labs are now well-equipped to carry out small-to-moderate scale HCS research. In such academic screenings, most labs used libraries of compounds with known substrate-protein interactions. Each compound may serve as a probe for a specific pathway, so that the screening may not only identify potential drug candidate but also represents a ‘‘chemical genetics’’ approach to investigate the complete repertoire of signaling pathways underlying stem cell biology [51, 52]. Moreover, by screening compounds that have already been assessed toxicologically and pharmacokinetically, the ‘‘drug repositioning’’ strategy also greatly reduces the cost of drug development, enables faster to market production, and quickly translates the scientific discovery to the patient bedside [53]. To facilitate such screening, a number of compound libraries are now available either commercially or through nonprofit academic institutes. These libraries consist of hundreds to thousands of compounds that can be readily screened with academic setting. A list of such libraries that have been previously used in stem cell research is shown in Table 2. The potential of HCS for stem cell mechanism research has already been demonstrated by the works reviewed above, with growing access HCS is expected to be used by more and more labs for basic biomedical research on stem cells.

FUTURE DEVELOPMENT OF HCS ON STEM CELLS Previous work on stem cell HCS has only used its minimal function and the full capacity is yet to be explored. It is expected that HCS techniques previously developed for other applications will gradually be adopted for stem cell research. Meanwhile, new techniques including novel software, reagents, and devices are being developed for stem cell HCS. Critical to HCS is the image processing programs that automatically manage and interpret the complex cellular

DAPI staining and counting Hoechst/TRA-1-60 immunofluorescence Oct4 immunofluorescence MYH6-mCherry signal Hoechst/SSEA3 immunofluorescence Nestin, sox2 staining and morphology Dye efflux

SA461 hESC HESC line H9 and H1 hESC MYH6-mCherry hESC Human EC line NTERA2 GBM stem cells NCI H460

hESC survival in feeder-free condition hESC survival and differentiation hESC pluripotency hESC Cardiomyocyte differentiation hESC survival and differentiation GBM stem cell proliferation and differentiation High drug-efflux cancer cells

20,987 compounds 1,040 compounds 806 purified proteins 549 compounds 80 compounds shRNAs targeting 61 CTA members 1,280 compounds

2,000 compounds 3,520 compounds 3,420 compounds 20,000 compounds 800 compounds 2,128 compounds 1,160 compounds

50,000 compounds 50,000 compounds 6,500 genes 2,880 compounds

Screening library

PRL-3 inhibitor and fluspirilene

Neuropathiazol SC1 Eight known and seven new genes Theanine, sinomenine, gatifloxacin, and flurbiprofen BIX-01294 and BayK8644 ()-indolactam V IDE1 and IDE2 Stauprimide RepSox Rho-kinase or protein kinase C inhibitors Vanilloid receptor 1 agonist; Y-27632, rho-kinase inhibitor IV, HA-1077 dihydrochloride ROCK inhibitors, etc. Steroids; pinacidil, etc. PEDF IWR-1 Y-27632, HA-1077; GF109203x, rottlerin CTAs leading to multiple phenotypes

Hits identified

Abbreviations: CTA, cancer testis antigen; DAPI, 40 ,6-diamidino-2-phenylindole; GBM, glioblastoma; GFP, green fluorescent protein; GFAP, glial fibrillary acidic protein; HCN, human cortical neuronal cell line; hESC, human embryonic stem cell; iPSC, induced pluripotent stem cell; mESC, mouse embryonic stem cell; MEF, mouse embryonic fibroblast; PEDF, pigment epitheliumderived factor.

ALP staining and morphology Pdx1 immunofluorescence Sox17-dsRed signal Sox17 immunofluorescence Oct4-GFP signal Hoechst/Oct4 immunofluorescence Cell confluence

MEF HUES9 hESC derivative Sox17/dsRed mESC R1 mESC Oct4-GFP MEF HSF1 hESC iPS derivative; G179

Mouse iPSC induction hESC pancreatic differentiation mESC endodermal differentiation mESC Endodermal differentiation sox2 replacement in iPSC induction hESC survival Neural stem cell proliferation

bIII tubulin and GFAP immunofluorescence Oct4-GFP signal Ta1-eGFP signal Oct4 immunofluorescence

Assay readout

HCN Oct4-GFP mESC Ta1-eGFP mESC H9 and H1 hESC

Assay cell

Neural progenitor neuronal differentiation mESC self-renewal mESC neuronal differentiation hESC self-renewal and differentiation

Screening target

Table 1. Recent work on high-content screening with stem cells

[9]

[30] [31] [34] [40] [32] [46]

[43] [37] [35] [36] [42] [29] [49]

[38] [27] [39] [28]

Ref.

