1
|
Barrett D, Wyatt M, Bar H, Haynes MK, Edwards BS, Sklar LA, Zweifach A. A phenotypic screen for compounds that reverse cAMP-mediated suppression of T cell functions. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:314-322. [PMID: 35385793 DOI: 10.1016/j.slasd.2022.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/14/2022] [Accepted: 03/31/2022] [Indexed: 06/14/2023]
Abstract
The solid tumor microenvironment (TME) suppresses immune responses. Three alterations in the TME converge on a pathway triggered by elevated cyclic AMP (cAMP) that suppresses T cell receptor (TCR) signaling. We developed a phenotypic assay to screen for small molecules that interfere with this pathway using TALL-104 human leukemic cytotoxic T lymphocytes pretreated with prostaglandin E2 to elevate cAMP. Beads coated with anti-CD3 antibodies stimulate lytic granule exocytosis, which is detected via binding of an antibody against lysosome associated membrane protein 1 (LAMP-1) measured with flow cytometry. Confirming that the assay can find compounds with desired activity, treating cells with a phorbol ester restores exocytosis. The assay behaves well in 96-well format and we screened a collection of compounds expected to have effects on epigenetic regulatory proteins. Compounds in this collection affected lytic granule exocytosis after 24-hour treatment, but none prevented cAMP from suppressing lytic granule exocytosis. We used a fully automated 384-well version of the assay to screen the Prestwick Compound Library but obtained no confirmed hits. Analyzing this assay's performance reveals two points of interest. First, cytometry offers multiple ways to quantify signals. Z' was higher using percent positive cells than mean fluorescence because the relationship between the two measures saturates, but using percent positive could make it harder to find hits in some assays. Second, variance was higher in positive controls than in negative controls in this assay, which degrades assay performance less than if variance was higher in negative controls.
Collapse
Affiliation(s)
- David Barrett
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, United States
| | - Meghan Wyatt
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, United States
| | - Haim Bar
- Department of Statistics, University of Connecticut, Storrs, CT, United States
| | - Mark K Haynes
- University of New Mexico Center for Molecular Discovery and Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Bruce S Edwards
- University of New Mexico Center for Molecular Discovery and Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Larry A Sklar
- University of New Mexico Center for Molecular Discovery and Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Adam Zweifach
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, United States.
| |
Collapse
|
2
|
Ding M, Baker D. Recent advances in high-throughput flow cytometry for drug discovery. Expert Opin Drug Discov 2020; 16:303-317. [PMID: 33054417 DOI: 10.1080/17460441.2021.1826433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION High-throughput flow cytometry (HTFC) has proven to be an important technology in drug discovery. The use of HTFC enables multi-parametric screening of suspension cells containing heterogenous cell populations and coated particles for screening proteins of interest. Novel targets, novel cell markers and compound clusters for drug development have been identified from HTFC screens. AREAS COVERED In this article, the authors focus on reviewing the recent HTFC applications reported during the last 5-6 years, including drug discovery screens and studies for immune, immune-oncology, infectious and inflammatory diseases. The main HTFC approaches, development of HTFC systems, and automated sample preparation systems for HTFC are also discussed. EXPERT OPINION The advance of HTFC technology coupled with automated sample acquisition and sample preparation has demonstrated its utility in screening large numbers of compounds using suspension cells, facilitated screening of disease-relevant human primary cells, and enabled deep understanding of mechanism of action by analyzing multiple parameters. The authors see HTFC as a very valuable tool in immune, immune-oncology, infectious and inflammatory diseases where immune cells play essential roles.
Collapse
Affiliation(s)
- Mei Ding
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - David Baker
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
3
|
Cannone Z, Shaqra AM, Lorenc C, Henowitz L, Keshipeddy S, Robinson VL, Zweifach A, Wright D, Peczuh MW. Post-Glycosylation Diversification (PGD): An Approach for Assembling Collections of Glycosylated Small Molecules. ACS COMBINATORIAL SCIENCE 2019; 21:192-197. [PMID: 30607941 DOI: 10.1021/acscombsci.8b00139] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Many small molecule natural products with antibiotic and antiproliferative activity are adorned with a carbohydrate residue as part of their molecular structure. The carbohydrate moiety can act to mediate key interactions with the target, attenuate physicochemical properties, or both. Facile incorporation of a carbohydrate group on de novo small molecules would enable these valuable properties to be leveraged in the evaluation of focused compound libraries. While there is no universal way to incorporate a sugar on small molecule libraries, techniques such as glycorandomization and neoglycorandomization have made signification headway toward this goal. Here, we report a new approach for the synthesis of glycosylated small molecule libraries. It puts the glycosylation early in the synthesis of library compounds. Functionalized aglycones subsequently participate in chemoselective diversification reactions distal to the carbohydrate. As a proof-of-concept, we prepared several desosaminyl glycosides from only a few starting glycosides, using click cycloadditions, acylations, and Suzuki couplings as diversification reactions. New compounds were then characterized for their inhibition of bacterial protein translation, bacterial growth, and in a T-cell activation assay.
