Cell-Based Assays in High-Throughput Screening for Drug Discovery

Authors

  • Ru Zang William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH 43210, USA
  • Ding Li William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH 43210, USA
  • I-Ching Tang Bioprocessing Innovative Company, 4734 Bridle Path Ct., Dublin, OH 43017, USA
  • Jufang Wang School of Biosciences and Bioengineering, South China University of Technology, Guangzhou, China
  • Shang-Tian Yang William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH 43210, USA

DOI:

https://doi.org/10.6000/1927-3037.2012.01.01.02

Keywords:

Cell-based high throughput screening, drug discovery, cell culture, herbal medicine, phytochemicals

Abstract

Drug screening is a long and costly process confronted with low productivity and challenges in using animals, which limit the discovery of new drugs. To improve drug screening efficacy and minimize animal testing, recent efforts have been dedicated to developing cell-based high throughput screening (HTS) platforms that can provide more relevant in vivo biological information than biochemical assays and thus reduce the number of animal tests and accelerate the drug discovery process. Today, cell-based assays are used in more than half of all high-throughput drug screenings for target validation and ADMET (absorption, distribution, metabolism, elimination and toxicity) in the early stage of drug discovery. In this review, we discuss the uses of different types of cells and cell culture systems, including 2D, 3D and perfusion cell cultures, in cell-based HTS for drug discovery. Optical and electrochemical methods for online, non-invasive detection and quantification of cells or cellular activities are discussed. Recent progresses and applications of 3D cultures and microfluidic systems for cell-based HTS are also discussed, followed with several successful examples of using cell-based HTS in commercial development of new drugs. Finally, a brief discussion on potential applications of cell-based HTS for screening phytochemicals and herbal medicines is provided in this review.

References

Anonymous. 2nd Annual Cell-Based Assays - Optimizing methods and integrating new platforms for drug discovery, development and toxicity testing. 2011; http://www.visiongain.com/Conference/299/2nd-Annual-Cell-Based-Assays-2011

Barberis A. Cell-based high-throughput screens for drug discovery. Eur Biopharmaceut Rev 2002.

An WF, Tolliday N. Cell-based assays for high throughput screening. Mol Biotechnol 2010; 45:180-186. http://dx.doi.org/10.1007/s12033-010-9251-z

Sundberg SA. High-throughput and ultra-high-throughput screening: solution- and cell-based approaches. Curr Opin Biotechnol 2000, 11: 47–53. http://dx.doi.org/10.1016/S0958-1669(99)00051-8

Denyer J, Worley J, Cox B, Allenby G, Banks M. HTS approaches to voltage-gated ion channel drug discovery. Drug Discov Today 1998; 3: 323-332. http://dx.doi.org/10.1016/S1359-6446(98)01199-4

Gonzales J, Oades K, Leychkis Y, Harootunian A, Negulescu P. Cell based assays and instrumentation for screening ion-channel targets. Drug Discov Today 1999; 4: 431-439. http://dx.doi.org/10.1016/S1359-6446(99)01383-5

Sharif T, Sharif M. A novel approach for examining the antiproliferative effect of protein kinase C inhibitors against human astrocytoma cells. Int J Oncol 1998; 13: 685-692.

Sharif T, Sharif M. A high throughput system for the evaluation of protein kinase C inhibitors based on Elk1 transcriptional activation in human astrocytoma cells. Int J Oncol 1999; 14: 327-335.

DeBasio R, Guiliano K, Zhou L, Demarest K. Quantification of G-protein coupled receptor internalization using G-protein coupled receptor–green fluorescent protein conjugates with the ArrayScanTM high-content screening system. J Biomol Screening 1999; 4: 75-86. http://dx.doi.org/10.1177/108705719900400207

Bedard J, May S, Barbeau D, Yuen L, Rando R, Bowlin T. A high throughput colorimetric cell proliferation assay for the identification of human cytomegalovirus inhibitors. Antiviral Res 1999; 41: 35-43. http://dx.doi.org/10.1016/S0166-3542(98)00061-8

Bharat Book Bureau. Cell-based Assays: Technologies and Global Markets. http://robotics.tmcnet.com/news/2011/12/29/ 6022837.htm

Kunapuli P, Lee S, Zheng W, et al. Identification of small molecule antagonists of the human mas-related gene-X1 receptor. Analytical Biochem 2006; 351: 50–61. http://dx.doi.org/10.1016/j.ab.2006.01.014

Van der Putten HHAGM, Joosten BJLJ, Klaren PHM, Everts ME. Uptake of tri-iodothyronine and thyroxine in myoblasts and myotubes of the embryonic heart cell line H9c2. J Endocrinol 2002; 175: 587-96. http://dx.doi.org/10.1677/joe.0.1750587

Ochsenbauer-Jambor C, Jones J, Heil M, Zammit KP, Kutsch O. T-cell line for HIV drug screening using EGFP as a quantitative marker of HIV-1 replication. BioTechniques 2006; 40: 91-100. http://dx.doi.org/10.2144/000112072

Shoemaker RH. The NCI60 human tumor cell line anticancer drug screen. Nat Rev 2006; 6: 813-23. http://dx.doi.org/10.1038/nrc1951

Salmon SE, Meyskens FL Jr, Alberts DS, Soehnlen B, Young L, New drugs in ovarian cancer and malignant melanoma: in vitro phase II screening with the human tumor stem cell assay. Cancer Treat Rep 1981; 65: 1-12.

