WO2000017643A2 - A system for cell-based screening - Google Patents

A system for cell-based screening Download PDF

Info

Publication number
WO2000017643A2
WO2000017643A2 PCT/US1999/021561 US9921561W WO0017643A2 WO 2000017643 A2 WO2000017643 A2 WO 2000017643A2 US 9921561 W US9921561 W US 9921561W WO 0017643 A2 WO0017643 A2 WO 0017643A2
Authority
WO
WIPO (PCT)
Prior art keywords
cell
cells
protein kinase
protein
microtubule
Prior art date
Application number
PCT/US1999/021561
Other languages
French (fr)
Other versions
WO2000017643A8 (en
WO2000017643A3 (en
WO2000017643A9 (en
Inventor
R. Terry Dunlay
D. Lansing Taylor
Albert H. Gough
Kenneth A. Guiliano
Richard A. Rubin
Original Assignee
Cellomics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cellomics, Inc. filed Critical Cellomics, Inc.
Priority to AU60485/99A priority Critical patent/AU6048599A/en
Priority to CA002344567A priority patent/CA2344567A1/en
Priority to EP99969495A priority patent/EP1114320A2/en
Priority to JP2000571253A priority patent/JP2002525603A/en
Publication of WO2000017643A2 publication Critical patent/WO2000017643A2/en
Publication of WO2000017643A8 publication Critical patent/WO2000017643A8/en
Publication of WO2000017643A9 publication Critical patent/WO2000017643A9/en
Publication of WO2000017643A3 publication Critical patent/WO2000017643A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1089Design, preparation, screening or analysis of libraries using computer algorithms
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5076Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving cell organelles, e.g. Golgi complex, endoplasmic reticulum
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5076Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving cell organelles, e.g. Golgi complex, endoplasmic reticulum
    • G01N33/5079Mitochondria
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6872Intracellular protein regulatory factors and their receptors, e.g. including ion channels
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00279Features relating to reactor vessels
    • B01J2219/00306Reactor vessels in a multiple arrangement
    • B01J2219/00313Reactor vessels in a multiple arrangement the reactor vessels being formed by arrays of wells in blocks
    • B01J2219/00315Microtiter plates
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00279Features relating to reactor vessels
    • B01J2219/00306Reactor vessels in a multiple arrangement
    • B01J2219/00313Reactor vessels in a multiple arrangement the reactor vessels being formed by arrays of wells in blocks
    • B01J2219/00315Microtiter plates
    • B01J2219/00317Microwell devices, i.e. having large numbers of wells
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00585Parallel processes
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00596Solid-phase processes
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/0061The surface being organic
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00614Delimitation of the attachment areas
    • B01J2219/00621Delimitation of the attachment areas by physical means, e.g. trenches, raised areas
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00632Introduction of reactive groups to the surface
    • B01J2219/00637Introduction of reactive groups to the surface by coating it with another layer
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00659Two-dimensional arrays
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/0068Means for controlling the apparatus of the process
    • B01J2219/00686Automatic
    • B01J2219/00689Automatic using computers
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/0068Means for controlling the apparatus of the process
    • B01J2219/00702Processes involving means for analysing and characterising the products
    • B01J2219/00707Processes involving means for analysing and characterising the products separated from the reactor apparatus
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00718Type of compounds synthesised
    • B01J2219/0072Organic compounds
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B60/00Apparatus specially adapted for use in combinatorial chemistry or with libraries
    • C40B60/14Apparatus specially adapted for use in combinatorial chemistry or with libraries for creating libraries
    • G01N15/1433
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N15/00Investigating characteristics of particles; Investigating permeability, pore-volume, or surface-area of porous materials
    • G01N15/10Investigating individual particles
    • G01N2015/1006Investigating individual particles for cytology
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • G01N2333/4701Details
    • G01N2333/4703Regulators; Modulating activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/91Transferases (2.)
    • G01N2333/912Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • G01N2333/91205Phosphotransferases in general
    • G01N2333/9121Phosphotransferases in general with an alcohol group as acceptor (2.7.1), e.g. general tyrosine, serine or threonine kinases
    • G01N2333/91215Phosphotransferases in general with an alcohol group as acceptor (2.7.1), e.g. general tyrosine, serine or threonine kinases with a definite EC number (2.7.1.-)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2510/00Detection of programmed cell death, i.e. apoptosis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N35/02Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor using a plurality of sample containers moved by a conveyor system past one or more treatment or analysis stations
    • G01N35/028Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor using a plurality of sample containers moved by a conveyor system past one or more treatment or analysis stations having reaction cells in the form of microtitration plates

Definitions

  • This invention is in the field of fluorescence-based cell and molecular biochemical assays for drug discovery.
  • Drug discovery is a long, multiple step process involving identification of specific disease targets, development of an assay based on a specific target, validation of the assay, optimization and automation of the assay to produce a screen, high throughput screening of compound libraries using the assay to identify "hits", hit validation and hit compound optimization.
  • the output of this process is a lead compound that goes into pre-clinical and, if validated, eventually into clinical trials.
  • the screening phase is distinct from the assay development phases, and involves testing compound efficacy in living biological systems.
  • genomic targets as points for therapeutic intervention using the existing methods and protocols has become a bottleneck in the drug discovery process due to the slow, manual methods employed, such as in vivo functional models, functional analysis of recombinant proteins, and stable cell line expression of candidate genes.
  • Primary DNA sequence data acquired through automated sequencing does not permit identification of gene function, but can provide information about common "motifs" and specific gene homology when compared to known sequence databases.
  • Genomic methods such as subtraction hybridization and RADE (rapid amplification of differential expression) can be used to identify genes that are up or down regulated in a disease state model. However, identification and validation still proceed down the same pathway.
  • proteomic methods use protein identification (global expression arrays, 2D electrophoresis, combinatorial libraries) in combination with reverse genetics to identify candidate genes of interest.
  • protein identification global expression arrays, 2D electrophoresis, combinatorial libraries
  • DAS isolated as intact cDNA are a great advantage to these methods, but they are identified by the hundreds without providing any information regarding type, activity, and distribution of the encoded protein.
  • Choosing a subset of DAS as drug screening targets is "random", and thus extremely inefficient, without functional data to provide a mechanistic link with disease. It is necessary, therefore, to provide new technologies to rapidly screen DAS to establish biological function, thereby improving target validation and candidate optimization in drug discovery.
  • Fluorescence-based reagents can yield more powerful, multiple parameter assays that are higher in throughput and information content and require lower volumes of reagents and test compounds. Fluorescence is also safer and less expensive than radioactivity-based methods. Screening of cells treated with dyes and fluorescent reagents is well known in the art. There is a considerable body of literature related to genetic engineering of cells to produce fluorescent proteins, such as modified green fluorescent protein (GFP), as a reporter molecule. Some properties of wild-type GFP are disclosed by Morise et al. (Biochemistry 13 (1974), p.
  • the GFP of the jellyfish Aequorea victoria has an excitation maximum at 395 nm and an emission maximum at 510 nm, and does not require an exogenous factor for fluorescence activity.
  • Uses for GFP disclosed in the literature are widespread and include the study of gene expression and protein localization (Chalfie et al, Science 263 (1994), p. 12501-12504)), as a tool for visualizing subcellular organelles (Rizzuto et al., Curr. Biology 5 (1995), p.
  • WO96/23898 relates to methods of detecting biologically active substances affecting intracellular processes by utilizing a GFP construct having a protein kinase activation site.
  • WO 96/09598 describes a system for isolating cells of interest utilizing the expression of a GFP like protein.
  • WO 96/27675 describes the expression of GFP in plants.
  • WO 95/21191 describes modified GFP protein expressed in transformed organisms to detect mutagenesis.
  • U. S. Patents 5,401,629 and 5,436,128 describe assays and compositions for detecting and evaluating the intracellular transduction of an extracellular signal using recombinant cells that express cell surface receptors and contain reporter gene constructs that include transcriptional regulatory elements that are responsive to the activity of cell surface receptors.
  • Standard high throughput screens use mixtures of compounds and biological reagents along with some indicator compound loaded into arrays of wells in standard microtiter plates with 96 or 384 wells.
  • the signal measured from each well either fluorescence emission, optical density, or radioactivity, integrates the signal from all the material in the well giving an overall population average of all the molecules in the well.
  • SAIC Science Applications International Corporation 130 Fifth Avenue, Seattle, WA. 98109
  • This system uses a CCD camera to image the whole area of a 96 well plate. The image is analyzed to calculate the total fluorescence per well for all the material in the well.
  • Molecular Devices, Inc. (Sunnyvale, CA) describes a system (FLIPR) which uses low angle laser scanning illumination and a mask to selectively excite fluorescence within approximately 200 microns of the bottoms of the wells in standard 96 well plates in order to reduce background when imaging cell monolayers.
  • FLIPR Molecular Devices, Inc.
  • This system uses a CCD camera to image the whole area of the plate bottom. Although this system measures signals originating from a cell monolayer at the bottom of the well, the signal measured is averaged over the area of the well and is therefore still considered a measurement of the average response of a population of cells. The image is analyzed to calculate the total fluorescence per well for cell-based assays.
  • Fluid delivery devices have also been incorporated into cell based screening systems, such as the FLIPR system, in order to initiate a response, which is then observed as a whole well population average response using a macro-imaging system.
  • High-content screens automate the extraction of multicolor fluorescence information derived from specific fluorescence-based reagents incorporated into cells (Giuliano and Taylor (1995), Curr. Op. Cell Biol. 7:4; Giuliano et al. (1995) Ann. Rev. Biophys. Biomol. Struct. 24:405). Cells are analyzed using an optical system that can measure spatial, as well as temporal dynamics. (Farkas et al. (1993) Ann. Rev. Physiol 55:785; Giuliano et al.
  • the types of biochemical and molecular information now accessible through fluorescence-based reagents applied to cells include ion concentrations, membrane potential, specific translocations, enzyme activities, gene expression, as well as the presence, amounts and patterns of metabolites, proteins, lipids, carbohydrates, and nucleic acid sequences (DeBiasio et al., (1996) Mol. Biol. Cell. 7:1259;Giuliano et al., (1995) Ann. Rev. Biophys. Biomol. Struct. 24:405; Heim and Tsien, (1996) Curr. Biol. 6:178).
  • High-content screens can be performed on either fixed cells, using fluorescently labeled antibodies, biological ligands, and/or nucleic acid hybridization probes, or live cells using multicolor fluorescent indicators and "biosensors.”
  • the choice of fixed or live cell screens depends on the specific cell-based assay required.
  • Live cell assays are more sophisticated and powerful, since an array of living cells containing the desired reagents can be screened over time, as well as space.
  • Environmental control of the cells is required during measurement, since the physiological health of the cells must be maintained for multiple fluorescence measurements over time.
  • fluorescent physiological indicators and “biosensors” that can report changes in biochemical and molecular activities within cells (Giuliano et al., (1995) Ann. Rev. Biophys. Biomol Struct. 24:405; Hahn et al., (1993) In Fluorescent and Luminescent Probes for Biological Activity. W.T. Mason, (ed.), pp. 349-359, Academic Press, San Diego).
  • the system consists of an epifluorescence inverted microscope with a motorized stage, video camera, image intensifier, and a microcomputer with a PC- Vision digitizer.
  • Turbo Pascal software controls the stage and scans the plate taking multiple images per well.
  • the software calculates total fluorescence per well, provides for daily calibration, and configures easily for a variety of tissue culture plate formats. Thresholding of digital images and reagents which fluoresce only when taken up by living cells are used to reduce background fluorescence without removing excess fluorescent reagent.
  • the instant invention provides systems, methods, and screens that combine high throughput screening (HTS) and high content screening (HCS) that significantly improve target validation and candidate optimization by combining many cell screening formats with fluorescence-based molecular reagents and computer-based feature extraction, data analysis, and automation, resulting in increased quantity and speed of data collection, shortened cycle times, and, ultimately, faster evaluation of promising drug candidates.
  • HTS high throughput screening
  • HCS high content screening
  • the instant invention also provides for miniaturizing the methods, thereby allowing increased throughput, while decreasing the volumes of reagents and test compounds required in each assay.
  • the present invention relates to a method for analyzing cells comprising
  • the method rapidly determines the distribution, environment, or activity of fluorescently labeled reporter molecules in cells for the purpose of screening large numbers of compounds for those that specifically affect particular biological functions.
  • the array of locations may be a microtiter plate or a microchip which is a microplate having cells in an array of locations.
  • the method includes computerized means for acquiring, processing, displaying and storing the data received.
  • the method further comprises automated fluid delivery to the arrays of cells.
  • the information obtained from high throughput measurements on the same plate are used to selectively perform high content screening on only a subset of the cell locations on the plate.
  • a cell screening system is provided that comprises:
  • a high magnification fluorescence optical system having a microscope objective, • an XY stage adapted for holding a plate containing an array of cells and having a means for moving the plate for proper alignment and focusing on the cell arrays;
  • a light source having optical means for directing excitation light to cell arrays and a means for directing fluorescent light emitted from the cells to the digital camera;
  • a computer means for receiving and processing digital data from the digital camera
  • the computer means includes a digital frame grabber for receiving the images from the camera, a display for user interaction and display of assay results, digital storage media for data storage and archiving, and a means for control, acquisition, processing and display of results.
  • the cell screening system further comprises a computer screen operatively associated with the computer for displaying data.
  • the computer means for receiving and processing digital data from the digital camera stores the data in a bioinformatics data base.
  • the cell screening system further comprises a reader that measures a signal from many or all the wells in parallel.
  • the cell screening system further comprises a mechanical-optical means for changing the magnification of the system, to allow changing modes between high throughput and high content screening.
  • the cell screening system further comprises a chamber and control system to maintain the temperature, CO 2 concentration and humidity surrounding the plate at levels required to keep cells alive.
  • the cell screening system utilizes a confocal scanning illumination and detection system.
  • a machine readable storage medium comprising a program containing a set of instructions for causing a cell screening system to execute procedures for defining the distribution and activity of specific cellular constituents and processes is provided.
  • the cell screening system comprises a high magnification fluorescence optical system with a stage adapted for holding cells and a means for moving the stage, a digital camera, a light source for receiving and processing the digital data from the digital camera, and a computer means for receiving and processing the digital data from the digital camera.
  • Preferred embodiments of the machine readable storage medium comprise programs consisting of a set of instructions for causing a cell screening system to execute the procedures set forth in Figures 9, 11, 12, 13, 14 or 15.
  • Another preferred embodiment comprises a program consisting of a set of instructions for causing a cell screening system to execute procedures for detecting the distribution and activity of specific cellular constituents and processes.
  • the cellular processes include, but are not limited to, nuclear translocation of a protein, cellular hypertrophy, apoptosis, and protease-induced translocation of a protein.
  • a variety of automated cell screening methods are provided, including screens to identify compounds that affect transcription factor activity, protein kinase activity, cell morphology, microtubule structure, apoptosis, receptor internalization, and protease-induced translocation of a protein.
  • Figure 1 shows a diagram of the components of the cell-based scanning system.
  • Figure 2 shows a schematic of the microscope subassembly.
  • Figure 3 shows the camera subassembly.
  • Figure 4 illustrates cell scanning system process.
  • Figure 5 illustrates a user interface showing major functions to guide the user.
  • Figure 6 is a block diagram of the two platform architecture of the Dual Mode System for Cell Based Screening in which one platform uses a telescope lens to read all wells of a microtiter plate and a second platform that uses a higher magnification lens to read individual cells in a well.
  • Figure 7 is a detail of an optical system for a single platform architecture of the Dual Mode System for Cell Based Screening that uses a moveable 'telescope' lens to read all wells of a microtiter plate and a moveable higher magnification lens to read individual cells in a well.
  • Figure 8 is an illustration of the fluid delivery system for acquiring kinetic data on the
  • Figure 9 is a flow chart of processing step for the cell-based scanning system.
  • Figure 10 A-J illustrates the strategy of the Nuclear Translocation Assay.
  • Figure 11 is a flow chart defining the processing steps in the Dual Mode System for
  • Figure 12 is a flow chart defining the processing steps in the High Throughput mode of the System for Cell Based Screening.
  • Figure 13 is a flow chart defining the processing steps in the High Content mode of the
  • Figure 14 is a flow chart defining the processing steps required for acquiring kinetic data in the High Content mode of the System for Cell Based Screening.
  • Figure 15 is a flow chart defining the processing steps performed within a well during the acquisition of kinetic data.
  • Figure 16 is an example of data from a known inhibitor of translocation.
  • Figure 17 is an example of data from a known stimulator of translocation.
  • Figure 18 illustrates data presentation on a graphical display.
  • Figure 19 is an illustration of the data from the High Throughput mode of the System for Cell Based Screening, an example of the data passed to the High Content mode, the data acquired in the high content mode, and the results of the analysis of that data.
  • Figure 20 shows the measurement of a drug-induced cytoplasm to nuclear translocation.
  • Figure 21 illustrates a graphical user interface of the measurement shown in Figure 20.
  • Figure 22 illustrates a graphical user interface, with data presentation, of the measurement shown in Fig. 20.
  • Figure 23 is a graph representing the kinetic data obtained from the measurements depicted in Fig. 20.