Collapse
Affiliation(s)
- Zachary Cannone
- Department of Chemistry, University of Connecticut, 55 N. Eagleville Road, U3060, Storrs, Connecticut 06269, United States
| | - Ala M. Shaqra
- Department of Molecular & Cellular Biology, University of Connecticut, 91 N. Eagleville Road, U3125, Storrs, Connecticut 06269, United States
| | - Chris Lorenc
- Department of Chemistry, University of Connecticut, 55 N. Eagleville Road, U3060, Storrs, Connecticut 06269, United States
| | - Liza Henowitz
- Department of Molecular & Cellular Biology, University of Connecticut, 91 N. Eagleville Road, U3125, Storrs, Connecticut 06269, United States
| | - Santosh Keshipeddy
- Department of Pharmaceutical Sciences, School of Pharmacy, 69 N.
Eagleville Road U3092, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Victoria L. Robinson
- Department of Molecular & Cellular Biology, University of Connecticut, 91 N. Eagleville Road, U3125, Storrs, Connecticut 06269, United States
| | - Adam Zweifach
- Department of Molecular & Cellular Biology, University of Connecticut, 91 N. Eagleville Road, U3125, Storrs, Connecticut 06269, United States
| | - Dennis Wright
- Department of Chemistry, University of Connecticut, 55 N. Eagleville Road, U3060, Storrs, Connecticut 06269, United States
- Department of Pharmaceutical Sciences, School of Pharmacy, 69 N.
Eagleville Road U3092, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Mark W. Peczuh
- Department of Chemistry, University of Connecticut, 55 N. Eagleville Road, U3060, Storrs, Connecticut 06269, United States
| |
Collapse
|
4
|
Yang XA, Zweifach A. Temperature-Dependent Expression of a CFP-YFP FRET Diacylglycerol Sensor Enables Multiple-Read Screening for Compounds That Affect C1 Domains. SLAS DISCOVERY 2019; 24:682-692. [PMID: 30802416 DOI: 10.1177/2472555219830086] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Intramolecular CFP-YFP fluorescence resonance energy transfer (FRET) sensors expressed in cells are powerful research tools but have seen relatively little use in screening. We exploited the discovery that the expression of a CFP-YFP FRET diacylglycerol sensor (DAGR) increases over time when cells are incubated at room temperature to assess requirements for robust measurements using a Molecular Devices Spectramax i3x fluorescence plate reader. Expression levels resulting in YFP fluorescence >10-fold higher than untransfected cells and phorbol ester-stimulated FRET ratio changes of 60% or more were required to consistently give robust Z' > 0.5. As a means of confirming that these conditions are suitable for screening, we developed a novel multiple-read protocol to assay the NCI's Mechanistic Set III for agonists and antagonists of C1 domain activation. Sixteen compounds prevented C1 domain translocation. However, none blocked phorbol ester-stimulated protein kinase C (PKC) activity assessed using a phospho-specific antibody-six actually stimulated PKC activity. Cytometry, which produces higher Z' for a given FRET ratio change, might have been a better approach for discovering antagonists, as it would have allowed lower phorbol ester concentrations to be used. We conclude that CFP-YFP FRET measured in a Spectramax i3x plate reader can be used for screening under the conditions we defined. Our strategy of varying expression level and FRET ratio could be useful to others for determining conditions needed for robust cell-based intramolecular CFP-YFP FRET measurements on their instrumentation.