Wu X, Ding S, Ding Q, Gray NS, Schultz PG. A small molecule with osteogenesis-inducing activity in multipotent mesenchymal progenitor cells. J Am Chem Soc 2002; 124: 14520-21. http://dx.doi.org/10.1021/ja0283908

Caspi O, Itzhaki I, Kehat I, et al. In vitro electrophysiological drug testing using human embryonic stem cell derived cardiomyocytes. Stem Cells Dev 2009; 18: 161-72. http://dx.doi.org/10.1089/scd.2007.0280

Mcneish J, Roach M, Hambor J, et al. High-throughput screening in embryonic stem cell-derived neurons identifies potentiators of α-amino-3-hydroxyl-5-methyl-4-isoxazolepropionate-type glutamate receptors. J Biol Chem 2010; 285: 17209-17. http://dx.doi.org/10.1074/jbc.M109.098814

Mett H, Hölscher K, Degen H, et al. Identification of inhibitors for a virally encoded protein kinase by 2 different screening systems: In vitro kinase assay and in-cell activity assay. J Biomol Screen 2005; 10: 36-45. http://dx.doi.org/10.1177/1087057104270269

Thurow K, Entzian K, Eberlein G. Toxicological and pharmacological evaluation of new drug candidates by in vitro robotic high throughput cell assays. J Assoc Lab Automat 2004; 9: 159-62. http://dx.doi.org/10.1016/j.jala.2004.04.009

Allison D. Ebert and Clive N. Svendsen. Human stem cells and drug screening: opportunities and challenges. Nat Rev Drug Discov 2010; 9: 1-6.

Janicke RU. MCF-7 breast carcinoma cells do not express caspase-3. Breast Cancer Res Treatment 2009; 117: 219-21. http://dx.doi.org/10.1007/s10549-008-0217-9

Sharma SV, Haber DA, Settleman J. Cell line-based platforms to evaluate the therapeutic efficacy of candidate anticancer agents. Nat Rev Cancer 2010; 10: 241-53.

Sabisz M, Skladanowski A. Cancer stem cells in drug resistance and drug screening: can we exploit the cancer stem cell paradigm in search for new antitumor agents? Cancer stem cells theories and practice 424-44.

Wen Y, Yang S-T. The future of microfluidic assays in drug development. Expert Opin Drug Discov 2008; 3: 1237-53. http://dx.doi.org/10.1517/17460441.3.10.1237

Yang ST, Ng R. 3D cultures for growing and studying cells. Materials Today 2007; 10(3): 56.

Yang ST, Zhang X, Wen Y. Microbioreactors for high-throughput cytotoxicity assays. Curr Opin Drug Discov Develop 2008; 11: 111-27.

Basu S, Yang ST. Astrocyte growth and glial cell line-derived neurotrophic factor secretion in three-dimensional polyethylene terephthalate fibrous matrices. Tissue Eng 2005; 11: 940-52. http://dx.doi.org/10.1089/ten.2005.11.940

Luo J, Yang ST. Effects of three-dimensional culturing in a fibrous matrix on cell cycle, apoptosis, and mab production by hybridoma cells. Biotechnol Prog 2004; 26: 306-15.

Kim J. Three-dimensional tissue culture models in cancer biology. Semin. Cancer Biol 2005; 15: 365-77. http://dx.doi.org/10.1016/j.semcancer.2005.05.002

Cukierman E. Taking cell-matrix adhesions to the third dimension. Science 2001; 294: 1708–12. http://dx.doi.org/10.1126/science.1064829

Smitskamp-Wilms E, Pinedo HM, Veerman G, Ruiz van Haperen VW, Peters GJ. Postconfluent multilayered cell line cultures for selective screening of gemcitabine. Eur J Cancer 1998; 34: 921-26. http://dx.doi.org/10.1016/S0959-8049(97)10125-3

Fischbach C, Chen R, Matsumoto T, et al. Engineering tumors with 3D scaffolds. Nature Methods. 2007; 4: 855-860. http://dx.doi.org/10.1038/nmeth1085

Gurski LA. 3D matrices for anti-cancer drug testing and development. Oncology 2010; (Jan/Feb) 20–5.