  • Figure 24 details a high-content screen of drug-induced apoptosis.
  • Luminescent probes that have high affinity for specific cellular constituents including specific organelles or molecules. These probes can either be small luminescent molecules or fluorescently tagged macromolecules used as "labeling reagents", “environmental indicators”, or “biosensors.”
  • Labeling reagents include, but are not limited to, luminescently labeled macromolecules including fluorescent protein analogs and biosensors, luminescent macromolecular chimeras including those formed with the green fluorescent protein and mutants thereof, luminescently labeled primary or secondary antibodies that react with cellular antigens involved in a physiological response, luminescent stains, dyes, and other small molecules.
  • Translocation markers can either simply report location relative to the markers of cellular domains or they can also be "biosensors" that report some biochemical or molecular activity as well.
  • Biosensors Macromolecules consisting of a biological functional domain and a luminescent probe or probes that report the environmental changes that occur either internally or on their surface.
  • a class of luminescently labeled macromolecules designed to sense and report these changes have been termed "fluorescent-protein biosensors".
  • the protein component of the biosensor provides a highly evolved molecular recognition moiety.
  • a fluorescent molecule attached to the protein component in the proximity of an active site transduces environmental changes into fluorescence signals that are detected using a system with an appropriate temporal and spatial resolution such as the cell scanning system of the present invention. Because the modulation of native protein activity within the living cell is reversible, and because fluorescent-protein biosensors can be designed to sense reversible changes in protein activity, these biosensors are essentially reusable.
  • DAS Disease associated sequences
  • High content screening can be used to measure the effects of drugs on complex molecular events such as signal transduction pathways, as well as cell functions including, but not limited to, apoptosis, cell division, cell adhesion, locomotion, exocytosis, and cell-cell communication.
  • Multicolor fluorescence permits multiple targets and cell processes to be assayed in a single screen. Cross-co ⁇ elation of cellular responses will yield a wealth of information required for target validation and lead optimization.
  • a cell screening system comprising a high magnification fluorescence optical system having a microscope objective, an XY stage adapted for holding a plate with an array of locations for holding cells and having a means for moving the plate to align the locations with the microscope objective and a means for moving the plate in the direction to effect focusing; a digital camera; a light source having optical means for directing excitation light to cells in the a ⁇ ay of locations and a means for directing fluorescent light emitted from the cells to the digital camera; and a computer means for receiving and processing digital data from the digital camera wherein the computer means includes: a digital frame grabber for receiving the images from the camera, a display for user interaction and display of assay results, digital storage media for data storage and archiving, and means for control, acquisition, processing and display of results.
  • FIG. 1 is a schematic diagram of a prefe ⁇ ed embodiment of the cell scanning system.
  • An inverted fluorescence microscope is used J_, such as a Zeiss Axiovert inverted fluorescence microscope which uses standard objectives with magnification of 1-1 OOx to the camera, and a white light source (e.g. 100W mercury-arc lamp or 75 W xenon lamp) with power supply 2.
  • J_ an inverted fluorescence microscope
  • J_ such as a Zeiss Axiovert inverted fluorescence microscope which uses standard objectives with magnification of 1-1 OOx to the camera, and a white light source (e.g. 100W mercury-arc lamp or 75 W xenon lamp) with power supply 2.
  • XY stage 3 to move the plate 4 in the XY direction over the microscope objective.
  • a Z-axis focus drive 5 moves the objective in the Z direction for focusing.
  • a joystick 6 provides for manual movement of the stage in the XYZ direction.
  • FIG. 1 is a schematic of one embodiment of the microscope assembly 1 of the invention, showing in more detail the XY stage 3, Z-axis focus drive 5, joystick 6, light source 2, and automation controller 9. Cables to the computer 15 and microscope 16, respectively, are provided.
  • Figure 2 shows a 96 well microtiter plate 17 which is moved on the XY stage 3 in the XY direction. Light from the light source 2 passes through the PC controlled shutter 18 to a motorized filter wheel 19 with excitation filters 20. The light passes into filter cube 25 which has a dichroic minor 26 and an emission filter 27.
  • Excitation light reflects off the dichroic minor to the wells in the microtiter plate 17 and fluorescent light 28 passes through the dichroic minor 26 and the emission filter 27 and to the digital camera 7.
  • Figure 3 shows a schematic drawing of a prefe ⁇ ed camera assembly.
  • a digital cable 30 transports digital signals to the computer.
  • the standard optical configurations described above use microscope optics to directly produce an enlarged image of the specimen on the camera sensor in order to capture a high resolution image of the specimen. This optical system is commonly refe ⁇ ed to as 'wide field' microscopy.
  • a high resolution image of the specimen can be created by a variety of other optical systems, including, but not limited to, standard scanning confocal detection of a focused point or line of illumination scanned over the specimen (Go et al. 1997, supra), and multi-photon scanning confocal microscopy (Denk et al., 1990, supra), both of which can form images on a CCD detector or by synchronous digitization of the analog output of a photomultiplier tube.
  • standard scanning confocal detection of a focused point or line of illumination scanned over the specimen Go et al. 1997, supra
  • multi-photon scanning confocal microscopy Digik confocal microscopy
  • HEK 293 ATCC CRL-1573
  • An optical system that can acquire images of single cell layers in multilayer preparations is required for use with cell lines that tend to form layers.
  • the large depth of field of wide field microscopes produces an image that is a projection through the many layers of cells, making analysis of subcellular spatial distributions extremely difficult in layer-forming cells.
  • the very shallow depth of field that can be achieved on a confocal microscope (about one micron), allows discrimination of a single cell layer at high resolution, simplifying the determination of the subcellular spatial distribution.
  • confocal imaging is preferable when detection modes such as fluorescence lifetime imaging are required.
  • the output of a standard confocal imaging attachment for a microscope is a digital image that can be converted to the same format as the images produced by the other cell screening system embodiments described above, and can therefore be processed in exactly the same way as those images.
  • the overall control, acquisition and analysis in this embodiment is essentially the same.
  • the optical configuration of the confocal microscope system is essentially the same as that described above, except for the illuminator and detectors. Illumination and detection systems required for confocal microscopy have been designed as accessories to be attached to standard microscope optical systems such as that of the present invention (Zeiss, Germany). These alternative optical systems therefore can be easily integrated into the system as described above.
  • Figure 4 illustrates an alternative embodiment of the invention in which cell a ⁇ ays are in micro wells 40 on a microplate 4J_, described ion co-pending U.S. Application S/N 08/865,341, inco ⁇ orated by reference herein in its entirety.
  • the microplate is 20 mm by 30 mm as compared to a standard 96 well microtiter plate which is 86 mm by 129 mm.
  • the higher density a ⁇ ay of cells on a microplate allows the microplate to be imaged at a low resolution of a few microns per pixel for high throughput and particular locations on the microplate to be imaged at a higher resolution of less than 0.5 microns per pixel. These two resolution modes help to improve the overall throughput of the system.
  • the microplate chamber 42 serves as a microfluidic delivery system for the addition of compounds to cells.
  • the microplate 4 . in the microplate chamber 42 is placed in an XY microplate reader 43.
  • Digital data is processed as described above.
  • the small size of this microplate system increases throughput, minimizes reagent volume and allows control of the distribution and placement of cells for fast and precise cell-based analysis.
  • Processed data can be displayed on a PC screen J_l and made part of a bioinformatics data base 44. This data base not only permits storage and retrieval of data obtained through the methods of this invention, but also permits acquisition and storage of external data relating to cells.
  • Figure 5 is a PC display which illustrates the operation of the software.
  • a high throughput system is directly coupled with the HCS either on the same platform or on two separate platforms connected electronically (e.g. via a local area network).
  • This embodiment of the invention referred to as a dual mode optical system, has the advantage of increasing the throughput of a HCS by coupling it with a HTS and thereby requiring slower high resolution data acquisition and analysis only on the small subset of wells that show a response in the coupled HTS.
  • High throughput 'whole plate' reader systems are well known in the art and are commonly used as a component of an HTS system used to screen large numbers of compounds (Beggs (1997), J. of Biomolec. Screening 2:71-78; Macaffrey et al, (1996) J. Biomolec. Screening 1 :187-190).
  • dual mode cell based screening a two platform architecture in which high throughput acquisition occurs on one platform and high content acquisition occurs on a second platform is provided (Figure 6). Processing occurs on each platform independently, with results passed over a network interface, or a single controller is used to process the data from both platforms.
  • an exemplified two platform dual mode optical system consists of two light optical instruments, a high throughput platform 60 and a high content platform 65 ⁇ which read fluorescent signals emitted from cells cultured in microtiter plates or microwell a ⁇ ays on a microplate, and communicate with each other via an electronic connection 64.
  • the high throughput platform 60 analyzes all the wells in the whole plate either in parallel or rapid serial fashion.
  • the high content platform 65 scans from well to well and acquires and analyzes high resolution image data collected from individual cells within a well.
  • the HTS software residing on the system's computer 62, controls the high throughput instrument, and results are displayed on the monitor 6J_.
  • the HCS software residing on it's computer system 67, controls the high content instrument hardware 65, optional devices (e.g. plate loader, environmental chamber, fluid dispenser), analyzes digital image data from the plate, displays results on the monitor 66 and manages data measured in an integrated database.
  • the two systems can also share a single computer, in which case all data would be collected, processed and displayed on that computer, without the need for a local area network to transfer the data.
  • Microtiter plates are transfened from the high throughput system to the high content system 63 either manually or by a robotic plate transfer device, as is well known in the art (Beggs ( 1997), supra; Mcaffrey ( 1996), supra).
  • the dual mode optical system utilizes a single platform system ( Figure 7). It consists of two separate optical modules, an HCS module 203 and an HTS module 209 that can be independently or collectively moved so that only one at a time is used to collect data from the microtiter plate 201.
  • the microtiter plate 201 is mounted in a motorized X,Y stage so it can be positioned for imaging in either HTS or HCS mode. After collecting and analyzing the HTS image data as described below, the HTS optical module 209 is moved out of the optical path and the HCS optical module 203 is moved into place.
  • the optical module for HTS 209 consists of a projection lens 214, excitation wavelength filter 213 and dichroic minor 210 which are used to illuminate the whole bottom of the plate with a specific wavelength band from a conventional microscope lamp system (not illustrated).
  • the fluorescence emission is collected through the dichroic minor 210 and emission wavelength filter 21 1 by a lens 212 which forms an image on the camera 216 with sensor 215.
  • the optical module for HCS 203 consists of a projection lens 208, excitation wavelength filter 207 and dichroic minor 204 which are used to illuminate the back aperture of the microscope objective 202, and thereby the field of that objective, from a standard microscope illumination system (not shown).
  • the fluorescence emission is collected by the microscope objective 202, passes through the dichroic minor 204 and emission wavelength filter 205 and is focused by a tube lens 206 which forms an image on the same camera 216 with sensor 215.
  • the cell screening system further comprises a fluid delivery device for use with the live cell embodiment of the method of cell screening (see below).
  • Figure 8 exemplifies a fluid delivery device for use with the system of the invention. It consists of a bank of 12 syringe pumps 701 driven by a single motor drive. Each syringe 702 is sized according to the volume to be delivered to each well, typically between 1 and 100 ⁇ L. Each syringe is attached via flexible tubing 703 to a similar bank of connectors which accept standard pipette tips
  • the bank of pipette tips are attached to a drive system so they can be lowered and raised relative to the microtiter plate 706 to deliver fluid to each well.
  • the plate is mounted on an X,Y stage, allowing movement relative to the optical system 707 for data collection pu ⁇ oses. This set-up allows one set of pipette tips, or even a single pipette tip, to deliver reagent to all the wells on the plate.
  • the bank of syringe pumps can be used to deliver fluid to 12 wells simultaneously, or to fewer wells by removing some of the tips.
  • the present invention provides a method for analyzing cells comprising providing an anay of locations which contain multiple cells wherein the cells contain one or more fluorescent reporter molecules; scanning multiple cells in each of the locations containing cells to obtain fluorescent signals from the fluorescent reporter molecule in the cells; converting the fluorescent signals into digital data; and utilizing the digital data to dete ⁇ nine the distribution, environment or activity of the fluorescent reporter molecule within the cells.
  • Standard 96 well microtiter plates which are 86 mm by 129 mm, with 6mm diameter wells on a 9mm pitch, are used for compatibility with cunent automated loading and robotic handling systems.
  • the microplate is typically 20 mm by 30 mm, with cell locations that are 100-200 microns in dimension on a pitch of about 500 microns. Methods for making microplates are described in U.S. Patent Application Serial No. 08/865,341, inco ⁇ orated by reference herein in its entirety.
  • Microplates may consist of coplanar layers of materials to which cells adhere, patterned with materials to which cells will not adhere, or etched 3-dimensional surfaces of similarly pattered materials.
  • the terms 'well' and 'micro well' refer to a location in an a ⁇ ay of any construction to which cells adhere and within which the cells are imaged.
  • Microplates may also include fluid delivery channels in the spaces between the wells. The smaller format of a microplate increases the overall efficiency of the system by minimizing the quantities of the reagents, storage and handling during preparation and the overall movement required for the scanning operation. In addition, the whole area of the microplate can be imaged more efficiently, allowing a second mode of operation for the microplate reader as described later in this document.
  • Fluorescence Reporter Molecules A major component of the new drug discovery paradigm is a continually growing family of fluorescent and luminescent reagents that are used to measure the temporal and spatial distribution, content, and activity of intracellular ions, metabolites, macromolecules, and organelles. Classes of these reagents include labeling reagents that measure the distribution and amount of molecules in living and fixed cells, environmental indicators to report signal transduction events in time and space, and fluorescent protein biosensors to measure target molecular activities within living cells. A multiparameter approach that combines several reagents in a single cell is a powerful new tool for drug discovery. The method of the present invention is based on the high affinity of fluorescent or luminescent molecules for specific cellular components.
  • the affinity for specific components is governed by physical forces such as ionic interactions, covalent bonding (which includes chimeric fusion with protein-based chromophores, fluorophores, and lumiphores), as well as hydrophobic interactions, electrical potential, and, in some cases, simple entrapment within a cellular component.
  • the luminescent probes can be small molecules, labeled macromolecules, or genetically engineered proteins, including, but not limited to green fluorescent protein chimeras.
  • fluorescent reporter molecules that can be used in the present invention, including, but not limited to, fluorescently labeled biomolecules such as proteins, phospholipids and DNA hybridizing probes.
  • fluorescent reagents specifically synthesized with particular chemical properties of binding or association have been used as fluorescent reporter molecules (Barak et al., (1997), J. Biol. Chem. 272:27497-27500; Southwick et al., (1990), Cytometry 1 1 :418-430; Tsien (1989) in Methods in Cell Biology, Vol. 29 Taylor and Wang (eds.), pp. 127-156).
  • Fluorescently labeled antibodies are particularly useful reporter molecules due to their high degree of specificity for attaching to a single molecular target in a mixture of molecules as complex as a cell or tissue.
  • the luminescent probes can be synthesized within the living cell or can be transported into the cell via several non-mechanical modes including diffusion, facilitated or active transport, signal-sequence-mediated transport, and endocytotic or pinocytotic uptake.
  • Mechanical bulk loading methods which are well known in the art, can also be used to load luminescent probes into living cells (Barber et al. (1996), Neuroscience Letters 207:17-20; Bright et al. (1996), Cytometry 24:226-233; McNeil (1989) in Methods in Cell Biology, Vol. 29, Taylor and Wang (eds.), pp. 153-173).
  • cells can be genetically engineered to express reporter molecules, such as GFP, coupled to a protein of interest as previously described (Chalfie and Prasher U.S. Patent No. 5,491,084; Cubitt et al. (1995), Trends in Biochemical Science 20:448-455).
  • the luminescent probes accumulate at their target domain as a result of specific and high affinity interactions with the target domain or other modes of molecular targeting such as signal-sequence-mediated transport.
  • Fluorescently labeled reporter molecules are useful for determining the location, amount and chemical environment of the reporter. For example, whether the reporter is in a lipophilic membrane environment or in a more aqueous environment can be determined (Giuliano et al. (1995), Ann. Rev. of Biophysics and Biomolecular Structure 24:405-434; Giuliano and Taylor (1995), Methods in Neuroscience 27:1-16). The pH environment of the reporter can be determined (Bright et al. (1989), J.
  • certain cell types within an organism may contain components that can be specifically labeled that may not occur in other cell types.
  • epithelial cells often contain polarized membrane components. That is, these cells asymmetrically distribute macromolecules along their plasma membrane.
  • Connective or supporting tissue cells often contain granules in which are trapped molecules specific to that cell type (e.g., heparin, histamine, serotonin, etc.).
  • Most muscular tissue cells contain a sarcoplasmic reticulum, a specialized organelle whose function is to regulate the concentration of calcium ions within the cell cytoplasm.
  • Many nervous tissue cells contain secretory granules and vesicles in which are trapped neurohormones or neurotransmitters. Therefore, fluorescent molecules can be designed to label not only specific components within specific cells, but also specific cells within a population of mixed cell types.