1804

Stem Cell High-Content Screening

Xia and Wong

1805

Table 2. Commercial and academic compound libraries commonly used for stem cell screening Library

Provider

The spectrum collection Microsource U.S.-Drug collection Microsource Prestwick Chemical library Prestwick Greenpharma Natural compound Prestwick Prestwick rare phytochemicals Prestwick library Library of pharmacologically Sigma active compounds Tocriscreen mini library Tocris FDA approved drug library Enzo Life Sciences Screen-well kinase inhibitor Enzo Life Sciences library InhibitorSelect protein kinase EMD Biosciences inhibitor library StemSelect small molecule EMD Biosciences regulator Bio-active lipid screening library BioMol Johns Hopkins custom collection Johns Hopkins

No. of compounds

2,056 1,040 1,200 240 320 1,230 815 640 80 244 303 928 1,503

information. In the past, advanced software tools have been developed to address the most common challenges. A typical challenge for stem cell research is heterogeneity. Stem cell cultures consist of various cell subtypes, including undifferentiated cells and various derivatives. Even within the same subtype, increasing evidence has shown that an individual cell may respond differently to the same perturbation and the cellular heterogeneity serves an important biological function, possibly representing distinct stereotyped signaling states [54– 56]. Tools have already been developed for single-cell analysis and the multivariate profiles generated from immunocytochemistry images can be used to systematically characterize the compound effect [13, 14]. It is expected that these tools will be increasingly used for stem cell HCS and lead to more accurate evaluation of compound effects as well as provide deeper insight into the signaling states of stem cells. Also, while we have discussed the many advantages of HCS, it has to be noted that HCS is typically slower than traditional HTS by 2 orders of magnitude so that its screening capacity is limited. As the rate-limiting step of HCS is image processing, it is anticipated that novel computing methods will be kept developed to significantly increase the throughput of HCS. Novel probing techniques are being developed to enable the fast detection and quantification of specific cellular states. For example, a recent work has shown that small molecules can be used to distinguish distinct states resulting from cellular differentiation from myoblasts to myotubes [57]. Such small molecules represent a significant advance over the traditionally used immunocytochemistry method by offering faster, cheaper, and easier to use reagents to distinguish the differentiation process. Novel microscopy techniques are also emerging for the detection of specific biochemical molecules, providing possible opportunities for cell lineage differentiation. In a recent work, screening was carried out for fat regulating genes using stimulated-Raman scattering microscopy, which can specifically detect the CH2 stretching vibration of fatty acid chains [58]. Such a label-free technique is specific, quantitative, and avoids the complications associated with immunostaining. It can also be applied to live cells or even animals, allowing time-lapse studies, so that the effects can be studied more thoroughly and accurately.

www.StemCells.com

Most current HCS use monolayer cultured stem cells for their convenience and low cost. However, three-dimensional (3D) cell culture technique is receiving increased interest with evidence showing that the cellular phenotype and biological response of 3D cultured cells are closer to cells in vivo [59]. It is probably not surprising as the 3D methods facilitate greater cell-cell and cell-matrix interactions allowing cells to adapt to their native morphology as well as physiological function. Various techniques have been developed for 3D cell culture, including the use of extracellular matrix protein gels, agitationbased bioreactors, porous polymeric materials, and nanoparticle materials [60–62]. 3D culture is not new to stem cells. In fact, several types of stem cells such as neural stem cells and mammary stem cells were originally cultured in 3D in spheroid form [63, 64]. Using 3D culture technique, a recent report even showed that organoid culture can be established from pure stem cells that faithfully resemble the living tissue [65]. Apparently, this will make stem cell more valuable for biomedical research and drug discovery. However, although pioneer high-throughput studies have already been carried out [66], a number of challenges still remain for the 3D culture to be used in HCS. Culture cells in 3D not only increase the cost but also complicate the result, leading to large standard deviation and poor screening performance. Perhaps more importantly, 3D culture poses a significant challenge for imaging as well as image processing. Since cells are not grown in a single focal plane, confocal sections followed by 3D reconstruction will be needed to evaluate the cell status. With these challenges addressed by further improvement of the hardware and software systems, it is anticipated that HCS on 3D cultured cells will lead to more faithful mechanism and drug discovery. Another recent innovation is gene screening using RNAi libraries. By facilitating the systematic study of the role of individual genes in any biological process, RNAi screening opens new prospects in biological research [10, 67]. Customized RNAi libraries, either in the form of virus or synthetic oligos, can now be obtained commercially. In addition, siRNA microarrays have been printed on chips and used for genome-wide RNAi screening [47, 68]. These techniques, together with other microfluidic devices that are being quickly developed, will greatly increase the throughput and reduce the cost of screening [69, 70]. With the capability of HCS in combination with RNAi screening to query the role of genes in individual cells at a subceullular resolution, RNAi screening will emerge as a powerful tool for understanding the molecular mechanism that controls the self-renewal and pluripotency of stem cells [71–75]. In return, the insightful knowledge of stem cell biology will eventually translate into novel medicine and clinical applications.