Suna T, Jackson S, Haycock JW, MacNeil S. Culture of skin cells in 3D rather than 2D improves their ability to survive exposure to cytotoxic agents. J Biotechnol 2006; 122: 372–381. http://dx.doi.org/10.1016/j.jbiotec.2005.12.021

Sittinger M, Schultz O, Keyszer G, Minuth WW, Burmester G. R. Artificial tissues in perfusion culture. Int J Artif Organs 1997; 20: 57-62.

Wu MH, Urban JPG, Cui Z, Cui ZF. Development of PDMS microbioreactor with well-defined and homogenous culture environment for chondrocyte 3-D culture. Biomed Microdevices 2006; 8: 331-40. http://dx.doi.org/10.1007/s10544-006-9597-y

Wu MH, Huang SB, Lee GB. Microfluidic cell culture systems for drug research. Lab Chip 2010; 10: 939–56. http://dx.doi.org/10.1039/b921695b

Lob V, Geisler T, Brischwein M, Uhl R, Wolf B. Automated live cell screening system based on a 24-well-microplate with integrated micro fluidics. Med Biol Eng Comput 2007; 45: 1023–28. http://dx.doi.org/10.1007/s11517-007-0260-4

Chen SYC, Hung PJ, Lee P. J Microfluidic array for three-dimensional perfusion culture of human mammary epithelial cells. Biomed Microdevices 2011; 13: 753-58. http://dx.doi.org/10.1007/s10544-011-9545-3

Pazzano D, Mercier KA, Moran JM, et al. Comparison of chondrogenesis in static and perfused bioreactor culture. Biotechnol Prog 2000; 16: 893–96. http://dx.doi.org/10.1021/bp000082v

Leclerc E, Sakai Y, Fujii T. Microfluidic PDMS (polydimethylsiloxane) bioreactor for large-scale culture of hepatocytes. Biotechnol Prog 2004; 20: 750–55. http://dx.doi.org/10.1021/bp0300568

Hughes JD, Blagg J, Price DA, et al. Physiochemical drug properties associated with in vivo toxicological outcomes. Bioorg Med Chem Lett 2008; 18: 4872–5. http://dx.doi.org/10.1016/j.bmcl.2008.07.071

Betts JI, Baganz F. Miniature bioreactors: Current practices and future opportunities. Microb Cell Fact 2006; 5: 21. http://dx.doi.org/10.1186/1475-2859-5-21

Kumar S, Wittmann C, Heinzle E. Minibioreactors. Biotechnol Lett 2004; 26(1): 1-10. http://dx.doi.org/10.1023/B:BILE.0000009469.69116.03

Ding L, Du D, Zhang X, Ju H. Trends in cell-based electrochemical biosensors. Curr Medicinal Chem 2008; 15: 3160-70. http://dx.doi.org/10.2174/092986708786848514

Bery MN, Grivell MB. In Bioelectrochemistry of cells and tissues, Walz, D, Berg H, Milazzo G, eds, Birkhauser Verlag: Basel, Switzerland 1995, pp. 134-158. http://dx.doi.org/10.1007/978-3-0348-9063-2_4

Nonner W, Eisenberg B, Electrodiffusion in ionic channels of biological membranes. J Mol Liq 2000; 87: 149-62. http://dx.doi.org/10.1016/S0167-7322(00)00118-5

Ding L, Hao C, Xue YD, Ju HX, A bio-inspired support of gold nanoparticles-chitosan nanocomposites gel for immobilization and electrochemical study of K 562 leukemia cells. Biomacromolecules 2007; 8: 1341-46. http://dx.doi.org/10.1021/bm061224y

Ivnitski D, Abdel-Hamid I, Atanasov P, Wilkins E, Stricker S. Application of electrochemical biosensors for detection of food pathogenic bacteria. Electroanalysis 2000; 12: 317-25. http://dx.doi.org/10.1002/(SICI)1521-4109(20000301)12:5<317::AID-ELAN317>3.0.CO;2-A

Borgmann S, Radtke I, Erichsen T, Blöchl A, Heumann R, Schuhmann W. Electrochemical high-content screening of nitric oxide release from endothelial cells. ChemBioChem 2006; 7: 662-8. http://dx.doi.org/10.1002/cbic.200500399

Kamei K, Haruyama T, Mie M, Yanagida Y, Aizawa M, Kobatake E. The construction of endothelial cellular biosensing system for the control of blood pressure drugs. Biosens Bioelectron 2004; 19: 1121-4. http://dx.doi.org/10.1016/j.bios.2003.06.001

May KML, Wang Y, Bachas LG, Anderson KW. Development of a whole-cell-based biosensor for detecting histamine as a model toxin. Anal Chem 2004; 76: 4156-61. http://dx.doi.org/10.1021/ac049810+

Rabinowitz JD, Rigler P, Carswell-Crumpton C, Beeson C, McConnel HM. Screening for novel drug effects with a microphysiometer: A potent effect of clofilium unrelated to potassium channel blockade. Life Sci 1997; 61: 87-94. http://dx.doi.org/10.1016/S0024-3205(97)00543-2

Braun RD, Lanzen JL, Snyder SA, Dewhirst MW. Comparison of tumor and normal tissue oxygen tension measurements using OxyLite or microelectrodes in rodents. Am J Physiol Heart Circ Physiol 2001; 280: H2533-H2544.