  • fluorescent reporter molecules exhibit a change in excitation or emission spectra, some exhibit resonance energy transfer where one fluorescent reporter loses fluorescence, while a second gains in fluorescence, some exhibit a loss (quenching) or appearance of fluorescence, while some report rotational movements (Giuliano et al. (1995), Ann. Rev. of Biophysics and Biomol Structure 24:405-434; Giuliano et al. (1995), Methods in Neuroscience 27:1-16). Scanning cell arrays
  • a prefened embodiment is provided to analyze cells that comprises operator-directed parameters being selected based on the assay being conducted, data acquisition by the cell screening system on the distribution of fluorescent signals within a sample, and interactive data review and analysis.
  • the operator enters information 100 that describes the sample, specifies the filter settings and fluorescent channels to match the biological labels being used and the information sought, and then adjusts the camera settings to match the sample brightness.
  • the software allows selection of various parameter settings used to identify nuclei and cytoplasm, and selection of different fluorescent reagents, identification of cells of interest based on mo ⁇ hology or brightness, and cell numbers to be analyzed per well. These parameters are stored in the system's for easy retrieval for each automated run.
  • the system's interactive cell identification mode simplifies the selection of mo ⁇ hological parameter limits such as the range of size, shape, and intensity of cells to be analyzed.
  • the user specifies which wells of the plate the system will scan and how many fields or how many cells to analyze in each well.
  • the system either automatically pre-focuses the region of the plate to be scanned using an autofocus procedure to "find focus" of the plate 102 or the user interactively pre-focuses 103 the scanning region by selecting three "tag" points which define the rectangular area to be scanned.
  • a least-squares fit "focal plane model” is then calculated from these tag points to estimate the focus of each well during an automated scan.
  • the focus of each well is estimated by inte ⁇ olating from the focal plane model during a scan.
  • the software dynamically displays the scan status, including the number of cells analyzed, the cunent well being analyzed, images of each independent wavelength as they are acquired, and the result of the screen for each well as it is determined.
  • the plate 4 ( Figure 1) is scanned in a se ⁇ entine style as the software automatically moves the motorized microscope XY stage 3 from well to well and field to field within each well of a 96-well plate.
  • Those skilled in the programming art will recognize how to adapt software for scanning of other microplate formats such as 24, 48, and 384 well plates.
  • the scan pattern of the entire plate as well as the scan pattern of fields within each well are programmed.
  • the system adjusts sample focus with an autofocus procedure 104 ( Figure 9) through the Z axis focus drive 5, controls filter selection via a motorized filter wheel 19, and acquires and analyzes images of up to four different colors ("channels" or "wavelengths").
  • the autofocus procedure is called at a user selected frequency, typically for the first field in each well and then once every 4 to 5 fields within each well.
  • the autofocus procedure calculates the starting Z-axis point by inte ⁇ olating from the pre-calculated plane focal model. Starting a programmable distance above or below this set point, the procedure moves the mechanical Z-axis through a number of different positions, acquires an image at each position, and finds the maximum of a calculated focus score that estimates the contrast of each image.
  • the Z position of the image with the maximum focus score determines the best focus for a particular field.
  • Those skilled in the art will recognize this as a variant of automatic focusing methods as described in Harms et al. in Cytometry 5 (1984), 236-243, Groen et al. in Cytometry 6 (1985), 81-91, and Firestone et al. in Cytometry 12 (1991), 195-206.
  • the camera's exposure time is separately adjusted for each dye to ensure a high-quality image from each channel.
  • Software procedures can be called, at the user's option, to conect for registration shifts between wavelengths by accounting for linear (X and Y) shifts between wavelengths before making any further measurements.
  • the electronic shutter 18 is controlled so that sample photo-bleaching is kept to a minimum. Background shading and uneven illumination can be co ⁇ ected by the software using methods known in the art (Bright et al. (1987), J. Cell Biol. 104: 1019-1033).
  • images are acquired of a primary marker 105 ( Figure 9) (typically cell nuclei counterstained with DAPI or PI fluorescent dyes) which are segmented ("identified") using an adaptive thresholding procedure.
  • the adaptive thresholding procedure 106 is used to dynamically select the threshold of an image for separating cells from the background.
  • the staining of cells with fluorescent dyes can vary to an unknown degree across cells in a microtiter plate sample as well as within images of a field of cells within each well of a microtiter plate. This variation can occur as a result of sample preparation and/or the dynamic nature of cells.
  • a global threshold is calculated for the complete image to separate the cells from background and account for field to field variation.
  • An alternative adaptive thresholding method utilizes local region thresholding in contrast to global image thresholding. Image analysis of local regions leads to better overall segmentation since staining of cell nuclei (as well as other labeled components) can vary across an image. Using this global/local procedure, a reduced resolution image (reduced in size by a factor of 2 to 4) is first globally segmented (using adaptive thresholding) to find regions of interest in the image. These regions then serve as guides to more fully analyze the same regions at full resolution.
  • a more localized threshold is then calculated (again using adaptive thresholding) for each region of interest.
  • the output of the segmentation procedure is a binary image wherein the objects are white and the background is black.
  • This binary image also called a mask in the art, is used to determine if the field contains objects 107.
  • the mask is labeled with a blob labeling method whereby each object (or blob) has a unique number assigned to it.
  • Mo ⁇ hological features, such as area and shape, of the blobs are used to differentiate blobs likely to be cells from those that are considered artifacts.
  • the user pre-sets the mo ⁇ hological selection criteria by either typing in known cell mo ⁇ hological features or by using the interactive training utility.
  • the software determines if the object meets the criteria for a valid cell nucleus 111 by measuring its mo ⁇ hological features (size and shape). For each valid cell, the XYZ stage location is recorded, a small image of the cell is stored, and features are measured 112.
  • the cell scanning method of the present invention can be used to perform many different assays on cellular samples by applying a number of analytical methods simultaneously to measure features at multiple wavelengths. An example of one such assay provides for the following measurements: 1. The total fluorescent intensity within the cell nucleus for colors 1 -4
  • the shape of the cell nucleus for color 1 is described by three shape features: a) perimeter squared area b) box area ratio c) height width ratio
  • the average fluorescent intensity of the cytoplasmic mask for colors 2-4 (i.e. #5 divided by #6) 8.
  • the ratio of the average fluorescent intensity of the cytoplasmic mask to average fluorescent intensity within the cell nucleus for colors 2-4 i.e. #7 divided by #4
  • Features 5-9 have been developed specifically to provide measurements of a cell's fluorescent molecules within the local cytoplasmic region of the cell and the translocation (i.e. movement) of fluorescent molecules from the cytoplasm to the nucleus. These features (steps 5-9) are used for analyzing cells in microplates for the inhibition of nuclear translocation. For example, inhibition of nuclear translocation of transcription factors provides a novel approach to screening intact cells (detailed examples of other types of screens will be provided below).
  • a specific method measures the amount of probe in the nuclear region (feature 4) versus the local cytoplasmic region (feature 7) of each cell. Quantification of the difference between these two sub-cellular compartments provides a measure of cytoplasm-nuclear translocation (feature 9).
  • Feature 10 describes a screen used for counting of DNA or RNA probes within the nuclear region in colors 2-4.
  • probes are commercially available for identifying chromosome-specific DNA sequences (Life Technologies, Gaithersburg, MD; Genosys, Woodlands, TX; Biotechnologies, Inc., Richmond, CA; Bio 101, Inc., Vista, CA) Cells are three-dimensional in nature and when examined at a high magnification under a microscope one probe may be in-focus while another may be completely out-of-focus.
  • the cell screening method of the present invention provides for detecting three-dimensional probes in nuclei by acquiring images from multiple focal planes.
  • the software moves the Z-axis motor drive 5 ( Figure 1) in small steps where the step distance is user selected to account for a wide range of different nuclear diameters.
  • an image is acquired.
  • the maximum gray-level intensity from each pixel in each image is found and stored in a resulting maximum projection image.
  • the maximum projection image is then used to count the probes.
  • the above method works well in counting probes that are not stacked directly above or below another one.
  • users can select an option to analyze probes in each of the focal planes acquired. In this mode, the scanning system performs the maximum plane projection method as discussed above, detects probe regions of interest in this image, then further analyzes these regions in all the focal plane images.
  • the system After measuring cell features 112 ( Figure 9), the system checks if there are any unprocessed objects in the cunent field U3. If there are any unprocessed objects, it locates the next object 110 and determines whether it meets the criteria for a valid cell nucleus 11 1, and measures its features. Once all the objects in the cunent field are processed, the system determines whether analysis of the cu ⁇ ent plate is complete 1 14; if not, it determines the need to find more cells in the cunent well 115. If the need exists, the system advances the XYZ stage to the next field within the cunent well 109 or advances the stage to the next well 1 16 of the plate.
  • images and data can be reviewed with the system's image review, data review, and summary review facilities. All images, data, and settings from a scan are archived in the system's database for later review or for interfacing with a network information management system. Data can also be exported to other third-party statistical packages to tabulate results and generate other reports. Users can review the images alone of every cell analyzed by the system with an interactive image review procedure 117. The user can review data on a cell-by-cell basis using a combination of interactive graphs, a data spreadsheet of measured features, and images of all the fluorescence channels of a cell of interest with the interactive cell-by-cell data review procedure 118.
  • Graphical plotting capabilities are provided in which data can be analyzed via interactive graphs such as histograms and scatter plots. Users can review summary data that are accumulated and summarized for all cells within each well of a plate with an interactive well-by-well data review procedure 119. Hard copies of graphs and images can be printed on a wide range of standard printers.
  • reports can be generated on one or more statistics of the measured features.
  • Users can generate a graphical report of data summarized on a well-by-well basis for the scanned region of the plate using an interactive report generation procedure 120.
  • This report includes a summary of the statistics by well in tabular and graphical format and identification information on the sample.
  • the report window allows the operator to enter comments about the scan for later retrieval.
  • Multiple reports can be generated on many statistics and be printed with the touch of one button. Reports can be previewed for placement and data before being printed.
  • the above-recited embodiment of the method operates in a single high resolution mode refened to as the high content screening (HCS) mode.
  • HCS high content screening
  • the HCS mode provides sufficient spatial resolution within a well (on the order of 1 ⁇ m) to define the distribution of material within the well, as well as within individual cells in the well.
  • the high degree of information content accessible in that mode comes at the expense of speed and complexity of the required signal processing.
  • a high throughput system is directly coupled with the HCS either on the same platform or on two separate platforms connected electronically (e.g. via a local area network).
  • This embodiment of the invention refe ⁇ ed to as a dual mode optical system, has the advantage of increasing the throughput of an HCS by coupling it with an HTS and thereby requiring slower high resolution data acquisition and analysis only on the small subset of wells that show a response in the coupled HTS.
  • High throughput 'whole plate' reader systems are well known in the art and are commonly used as a component of an HTS system used to screen large numbers of compounds (Beggs et al. (1997), supra; McCaffrey et al. (1996), supra ).
  • the HTS of the present invention is carried out on the microtiter plate or microwell anay by reading many or all wells in the plate simultaneously with sufficient resolution to make determinations on a well-by-well basis. That is, calculations are made by averaging the total signal output of many or all the cells or the bulk of the material in each well. Wells that exhibit some defined response in the HTS (the 'hits') are flagged by the system. Then on the same microtiter plate or microwell anay, each well identified as a hit is measured via HCS as described above.
  • the dual mode process involves:
  • An optional environmental chamber 304 is controlled by the system to maintain the temperature, humidity and CO levels in the air su ⁇ ounding live cells in the microtiter plate or microwell anay.
  • An optional fluid delivery device 305 (see Figure 8) is controlled by the system to dispense fluids into the wells during the scan.
  • High throughput processing 306 is first performed on the microtiter plate or microwell anay by acquiring and analyzing the signal from each of the wells in the plate.
  • the processing performed in high throughput mode 307 is illustrated in Figure 12 and described below.
  • Wells that exhibit some selected intensity response in this high throughput mode (“hits") are identified by the system.
  • the system performs a conditional operation 308 that tests for hits. If hits are found, those specific hit wells are further analyzed in high content (micro level) mode 309.
  • the processing performed in high content mode 312 is illustrated in Figure 13.
  • the system updates 310 the informatics database 31 1 with results of the measurements on the plate. If there are more plates to be analyzed 313 the system loads the next plate 303; otherwise the analysis of the plates terminates 314.
  • FIG. 12 The prefened embodiment of the system, the single platfo ⁇ n dual mode screening system, will be described. Those skilled in the art will recognize that operationally the dual platform system simply involves moving the plate between two optical systems rather than moving the optics.
  • the system begins the HTS acquisition and analysis 40L
  • the HTS optical module is selected by controlling a motorized optical positioning device 402 on the dual mode system.
  • data from a primary marker on the plate is acquired 403 and wells are isolated from the plate background using a masking procedure 404. Images are also acquired in other fluorescence channels being used 405. The region in each image conesponding to each well 406 is measured 407.
  • a feature calculated from the measurements for a particular well is compared with a predefined threshold or intensity response 408, and based on the result the well is either flagged as a "hit" 409 or not.
  • the locations of the wells flagged as hits are recorded for subsequent high content mode processing. If there are wells remaining to be processed 410 the program loops back 406 until all the wells have been processed 411 and the system exits high throughput mode.
  • the system starts the high content mode processing 501 defined in Figure 13.
  • the system selects the HCS optical module 502 by controlling the motorized positioning system.
  • the XY stage location of the well is retrieved from memory or disk and the stage is then moved to the selected stage location 503.
  • the autofocus procedure 504 is called for the first field in each hit well and then once every 5 to 8 fields within each well.
  • images are acquired of the primary marker 505 (typically cell nuclei counterstained with DAPI, Hoechst or PI fluorescent dye). The images are then segmented (separated into regions of nuclei and non-nuclei) using an adaptive thresholding procedure 506.
  • the primary marker 505 typically cell nuclei counterstained with DAPI, Hoechst or PI fluorescent dye
  • the output of the segmentation procedure is a binary mask wherein the objects are white and the background is black.
  • This binary image also called a mask in the art, is used to determine if the field contains objects 507.
  • the mask is labeled with a blob labeling method whereby each object (or blob) has a unique number assigned to it. If objects are found in the field, images are acquired for all other active channels 508. otherwise the stage is advanced to the next field 514 in the cunent well. Each object is located in the image for further analysis 509.
  • Mo ⁇ hological features such as area and shape of the objects, are used to select objects likely to be cell nuclei 510, and discard (do no further processing on) those that are considered artifacts.
  • the XYZ stage location is recorded, a small image of the cell is stored, and assay specific features are measured 511.
  • the system then performs multiple tests on the cells by applying several analytical methods to measure features at each of several wavelengths.
  • the systems checks if there are any unprocessed objects in the cunent field 512. If there are any unprocessed objects, it locates the next object 509 and determines whether it meets the criteria for a valid cell nucleus 510, and measures its features. After processing all the objects in the cunent field, the system deteremines whether it needs to find more cells or fields in the cunent well 513.
  • the system checks whether it has any remaining hit wells to measure 515. If so, it advances to the next hit well 503 and proceeds through another cycle of acquisition and analysis, otherwise the HCS mode is finished 516.
  • a method of kinetic live cell screening is provided.
  • the previously described embodiments of the invention are used to characterize the spatial distribution of cellular components at a specific point in time, the time of chemical fixation.
  • these embodiments have limited utility for implementing kinetic based screens, due to the sequential nature of the image acquisition, and the amount of time required to read all the wells on a plate. For example, since a plate can require 30 - 60 minutes to read through all the wells, only very slow kinetic processes can be measured by simply preparing a plate of live cells and then reading through all the wells more than once. Faster kinetic processes can be measured by taking multiple readings of each well before proceeding to the next well, but the elapsed time between the first and last well would be too long, and fast kinetic processes would likely be complete before reaching the last well.
  • the kinetic live cell extension of the invention enables the design and use of screens in which a biological process is characterized by its kinetics instead of, or in addition to, its spatial characteristics.
  • a response in live cells can be measured by adding a reagent to a specific well and making multiple measurements on that well with the appropriate timing.
  • This dynamic live cell embodiment of the invention therefore includes apparatus for fluid delivery to individual wells of the system in order to deliver reagents to each well at a specific time in advance of reading the well. This embodiment thereby allows kinetic measurements to be made with temporal resolution of seconds to minutes on each well of the plate.
  • the acquisition control program is modified to allow repetitive data collection from sub-regions of the plate, allowing the system to read other wells between the time points required for an individual well.
  • Figure 8 describes an example of a fluid delivery device for use with the live cell embodiment of the invention and is described above.
  • This set-up allows one set of pipette tips 705, or even a single pipette tip, to deliver reagent to all the wells on the plate.
  • the bank of syringe pumps 701 can be used to deliver fluid to 12 wells simultaneously, or to fewer wells by removing some of the tips 705.
  • the temporal resolution of the system can therefore be adjusted, without sacrificing data collection efficiency, by changing the number of tips and the scan pattern as follows.
  • the data collection and analysis from a single well takes about 5 seconds. Moving from well to well and focusing in a well requires about 5 seconds, so the overall cycle time for a well is about 10 seconds.