ACKNOWLEDGMENTS This research was supported by NIH R01AG028928, and Ting Tsung and Wei Fong Chao Center for Bioinformatics Research and Imaging in Neurosciences (BRAIN) to S.T.W.

DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST The authors indicate no potential conflicts of interest.

Stem Cell High-Content Screening

1806

REFERENCES 32 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28

29 30 31

Posner BA. High-throughput screening-driven lead discovery: Meeting the challenges of finding new therapeutics. Curr Opin Drug Discov Dev 2005;8:487–494. Mayr LM, Bojanic D. Novel trends in high-throughput screening. Curr Opin Pharmacol 2009;9:580–588. Mayr LM, Fuerst P. The future of high-throughput screening. J Biomol Screen 2008;13:443–448. Butcher EC. Can cell systems biology rescue drug discovery? Nat Rev Drug Discov 2005;4:461–467. Clemons PA. Complex phenotypic assays in high-throughput screening. Curr Opin Chem Biol 2004;8:334–338. An WF, Tolliday NJ. Introduction: Cell-based assays for high-throughput screening. Methods Mol Biol 2009;486:1–12. Taylor DL. Past, present, and future of high content screening and the field of cellomics. Methods Mol Biol 2007;356:3–18. Zanella F, Lorens JB, Link W. High content screening: Seeing is believing. Trends Biotechnol 2010;28:237–245. Xia X, Yang J, Li F et al. Image-based chemical screening identifies drug efflux inhibitors in lung cancer cells. Cancer Res 2010;70: 7723–7733. Wang J, Zhou X, Bradley PL et al. Cellular phenotype recognition for high-content RNA interference genome-wide screening. J Biomol Screen 2008;13:29–39. Ofengeim D, Shi P, Miao B et al. Identification of small molecule inhibitors of neurite loss induced by abeta peptide using high content screening. J Biol Chem 2012;287:8714–8723. Zhang K, Xiong H, Zhou X et al. A confident scale-space shape representation framework for cell migration detection. J Microsc 2008;231: 395–407. Loo LH, Wu LF, Altschuler SJ. Image-based multivariate profiling of drug responses from single cells. Nat Methods 2007;4:445–453. Loo LH, Lin HJ, Steininger RJ, 3rd et al. An approach for extensibly profiling the molecular states of cellular subpopulations. Nat Methods 2009;6:759–765. Eglen RM, Gilchrist A, Reisine T. The use of immortalized cell lines in GPCR screening: The good, bad and ugly. Comb Chem High Throughput Screen 2008;11:560–565. Ebert AD, Svendsen CN. Human stem cells and drug screening: Opportunities and challenges. Nat Rev Drug Discov 2010;9:367–372. Giudice A, Trounson A. Genetic modification of human embryonic stem cells for derivation of target cells. Cell Stem Cell 2008;2: 422–433. Odorico JS, Kaufman DS, Thomson JA. Multilineage differentiation from human embryonic stem cell lines. Stem Cells 2001;19:193–204. Dimos JT, Rodolfa KT, Niakan KK et al. Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science 2008;321:1218–1221. Park IH, Arora N, Huo H et al. Disease-specific induced pluripotent stem cells. Cell 2008;134:877–886. Ebert AD, Yu J, Rose FF, Jr. et al. Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature 2009;457:277–280. Przyborski SA, Smith S, Wood A. Transcriptional profiling of neuronal differentiation by human embryonal carcinoma stem cells in vitro. Stem Cells 2003;21:459–471. Beqqali A, Kloots J, Ward-van Oostwaard D et al. Genome-wide transcriptional profiling of human embryonic stem cells differentiating to cardiomyocytes. Stem Cells 2006;24:1956–1967. Beqqali A, van Eldik W, Mummery C et al. Human stem cells as a model for cardiac differentiation and disease. Cell Mol Life Sci 2009; 66:800–813. Lee G, Papapetrou EP, Kim H et al. Modelling pathogenesis and treatment of familial dysautonomia using patient-specific iPSCs. Nature 2009;461:402–406. Bushway PJ, Mercola M. High-throughput screening for modulators of stem cell differentiation. Methods Enzymol 2006;414:300–316. Chen S, Do JT, Zhang Q et al. Self-renewal of embryonic stem cells by a small molecule. Proc Natl Acad Sci USA 2006;103: 17266–17271. Desbordes SC, Placantonakis DG, Ciro A et al. High-throughput screening assay for the identification of compounds regulating selfrenewal and differentiation in human embryonic stem cells. Cell Stem Cell 2008;2:602–612. Damoiseaux R, Sherman SP, Alva JA et al. Integrated chemical genomics reveals modifiers of survival in human embryonic stem cells. Stem Cells 2009;27:533–542. Andrews PD, Becroft M, Aspegren A et al. High-content screening of feeder-free human embryonic stem cells to identify pro-survival small molecules. Biochem J 2010;432:21–33. Barbaric I, Gokhale PJ, Jones M et al. Novel regulators of stem cell fates identified by a multivariate phenotype screen of small com-