Chen Y, Zhang J, Wang Y, et al. Real-time monitoring approach: assessment of effects of antibodies on the adhesion of NCI-H460 cancer cells to the extracellular matrix. Biosens Bioelectron 2008; 23: 1390-6. http://dx.doi.org/10.1016/j.bios.2007.12.006

Yeon JH, Park JK. Cytotoxicity test based on electrochemical impedance measurement of HepG2 cultured in microfabricated cell chip Anal Biochem 2005; 341: 308–15. http://dx.doi.org/10.1016/j.ab.2005.03.047

Hogg D, Boden P, Lawton G, Kozlowski R. Drug Discov World 2006; 7: 83.

Pui TS, Sudibya HG, Luan X, et al. Non-invasive detection of cellular bioelectricity based on carbon nanotube devices for high throughput drug screening. Advanced Materials 2010; 22: 3199-203. http://dx.doi.org/10.1002/adma.201000548

Wodnicka M, Guarino RD, Hemperly JJ, Timmins MR, Stitt D, Pitner JB. Novel fluorescent technology platform for high throughput cytotoxicity and proliferation assays. J Biomol Screen 2000; 5: 141-52. http://dx.doi.org/10.1177/108705710000500306

O'Brien J, Wilson I, Orton T, Pognan F. Investigation of the Alamar Blue (resazurin) fluorescent dye for the assessment of mammalian cell cytotoxicity. Eur J Biochem 2000; 267: 5421-26. http://dx.doi.org/10.1046/j.1432-1327.2000.01606.x

Derfus AM, Chan WC, Bhatia SN. Probing the cytotoxicity of semiconductor quantum dots. Nano Lett 2004; 4: 11-18. http://dx.doi.org/10.1021/nl0347334

Malich G, Markovic B, Winder C. The sensitivity and specificity of the MTS tetrazolium assay for detecting the in vitro cytotoxicity of 20 chemicals using human cell lines. Toxicology 1997; 124: 179–92. http://dx.doi.org/10.1016/S0300-483X(97)00151-0

Mahon FX, Belloc F, Lagarde V, et al. MDR1 gene overexpression confers resistance to imatinib mesylate in leukemia cell line models. Blood 2003; 101: 2368-73. http://dx.doi.org/10.1182/blood.V101.6.2368

Okubo Y, Siddle K, Firth H, et al. cell proliferation activities on skin fibroblasts from a short child with absence of one copy of the type 1 insulin-like growth factor receptor (IGF1R) gene and a tall child with three copies of the IGF1R gene. J Clin Endocr Metabol 2003; 88: 5981-88. http://dx.doi.org/10.1210/jc.2002-021080

Durick K, Negulescu P. Cellular biosensors for drug discovery. Biosens Bioelectron. 2001; 16: 587-92. http://dx.doi.org/10.1016/S0956-5663(01)00173-7

Fan F, Wood KV. Bioluminescent assays for high-throughput screening. Assay Drug Dev Technol 2007; 5: 127–36. http://dx.doi.org/10.1089/adt.2006.053

Meisenheimer PL, O’Brien MA, Cali JJ. Luminogenic enzyme substrates: The basis for a new paradigm in assay design. Promega Notes 2008; 100: 22–26.

Baldwin TO, Lee J, Jung E, et al. A cell-based system for screening hair growth-promoting agents. Arch Dermatol Res 2009; 301: 381–85. http://dx.doi.org/10.1007/s00403-009-0931-0

Inoue Y, Tojo A, Sekine R, et al. In vitro validation of bioluminescent monitoring of disease progression and therapeutic response in leukemia model animals. Eur J Nuclear Med Mol Imaging 2006; 33: 557–65. http://dx.doi.org/10.1007/s00259-005-0048-4

Gribbon P, Sewing A. Fluorescence readouts in HTS: no gain without pain?. Drug Discov Today 2003; 8: 1035–43. http://dx.doi.org/10.1016/S1359-6446(03)02895-2

Beske OE, Goldbard S. High-throughput cell analysis using multiplexed array technologies. Drug Disc Today 2002; 7(18Suppl): S131-S135.