  • the overall time to collect a kinetic data set from the plate is then simply the time to perform a single scan of the plate, multiplied by the number of time points required. Typically, 1 time point before addition of compounds and 2 or 3 time points following addition should be sufficient for screening pu ⁇ oses.
  • Figure 14 shows the acquisition sequence used for kinetic analysis.
  • the start of processing 801 is configuration of the system, much of which is identical to the standard HCS configuration.
  • the operator must enter information specific to the kinetic analysis being performed 802, such as the sub-region size, the number of time points required, and the required time increment.
  • a sub-region is a group of wells that will be scanned repetitively in order to accumulate kinetic data.
  • the size of the sub-region is adjusted so that the system can scan a whole sub-region once during a single time increment, thus minimizing wait times.
  • the optimum sub-region size is calculated from the setup parameters, and adjusted if necessary by the operator.
  • the acquisition sequence performed in each well is exactly the same as that required for the specific HCS being run in kinetic mode.
  • Figure 15 details a flow chart for that processing. All of the steps between the start 901 and the return 902 are identical to those described as steps 504 - 5_14 in Figure 13.
  • the system After processing each well in a sub-region, the system checks to see if all the wells in the sub-region have been processed 806 ( Figure 14), and cycles through all the wells until the whole region has been processed. The system then moves the plate into position for fluid addition, and controls fluidic system delivery of fluids to the entire sub-region 807. This may require multiple additions for sub-regions which span several rows on the plate, with the system moving the plate on the X,Y stage between additions. Once the fluids have been added, the system moves to the first well in the sub-region 808 to begin acquisition of time points. The data is acquired from each well 809 and as before the system cycles through all the wells in the sub-region 810.
  • the system After each pass through the sub-region, the system checks whether all the time points have been collected 811 and if not, pauses 813 if necessary 812 to stay synchronized with the requested time increment. Otherwise, the system checks for additional sub-regions on the plate 814 and either moves to the next sub-region 803 or finishes 815.
  • the kinetic analysis mode comprises operator identification of sub-regions of the microtiter plate or microwells to be screened, based on the kinetic response to be investigated, with data acquisitions within a sub-region prior to data acquisition in subsequent sub- regions.
  • the cell screening system comprises a high magnification fluorescence optical system with a stage adapted for holding cells and a means for moving the stage, a digital camera, a light source for receiving and processing the digital data from the digital camera, and a computer means for receiving and processing the digital data from the digital camera.
  • This aspect of the invention comprises programs that instruct the cell screening system to define the distribution and activity of specific cellular constituents and processes, using the luminescent probes, the optical imaging system, and the pattern recognition software of the invention.
  • Prefe ⁇ ed embodiments of the machine readable storage medium comprise programs consisting of a set of instructions for causing a cell screening system to execute the procedures set forth in Figures 9, 11, 12, 13, 14 or 15.
  • Another prefe ⁇ ed embodiment comprises a program consisting of a set of instructions for causing a cell screening system to execute procedures for detecting the distribution and activity of specific cellular constituents and processes.
  • the cellular processes include, but are not limited to, nuclear translocation of a protein, cellular mo ⁇ hology, apoptosis, receptor internalization, and protease-induced translocation of a protein.
  • the cell screening methods are used to identify compounds that modify the various cellular processes.
  • the cells can be contacted with a test compound, and the effect of the test compound on a particular cellular process can be analyzed.
  • the cells can be contacted with a test compound and a known agent that modifies the particular cellular process, to determine whether the test compound can inhibit or enhance the effect of the known agent.
  • the methods can be used to identify test compounds that increase or decrease a particular cellular response, as well as to identify test compounds that affects the ability of other agents to increase or decrease a particular cellular response.
  • the locations containing cells are analyzed using the above methods at low resolution in a high throughput mode, and only a subset of the locations containing cells are analyzed in a high content mode to obtain luminescent signals from the luminescently labeled reporter molecules in subcellular compartments of the cells being analyzed.
  • Regulation of transcription of some genes involves activation of a transcription factor in the cytoplasm, resulting in that factor being transported into the nucleus where it can initiate transcription of a particular gene or genes.
  • This change in transcription factor distribution is the basis of a screen for the cell-based screening system to detect compounds that inhibit or induce transcription of a particular gene or group of genes. A general description of the screen is given followed by a specific example.
  • the distribution of the transcription factor is determined by labeling the nuclei with a DNA specific fluorophore like Hoechst 33423 and the transcription factor with a specific fluorescent antibody. After autofocusing on the Hoechst labeled nuclei, an image of the nuclei is acquired in the cell-based screening system and used to create a mask by one of several optional thresholding methods, as described supra. The mo ⁇ hological descriptors of the regions defined by the mask are compared with the user defined parameters and valid nuclear masks are identified and used with the following method to extract transcription factor distributions. Each valid nuclear mask is eroded to define a slightly smaller nuclear region.
  • the original nuclear mask is then dilated in two steps to define a ring shaped region around the nucleus, which represents a cytoplasmic region.
  • the average antibody fluorescence in each of these two regions is determined, and the difference between these averages is defined as the NucCyt
  • Figure 10A illustrates an unstimulated cell with its nucleus 200 labeled with a blue fluorophore and a transcription factor in the cytoplasm
  • Figure 10B illustrates the nuclear mask 202 derived by the cell-based screening system.
  • Figure 10C illustrates the cytoplasm 203 of the unstimulated cell imaged at a green wavelength.
  • Figure 10D illustrates the nuclear mask 202 is eroded (reduced) once to define a nuclear sampling region 204 with minimal cytoplasmic distribution.
  • the nucleus boundary 202 is dilated (expanded) several times to form a ring that is 2-3 pixels wide that is used to define the cytoplasmic sampling region 205 for the same cell.
  • Figure 10E further illustrates a side view which shows the nuclear sampling region 204 and the cytoplasmic sampling region 205.
  • Figure 10F-J illustrates the strategy for determining nuclear translocation in a stimulated cell.
  • Figure 10F illustrates a stimulated cell with its nucleus 206 labeled with a blue fluorophore and a transcription factor in the cytoplasm 207 labeled with a green fluorophore.
  • the nuclear mask 208 in Figure 10G is derived by the cell based screening system.
  • Figure 10H illustrates the cytoplasm 209 of a stimulated cell imaged at a green wavelength.
  • Figure 101 illustrates the nuclear sampling region 211 and cytoplasmic sampling region 212 of the stimulated cell.
  • Figure 10 J further illustrates a side view which shows the nuclear sampling region 21 1 and the cytoplasmic sampling region 212.
  • a specific application of this method has been used to validate this method as a screen.
  • a human cell line was plated in 96 well microtiter plates. Some rows of wells were titrated with IL-1, a known inducer of the NF-KB transcription factor. The cells were then fixed and stained by standard methods with a fluorescein labeled antibody to the transcription factor, and Hoechst 33423. The cell-based screening system was used to acquire and analyze images from this plate and the NucCyt Difference was found to be strongly conelated with the amount of agonist added to the wells as illustrated in
  • Figure 18 is a representative display on a PC screen of data which was obtained in accordance with Example 1.
  • Graph 1 180 plots the difference between the average antibody fluorescence in the nuclear sampling region and cytoplasmic sampling region, NucCyt Difference verses Well #.
  • Graph 2 181 plots the average fluorescence of the antibody in the nuclear sampling region, NP 1 average, versus the Well #.
  • Graph 3 182 plots the average antibody fluorescence in the cytoplasmic sampling region, LIP1 average, versus Well #.
  • the software permits displaying data from each cell.
  • Figure 18 shows a screen display 183. the nuclear image 184. and the fluorescent antibody image 185 for cell #26.
  • NucCyt Difference refened to in graph 1 180 of Figure 18 is the difference between the average cytoplasmic probe (fluorescent reporter molecule) intensity and the average nuclear probe (fluorescent reporter molecule) intensity.
  • NP1 average refened to in graph 2 181 of Figure 18 is the average of cytoplasmic probe (fluorescent reporter molecule) intensity within the nuclear sampling region.
  • L1P1 average refened to in graph 3 182 of Figure 18 is the average probe (fluorescent reporter molecule) intensity within the cytoplasmic sampling region. It will be understood by one of skill in the art that this aspect of the invention can be performed using other transcription factors that translocate from the cytoplasm to the nucleus upon activation.
  • activation of the c-fos transcription factor was assessed by defining its spatial position within cells. Activated c-fos is found only within the nucleus, while inactivated c-fos resides within the cytoplasm.
  • 3T3 cells were plated at 5000-10000 cells per well in a Polyfiltronics 96-well plate. The cells were allowed to attach and grow overnight. The cells were rinsed twice with 100 ⁇ l serum-free medium, incubated for 24-30 hours in serum-free MEM culture medium, and then stimulated with platelet derived growth factor (PDGF-BB) (Sigma Chemical Co., St. Louis, MO) diluted directly into serum free medium at concentrations ranging from 1-50 ng/ml for an average time of 20 minutes.
  • PDGF-BB platelet derived growth factor
  • HBSS IX Hanks buffered saline solution
  • HBSS HBSS
  • 50 ⁇ l of the dilution was applied to each well.
  • Cells were incubated in the presence of primary antibody for one hour at room temperature, and then incubated for one hour at room temperature in a light tight container with goat anti-rabbit secondary antibody conjugated to ALEXATM 488 (Molecular Probes), diluted 1 :500 from a 100 ⁇ g/ml stock in HBSS.
  • Hoechst DNA dye (Molecular Probes) was then added at a 1 :1000 dilution of the manufacturer's stock solution (10 mg/ml).
  • the cells were then washed with HBSS, and the plate was sealed prior to analysis with the cell screening system of the invention.
  • the data from these experiments demonstrated that the methods of the invention could be used to measure transcriptional activation of c-fos by defining its spatial position within cells.
  • transcription factors include, but are not limited to fos and jun homologs, NF-KB (nuclear factor kappa from B cells), NFAT (nuclear factor of activated T-lymphocytes), and STATs (signal transducer and activator of transcription) factors (For example, see Strehlow, I., and Schindler, C. 1998. J. Biol. Chem. 273:28049-28056; Chow, et al. 1997 Science.
  • indicator cells are treated with test compounds and the distribution of luminescently labeled transcription factor is measured in space and time using a cell screening system, such as the one disclosed above.
  • the luminescently labeled transcription factor may be expressed by or added to the cells either before, together with, or after contacting the cells with a test compound.
  • the transcription factor may be expressed as a luminescently labeled protein chimera by transfected indicator cells.
  • the luminescently labeled transcription factor may be expressed, isolated, and bulk-loaded into the indicator cells as described above, or the transcription factor may be luminescently labeled after isolation.
  • the transcription factor is expressed by the indicator cell, which is subsequently contacted with a luminescent label, such as an antibody, that detects the transcription factor.
  • kits for analyzing transcription factor activation, comprising an antibody that specifically recognizes a transcription factor of interest, and instructions for using the antibody for carrying out the methods described above.
  • the transcription factor-specific antibody, or a secondary antibody that detects the transcription factor antibody is luminescently labeled.
  • the kit contains cells that express the transcription factor of interest, and/or the kit contains a compound that is known to modify activation of the transcription factor of interest, including but not limited to platelet derived growth factor (PDGF) and serum, which both modify fos activation; and interleukin l(IL-l) and tumor necrosis factor (TNF), which both modify NF-KB activation.
  • PDGF platelet derived growth factor
  • TNF tumor necrosis factor
  • the kit comprises a recombinant expression vector comprising a nucleic acid encoding a transcription factor of interest that translocates from the cytoplasm to the nucleus upon activation, and instructions for using the expression vector to identify compounds that modify transcription factor activation in a cell of interest.
  • the kits contain a purified, luminescently labeled transcription factor.
  • the transcription factor is expressed as a fusion protein with a luminescent protein, including but not limited to green fluorescent protein, luceriferase, or mutants or fragments thereof.
  • the kit further contains cells that are transfected with the expression vector, an antibody or fragment that specifically bind to the transcription factor of interest, and/or a compound that is known to modify activation of the transcription factor of interest (as above).
  • the cytoplasm to nucleus screening methods can also be used to analyze the activation of any protein kinase that is present in an inactive state in the cytoplasm and is transported to the nucleus upon activation, or that phosphorylates a substrate that translocates from the cytoplasm to the nucleus upon phosphorylation.
  • protein kinases include, but are not limited to extracellular signal-regulated protein kinases (ERKs), c-Jun amino-terminal kinases (JNKs), Fos regulating protein kinases (FRKs), p38 mitogen activated protein kinase (p38MAPK), protein kinase A (PKA), and mitogen activated protein kinase kinases (MAPKKs).
  • ERKs extracellular signal-regulated protein kinases
  • JNKs c-Jun amino-terminal kinases
  • FRKs Fos regulating protein kinases
  • p38MAPK p38 mitogen activated protein kinase
  • PKA protein kinase A
  • MAPKKs mitogen activated protein kinase kinases
  • protein kinase activity is assayed by monitoring translocation of a luminescently labeled protein kinase substrate from the cytoplasm to the nucleus after being phosphorylated by the protein kinase of interest.
  • the substrate is non-phosphorylated and cytoplasmic prior to phosphorylation, and is translocated to the nucleus upon phosphorylation by the protein kinase.
  • the protein kinase itself translocates from the cytoplasm to the nucleus in this embodiment.
  • substrates and the conesponding protein kinase
  • indicator cells are treated with test compounds and the distribution of luminescently labeled protein kinase or protein kinase substrate is measured in space and time using a cell screening system, such as the one disclosed above.
  • the luminescently labeled protein kinase or protein kinase substrate may be expressed by or added to the cells either before, together with, or after contacting the cells with a test compound.
  • the protein kinase or protein kinase substrate may be expressed as a luminescently labeled protein chimera by transfected indicator cells.
  • the luminescently labeled protein kinase or protein kinase substrate may be expressed, isolated, and bulk-loaded into the indicator cells as described above, or the protein kinase or protein kinase substrate may be luminescently labeled after isolation.
  • the protein kinase or protein kinase substrate is expressed by the indicator cell, which is subsequently contacted with a luminescent label, such as a labeled antibody, that detects the protein kinase or protein kinase substrate.
  • protein kinase activity is assayed by monitoring the phosphorylation state (ie: phosphorylated or not phosphorylated) of a protein kinase substrate.
  • phosphorylation state ie: phosphorylated or not phosphorylated
  • phosphorylation state is monitored by contacting the cells with an antibody that binds only to the phosphorylated form of the protein kinase substrate of interest (For example, as disclosed in U.S. Patent No. 5,599,681).
  • a biosensor of phosphorylation is used.
  • a luminescently labeled protein or fragment thereof can be fused to a protein that has been engineered to contain (a) a phosphorylation site that is recognized by a protein kinase of interest; and (b) a nuclear localization signal that is unmasked by the phosphorylation.
  • a biosensor will thus be translocated to the nucleus upon phosphorylation, and its translocation can be used as a measure of protein kinase activation.
  • kits for analyzing protein kinase activation, comprising a primary antibody that specifically binds to a protein kinase, a protein kinase substrate, or a phosphorylated form of the protein kinase substrate of interest and instructions for using the primary antibody to identify compounds that modify protein kinase activation in a cell of interest.
  • the primary antibody, or a secondary antibody that detects the primary antibody is luminescently labeled.
  • the kit further comprises cells that express the protein kinase of interest, and/or a compound that is known to modify activation of the protein kinase of interest, including but not limited to dibutyryl cAMP (modifies PKA), forskolin (PKA), and anisomycin (p38MAPK).
  • a compound that is known to modify activation of the protein kinase of interest including but not limited to dibutyryl cAMP (modifies PKA), forskolin (PKA), and anisomycin (p38MAPK).
  • kits comprise an expression vector encoding a protein kinase or a protein kinase substrate of interest that translocates from the cytoplasm to the nucleus upon activation and instructions for using the expression vector to identify compounds that modify protein kinase activation in a cell of interest.
  • the kits contain a purified, luminescently labeled protein kinase or protein kinase substrate.
  • the protein kinase or protein kinase substrate of interest is expressed as a fusion protein with a luminescent protein.
  • the kit further comprises cells that are transfected with the expression vector, an antibody or fragment thereof that specifically binds to the protein kinase or protein kinase substrate of interest, and/or a compound that is known to modify activation of the protein kinase of interest, (as above)
  • the present invention comprises a machine readable storage medium comprising a program containing a set of instructions for causing a cell screening system to execute the methods disclosed for analyzing transcription factor or protein kinase activation, wherein the cell screening system comprises an optical system with a stage adapted for holding a plate containing cells, a digital camera, a means for directing fluorescence or luminescence emitted from the cells to the digital camera, and a computer means for receiving and processing the digital data from the digital camera.
  • Changes in cell size are associated with a number of cellular conditions, such as hypertrophy, cell attachment and spreading, differentiation, growth and division, necrotic and programmed cell death, cell motility, mo ⁇ hogenesis, tube formation, and colony formation.
  • cellular hypertrophy has been associated with a cascade of alterations in gene expression and can be characterized in cell culture by an alteration in cell size, that is clearly visible in adherent cells growing on a coverslip.
  • Cell size can also be measured to determine the attachment and spreading of adherent cells.