Collapse
Affiliation(s)
- Xiuyi Alexander Yang
- 1 Department of Molecular and Cell Biology, University of Connecticut at Storrs, Storrs, CT, USA
| | - Adam Zweifach
- 1 Department of Molecular and Cell Biology, University of Connecticut at Storrs, Storrs, CT, USA
| |
Collapse
|
5
|
Chen EW, Brzostek J, Gascoigne NRJ, Rybakin V. Development of a screening strategy for new modulators of T cell receptor signaling and T cell activation. Sci Rep 2018; 8:10046. [PMID: 29968737 PMCID: PMC6030045 DOI: 10.1038/s41598-018-28106-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 06/15/2018] [Indexed: 12/31/2022] Open
Abstract
Activation of the T cell receptor (TCR) leads to the generation of a network of signaling events critical to the developmental decision making and activation of T cells. Various experimental approaches continue to identify new signaling molecules, adaptor proteins, and other regulators of TCR signaling. We propose a screening strategy for the identification of small molecules affecting TCR signaling based on the uncoupling of TCR stimulation from cellular responses in developing thymocytes. We demonstrate that this strategy successfully identifies inhibitors of kinases already shown to act downstream of TCR engagement, as well as new inhibitors. The proposed strategy is easily scalable for high throughput screening and will contribute to the identification of new druggable targets in T cell activation.
Collapse
Affiliation(s)
- Elijah W Chen
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2 Blk MD4, Singapore, 117545, Singapore
| | - Joanna Brzostek
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2 Blk MD4, Singapore, 117545, Singapore
| | - Nicholas R J Gascoigne
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2 Blk MD4, Singapore, 117545, Singapore.
| | - Vasily Rybakin
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2 Blk MD4, Singapore, 117545, Singapore. .,Department of Immunobiology, Rega Institute for Medical Research, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
6
|
Liu Z, O’Rourke J. Expediting Antibody Discovery with a Cell and Bead Multiplexed Competition Assay. SLAS DISCOVERY 2018; 23:667-675. [DOI: 10.1177/2472555218776308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
With advances in molecular engineering and humanization, monoclonal antibodies are one of the fastest-growing classes of biopharmaceuticals. During antibody discovery, antibody from hybridoma or primary B-cell supernatants is screened for the desired binding characteristics, and secondary screens measure antibody function and concentration, identify immunoglobulin G (IgG) isotype, and assess cell health. In order to expedite the antibody discovery process, we developed a high-throughput, multiplexed cell and bead-based competition assay that identifies and quantitates mouse IgG isotypes and assesses cell health. No differences in assay performance were observed between single and multiplex formats. The linear range of the assay was from 0.5 to 50 µg/mL, and washing was not required, decreasing assay time and variability. Slight modifications to the protocol allowed quantification of dilute antibody supernatants (0.1–5 µg/mL). Using hybridoma cultures, we showed that cell viability measurements in the assay did not interfere with the bead-based IgG measurements. The assay described here is a simple mix-and-read, no-dilution screen that can reduce the time to antibody cloning and production. The high-content data can differentiate monoclonal and polyclonal wells, determine IgG quantity for downstream functional assays, provide isotype information, and monitor cell proliferation and viability.
Collapse
Affiliation(s)
- Zhaoping Liu
- Intellicyt, A Sartorius Brand, Albuquerque, NM, USA
| | | |
Collapse
|
7
|
Ding M, Clark R, Bardelle C, Backmark A, Norris T, Williams W, Wigglesworth M, Howes R. Application of High-Throughput Flow Cytometry in Early Drug Discovery: An AstraZeneca Perspective. SLAS DISCOVERY 2018; 23:719-731. [PMID: 29787326 DOI: 10.1177/2472555218775074] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Flow cytometry is a powerful tool providing multiparametric analysis of single cells or particles. The introduction of faster plate-based sampling technologies on flow cytometers has transformed the technology into one that has become attractive for higher throughput drug discovery screening. This article describes AstraZeneca's perspectives on the deployment and application of high-throughput flow cytometry (HTFC) platforms for small-molecule high-throughput screening (HTS), structure-activity relationship (SAR) and phenotypic screening, and antibody screening. We describe the overarching HTFC workflow, including the associated automation and data analysis, along with a high-level overview of our HTFC assay portfolio. We go on to discuss the practical challenges encountered and solutions adopted in the course of our deployment of HTFC, as well as future enhancements and expansion of the technology to new areas of drug discovery.