Qdot nanocrystals: The vision of nanotechnology: Invitrogen Corp, Carlsbad, CA, USA. www.qdots.com

Wolff M, Wiedenmann J, Nienhaus GU, Valler M, Heilker R. Novel fluorescent proteins for high-content screening. Drug Discov Today 2006; 11: 1054-60. http://dx.doi.org/10.1016/j.drudis.2006.09.005

Klug MG, Soonpaa MH, Koh GY, Field LJ. Genetically selected cardiomyocytes from differentiating embryonic stem cells form stable intracardiac grafts. J Clin Invest 1996; 98: 216-24. http://dx.doi.org/10.1172/JCI118769

Zweigerdt R, Burg M, Willbold E, Abts H, Ruediger M. Generation of confluent cardiomyocyte monolayers derived from embryonic stem cells in suspension: A cell source for new therapies and screening strategies. Cytotherapy 2003; 5: 399-13. http://dx.doi.org/10.1080/14653240310003062

Misra RP, Bronson SK, Xiao Q, et al. Generation of single-copy transgenic mouse embryos directly from ES cells by tetraploid embryo complementation. BMC Biotechnol 2001; 1: 12. http://dx.doi.org/10.1186/1472-6750-1-12

Takahashi T, Lord B, Schulze PC, et al. Ascorbic acid enhances differentiation of embryonic stem cells into cardiac myocytes. Circulation 2003; 107: 1912-16. http://dx.doi.org/10.1161/01.CIR.0000064899.53876.A3

Axiogenesis AG, Ehlich A, Bohlen H, Schwengberg S. Secreted proteins as markers for cell differentiation. WO-2005005662. 2005.

Danalache BA, Paquin J, Donghao W, et al. Nitric oxide signaling in oxytocin mediated cardiomyogenesis. Stem Cells 2007; 25: 679-88. http://dx.doi.org/10.1634/stemcells.2005-0610

Huber I, Itzhaki I, Caspi O, et al. Identification and selection of cardiomyocytes during human embryonic stem cell differentiation. FASEB J 2007; 21: 2551-63. http://dx.doi.org/10.1096/fj.05-5711com

Meyer N, Jaconi M, Landopoulou A, Fort P, Pucéat M. A fluorescent reporter gene as a marker for ventricular specification in ES-derived cardiac cells. FEBS Lett 2000; 478: 151-58. http://dx.doi.org/10.1016/S0014-5793(00)01839-1

Müller M, Fleischmann BK, Selbert S, et al. Selection of ventricular-like cardiomyocytes from ES cells in vitro. FASEB J 2000; 14: 2540-48. http://dx.doi.org/10.1096/fj.00-0002com

Wobus AM, Kaomei G, Shan J, et al. Retinoic acid accelerates embryonic stem cell-derived cardiac differentiation and enhances development of ventricular cardiomyocytes. J Mol Cell Cardiol 1997; 29: 1525-39. http://dx.doi.org/10.1006/jmcc.1997.0433

Bremer S, Worth AP, Paparella M, et al. Establishment of an in vitro reporter gene assay for developmental cardiac toxicity. Toxicol Vitro 2001; 15: 215-23. http://dx.doi.org/10.1016/S0887-2333(01)00007-8

Kolossov E, Fleischmann BK, Liu Q, et al. Functional characteristics of ES cell-derived cardiac precursor cells identified by tissue-specific expression of the green fluorescent protein. J Cell Biol 1998; 143: 2045-56. http://dx.doi.org/10.1083/jcb.143.7.2045

Hescheler J, Fleischmann BK, Wartenberg M, et al. Establishment of ionic channels and signaling cascades in the embryonic stem cell-derived primitive endoderm and cardiovascular system. Cells Tissues Organs 1999; 165: 153-164. http://dx.doi.org/10.1159/000016695

Anderson D, Self T, Mellor IR, Goh G, Hill SJ, Denning C. Transgenic enrichment of cardiomyocytes from human embryonic stem cells. Mol Ther 2007; 15: 2027-36. http://dx.doi.org/10.1038/sj.mt.6300303

Fijnvandraat AC, van Ginneken AC, Schumacher CA, et al. Cardiomyocytes purified from differentiated embryonic stem cells exhibit characteristics of early chamber myocardium. J Mol Cell Cardiol 2003; 35: 1461-72. http://dx.doi.org/10.1016/j.yjmcc.2003.09.011

Ezekiel U. Embryoid body-based screen. US20070015210. 2007.