  • Cell spreading is the result of selective binding of cell surface receptors to substrate ligands and subsequent activation of signaling pathways to the cytoskeleton.
  • Cell attachment and spreading to substrate molecules is an important step for the metastasis of cancer cells, leukocyte activation during the inflammatory response, keratinocyte movement during wound healing, and endothelial cell movement during angiogenesis.
  • Compounds that affect these surface receptors, signaling pathways, or the cytoskeleton will affect cell spreading and can be screened by measuring cell size.
  • Total cellular area can be monitored by labeling the entire cell body or the cell cytoplasm using cytoskeletal markers, cytosolic volume markers, or cell surface markers, in conjunction with a DNA label.
  • cytoskeletal markers include cytoskeletal markers, cytosolic volume markers, or cell surface markers, in conjunction with a DNA label.
  • cytosolic volume markers include the following:
  • CMFDA Chloromethylfluorescein diacetate
  • Dihexadecyl tetramethylindocarbocyanine perchlorate (DilC 16) lipid dyes Triethylammonium propyl dibutylamino styryl pyridinium (FM 4-64, FM 1-43) lipid dyes
  • Lectins to oligosaccarides such as fluorescein concanavalin A or wheat germ agglutinin
  • Antibodies to various surface proteins such as epidermal growth factor
  • Cells are stained live or after fixation and the cell area can be measured.
  • live cells stained with DiIC16 have homogeneously labeled plasma membranes, and the projected cross-sectional area of the cell is uniformly discriminated from background by fluorescence intensity of the dye.
  • Live cells stained with cytosolic stains such as CMFDA produce a fluorescence intensity that is proportional to cell thickness.
  • cell labeling is dimmer in thin regions of the cell, total cell area can be discriminated from background.
  • Fixed cells can be stained with cytoskeletal markers such as ALEXATM 488 phalloidin that label polymerized actin. Phalloidin does not homogeneously stain the cytoplasm, but still permits discrimination of the total cell area from background.
  • Cellular hypertrophy A screen to analyze cellular hypertrophy is implemented using the following strategy.
  • Primary rat myocytes can be cultured in 96 well plates, treated with various compounds and then fixed and labeled with a fluorescent marker for the cell membrane or cytoplasm, or cytoskeleton, such as an antibody to a cell surface marker or a fluorescent marker for the cytoskeleton like rhodamine-phalloidin, in combination with a DNA label like Hoechst. After focusing on the Hoechst labeled nuclei, two images are acquired, one of the Hoechst labeled nuclei and one of the fluorescent cytoplasm image.
  • the nuclei are identified by thresholding to create a mask and then comparing the mo ⁇ hological descriptors of the mask with a set of user defined descriptor values.
  • Each non-nucleus image (or "cytoplasmic image") is then processed separately.
  • the original cytoplasm image can be thresholded, creating a cytoplasmic mask image.
  • Local regions containing cells are defined around the nuclei. The limits of the cells in those regions are then defined by a local dynamic threshold operation on the same region in the fluorescent antibody image.
  • a sequence of erosions and dilations is used to separate slightly touching cells and a second set of mo ⁇ hological descriptors is used to identify single cells.
  • the area of the individual cells is tabulated in order to define the distribution of cell sizes for comparison with size data from normal and hypertrophic cells.
  • the aggregate whole nucleus area is the number of nonzero pixels in the nuclear mask.
  • the average whole nucleus area is the aggregate whole nucleus area divided by the total number of nuclei.
  • the aggregate cytoplasm intensity is the sum of the intensities of all pixels in the cytoplasmic mask.
  • the cytoplasmic area per nucleus is the total cytoplasmic area divided by the total nucleus count.
  • the cytoplasmic intensity per nucleus is the aggregate cytoplasm intensity divided by the total nucleus count.
  • the average cytoplasm intensity is the aggregate cytoplasm intensity divided by the cytoplasm area.
  • the cytoplasm nucleus ratio is the total cytoplasm area divided by the total nucleus area.
  • one or more fluorescent antibodies to other cellular proteins can be included. Images of these additional labeled proteins can be acquired and stored with the above images, for later review, to identify anomalies in the distribution and mo ⁇ hology of these proteins in hypertrophic cells. This example of a multi-parametric screen allows for simultaneous analysis of cellular hypertrophy and changes in actin or myosin distribution.
  • Cell spreading is a measure of the response of cell surface receptors to substrate attachment ligands. Spreading is proportional to the ligand concentration or to the concentration of compounds that reduce receptor-ligand function.
  • One example of selective cell-substrate attachment is prostate carcinoma cell adhesion to the extracellular matrix protein collagen. Prostate carcinoma cells metastasize to bone via selective adhesion to collagen.
  • PC3 human prostate carcinoma cells were cultured in media with appropriate stimulants and are passaged to collagen coated 96 well plates.
  • Ligand concentration can be varied or inhibitors of cell spreading can be added to the wells.
  • Examples of compounds that can affect spreading are receptor antagonists such as integrin- or proteoglycan-blocking antibodies, signaling inhibitors including phosphatidyl inositol-3 kinase inhibitors, and cytoskeletal inhibitors such as cytochalasin D.
  • receptor antagonists such as integrin- or proteoglycan-blocking antibodies
  • signaling inhibitors including phosphatidyl inositol-3 kinase inhibitors
  • cytoskeletal inhibitors such as cytochalasin D.
  • the size of cells under these various conditions can be distinguished above background levels.
  • the number of cells per field is determined by measuring the number of nuclei stained with the Hoechst DNA dye.
  • the area per cell is found by dividing the cytoplasmic area (phalloidin image) by the cell number (Hoechst image).
  • the size of cells is proportional to the ligand-receptor function. Since the area is determined by ligand concentration and by the resultant function of the cell, drug efficacy, as well as drug potency, can be determined by this cell-based assay. Other measurements can be made as discussed above for cellular hypertrophy.
  • the methods for analyzing cellular mo ⁇ hology can be used in a combined high throughput-high content screen.
  • the high throughput mode scans the whole well for an increase in fluorescent phalloidin intensity.
  • a threshold is set above which both nuclei (Hoechst) and cells (phalloidin) are measured in a high content mode.
  • an environmental biosensor (examples include, but are not limited to, those biosensors that are sensitive to calcium and pH changes) is added to the cells, and the cells are contacted with a compound. The cells are scanned in a high throughput mode, and those wells that exceed a pre-determined threshold for luminescence of the biosensor are scanned in a high content mode.
  • kits for analyzing cellular mo ⁇ hology, comprising a luminescent compound that can be used to specifically label the cell cytoplasm, membrane, or cytoskeleton (such as those described above), and instructions for using the luminescent compound to identify test stimuli that induce or inhibit changes in cellular mo ⁇ hology according to the above methods.
  • the kit further comprises a luminescent marker for cell nuclei.
  • the kit comprises at least one compound that is known to modify cellular mo ⁇ hology, including, but not limited to integrin- or proteoglycan- blocking antibodies, signaling inhibitors including phosphatidyl inositol-3 kinase inhibitors, and cytoskeletal inhibitors such as cytochalasin D.
  • the present invention comprises a machine readable storage medium comprising a program containing a set of instructions for causing a cell screening system to execute the disclosed methods for analyzing cellular mo ⁇ hology
  • the cell screening system comprises an optical system with a stage adapted for holding a plate containing cells, a digital camera, a means for directing fluorescence or luminescence emitted from the cells to the digital camera, and a computer means for receiving and processing the digital data from the digital camera.
  • the following example is a screen for activation of a G-protein coupled receptor
  • GPCR GPCR as detected by the translocation of the GPCR from the plasma membrane to a proximal nuclear location.
  • This example illustrates how a high throughput screen can be coupled with a high-content screen in the dual mode System for Cell Based Screening.
  • G-protein coupled receptors are a large class of 7 trans-membrane domain cell surface receptors. Ligands for these receptors stimulate a cascade of secondary signals in the cell, which may include, but are not limited to, Ca ++ transients, cyclic AMP production, inositol triphosphate (IP ) production and phosphorylation. Each of these signals are rapid, occuring in a matter of seconds to minutes, but are also generic. For example, many different GPCRs produce a secondary Ca ++ signal when activated. Stimulation of a GPCR also results in the transport of that GPCR from the cell surface membrane to an internal, proximal nuclear compartment. This intemalization is a much more receptor-specific indicator of activation of a particular receptor than are the secondary signals described above.
  • Figure 19 illustrates a dual mode screen for activation of a GPCR.
  • Cells carrying a stable chimera of the GPCR with a blue fluorescent protein (BFP) would be loaded with the acetoxymethyl ester form of Fluo-3, a cell permeable calcium indicator (green fluorescence) that is trapped in living cells by the hydrolysis of the esters. They would then be deposited into the wells of a microtiter plate 601. The wells would then be treated with an anay of test compounds using a fluid delivery system, and a short sequence of Fluo-3 images of the whole microtiter plate would be acquired and analyzed for wells exhibiting a calcium response (i.e., high throughput mode).
  • BFP blue fluorescent protein
  • the images would appear like the illustration of the microtiter plate 601 in Figure 19.
  • a small number of wells such as wells C4 and E9 in the illustration, would fluoresce more brightly due to the Ca ++ released upon stimulation of the receptors.
  • the locations of wells containing compounds that induced a response 602, would then be transfened to the HCS program and the optics switched for detailed cell by cell analysis of the blue fluorescence for evidence of GPCR translocation to the perinuclear region.
  • the bottom of Figure 19 illustrates the two possible outcomes of the analysis of the high resolution cell data.
  • the camera images a sub-region 604 of the well area 603, producing images of the fluorescent cells 605.
  • the uniform distribution of the fluorescence in the cells indicates that the receptor has not internalized, implying that the Ca ++ response seen was the result of the stimulation of some other signalling system in the cell.
  • the cells in well E9 606 clearly indicate a concentration of the receptor in the perinuclear region clearly indicating the full activation of the receptor. Because only a few hit wells have to be analyzed with high resolution, the overall throughput of the dual mode system can be quite high, comparable to the high throughput system alone.
  • Example 4 Kinetic High Content Screen The following is an example of a screen to measure the kinetics of intemalization of a receptor.
  • the stimulation of a GPCR results in the intemalization of the receptor, with a time course of about 15 min.
  • Simply detecting the endpoint as internalized or not, may not be sufficient for defining the potency of a compound as a GPCR agonist or antagonist.
  • 3 time points at 5 min intervals would provide information not only about potency during the time course of measurement, but would also allow extrapolation of the data to much longer time periods.
  • the sub-region would be defined as two rows, the sampling interval as 5 minutes and the total number of time points 3.
  • HCS human glucocorticoid receptor
  • hGR human glucocorticoid receptor
  • the ligand-receptor complex translocates to the nucleus where transcriptional activation occurs (Htun et al., Proc. Natl. Acad. Sci. 93:4845, 1996).
  • hormone receptors are excellent drug targets because their activity lies at the apex of key intracellular signaling pathways. Therefore, a high-content screen of hGR translocation has distinct advantage over in vitro ligand-receptor binding assays.
  • the availability of up to two more channels of fluorescence in the cell screening system of the present invention permits the screen to contain two additional parameters in parallel, such as other receptors, other distinct targets or other cellular processes.
  • Plasmid construct A eukaryotic expression plasmid containing a coding sequence for a green fluorescent protein - human glucocorticoid receptor (GFP-hGR) chimera was prepared using GFP mutants (Palm et al., Nat. Struct. Biol. 4:361 (1997). The construct was used to transfect a human cervical carcinoma cell line (HeLa).
  • GFP-hGR green fluorescent protein - human glucocorticoid receptor
  • HeLa cells ATCC CCL-2
  • DMEM fetal bovine serum
  • C-DMEM penicillin-streptomycin
  • the medium was replaced, prior to transfection, with DMEM containing 5% charcoal/dextran-treated FBS.
  • Cells were incubated with the calcium phosphate-D ⁇ A precipitate for 4-5 hours at 37°C and 5% CO 2 , washed 3-4 times with DMEM to remove the precipitate, followed by the addition of C-DMEM.
  • Lipofectamine transfections were performed in serum-free DMEM without antibiotics according to the manufacturer's instructions (Life Technologies, Gaithersburg, MD). Following a 2-3 hour incubation with the D ⁇ A-liposome complexes, the medium was removed and replaced with C-DMEM. All transfected cells in 96-well microtiter plates were incubated at 33°C and 5% CO 2 for 24-48 hours prior to drug treatment. Experiments were performed with the receptor expressed transiently in HeLa cells.
  • Dexamethasone induction of GFP-hGR translocation To obtain receptor- ligand translocation kinetic data, nuclei of transfected cells were first labeled with 5 ⁇ g/ml Hoechst 33342 (Molecular Probes) in C-DMEM for 20 minutes at 33°C and 5% CO 2 . Cells were washed once in Hank's Balanced Salt Solution (HBSS) followed by the addition of 100 nM dexamethasone in HBSS with 1% charcoal/dextran-treated FBS. To obtain fixed time point dexamethasone titration data, transfected HeLa cells
  • Kinetic data were collected by acquiring fluorescence image pairs (GFP-hGR and Hoechst 33342-labeled nuclei) from fields of living cells at 1 min intervals for 30 min after the addition of dexamethasone. Likewise, image pairs were obtained from each well of the fixed time point screening plates 1 h after the addition of dexamethasone. In both cases, the image pairs obtained at each time point were used to define nuclear and cytoplasmic regions in each cell. Translocation of GFP-hGR was calculated by dividing the integrated fluorescence intensity of GFP-hGR in the nucleus by the integrated fluorescence intensity of the chimera in the cytoplasm or as a nuclear-cytoplasmic difference of GFP fluorescence.
  • this translocation ratio was calculated from data obtained from at least 200 cells at each concentration of dexamethasone tested. Drug-induced translocation of GFP-hGR from the cytoplasm to the nucleus was therefore conelated with an increase in the translocation ratio.
  • Figure 20 schematically displays the drag-induced cytoplasm 253 to nucleus 252 translocation of the human glucocorticoid receptor.
  • the upper pair of schematic diagrams depicts the localization of GFP-hGR within the cell before 250 (A) and after 251 (B) stimulation with dexamethasone.
  • the drug induces a large portion of the cytoplasmic GFP-hGR to translocate into the nucleus. This redistribution is quantified by determining the integrated intensities ratio of the cytoplasmic and nuclear fluorescence in treated 255 and untreated 254 cells.
  • the lower pair of fluorescence micrographs show the dynamic redistribution of GFP-hGR in a single cell, before 254 and after 255 treatment.
  • the HCS is performed on wells containing hundreds to thousands of transfected cells and the translocation is quantified for each cell in the field exhibiting GFP fluorescence. Although the use of a stably transfected cell line would yield the most consistently labeled cells, the heterogeneous levels of GFP-hGR expression induced by transient transfection did not interfere with analysis by the cell screening system of the present invention.
  • the cell screening system scans each well of the plate, images a population of cells in each, and analyzes cells individually.
  • two channels of fluorescence are used to define the cytoplasmic and nuclear distribution of the GFP-hGR within each cell.
  • Depicted in Figure 21 is the graphical user interface of the cell screening system near the end of a GFP-hGR screen.
  • the user interface depicts the parallel data collection and analysis capability of the system.
  • the windows labeled "Nucleus” 261 and "GFP-hGR" 262 show the pair of fluorescence images being obtained and analyzed in a single field.
  • the window labeled "Color Overlay" 260 is formed by pseudocoloring the above images and merging them so the user can immediately identify cellular changes.
  • 96 well plate depicted in the lower window of the screen 267 shows which wells have met a set of user-defined screening criteria. For example, a white-colored well 269 indicates that the drag-induced translocation has exceeded a predetermined threshold value of 50%. On the other hand, a black-colored well 270 indicates that the drag being tested induced less than 10% translocation. Gray-colored wells 268 indicate "hits" where the translocation value fell between 10% and 50%.
  • Row “E” on the 96 well plate being analyzed 266 shows a titration with a drag known to activate GFP-hGR translocation, dexamethasone. This example screen used only two fluorescence channels. Two additional channels (Channels 3 263 and 4 264) are available for parallel analysis of other specific targets, cell processes, or cytotoxicity to create multiple parameter screens.
  • the comprehensive data analysis package of the cell screening system allows the user to examine HCS data at multiple levels. Images 276 and detailed data in a spread sheet 279 for individual cells can be viewed separately, or summary data can be plotted. For example, the calculated results of a single parameter for each cell in a 96 well plate are shown in the panel labeled Graph 1 275. By selecting a single point in the graph, the user can display the entire data set for a particular cell that is recalled from an existing database.
  • the large graphical insert 278 shows the results of dexamethasone concentration on the translocation of GFP-hGR. Each point is the average of data from at least 200 cells.
  • the calculated EC 50 for dexamethasone in this assay is 2 nM.
  • FIG. 23 shows kinetic data for the dexamethasone- induced translocation of GFP-hGR in several cells within a single field.
  • Human HeLa cells transfected with GFP-hGR were treated with 100 nM dexamethasone and the translocation of GFP-hGR was measured over time in a population of single cells.