Collapse
Affiliation(s)
- Mei Ding
- 1 Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Roger Clark
- 2 Discovery Sciences, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | | | - Anna Backmark
- 1 Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Tyrrell Norris
- 1 Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Wendy Williams
- 4 Antibody Discovery and Protein Engineering, MedImmune, Cambridge, UK
| | - Mark Wigglesworth
- 3 Discovery Sciences, IMED Biotech Unit, AstraZeneca, Macclesfield, UK
| | - Rob Howes
- 1 Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
8
|
High-throughput flow cytometry for drug discovery: principles, applications, and case studies. Drug Discov Today 2017; 22:1844-1850. [DOI: 10.1016/j.drudis.2017.09.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/01/2017] [Accepted: 09/08/2017] [Indexed: 12/31/2022]
|
9
|
Gromek SM, deMayo JA, Maxwell AT, West AM, Pavlik CM, Zhao Z, Li J, Wiemer AJ, Zweifach A, Balunas MJ. Synthesis and biological evaluation of santacruzamate A analogues for anti-proliferative and immunomodulatory activity. Bioorg Med Chem 2016; 24:5183-5196. [PMID: 27614919 PMCID: PMC5065774 DOI: 10.1016/j.bmc.2016.08.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 08/17/2016] [Accepted: 08/22/2016] [Indexed: 01/17/2023]
Abstract
Santacruzamate A (SCA) is a natural product isolated from a Panamanian marine cyanobacterium, previously reported to have potent and selective histone deacetylase (HDAC) activity. To optimize the enzymatic and cellular activity, 40 SCA analogues were synthesized in a systematic exploration of the zinc-binding group (ZBG), cap terminus, and linker region. Two cap group analogues inhibited proliferation of MCF-7 breast cancer cells, with analogous increased degranulation of cytotoxic T cells (CTLs), while one cap group analogue reduced CTL degranulation, indicative of suppression of the immune response. Additional testing of these analogues resulted in reevaluation of the previously reported SCA mechanism of action. These analogues and the resulting structure-activity relationships will be of interest for future studies on cell proliferation and immune modulation.
Collapse
Affiliation(s)
- Samantha M Gromek
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, 69 N. Eagleville Rd, Storrs, CT 06269, USA
| | - James A deMayo
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, 69 N. Eagleville Rd, Storrs, CT 06269, USA
| | - Andrew T Maxwell
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, 69 N. Eagleville Rd, Storrs, CT 06269, USA
| | - Ashley M West
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, 69 N. Eagleville Rd, Storrs, CT 06269, USA
| | - Christopher M Pavlik
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, 69 N. Eagleville Rd, Storrs, CT 06269, USA
| | - Ziyan Zhao
- Department of Molecular and Cell Biology, University of Connecticut, 91 N. Eagleville Rd, Storrs, CT 06269, USA
| | - Jin Li
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, 69 N. Eagleville Rd, Storrs, CT 06269, USA
| | - Andrew J Wiemer
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, 69 N. Eagleville Rd, Storrs, CT 06269, USA
| | - Adam Zweifach
- Department of Molecular and Cell Biology, University of Connecticut, 91 N. Eagleville Rd, Storrs, CT 06269, USA
| | - Marcy J Balunas
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, 69 N. Eagleville Rd, Storrs, CT 06269, USA.
| |
Collapse
|
10
|
Zhao Z, deMayo JA, West AM, Balunas MJ, Zweifach A. Development of an Enhanced Phenotypic Screen of Cytotoxic T-Lymphocyte Lytic Granule Exocytosis Suitable for Use with Synthetic Compound and Natural Product Collections. JOURNAL OF BIOMOLECULAR SCREENING 2016; 21:556-66. [PMID: 27048485 DOI: 10.1177/1087057116643260] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 03/15/2016] [Indexed: 01/11/2023]
Abstract
We previously developed an assay of cytotoxic T-lymphocyte lytic granule exocytosis based on externalization of LAMP-1/CD107A using nonphysiological stimuli to generate maximal levels of exocytosis. Here, we used polystyrene beads coated with anti-CD3 antibodies to stimulate cells. Light scatter let us distinguish cells that contacted beads from cells that had not, allowing comparison of signaling events and exocytosis from stimulated and unstimulated cells in one sample. Bead stimulation resulted in submaximal exocytosis, making it possible to detect compounds that either augment or inhibit lytic granule exocytosis. Coupled with the assay's ability to distinguish responses in cells that have and have not contacted a stimulatory bead, it is possible to detect three kinds of compounds: inhibitors, stimulators, which cause exocytosis, and augmenters, which enhance receptor-stimulated exocytosis. To validate the assay, we screened a set of synthetic compounds identified using our previous assay and a library of 320 extracts prepared from tunicate-associated bacteria. One of the extracts augmented exocytosis threefold. Activity-guided fractionation and structure elucidation revealed that this compound is the known PKC activator teleocidin A-1. We conclude that our modified assay is suitable for screening synthetic compound plates and natural product collections, and will be useful for identifying immunologically active small molecules.