Parmar M, Li M. Early specification of dopaminergic phenotype during ES cell differentiation. BMC Dev Biol 2007; 7: 86. http://dx.doi.org/10.1186/1471-213X-7-86

Veraitch FS, Scott R, Wong JW, Lye GJ, Mason C. The impact of manual processing on the expansion and directed differentiation of embryonic stem cells. Biotechnol Bioeng 2007; 99: 1216-29. http://dx.doi.org/10.1002/bit.21673

Andressen C, Stöcker E, Klinz FJ, et al. Nestin-specific green fluorescent protein expression in embryonic stem cell-derived neural precursor cells used for transplantation. Stem Cells 2001; 19: 419-24. http://dx.doi.org/10.1634/stemcells.19-5-419

Hedlund E, Pruszak J, Ferree A, et al. Selection of embryonic stem cell-derived enhanced green fluorescent protein-positive dopamine neurons using the tyrosine hydroxylase promoter is confounded by reporter gene expression in immature cell populations. Stem Cells 2007; 25: 1126-35. http://dx.doi.org/10.1634/stemcells.2006-0540

Yoshizaki T, Inaji M, Kouike H, et al. Isolation and transplantation of dopaminergic neurons generated from mouse embryonic stem cells. Neurosci Lett 2004; 363: 33-37. http://dx.doi.org/10.1016/j.neulet.2004.03.074

Wischhusen J, Melino G, Weller M. p53 and its family members – reporter genes may not see the difference. Nat Cell Death Different 2004; 11: 1150–52. http://dx.doi.org/10.1038/sj.cdd.4401477

Marchetti S, Gimond C, Iljin K, et al. Endothelial cells genetically selected from differentiating mouse embryonic stem cells incorporate at sites of neovascularization in vivo. J Cell Sci 2002; 115: 2075-85.

Hirai H, Ogawa M, Suzuki N, et al. Hemogenic and nonhemogenic endothelium can be distinguished by the activity of fetal liver kinase (Flk)-1 promoter/enhancer during mouse embryogenesis. Blood 2003; 101: 886-93. http://dx.doi.org/10.1182/blood-2002-02-0655

Asahina K, Fujimori H, Shimizu-Saito K, et al. Expression of the liver-specific gene CYP7A1 reveals hepatic differentiation in embryoid bodies derived from mouse embryonic stem cells. Genes Cells 2004; 9: 1297-308. http://dx.doi.org/10.1111/j.1365-2443.2004.00809.x

Pla P, Solov'eva O, Moore R, Alberti C, Kunisada T, Larue L. Dct::lacZ ES cells: A novel cellular model to study melanocyte determination and differentiation. Pigment Cell Res 2004; 17: 142-49. http://dx.doi.org/10.1046/j.1600-0749.2003.00121.x

Vigneau C, Polgar K, Striker G, et al. Mouse embryonic stem cell-derived embryoid bodies generate progenitors that integrate long term into renal proximal tubules in vivo. J Am Soc Nephrol 2007; 18: 1709-20. http://dx.doi.org/10.1681/ASN.2006101078

Gerrard L, Zhao D, Clark AJ, Cui W. Stably transfected human embryonic stem cell clones express OCT4-specific green fluorescent protein and maintain self-renewal and pluripotency. Stem Cells 2005; 23: 124-33. http://dx.doi.org/10.1634/stemcells.2004-0102

Eiges R, Schuldiner M, Drukker M, Yanuka O, Itskovitz-Eldor J, Benvenisty N. Establishment of human embryonic stem cell transfected clones carrying a marker for undifferentiated cells. Curr Biol 2001; 11: 514-18. http://dx.doi.org/10.1016/S0960-9822(01)00144-0

Mahajan NP, Harrison-Shostak DC, Michaux J, Herman B. Novel mutant green fluorescent protein protease substrates reveal the activation of specific caspases during apoptosis. Chem Biol 1999; 6(6): 401-409. http://dx.doi.org/10.1016/S1074-5521(99)80051-9

Xu X, Gerard AL, Huang BC, Anderson DC, Payan DG, Luo Y. Detection of programmed cell death using fluorescence energy transfer. Nucleic Acids Res 1998; 26(8): 2034-35. http://dx.doi.org/10.1093/nar/26.8.2034

Cellomics Inc, Pittsburg, PA, USA. www.cellomics.com

BD Biosciences, San Jose, CA, USA. www.bdbiosciences.com

GE Healthcare Biosciences AB, Uppsala, Sweden. www.gelifesciences.com

Abraham VC, Taylor DL, Haskins JR. High content screening applied to large-scale cell biology. Trends Biotechnol 2004; 22(1): 15-22. http://dx.doi.org/10.1016/j.tibtech.2003.10.012

Haney SA, LaPan P, Pan J, Zhang J. High-content screening moves to the front of the line. Drug Discov Today 2006; 11(19-20): 889-94. http://dx.doi.org/10.1016/j.drudis.2006.08.015

Ashcroft RG, Lopez PA. Commercial high-speed machines open new opportunities in high throughput flow cytometry (HTFC). J Immunol Methods 2000; 243: 13-24. http://dx.doi.org/10.1016/S0022-1759(00)00219-2

Zhang X, Yang ST. High-throughput 3-D cell-based proliferation and cytotoxicity assays for drug screening and bioprocess development. J Biotechnol 2011; 151: 186-93. http://dx.doi.org/10.1016/j.jbiotec.2010.11.012