  • the graph shows the response of transfected cells 285, 286, 287, and 288 and non- transfected cells 289. These data also illustrate the ability to analyze cells with different expression levels.
  • Apoptosis is a complex cellular program that involves myriad molecular events and pathways. To understand the mechanisms of drug action on this process, it is essential to measure as many of these events within cells as possible with temporal and spatial resolution. Therefore, an apoptosis screen that requires little cell sample preparation yet provides an automated readout of several apoptosis-related parameters would be ideal.
  • a cell-based assay designed for the cell screening system has been used to simultaneously quantify several of the mo ⁇ hological, organellar, and macromolecular hallmarks of paclitaxel -induced apoptosis. Cell preparation.
  • mice connective tissue fibroblasts L-929; ATCC CCL-1
  • SNB- 19 highly invasive glioblastoma cell line
  • 3500 cells were placed into each well
  • each well received fresh medium containing 750 nM MitoTracker Red (Molecular Probes; Eugene, OR) and 3 ⁇ g/ml Hoechst 33342 DNA-binding dye (Molecular Probes) and was incubated as above for 20 min.
  • fluorescence reagents can be designed to convert this assay into a live cell high-content screen.
  • the average nuclear area ( ⁇ m 2 ) was calculated by dividing the total nuclear area in a field by the number of nuclei detected.
  • the average nuclear perimeter ( ⁇ m) was calculated by dividing the sum of the perimeters of all nuclei in a field by the number of nuclei detected in that field. Highly convoluted apoptotic nuclei had the largest nuclear perimeter values.
  • the average nuclear brightness was calculated by dividing the integrated intensity of the entire field of nuclei by the number of nuclei in that field.
  • the average cellular brightness was calculated by dividing the integrated intensity of an entire field of cells stained with MitoTracker dye by the number of nuclei in that field. Because the amount of MitoTracker dye that accumulates within the mitochondria is proportional to the mitochondrial potential, an increase in the average cell brightness is consistent with an increase in mitochondrial potential.
  • the average cellular brightness was also calculated by dividing the integrated intensity of an entire field of cells stained with Bodipy FL phallacidin dye by the number of nuclei in that field. Because the phallotoxins bind with high affinity to the polymerized form of actin, the amount of Bodipy FL phallacidin dye that accumulates within the cell is proportional to actin polymerization state.
  • Figure 24 shows the changes paclitaxel induced in the nuclear mo ⁇ hology of L-929 cells. Increasing amounts of paclitaxel caused nuclei to enlarge and fragment 293, a hallmark of apoptosis. Quantitative analysis of these and other images obtained by the cell screening system is presented in the same figure. Each parameter measured showed that the L-929 cells 296 were less sensitive to low concentrations of paclitaxel than were SNB-19 cells 297. At higher concentrations though, the L-929 cells showed a response for each parameter measured. The multiparameter approach of this assay is useful in dissecting the mechanisms of drag action.
  • the area, brightness, and fragmentation of the nucleus 298 and actin polymerization values 294 reached a maximum value when SNB-19 cells were treated with 10 nM paclitaxel (Figure 24; top and bottom graphs).
  • mitochondrial potential 295 was minimal at the same concentration of paclitaxel ( Figure 24; middle graph).
  • the fact that all the parameters measured approached control levels at increasing paclitaxel concentrations (>10 nM) suggests that SNB-19 cells have low affinity drag metabolic or clearance pathways that are compensatory at sufficiently high levels of the drag. Contrasting the drag sensitivity of SNB-19 cells 297. L-929 showed a different response to paclitaxel 296.
  • fibroblastic cells showed a maximal response in many parameters at 5 ⁇ M paclitaxel, a 500-fold higher dose than SNB-19 cells. Furthermore, the L-929 cells did not show a sha ⁇ decrease in mitochondrial potential 295 at any of the paclitaxel concentrations tested. This result is consistent with the presence of unique apoptosis pathways between a normal and cancer cell line. Therefore, these results indicate that a relatively simple fluorescence labeling protocol can be coupled with the cell screening system of the present invention to produce a high-content screen of key events involved in programmed cell death.
  • Example 7 Protease induced translocation of a signaling enzyme containing a disease-associated sequence from cytoplasm to nucleus.
  • Plasmid construct A eukaryotic expression plasmid containing a coding sequence for a green fluorescent protein - caspase (Cohen (1997), Biochemical J. 326:1-16; Liang et al. (1997), J. ofMolec. Biol. 274:291-302) chimera is prepared using GFP mutants. The construct is used to transfect eukaryotic cells.
  • Cell preparation and transfection Cells are trypsinized and plated 24 h prior to transfection and incubated at 37°C and 5% CO 2 . Transfections are performed by methods including, but not limited to calcium phosphate coprecipitation or lipofection. Cells are incubated with the calcium phosphate-DNA precipitate for 4-5 hours at 37°C and 5% CO 2 , washed 3-4 times with DMEM to remove the precipitate, followed by the addition of C-DMEM. Lipofectamine transfections are performed in serum-free DMEM without antibiotics according to the manufacturer's instructions. Following a 2-3 hour incubation with the DNA-liposome complexes, the medium is removed and replaced with C-DMEM.
  • Caspase-GFP translocation kinetic data nuclei of transfected cells are first labeled with 5 ⁇ g/ml Hoechst 33342 (Molecular Probes) in C-DMEM for 20 minutes at 37°C and 5% CO 2 . Cells are washed once in Hank's Balanced Salt Solution (HBSS) followed by the addition of compounds that induce apoptosis. These compounds include, but are not limited to paclitaxel, staurosporine, ceramide, and tumor necrosis factor. To obtain fixed time point titration data, transfected cells are first washed with DMEM and then
  • DMEM fetal calf serum
  • Kinetic data are collected by acquiring fluorescence image pairs (Caspase-GFP and Hoechst 33342-labeled nuclei) from fields of living cells at 1 min intervals for 30 min after the addition of compound. Likewise, image pairs are obtained from each well of the fixed time point screening plates 1 h after the addition of compound. In both cases, the image pairs obtained at each time point are used to define nuclear and cytoplasmic regions in each cell. Translocation of Caspase-GFP is calculated by dividing the integrated fluorescence intensity of Caspase- GFP in the nucleus by the integrated fluorescence intensity of the chimera in the cytoplasm or as a nuclear-cytoplasmic difference of GFP fluorescence.
  • this translocation ratio is calculated from data obtained from at least 200 cells at each concentration of compound tested. Drag-induced translocation of Caspase-GFP from the cytoplasm to the nucleus is therefore conelated with an increase in the translocation ratio.
  • Molecular interaction libraries including, but not limited to those comprising putative activators or inhibitors of apoptosis-activated enzymes are use to screen the indicator cell lines and identify a specific ligand for the DAS, and a pathway activated by compound activity.
  • Example 8 Identification of novel steroid receptors from DAS Two sources of material and/or information are required to make use of this embodiment, which allows assessment of the function of an uncharacterized gene.
  • disease associated sequence bank(s) containing cDNA sequences suitable for transfection into mammalian cells can be used. Because every RADE or differential expression experiment generates up to several hundred sequences, it is possible to generate an ample supply of DAS.
  • information from primary sequence database searches can be used to place DAS into broad categories, including, but not limited to, those that contain signal sequences, seven trans-membrane motifs, conserved protease active site domains, or other identifiable motifs. Based on the information acquired from these sources, method types and indicator cell lines to be transfected are selected. A large number of motifs are already well characterized and encoded in the linear sequences contained within the large number genes in existing genomic databases.
  • Information from the DAS identification experiment (including database searches) is used as the basis for selecting the relevant biological processes, (for example, look at the DAS from a tumor line for cell cycle modulation, apoptosis, metastatic proteases, etc.)
  • This initial grouping will determine fluorescent tagging strategies, host cell lines, indicator cell lines, and banks of bioactive molecules to be screened, as described supra.
  • DAS ligate DAS into an expression vector designed for this pu ⁇ ose.
  • Generalized expression vectors contain promoters, enhancers, and terminators for which to deliver target sequences to the cell for transient expression. Such vectors may also contain antibody tagging sequences, direct association sequences, chromophore fusion sequences like GFP, etc. to facilitate detection when expressed by the host.
  • DAS shown to possess a motif(s) suggestive of transcriptional activation potential are utilized to identify novel steroid receptors.
  • Defining the fluorescent tags for this experiment involves identification of the nucleus through staining, and tagging the DAS by creating a GFP chimera via insertion of DAS into an expression vector, proximally fused to the gene encoding GFP.
  • a single chain antibody fragment with high affinity to some portion of the expressed DAS could be constructed using technology available in the art (Cambridge Antibody Technologies) and linked to a fluorophore (FITC) to tag the putative transcriptional activator/receptor in the cells. This alternative would provide an external tag requiring no DNA transfection and therefore would be useful if distribution data were to be gathered from the original primary cultures used to generate the DAS.
  • Plasmid construct A eukaryotic expression plasmid containing a coding sequence for a green fluorescent protein - DAS chimera is prepared using GFP mutants. The construct is used to transfect HeLa cells. The plasmid, when transfected into the host cell, produces a GFP fused to the DAS protein product, designated GFP- DASpp. Cell preparation and transfection. HeLa cells are trypsinized and plated using DMEM containing 5% charcoal/dextran-treated fetal bovine serum (FBS) (Hyclone) and 1%) penicillin-streptomycin (C-DMEM) 12-24 hours prior to transfection and incubated at 37°C and 5% CO .
  • FBS charcoal/dextran-treated fetal bovine serum
  • C-DMEM penicillin-streptomycin
  • Transfections are performed by calcium phosphate coprecipitation or with Lipofectamine (Life Technologies).
  • the medium is replaced, prior to transfection, with DMEM containing 5% charcoal/dextran-treated FBS.
  • Cells are incubated with the calcium phosphate-DNA precipitate for 4-5 hours at 37°C and 5% CO 2 , and washed 3-4 times with DMEM to remove the precipitate, followed by the addition of C-DMEM.
  • Lipofectamine transfections are performed in seram-free DMEM without antibiotics according to the manufacturer's instructions. Following a 2-3 hour incubation with the DNA-liposome complexes, the medium is removed and replaced with C-DMEM. All transfected cells in 96-well microtiter plates are incubated at 33°C and 5% CO 2 for 24-48 hours prior to drug treatment. Experiments are performed with the receptor expressed transiently in HeLa cells.
  • nuclei of transfected cells are first labeled with 5 ⁇ g/ml Hoechst 33342 (Molecular Probes) in C-DMEM for 20 minutes at 33°C and 5% CO . Cells are washed once in Hank's Balanced Salt Solution (HBSS). The cells are analyzed live or they are rinsed with HBSS, fixed for 15 min with 3.7% formaldehyde in HBSS, stained with Hoechst 33342, and washed before analysis.
  • Hoechst 33342 Molecular Probes
  • GFP-DASpp fluorescence signal is collected by acquiring fluorescence image pairs (GFP-DASpp and Hoechst 33342-labeled nuclei) from field cells.
  • the image pairs obtained at each time point are used to define nuclear and cytoplasmic regions in each cell. Data demonstrating dispersed signal in the cytoplasm would be consistent with known steroid receptors that are DNA transcriptional activators. Screening for induction of GFP-DASpp translocation.
  • a screen of various ligands is performed using a series of steroid type ligands including, but not limited to: estrogen, progesterone, retinoids, growth factors, androgens, and many other steroid and steroid based molecules.
  • Image acquisition and analysis are performed using the cell screening system of the invention.
  • the intracellular GFP-DASpp fluorescence signal is collected by acquiring fluorescence image pairs (GFP-DASpp and Hoechst 33342-labeled nuclei) from fields cells. The image pairs obtained at each time point are used to define nuclear and cytoplasmic regions in each cell.
  • Translocation of GFP-DASpp is calculated by dividing the integrated fluorescence intensity of GFP-DASpp in the nucleus by the integrated fluorescence intensity of the chimera in the cytoplasm or as a nuclear-cytoplasmic difference of GFP fluorescence.
  • a translocation from the cytoplasm into the nucleus indicates a ligand binding activation of the DASpp thus identifying the potential receptor class and action. Combining this data with other data obtained in a similar fashion using known inhibitors and modifiers of steroid receptors, would either validate the DASpp as a target, or more data would be generated from various sources.
  • a fluorescent protein biosensor of profilin membrane binding is prepared by labeling purified profilin (Federov et al.(1994), J. Molec. Biol. 241 :480-482; Lanbrechts et al. (1995), Eur. J. Biochem. 230:281-286) with a probe possessing a fluorescence lifetime in the range of 2-300 ns.
  • the labeled profilin is introduced into living indicator cells using bulk loading methodology and the indicator cells are treated with test compounds. Fluorescence anisotropy imaging microscopy (Gough and Taylor (1993), J. Cell Biol. 121:1095-1107) is used to measure test- compound dependent movement of the fluorescent derivative of profilin between the cytoplasm and membrane for a period of time after treatment ranging from 0.1 s to 10 h.
  • Rho-RhoGDI complex translocation to the membrane In another embodiment, indicator cells are treated with test compounds and then fixed, washed, and permeabilized. The indicator cell plasma membrane, cytoplasm, and nucleus are all labeled with distinctly colored markers followed by immunolocalization of Rho protein (Self et al. (1995), Methods in Enzymology 256:3-10; Tanaka et al. (1995), Methods in Enzymology 256:41-49) with antibodies labeled with a fourth color. Each of the four labels is imaged separately using the cell screening system, and the images used to calculate the amount of inhibition or activation of translocation effected by the test compound.
  • the images of the probes used to mark the plasma membrane and cytoplasm are used to mask the image of the immunological probe marking the location of intracellular Rho protein.
  • the integrated brightness per unit area under each mask is used to form a translocation quotient by dividing the plasma membrane integrated brightness/area by the cytoplasmic integrated brightness/area. By comparing the translocation quotient values from control and experimental wells, the percent translocation is calculated for each potential lead compound.
  • the indicator cells contain luminescent markers consisting of a green fluorescent protein
  • GFP- ⁇ -a ⁇ estin ⁇ -anestin (GFP- ⁇ -a ⁇ estin) protein chimera
  • the time scale is 1 h.
  • the output of the high- content screen relates quantitative data describing the magnitude of the translocation within a large number of individual cells that have been treated with test compounds of interest.
  • an endoplasmic reticulum to Golgi translocation high- content screen the translocation of a VSVG protein from the ts045 mutant strain of vesicular stomatitis virus (Ellenberg et al. (1997), J. Cell Biol 138:1193-1206; Presley et al. (1997) Nature 389:81-85) from the endoplasmic reticulum to the Golgi domain is measured in response to cell treatment.
  • indicator cells containing luminescent reporters are treated with test compounds and the movement of the reporters is measured in space and time using the cell screening system of the present invention.
  • the indicator cells contain luminescent reporters consisting of a GFP-VSVG protein chimera that is expressed by the indicator cell through the use of transient or stable cell transfection and other domain markers used to measure the localization of the endoplasmic reticulum and Golgi domains.
  • the GFP-VSVG protein chimera molecules are partitioned predominately in the endoplasmic reticulum.
  • domain markers of distinct colors used to delineate the endoplasmic reticulum and the Golgi domains in distinct channels of fluorescence.
  • the dynamic redistribution of the GFP-VSVG protein chimera is recorded as a series of images over a time scale ranging from 0.1 s to 10 h. Each image is analyzed by a method that quantifies the movement of the GFP-VSVG protein chimera between the endoplasmic reticulum and the Golgi domains. To do this calculation, the images of the probes used to mark the endoplasmic reticulum and the Golgi domains are used to mask the image of the GFP-VSVG probe marking the location of intracellular GFP- VSVG protein.
  • the integrated brightness per unit area under each mask is used to form a translocation quotient by dividing the endoplasmic reticulum integrated brightness/area by the Golgi integrated brightness/area. By comparing the translocation quotient values from control and experimental wells, the percent translocation is calculated for each potential lead compound.
  • the output of the high-content screen relates quantitative data describing the magnitude of the translocation within a large number of individual cells that have been treated with test compounds of interest at final concentrations ranging from IO "12 M to 10 3 M for a period ranging from 1 min to 10 h.
  • an automated method for identifying compounds that modify microtubule structure is provided.
  • indicator cells are treated with test compounds and the distribution of luminescent microtubule-labeling molecules is measured in space and time using a cell screening system, such as the one disclosed above.
  • the luminescent microtubule-labeling molecules may be expressed by or added to the cells either before, together with, or after contacting the cells with a test compound.
  • living cells express a luminescently labeled protein biosensor of microtubule dynamics, comprising a protein that labels microtubules fused to a luminescent protein.
  • a luminescently labeled protein biosensor of microtubule dynamics comprising a protein that labels microtubules fused to a luminescent protein.
  • labeling proteins for this aspect of the invention include, but are not limited to ⁇ and ⁇
  • the method involves transfecting cells with a microtubule labeling luminescent protein, wherein the microtubule labeling protein can be, but is
  • ⁇ -tubulin not limited to, ⁇ -tubulin, ⁇ -tubulin, or microtubule-associated protein 4 (MAP4).