33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61

pounds on human embryonic stem cell colonies. Stem Cell Res 2010; 5:104–119. Barbaric I, Jones M, Harley DJ et al. High-content screening for chemical modulators of embryonal carcinoma cell differentiation and survival. J Biomol Screen 2011;16:603–617. Kim MO, Kim SH, Cho YY et al. ERK1 and ERK2 regulate embryonic stem cell self-renewal through phosphorylation of Klf4. Nat Struct Mol Biol 2012;19:283–290. Gonzalez R, Jennings LL, Knuth M et al. Screening the mammalian extracellular proteome for regulators of embryonic human stem cell pluripotency. Proc Natl Acad Sci USA 2010;107:3552–3557. Borowiak M, Maehr R, Chen S et al. Small molecules efficiently direct endodermal differentiation of mouse and human embryonic stem cells. Cell Stem Cell 2009;4:348–358. Zhu S, Wurdak H, Wang J et al. A small molecule primes embryonic stem cells for differentiation. Cell Stem Cell 2009;4:416–426. Chen S, Borowiak M, Fox JL et al. A small molecule that directs differentiation of human ESCs into the pancreatic lineage. Nat Chem Biol 2009;5:258–265. Warashina M, Min KH, Kuwabara T et al. A synthetic small molecule that induces neuronal differentiation of adult hippocampal neural progenitor cells. Angew Chem Int Ed Engl 2006;45:591–593. Falk A, Karlsson TE, Kurdija S et al. High-throughput identification of genes promoting neuron formation and lineage choice in mouse embryonic stem cells. Stem Cells 2007;25:1539–1545. Willems E, Spiering S, Davidovics H et al. Small-molecule inhibitors of the Wnt pathway potently promote cardiomyocytes from human embryonic stem cell-derived mesoderm. Circ Res 2011;109:360–364. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006;126:663–676. Ichida JK, Blanchard J, Lam K et al. A small-molecule inhibitor of tgf-Beta signaling replaces sox2 in reprogramming by inducing nanog. Cell Stem Cell 2009;5:491–503. Shi Y, Desponts C, Do JT et al. Induction of pluripotent stem cells from mouse embryonic fibroblasts by Oct4 and Klf4 with small-molecule compounds. Cell Stem Cell 2008;3:568–574. Al-Hajj M, Wicha MS, Benito-Hernandez A et al. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA 2003;100:3983–3988. Singh SK, Hawkins C, Clarke ID et al. Identification of human brain tumour initiating cells. Nature 2004;432:396–401. Low J, Dowless M, Shiyanova T et al. Knockdown of cancer testis antigens modulates neural stem cell marker expression in glioblastoma tumor stem cells. J Biomol Screen 2010;15:830–839. Neumann B, Held M, Liebel U et al. High-throughput RNAi screening by time-lapse imaging of live human cells. Nat Methods 2006;3: 385–390. Held M, Schmitz MH, Fischer B et al. CellCognition: Time-resolved phenotype annotation in high-throughput live cell imaging. Nat Methods 2010;7:747–754. Danovi D, Falk A, Humphreys P et al. Imaging-based chemical screens using normal and glioma-derived neural stem cells. Biochem Soc Trans 2010;38:1067–1071. Pepperkok R, Ellenberg J. High-throughput fluorescence microscopy for systems biology. Nat Rev Mol Cell Biol 2006;7:690–696. Walsh DP, Chang YT. Chemical genetics. Chem Rev 2006;106: 2476–2530. Diamandis P, Wildenhain J, Clarke ID et al. Chemical genetics reveals a complex functional ground state of neural stem cells. Nat Chem Biol 2007;3:268–273. Chong CR, Sullivan DJ, Jr. New uses for old drugs. Nature 2007;448: 645–646. Slack MD, Martinez ED, Wu LF et al. Characterizing heterogeneous cellular responses to perturbations. Proc Natl Acad Sci USA 2008; 105:19306–19311. Altschuler SJ, Wu LF. Cellular heterogeneity: Do differences make a difference? Cell 2010;141:559–563. Singh DK, Ku CJ, Wichaidit C et al. Patterns of basal signaling heterogeneity can distinguish cellular populations with different drug sensitivities. Mol Syst Biol 2010;6:369. Wagner BK, Carrinski HA, Ahn YH et al. Small-molecule fluorophores to detect cell-state switching in the context of high-throughput screening. J Am Chem Soc 2008;130:4208–4209. Wang MC, Min W, Freudiger CW et al. RNAi screening for fat regulatory genes with SRS microscopy. Nat Methods 2011;8:135–138. Cukierman E, Pankov R, Stevens DR et al. Taking cell-matrix adhesions to the third dimension. Science 2001;294:1708–1712. Griffith LG, Swartz MA. Capturing complex 3D tissue physiology in vitro. Nat Rev Mol Cell Biol 2006;7:211–224. Pampaloni F, Reynaud EG, Stelzer EH. The third dimension bridges the gap between cell culture and live tissue. Nat Rev Mol Cell Biol 2007;8:839–845.