Hunt L, Jordan M, De Jesus M, Wurm FM. GFP-expressing mammalian cells for fast, sensitive, noninvasive cell growth assessment in a kinetic mode. Biotechnol Bioeng 1999; 65(2): 201-205. http://dx.doi.org/10.1002/(SICI)1097-0290(19991020)65:2<201::AID-BIT10>3.0.CO;2-H

Girard P, Jordan M, Tsao M, Wurm FM. Small-scale bioreactor system for process development and optimization. Biochem Eng J 2001; 7(2): 117-19. http://dx.doi.org/10.1016/S1369-703X(00)00110-8

Leclerc, E, Sakai, Y, Fujii T. Cell culture in 3-D microfluidic structure of PDMS. Biomed Microdev 2003; 5: 109-14. http://dx.doi.org/10.1023/A:1024583026925

Viravaidya K, Sin A, Shuler ML. Development of a microscale cell culture analog to probe naphthalene toxicity. Biotechnol Prog 2004; 20: 316-23. http://dx.doi.org/10.1021/bp0341996

Walker GM, Ozers MS, Beebe DJ. Insect cell culture in microfluidic channels. Biomed Microdev 2002; 4: 161-66. http://dx.doi.org/10.1023/A:1016088128057

Hung PJ, Lee PJ, Sabounchi P, Lin R, Lee LP. Continuous perfusion microfluidic cell culture array for high-throughput cell-based assays. Biotechnol Bioeng 2005; 89: 1-8. http://dx.doi.org/10.1002/bit.20289

Su X, Young EWK, Underkofler HAS, Kamp TJ, January CT, Beebe DJ. Microfluidic Cell Culture and Its Application in High-Throughput Drug Screening: Cardiotoxicity Assay for hERG channels. J Biomol Screen 2011; 16: 101-11. http://dx.doi.org/10.1177/1087057110386218

Barbulovic-Nad I, Auab SH, Wheeler AR. A microfluidic platform for complete mammalian cell culture. Lab Chip 2010; 10: 1536–42. http://dx.doi.org/10.1039/c002147d

Forry SP, Locascio LE. On-chip CO2 control for microfluidic cell culture. Lab Chip 2011; 11: 4041–46. http://dx.doi.org/10.1039/c1lc20505f

Vickerman V, Blundo J, Kamm R. Design, fabrication and implementation of a novael multi-parameter control microfluidic platform for 3-D cell culture and real time imaging. Lab Chip 2008; 8: 1468-77. http://dx.doi.org/10.1039/b802395f

Toh YC, Zhang C, Yu H. A novel 3D mammalian cell perfusion culture system in microfluidic channels. Lab Chip 2007; 7: 302-309. http://dx.doi.org/10.1039/b614872g

Huang CP, Lu J, Seon H, et al. Engineering microscale cellular niches for three-dimensional multicellular co-cultures Lab Chip 2009; 9: 1740–48. http://dx.doi.org/10.1039/b818401a

Wen Y, Zhang X, Yang ST. Microplate-reader compatible perfusion microbioreactor array for modular tissue culture and cytotoxicity assays. Biotechnol Progr 2010; 26: 1135-44.

Macarron R, Banks MN, Bojanic D, et al. Impact of high-throughput screening in biomedical research. Nat Rev Drug Discov 2011; 10: 188-195. http://dx.doi.org/10.1038/nrd3368

Duffy KJ. Hydrazinonaphthalene and azonaphthalene thrombopoietin mimics are nonpeptidyl promoters of megakaryocytopoiesis. J Med Chem 2001; 44: 3730–45. http://dx.doi.org/10.1021/jm010283l

Duffy KJ. Identification of a pharmacophore for thrombopoietic activity of small, non-peptidyl molecules. 1. Discovery and optimization of salicylaldehyde thiosemicarbazone thrombopoietin mimics. J Med Chem 2002; 45: 3573–75. http://dx.doi.org/10.1021/jm025535c

Duffy KJ. Identification of a pharmacophore for thrombopoietic activity of small, non-peptidyl molecules. 2. Rational design of naphtho[1,2-d]imidazole thrombopoietin mimics. J Med Chem 2002; 45: 3576–78. http://dx.doi.org/10.1021/jm0255365

Luengo JI, Duffy KJ, Shaw AN, et al. Discovery of SB-497115, a small-molecule thrombopoietin (TPO) receptor agonist for the treatment of thrombocytopenia. Blood (ASH Annual Meeting Abstracts) 2004; 104: 2910.