  • MAP4 microtubule-associated protein 4
  • MAP4 is fused to a modified version of the Aequorea victoria green fluorescent protein (GFP).
  • GFP Aequorea victoria green fluorescent protein
  • a DNA construct has been made which consists of a fusion between the EGFP coding sequence (available from Clontech) and the coding sequence for mouse MAP4. (Olson et al., (1995), J. Cell Biol. 130(3): 639-650).
  • MAP4 is a ubiquitous microtubule-associated protein that is known to interact with microtubules in inte ⁇ hase as well as mitotic cells (Olmsted and Murofushi, (1993), MAP4. In "Guidebook to the Cytoskeleton and Motor Proteins.”
  • the luminescently labeled protein biosensor of microtubule dynamics is expressed, isolated, and added to the cells to be analyzed via bulk loading techniques, such as microinjection, scrape loading, and impact-mediated loading.
  • bulk loading techniques such as microinjection, scrape loading, and impact-mediated loading.
  • ⁇ and ⁇ tubulin isoforms, MAP4, MAP2 and/or tau can all be used.
  • the protein biosensor is expressed by the cell, and the cell is subsequently contacted with a luminescent label, such as a labeled antibody, that detects the protein biosensor, endogenous levels of a protein antigen, or both.
  • a luminescent label such as a labeled antibody
  • a luminescent label that detects ⁇ and ⁇ tubulin isoforms, MAP4, MAP2
  • GFP mutants can be used.
  • GFP mutants are available, all of which would be effective in this invention, including, but not limited to, GFP mutants which are commercially available ⁇ (Clontech, California).
  • the MAP4 construct has been introduced into several mammalian cell lines (BHK-21, Swiss 3T3, HeLa, HEK 293, LLCPK) and the organization and localization of tubulin has been visualized in live cells by virtue of the GFP fluorescence as an indicator of MAP4 localization.
  • the construct can be expressed transiently or stable cell lines can be prepared by standard methods. Stable HeLa cell lines expressing the EGFP-MAP4 chimera have been obtained, indicating that expression of the chimera is not toxic and does not interfere with mitosis.
  • Possible selectable markers for establishment and maintenance of stable cell lines include, but are not limited to the neomycin resistance gene, hygromycin resistance gene, zeocin resistance gene, puromycin resistance gene, bleomycin resistance gene, and blastacidin resistance gene.
  • the utility of this method for the monitoring of microtubule assembly, disassembly, and reanangement has been demonstrated by treatment of transiently and stably transfected cells with microtubule drags such as paclitaxel, nocodazole, vincristine, or vinblastine.
  • the present method provides high-content and combined high throughput-high content cell-based screens for anti-microtubule drags, particularly as one parameter in a multi-parametric cancer target screen.
  • the EGFP-MAP4 construct used herein can also be used as one of the components of a high-content screen that measures multiple signaling pathways or physiological events.
  • a combined high throughput and high content screen is employed, wherein multiple cells in each of the locations containing cells are analyzed in a high throughput mode, and only a subset of the locations containing cells are analyzed in a high content mode.
  • the high throughput screen can be any screen that would be useful to identify those locations containing cells that should be further analyzed, including, but not limited to, identifying locations with increased luminescence intensity, those exhibiting expression of a reporter gene, those undergoing calcium changes, and those undergoing pH changes.
  • the present invention may be applied to clinical diagnostics, the detection of chemical and biological warfare weapons, and the basic research market since fundamental cell processes, such as cell division and motility, are highly dependent upon microtubule dynamics.
  • Image data can be obtained from either fixed or living indicator cells.
  • mo ⁇ hometric data from each of the images obtained the following method of analysis is used:
  • Microtubule mo ⁇ hology is defined using a set of classifiers to quantify aspects of microtubule shape, size, aggregation state, and polymerization state.
  • classifiers can be based on approaches that include co-occunence matrices, texture measurements, spectral methods, structural methods, wavelet transforms, statistical methods, or combinations thereof. Examples of such classifiers are as follows:
  • a classifier to quantify microtubule length and width using edge detection methods such as that discussed in Kolega et al. ((1993). Biolmaging 1 :136- 150), which discloses a non-automated method to determine edge strength in individual cells), to calculate the total edge strength within each cell.
  • the total edge strength can be divided by the cell area to give a "microtubule mo ⁇ hology" value.
  • Large microtubule mo ⁇ hology values are associated with strong edge strength values and are therefore maximal in cells containing distinct microtubule structures.
  • small microtubule mo ⁇ hology values are associated with weak edge strength and are minimal in cells with depolymerized microtubules.
  • the physiological range of microtubule mo ⁇ hology values is set by treating cells with either the microtubule stabilizing drug paclitaxel (10 ⁇ M) or the microtubule depolymerizing drag nocodazole (10 ⁇ g/ml).
  • a classifier to quantify microtubule depolymerization using a measure of image texture 4.
  • kits for analyzing microtubule stability, comprising an expression vector comprising a nucleic acid that encodes a microtubule labeling protein and instructions for using the expression vector for carrying out the methods described above.
  • the expression vector further comprises a nucleic acid that encodes a luminescent protein, wherein the microtubule binding protein and the luminescent protein thereof are expressed as a fusion protein.
  • the kit may contain an antibody that specifically binds to the microtubule-labeling protein.
  • the kit includes cells that express the microtubule labeling protein.
  • the cells are transfected with the expression vector.
  • kits further contain a compound that is known to disrapt microtubule structure, including but not limited to curacin, nocodazole, vincristine, or vinblastine.
  • kits further comprise a compound that is known to stabilize microtubule structure, including but not limited to taxol (paclitaxel), and discodermolide.
  • the present invention comprises a machine readable storage medium comprising a program containing a set of instructions for causing a cell screening system to execute the disclosed methods for analyzing microtubule stability
  • the cell screening system comprises an optical system with a stage adapted for holding a plate containing cells, a digital camera, a means for directing fluorescence or luminescence emitted from the cells to the digital camera, and a computer means for receiving and processing the digital data from the digital camera.
  • Glycolytic enzyme activity regulation In a prefened embodiment of a cellular enzyme activity high-content screen, the activity of key glycolytic regulatory enzymes are measured in treated cells. To measure enzyme activity, indicator cells containing luminescent labeling reagents are treated with test compounds and the activity of the reporters is measured in space and time using cell screening system of the present invention.
  • the reporter of intracellular enzyme activity is fructose-6- phosphate, 2-kinase/fructose-2,6-bisphosphatase (PFK-2), a regulatory enzyme whose phosphorylation state indicates intracellular carbohydrate anabolism or catabolism (Deprez et al. (1997) J. Biol. Chem. 272:17269-17275; Kealer et al. (1996) FEBS Letters 395:225-227; Lee et al. (1996), Biochemisti ⁇ 35:6010-6019).
  • the indicator cells contain luminescent reporters consisting of a fluorescent protein biosensor of PFK-2 phosphorylation.
  • the fluorescent protein biosensor is constructed by introducing an environmentally sensitive fluorescent dye near to the known phosphorylation site of the enzyme (Deprez et al. (1997), supra; Giuliano et al. (1995), supra).
  • the dye can be of the ketocyanine class (Kessler and Wolfbeis (1991), Spectrochimica Acta 47A:187-192 ) or any class that contains a protein reactive moiety and a fluorochrome whose excitation or emission spectrum is sensitive to solution polarity.
  • the fluorescent protein biosensor is introduced into the indicator cells using bulk loading methodology.
  • Living indicator cells are treated with test compounds, at final concentrations ranging from IO "12 M to IO "3 M for times ranging from 0.1 s to 10 h.
  • ratio image data are obtained from living treated indicator cells by collecting a spectral pair of fluorescence images at each time point. To extract mo ⁇ hometric data from each time point, a ratio is made between each pair of images by numerically dividing the two spectral images at each time point, pixel by pixel. Each pixel value is then used to calculate the fractional phosphorylation of PFK-2. At small fractional values of phosphorylation, PFK-2 stimulates carbohydrate catabolism. At high fractional values of phosphorylation, PFK-2 stimulates carbohydrate anabolism.
  • Protein kinase A activity and localization of sub units are measured in response to treatment with test compounds.
  • PKA protein kinase A
  • the indicator cells contain luminescent reporters including a fluorescent protein biosensor of PKA activation.
  • the fluorescent protein biosensor is constructed by introducing an environmentally sensitive fluorescent dye into the catalytic subunit of PKA near the site known to interact with the regulatory subunit of PKA (Harootunian et al. (1993), Mol. Biol. of the Cell 4:993-1002; Johnson et al. (1996), Cell 85: 149-158; Giuliano et al. (1995), supra).
  • the dye can be of the ketocyanine class (Kessler, and Wolfbeis (1991), Spectrochimica Acta 47A:187-192) or any class that contains a protein reactive moiety and a fluorochrome whose excitation or emission spectrum is sensitive to solution polarity.
  • the fluorescent protein biosensor of PKA activation is introduced into the indicator cells using bulk loading methodology.
  • living indicator cells are treated with test compounds, at final concentrations ranging from IO "12 M to 10 "3 M for times ranging from 0.1 s to 10 h.
  • ratio image data are obtained from living treated indicator cells. To extract biosensor data from each time point, a ratio is made between each pair of images, and each pixel value is then used to calculate the fractional activation of PKA (e.g., separation of the catalytic and regulatory subunits after cAMP binding). At high fractional values of activity, PFK-2 stimulates biochemical cascades within the living cell.
  • indicator cells containing luminescent reporters are treated with test compounds and the movement of the reporters is measured in space and time using the cell screening system.
  • the indicator cells contain luminescent reporters consisting of domain markers used to measure the localization of the cytoplasmic and nuclear domains.
  • the dynamic redistribution of a PKA fluorescent protein biosensor is recorded intracellularly as a series of images over a time scale ranging from 0.1 s to 10 h. Each image is analyzed by a method that quantifies the movement of the PKA between the cytoplasmic and nuclear domains.
  • the images of the probes used to mark the cytoplasmic and nuclear domains are used to mask the image of the PKA fluorescent protein biosensor.
  • the integrated brightness per unit area under each mask is used to form a translocation quotient by dividing the cytoplasmic integrated brightness/area by the nuclear integrated brightness/area.
  • the percent translocation is calculated for each potential lead compound.
  • the output of the high-content screen relates quantitative data describing the magnitude of the translocation within a large number of individual cells that have been treated with test compound in the concentration range of 10 " M to 10 " M. High-content screens involving the induction or inhibition of gene expression RNA-based fluorescent biosensors
  • Cytoskeletal protein transcription and message localization Regulation of the general classes of cell physiological responses including cell-substrate adhesion, cell-cell adhesion, signal transduction, cell-cycle events, intermediary and signaling molecule metabolism, cell locomotion, cell-cell communication, and cell death can involve the alteration of gene expression. High-content screens can also be designed to measure this class of physiological response.
  • the reporter of intracellular gene expression is an oligonucleotide that can hybridize with the target mRNA and alter its fluorescence signal.
  • the oligonucleotide is a molecular beacon (Tyagi and Kramer (1996) Nat. Biotechnol. 14:303-308), a luminescence-based reagent whose fluorescence signal is dependent on intermolecular and intramolecular interactions.
  • the fluorescent biosensor is constructed by introducing a fluorescence energy transfer pair of fluorescent dyes such that there is one at each end (5' and 3') of the reagent.
  • the dyes can be of any class that contains a protein reactive moiety and fluorochromes whose excitation and emission spectra overlap sufficiently to provide fluorescence energy transfer between the dyes in the resting state, including, but not limited to, fluorescein and rhodamine (Molecular Probes, Inc.).
  • fluorescein and rhodamine Molecular Probes, Inc.
  • the complete fluorescent biosensor is introduced into the indicator cells using bulk loading methodology.
  • living indicator cells are treated with test compounds, at final concentrations ranging from IO "12 M to IO "3 M for times ranging from 0.1 s to 10 h.
  • ratio image data are obtained from living treated indicator cells. To extract mo ⁇ hometric data from each time point, a ratio is made between each pair of images, and each pixel value is then used to calculate the fractional hybridization of the labeled nucleotide. At small fractional values of
  • 270:C1438-C1446) that are labeled with a luminescent probe of a different color for an appropriate time under the appropriate conditions.
  • a luminescent probe of a different color for an appropriate time under the appropriate conditions.
  • unbound insulin molecules are washed away, the cells fixed and the distribution and concentration of the insulin on the plasma membrane is measured.
  • the cell membrane image is used as a mask for the insulin image.
  • the integrated intensity from the masked insulin image is compared to a set of images containing known amounts of labeled insulin.
  • the amount of insulin bound to the cell is determined from the standards and used in conjunction with the total concentration of insulin incubated with the cell to calculate a dissociation constant or insulin to its cell surface receptor.
  • Labeling of cellular compartments Whole cell labeling is accomplished by labeling cellular components such that dynamics of cell shape and motility of the cell can be measured over time by analyzing fluorescence images of cells.
  • small reactive fluorescent molecules are introduced into living cells. These membrane-permeant molecules both diffuse through and react with protein components in the plasma membrane. Dye molecules react with intracellular molecules to both increase the fluorescence signal emitted from each molecule and to entrap the fluorescent dye within living cells. These molecules include reactive chloromethyl derivatives of aminocoumarins, hydroxycoumarins, eosin diacetate, fluorescein diacetate, some Bodipy dye derivatives, and tetramethylrhodamine. The reactivity of these dyes toward macromolecules includes free primary amino groups and free sulfhydryl groups.
  • the cell surface is labeled by allowing the cell to interact with fluorescently labeled antibodies or lectins (Sigma Chemical Company, St. Louis, MO) that react specifically with molecules on the cell surface.
  • fluorescently labeled antibodies or lectins Sigma Chemical Company, St. Louis, MO
  • Cell surface protein chimeras expressed by the cell of interest that contain a green fluorescent protein, or mutant thereof, component can also be used to fluorescently label the entire cell surface.
  • labeling the whole plasma membrane employs some of the same methodology described above for labeling the entire cells.
  • Luminescent molecules that label the entire cell surface act to delineate the plasma membrane.
  • subdomains of the plasma membrane, the extracellular surface, the lipid bilayer, and the intracellular surface can be labeled separately and used as components of high content screens.
  • the extracellular surface is labeled using a brief treatment with a reactive fluorescent molecule such as the succinimidyl ester or iodoacetamde derivatives of fluorescent dyes such as the fluoresceins, rhodamines, cyanines, and Bodipys.
  • the extracellular surface is labeled using fluorescently labeled macromolecules with a high affinity for cell surface molecules.
  • fluorescently labeled macromolecules such as the fluorescein, rhodamine, and cyanine derivatives of lectins derived from jack bean (Con A), red kidney bean
  • fluorescently labeled antibodies with a high affinity for cell surface components are used to label the extracellular region of the plasma membrane.
  • Extracellular regions of cell surface receptors and ion channels are examples of proteins that can be labeled with antibodies.
  • the lipid bilayer of the plasma membrane is labeled with fluorescent molecules.
  • fluorescent molecules include fluorescent dyes attached to long chain hydrophobic molecules that interact strongly with the hydrophobic region in the center of the plasma membrane lipid bilayer. Examples of these dyes include the PKH series of dyes (U.S. 4,783,401, 4,762701, and 4,859,584; available commercially from Sigma Chemical Company, St.
  • fluorescent phospholipids such as nitrobenzoxadiazole glycerophosphoethanolamine and fluorescein-derivatized dihexadecanoylglycerophosphoetha-nolamine, fluorescent fatty acids such as 5-butyl- 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene-3-nonanoic acid and 1 -pyrenedecanoic acid (Molecular Probes, Inc.), fluorescent sterols including cholesteryl 4,4-difluoro-5,7- dimethyl-4-bora-3a,4a-diaza-s-indacene-3-dodecanoate and cholesteryl 1- pyrenehexanoate, and fluorescently labeled proteins that interact specifically with lipid bilayer components such as the fluorescein derivative of annexin V (Caltag Antibody Co, Burlingame, CA).
  • fluorescent phospholipids such as nitrobenzoxadiazole
  • the intracellular component of the plasma membrane is labeled with fluorescent molecules.
  • these molecules are the intracellular components of the trimeric G-protein receptor, adenylyl cyclase, and ionic transport proteins. These molecules can be labeled as a result of tight binding to a fluorescently labeled specific antibody or by the inco ⁇ oration of a fluorescent protein chimera that is comprised of a membrane-associated protein and the green fluorescent protein, and mutants thereof.
  • ligands that are transported into cells by receptor-mediated endocytosis are used to trace the dynamics of endosomal organelles.
  • labeled ligands include Bodipy FL-labeled low density lipoprotein complexes, tetramethylrhodamine transferrin analogs, and fluorescently labeled epidermal growth factor (Molecular Probes, Inc.)
  • fluorescently labeled primary or secondary antibodies (Sigma Chemical Co. St. Louis, MO; Molecular Probes, Inc. Eugene, OR; Caltag Antibody Co.) that specifically label endosomal ligands are used to mark the endosomal compartment in cells.
  • endosomes are fluorescently labeled in cells expressing protein chimeras formed by fusing a green fluorescent protein, or mutants thereof, with a receptor whose intemalization labels endosomes.