Collapse
Affiliation(s)
- Ziyan Zhao
- Department of Molecular and Cell Biology, University of Connecticut at Storrs, Storrs, CT, USA
| | - James A deMayo
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Ashley M West
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Marcy J Balunas
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Adam Zweifach
- Department of Molecular and Cell Biology, University of Connecticut at Storrs, Storrs, CT, USA
| |
Collapse
|
11
|
Doucette J, Zhao Z, Geyer RJ, Barra MM, Balunas MJ, Zweifach A. Flow Cytometry Enables Multiplexed Measurements of Genetically Encoded Intramolecular FRET Sensors Suitable for Screening. ACTA ACUST UNITED AC 2016; 21:535-47. [DOI: 10.1177/1087057116634007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 01/31/2016] [Indexed: 11/16/2022]
Abstract
Genetically encoded sensors based on intramolecular FRET between CFP and YFP are used extensively in cell biology research. Flow cytometry has been shown to offer a means to measure CFP-YFP FRET; we suspected it would provide a unique way to conduct multiplexed measurements from cells expressing different FRET sensors, which is difficult to do with microscopy, and that this could be used for screening. We confirmed that flow cytometry accurately measures FRET signals using cells transiently transfected with an ERK activity reporter, comparing responses measured with imaging and cytometry. We created polyclonal long-term transfectant lines, each expressing a different intramolecular FRET sensor, and devised a way to bar-code four distinct populations of cells. We demonstrated the feasibility of multiplexed measurements and determined that robust multiplexed measurements can be conducted in plate format. To validate the suitability of the method for screening, we measured responses from a plate of bacterial extracts that in unrelated experiments we had determined contained the protein kinase C (PKC)–activating compound teleocidin A-1. The multiplexed assay correctly identifying the teleocidin A-1-containing well. We propose that multiplexed cytometric FRET measurements will be useful for analyzing cellular function and for screening compound collections.
Collapse
Affiliation(s)
- Jaimee Doucette
- Department of Molecular and Cell Biology, University of Connecticut at Storrs, Storrs, CT
| | - Ziyan Zhao
- Department of Molecular and Cell Biology, University of Connecticut at Storrs, Storrs, CT
| | - Rory J. Geyer
- Department of Molecular and Cell Biology, University of Connecticut at Storrs, Storrs, CT
| | - Melanie M. Barra
- Department of Molecular and Cell Biology, University of Connecticut at Storrs, Storrs, CT
| | - Marcy J. Balunas
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut at Storrs, Storrs, CT
| | - Adam Zweifach
- Department of Molecular and Cell Biology, University of Connecticut at Storrs, Storrs, CT
| |
Collapse
|
12
|
Edwards BS, Sklar LA. Flow Cytometry: Impact on Early Drug Discovery. JOURNAL OF BIOMOLECULAR SCREENING 2015; 20:689-707. [PMID: 25805180 PMCID: PMC4606936 DOI: 10.1177/1087057115578273] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 02/27/2015] [Indexed: 12/15/2022]
Abstract
Modern flow cytometers can make optical measurements of 10 or more parameters per cell at tens of thousands of cells per second and more than five orders of magnitude dynamic range. Although flow cytometry is used in most drug discovery stages, "sip-and-spit" sampling technology has restricted it to low-sample-throughput applications. The advent of HyperCyt sampling technology has recently made possible primary screening applications in which tens of thousands of compounds are analyzed per day. Target-multiplexing methodologies in combination with extended multiparameter analyses enable profiling of lead candidates early in the discovery process, when the greatest numbers of candidates are available for evaluation. The ability to sample small volumes with negligible waste reduces reagent costs, compound usage, and consumption of cells. Improved compound library formatting strategies can further extend primary screening opportunities when samples are scarce. Dozens of targets have been screened in 384- and 1536-well assay formats, predominantly in academic screening lab settings. In concert with commercial platform evolution and trending drug discovery strategies, HyperCyt-based systems are now finding their way into mainstream screening labs. Recent advances in flow-based imaging, mass spectrometry, and parallel sample processing promise dramatically expanded single-cell profiling capabilities to bolster systems-level approaches to drug discovery.
Collapse
Affiliation(s)
- Bruce S Edwards
- Center for Molecular Discovery, Innovation Discovery and Training Center, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| | - Larry A Sklar
- Center for Molecular Discovery, Innovation Discovery and Training Center, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|