Erickson-Miller CL. Discovery and characterization of a selective, nonpeptidyl thrombopoietin receptor agonist. Exp Hematol 2005; 33: 85–93. http://dx.doi.org/10.1016/j.exphem.2004.09.006

O’Boyle II DR, Nower PT, Lemm JA, et al. Development of a cell-based high-throughput specificity screen using a Hepatitis C virus-bovine viral diarrhea virus dual replicon assay. Antimicrob Agents Chemother 2005; 49: 1346–53. http://dx.doi.org/10.1128/AAC.49.4.1346-1353.2005

Lemm JA, O’Boyle II D, Liu M, et al. Identification of Hepatitis C virus NS5A inhibitors. J Virol 2010; 84: 482-91. http://dx.doi.org/10.1128/JVI.01360-09

Lee C. Discovery of Hepatitis C virus HS5A inhibitors as a new class of anti-HCV therapy. Arch Pharm Res 2011; 34: 1403-407. http://dx.doi.org/10.1007/s12272-011-0921-6

Paull KD. Display and analysis of patterns of differential activity of drugs against human tumor cell lines: development of mean graph and COMPARE algorithm. J Natl Cancer Inst 1989; 81: 1088-92. http://dx.doi.org/10.1093/jnci/81.14.1088

Harborne JB. Classes and functions of secondary products from plants, in Chemicals from Plants – Perspectives on Plant Secondary Products, ed by Walton JN and Brown DE, Imperial College Press, London, UK 1999; pp. 1-25. http://dx.doi.org/10.1142/9789812817273_0001

Dillard CJ, German JB. Phytochemicals: nutraceuticals and human health, J Sci Food Agric 2000; 80: 1744-56. http://dx.doi.org/10.1002/1097-0010(20000915)80:12<1744::AID-JSFA725>3.0.CO;2-W

Steinmetz KA, Potter JD. Vegetables, fruit, and cancer prevention: a review, J Am Diet Assoc 1996; 96: 1027-39. http://dx.doi.org/10.1016/S0002-8223(96)00273-8

Lampe JW. Spicing up a vegetarian diet: chemopreventive effects of phytochemicals, Am J Clin Nutr 2003; 78: 579S-583S.

Mo H, Elson CE. Apoptosis and cell-cycle arrest in human and murine tumor cells are initiated by isoprenoids. J Nutr 1999; 129: 804-813.

Zhou JR, Mukherjee P, Gugger ET, Tanaka T, Blackburn GL, Clinton SK. Inhibition of murine bladder tumorigenesis by soy sioflavones via alternations in the cell cycle, apoptosis, and angiogenesis. Cancer Res 1998; 58: 5231-38.

Ellington AA, Berhow M, Singletary KW. Induction of macroautophagy in human colon cancer cells by soybean B-group triterpenoid saponins. Carcinogenesis 2005; 26: 159-67. http://dx.doi.org/10.1093/carcin/bgh297

Slvin J, Jacobs D, Marquart L. Whole grain consumption and chronic disease: protective mechanisms, Nutr Cancer 1997; 27: 14-21. http://dx.doi.org/10.1080/01635589709514495

Qu H, Madl RL, Takemoto DJ, Baybutt RC, Wang W. Lignans are involved in the antitumor activity of wheat bran in colon cancer SW480 cells, J Nutr 2005; 135: 598-602.

Joe AK, Liu H, Suzui M, Vural ME, Xiao D, Weinstein IB. Resveratrol induces growth inhibition, S-phase arrest, apoptosis, and changes in biomarker expression in several cancer cell lines. Clin Cancer Res 2002; 8: 893-903.

Chang YS, Seo EK, Gyllenhaal C, Block KI. Panax ginseng: a role in cancer therapy? Integrative Cancer Therapies 2003; 2: 13-33. http://dx.doi.org/10.1177/1534735403251167

Yueh MF, Kawahara M, Raucy J. Cell-based high-throughput bioassays to assess induction and inhibition of CYP1A enzymes, Toxicol In Vitro 2005; 19: 275-87. http://dx.doi.org/10.1016/j.tiv.2004.10.003

Pan SY, Chen SB, Dong HG, et al. New perspectives on Chinese herbal medicine (Zhong-Yao) research and development. Evidence-Based Compl Alt Med 2011; 2011: 403709 epub doi: 10.1093/ecam/neq056.

Okamoto H. Reconsideration of Japanese traditional herbal medicine: new field of research and clinical medicine.

Mini-Rev Med Chem 2006; 6: 543-47. http://dx.doi.org/10.2174/138955706776876258

Firenzuoli F, Gori L. Herbal medicine today: clinical and research issues. Evidence-Based Compl Alt Med 2007; 4: 37-40. http://dx.doi.org/10.1093/ecam/nem096

Anonymous, Position of the American Dietetic Association: Phytochemicals and functional foods, J Am Diet Assoc 1995; 95: 493-496.

Downloads

Published

2012-04-04

How to Cite

Zang, R., Li, D., Tang, I.-C., Wang, J., & Yang, S.-T. (2012). Cell-Based Assays in High-Throughput Screening for Drug Discovery. International Journal of Biotechnology for Wellness Industries, 1(1), 31–51. https://doi.org/10.6000/1927-3037.2012.01.01.02

Issue

Section

Articles