Xia and Wong

62 Souza GR, Molina JR, Raphael RM et al. Three-dimensional tissue culture based on magnetic cell levitation. Nat Nanotechnol 2010;5: 291–296. 63 Uchida N, Buck DW, He D et al. Direct isolation of human central nervous system stem cells. Proc Natl Acad Sci USA 2000;97: 14720–14725. 64 Dontu G, Abdallah WM, Foley JM et al. In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells. Genes Dev 2003;17:1253–1270. 65 Sato T, Vries RG, Snippert HJ et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 2009;459:262–265. 66 Fernandes TG, Kwon SJ, Bale SS et al. Three-dimensional cell culture microarray for high-throughput studies of stem cell fate. Biotechnol Bioeng 2010;106:106–118. 67 Echeverri CJ, Perrimon N. High-throughput RNAi screening in cultured cells: A user’s guide. Nat Rev Genet 2006;7:373–384. 68 Neumann B, Walter T, Heriche JK et al. Phenotypic profiling of the human genome by time-lapse microscopy reveals cell division genes. Nature 2010;464:721–727.

www.StemCells.com

1807

69 Gupta K, Kim DH, Ellison D et al. Lab-on-a-chip devices as an emerging platform for stem cell biology. Lab Chip 2010;10:2019–2031. 70 Fernandes TG, Diogo MM, Clark DS et al. High-throughput cellular microarray platforms: Applications in drug discovery, toxicology and stem cell research. Trends Biotechnol 2009;27:342–349. 71 Ding L, Paszkowski-Rogacz M, Nitzsche A et al. A genome-scale RNAi screen for Oct4 modulators defines a role of the Paf1 complex for embryonic stem cell identity. Cell Stem Cell 2009;4:403–415. 72 Hu G, Kim J, Xu Q et al. A genome-wide RNAi screen identifies a new transcriptional module required for self-renewal. Genes Dev 2009;23:837–848. 73 Chia NY, Chan YS, Feng B et al. A genome-wide RNAi screen reveals determinants of human embryonic stem cell identity. Nature 2010;468:316–320. 74 Chen T, Heller E, Beronja S et al. An RNA interference screen uncovers a new molecule in stem cell self-renewal and long-term regeneration. Nature 2012;485:104–108. 75 Schaniel C, Lee DF, Lemischka IR. Exploration of self-renewal and pluripotency in ES cells using RNAi. Methods Enzymol 2010;477: 351–365.