  • Chimeras of the EGF, transferrin, and low density lipoprotein receptors are examples of these molecules.
  • membrane permeant lysosome-specific luminescent reagents are used to label the lysosomal compartment of living and fixed cells. These reagents include the luminescent molecules neutral red, N-(3-((2,4- dinitrophenyl)amino)propyl)-N-(3-aminopropyl)methylamine, and the LysoTracker probes which report intralysosomal pH as well as the dynamic distribution of lysosomes (Molecular Probes, Inc.)
  • antibodies against lysosomal antigens are used to label lysosomal components that are localized in specific lysosomal domains.
  • these components are the degradative enzymes involved in cholesterol ester hydrolysis, membrane protein proteases, and nucleases as well as the ATP-driven lysosomal proton pump.
  • protein chimeras consisting of a lysosomal protein genetically fused to an intrinsically luminescent protein such as the green fluorescent protein, or mutants thereof, are used to label the lysosomal domain. Examples of these components are the degradative enzymes involved in cholesterol ester hydrolysis, membrane protein proteases, and nucleases as well as the ATP-driven lysosomal proton pump.
  • cell permeant fluorescent dyes (Molecular Probes, Inc.) with a reactive group are reacted with living cells.
  • Reactive dyes including monobromobimane, 5-chloromethylfluorescein diacetate, carboxy fluorescein diacetate succinimidyl ester, and chloromethyl tetramethylrhodamine are examples of cell permeant fluorescent dyes that are used for long term labeling of the cytoplasm of cells.
  • polar tracer molecules such as Lucifer yellow and cascade blue-based fluorescent dyes (Molecular Probes, Inc.) are introduced into cells using bulk loading methods and are also used for cytoplasmic labeling.
  • antibodies against cytoplasmic components are used to fluorescently label the cytoplasm.
  • cytoplasmic antigens are many of the enzymes involved in intermediary metabolism. Enolase, phosphofractokinase, and acetyl-CoA dehydrogenase are examples of uniformly distributed cytoplasmic antigens.
  • protein chimeras consisting of a cytoplasmic protein genetically fused to an intrinsically luminescent protein such as the green fluorescent protein, or mutants thereof, are used to label the cytoplasm.
  • fluorescent chimeras of uniformly distributed proteins are used to label the entire cytoplasmic domain. Examples of these proteins are many of the proteins involved in intermediary metabolism and include enolase, lactate dehydrogenase, and hexokinase.
  • antibodies against cytoplasmic antigens are used to label cytoplasmic components that are localized in specific cytoplasmic sub-domains.
  • these components are the cytoskeletal proteins actin, tubulin, and cytokeratin.
  • a population of these proteins within cells is assembled into discrete structures, which in this case, are fibrous. Fluorescence labeling of these proteins with antibody-based reagents therefore labels a specific sub-domain of the cytoplasm.
  • non-antibody-based fluorescently labeled molecules that interact strongly with cytoplasmic proteins are used to label specific cytoplasmic components.
  • cytoplasmic components include DNAse I (Molecular Probes, Inc.) Fluorescent analogs of this enzyme bind tightly and specifically to cytoplasmic actin, thus labeling a sub-domain of the cytoplasm.
  • fluorescent analogs of the mushroom toxin phalloidin or the drag paclitaxel are used to label components of the actin- and microtubule-cytoskeletons, respectively.
  • protein chimeras consisting of a cytoplasmic protein genetically fused to an intrinsically luminescent protein such as the green fluorescent protein, or mutants thereof, are used to label specific domains of the cytoplasm.
  • fluorescent chimeras of highly localized proteins are used to label cytoplasmic sub- domains. Examples of these proteins are many of the proteins involved in regulating the cytoskeleton. They include the structural proteins actin, tubulin, and cytokeratin as
  • microtubule associated protein 4 As well as the regulatory proteins microtubule associated protein 4 and ⁇ -actinin.
  • membrane permeant nuclei c-acid-specific luminescent reagents are used to label the nucleus of living and fixed cells.
  • reagents include cyanine-based dyes (e.g., TOTO ® , YOYO ® , and BOBOTM),
  • phenanthidines and acridines e.g., ethidium bromide, propidium iodide, and acridine orange
  • indoles and imidazoles e.g., Hoechst 33258, Hoechst 33342, and 4', 6- diamidino-2-phenylindole
  • other similar reagents e.g., 7-aminoactinomycin D, hydroxystilbamidine, and the psoralens.
  • antibodies against nuclear antigens are used to label nuclear components that are localized in specific nuclear domains.
  • nuclear antigens are the macromolecules involved in maintaining DNA structure and function.
  • DNA, RNA, histones, DNA polymerase, RNA polymerase, lamins, and nuclear variants of cytoplasmic proteins such as actin are examples of nuclear antigens.
  • protein chimeras consisting of a nuclear protein genetically fused to an intrinsically luminescent protein such as the green fluorescent protein, or mutants thereof, are used to label the nuclear domain.
  • these proteins are many of the proteins involved in maintaining DNA stracture and function. Histones, DNA polymerase, RNA polymerase, lamins, and nuclear variants of cytoplasmic proteins such as actin are examples of nuclear proteins.
  • membrane permeant mitochondrial-specific luminescent reagents are used to label the mitochondria of living and fixed cells. These reagents include rhodamine 123, tetramethyl rosamine, JC-1, and the MitoTracker reactive dyes.
  • antibodies against mitochondrial antigens are used to label mitochondrial components that are localized in specific mitochondrial domains.
  • these components are the macromolecules involved in maintaining mitochondrial DNA stracture and function.
  • DNA, RNA, histones, DNA polymerase, RNA polymerase, and mitochondrial variants of cytoplasmic macromolecules such as mitochondrial tRNA and rRNA are examples mitochondrial antigens.
  • Other examples of mitochondrial antigens are the components of the oxidative phosphorylation system found in the mitochondria (e.g., cytochrome c, cytochrome c oxidase, and succinate dehydrogenase).
  • protein chimeras consisting of a mitochondrial protein genetically fused to an intrinsically luminescent protein such as the green fluorescent protein, or mutants thereof, are used to label the mitochondrial domain.
  • these components are the macromolecules involved in maintaining mitochondrial DNA structure and function. Examples include histones, DNA polymerase, RNA polymerase, and the components of the oxidative phosphorylation system found in the mitochondria (e.g., cytochrome c, cytochrome c oxidase, and succinate dehydrogenase).
  • membrane permeant endoplasmic reticulum-specific luminescent reagents are used to label the endoplasmic reticulum of living and fixed cells.
  • These reagents include short chain carbocyanine dyes (e.g., DiOC and DiOC 3 ), long chain carbocyanine dyes (e.g., DiICi 6 and DiIC ⁇ ), and luminescently labeled lectins such as concanavalin A.
  • antibodies against endoplasmic reticulum antigens are provided.
  • endoplasmic reticulum components that are localized in specific endoplasmic reticulum domains. Examples of these components are the macromolecules involved in the fatty acid elongation systems, glucose-6-phosphatase, and HMG CoA-reductase.
  • protein chimeras consisting of a endoplasmic reticulum protein genetically fused to an intrinsically luminescent protein such as the green fluorescent protein, or mutants thereof, are used to label the endoplasmic reticulum domain.
  • these components are the macromolecules involved in the fatty acid elongation systems, glucose-6-phosphatase, and HMG CoA-reductase.
  • membrane permeant Golgi-specific luminescent reagents are used to label the Golgi of living and fixed cells.
  • These reagents include luminescently labeled macromolecules such as wheat germ agglutinin and Brefeldin A as well as luminescently labeled ceramide.
  • Golgi components that are localized in specific Golgi domains. Examples of these components are N- acetylglucosamine phosphotransferase, Golgi-specific phosphodiesterase, and mannose-6-phosphate receptor protein.
  • protein chimeras consisting of a Golgi protein genetically fused to an intrinsically luminescent protein such as the green fluorescent protein, or mutants thereof, are used to label the Golgi domain.
  • these components are N-acetylglucosamine phosphotransferase, Golgi-specific phosphodiesterase, and mannose-6-phosphate receptor protein.
  • the signaling pathway from the cell surface to target sites within the cell involves the translocation of plasma membrane-associated proteins to the cytoplasm.
  • plasma membrane-associated proteins For example, it is known that one of the src family of protein tyrosine kinases, pp60c-src (Walker et al (1993), J. Biol Chem. 268:19552-19558) translocates from the plasma membrane to the cytoplasm upon stimulation of fibroblasts with platelet-derived growth factor (PDGF).
  • PDGF platelet-derived growth factor
  • the targets for screening can themselves be converted into fluorescence- based reagents that report molecular changes including ligand-binding and post- translocational modifications.

Abstract

The present invention provides systems, methods, screens, and kits for optical system analysis of cells to rapidly determine the distribution, environment, or activity of fluorescently labeled reporter molecules in cells for the purpose of screening large numbers of compounds for those that specifically affect particular biological functions. The invention involves providing cells containing fluorescent reporter molecules in an array of locations and scanning numerous cells in each location with a high magnification fluorescence optical system, converting the optical information into digital data, and utilizing the digital data to determine the distribution, environment or activity of the fluorescently labeled reporter molecules in the cells. The array of locations may be an industry standard 96 well or 384 well microtiter plate or a microplate which is a microplate having cells in a micropatterned array of locations. The invention includes apparatus and computerized method for processing, displaying and storing the data.

Description

A SYSTEM FOR CELL-BASED SCREENING
Cross Reference
This application is a continuation-in-part of U.S. Applications for Patent S/N
60/100,973 filed September 18, 1998, and 09/031,271 filed February 27, 1998 which is a continuation in part of U.S. Application S/N 08/810983, filed on February 27, 1997.
Field of The Invention
This invention is in the field of fluorescence-based cell and molecular biochemical assays for drug discovery.
Background of the Invention
Drug discovery, as currently practiced in the art, is a long, multiple step process involving identification of specific disease targets, development of an assay based on a specific target, validation of the assay, optimization and automation of the assay to produce a screen, high throughput screening of compound libraries using the assay to identify "hits", hit validation and hit compound optimization. The output of this process is a lead compound that goes into pre-clinical and, if validated, eventually into clinical trials. In this process, the screening phase is distinct from the assay development phases, and involves testing compound efficacy in living biological systems.
Historically, drug discovery is a slow and costly process, spanning numerous years and consuming hundreds of millions of dollars per drug created. Developments in the areas of genomics and high throughput screening have resulted in increased capacity and efficiency in the areas of target identification and volume of compounds screened. Significant advances in automated DNA sequencing, PCR application, positional cloning, hybridization arrays, and bioinformatics have greatly increased the number of genes (and gene fragments) encoding potential drug screening targets. However, the basic scheme for drug screening remians the same.
Validation of genomic targets as points for therapeutic intervention using the existing methods and protocols has become a bottleneck in the drug discovery process due to the slow, manual methods employed, such as in vivo functional models, functional analysis of recombinant proteins, and stable cell line expression of candidate genes. Primary DNA sequence data acquired through automated sequencing does not permit identification of gene function, but can provide information about common "motifs" and specific gene homology when compared to known sequence databases. Genomic methods such as subtraction hybridization and RADE (rapid amplification of differential expression) can be used to identify genes that are up or down regulated in a disease state model. However, identification and validation still proceed down the same pathway. Some proteomic methods use protein identification (global expression arrays, 2D electrophoresis, combinatorial libraries) in combination with reverse genetics to identify candidate genes of interest. Such putative "disease associated sequences" or DAS isolated as intact cDNA are a great advantage to these methods, but they are identified by the hundreds without providing any information regarding type, activity, and distribution of the encoded protein. Choosing a subset of DAS as drug screening targets is "random", and thus extremely inefficient, without functional data to provide a mechanistic link with disease. It is necessary, therefore, to provide new technologies to rapidly screen DAS to establish biological function, thereby improving target validation and candidate optimization in drug discovery.
There are three major avenues for improving early drug discovery productivity. First, there is a need for tools that provide increased information handling capability. Bioinformatics has blossomed with the rapid development of DNA sequencing systems and the evolution of the genomics database. Genomics is beginning to play a critical role in the identification of potential new targets. Proteomics has become indispensible in relating structure and function of protein targets in order to predict drug interactions. However, the next level of biological complexity is the cell. Therefore, there is a need to acquire, manage and search multi-dimensional information from cells. Secondly, there is a need for higher throughput tools. Automation is a key to improving productivity as has already been demonstrated in DNA sequencing and high throughput primary screening. The instant invention provides for automated systems that extract multiple parameter information from cells that meet the need for higher throughput tools. The instant invention also provides for miniaturizing the methods, thereby allowing increased throughput, while decreasing the volumes of reagents and test compounds required in each assay.
Radioactivity has been the dominant read-out in early drug discovery assays. However, the need for more information, higher throughput and miniaturization has caused a shift towards using fluorescence detection. Fluorescence-based reagents can yield more powerful, multiple parameter assays that are higher in throughput and information content and require lower volumes of reagents and test compounds. Fluorescence is also safer and less expensive than radioactivity-based methods. Screening of cells treated with dyes and fluorescent reagents is well known in the art. There is a considerable body of literature related to genetic engineering of cells to produce fluorescent proteins, such as modified green fluorescent protein (GFP), as a reporter molecule. Some properties of wild-type GFP are disclosed by Morise et al. (Biochemistry 13 (1974), p. 2656-2662), and Ward et al. (Photochem. Photobiol 31 (1980), p. 611-615). The GFP of the jellyfish Aequorea victoria has an excitation maximum at 395 nm and an emission maximum at 510 nm, and does not require an exogenous factor for fluorescence activity. Uses for GFP disclosed in the literature are widespread and include the study of gene expression and protein localization (Chalfie et al, Science 263 (1994), p. 12501-12504)), as a tool for visualizing subcellular organelles (Rizzuto et al., Curr. Biology 5 (1995), p. 635-642)), visualization of protein transport along the secretory pathway (Kaether and Gerdes, FEBS Letters 369 (1995), p. 267-271)), expression in plant cells (Hu and Cheng, FEBS Letters 369 (1995), p. 331-334)) and Drosophila embryos (Davis et al, Dev. Biology 170 (1995), p. 726- 729)), and as a reporter molecule fused to another protein of interest (U. S. Patent 5,491,084). Similarly, WO96/23898 relates to methods of detecting biologically active substances affecting intracellular processes by utilizing a GFP construct having a protein kinase activation site. This patent, and all other patents referenced in this application are incoφorated by reference in their entirety Numerous references are related to GFP proteins in biological systems. For example, WO 96/09598 describes a system for isolating cells of interest utilizing the expression of a GFP like protein. WO 96/27675 describes the expression of GFP in plants. WO 95/21191 describes modified GFP protein expressed in transformed organisms to detect mutagenesis. U. S. Patents 5,401,629 and 5,436,128 describe assays and compositions for detecting and evaluating the intracellular transduction of an extracellular signal using recombinant cells that express cell surface receptors and contain reporter gene constructs that include transcriptional regulatory elements that are responsive to the activity of cell surface receptors. Performing a screen on many thousands of compounds requires parallel handling and processing of many compounds and assay component reagents. Standard high throughput screens ("HTS") use mixtures of compounds and biological reagents along with some indicator compound loaded into arrays of wells in standard microtiter plates with 96 or 384 wells. The signal measured from each well, either fluorescence emission, optical density, or radioactivity, integrates the signal from all the material in the well giving an overall population average of all the molecules in the well.
Science Applications International Corporation (SAIC) 130 Fifth Avenue, Seattle, WA. 98109) describes an imaging plate reader. This system uses a CCD camera to image the whole area of a 96 well plate. The image is analyzed to calculate the total fluorescence per well for all the material in the well.
Molecular Devices, Inc. (Sunnyvale, CA) describes a system (FLIPR) which uses low angle laser scanning illumination and a mask to selectively excite fluorescence within approximately 200 microns of the bottoms of the wells in standard 96 well plates in order to reduce background when imaging cell monolayers. This system uses a CCD camera to image the whole area of the plate bottom. Although this system measures signals originating from a cell monolayer at the bottom of the well, the signal measured is averaged over the area of the well and is therefore still considered a measurement of the average response of a population of cells. The image is analyzed to calculate the total fluorescence per well for cell-based assays. Fluid delivery devices have also been incorporated into cell based screening systems, such as the FLIPR system, in order to initiate a response, which is then observed as a whole well population average response using a macro-imaging system.
In contrast to high throughput screens, various high-content screens ("HCS") have been developed to address the need for more detailed information about the temporal-spatial dynamics of cell constituents and processes. High-content screens automate the extraction of multicolor fluorescence information derived from specific fluorescence-based reagents incorporated into cells (Giuliano and Taylor (1995), Curr. Op. Cell Biol. 7:4; Giuliano et al. (1995) Ann. Rev. Biophys. Biomol. Struct. 24:405). Cells are analyzed using an optical system that can measure spatial, as well as temporal dynamics. (Farkas et al. (1993) Ann. Rev. Physiol 55:785; Giuliano et al. (1990) In Optical Microscopy for Biology. B. Herman and K. Jacobson (eds.), pp. 543-557. Wiley-Liss, New York; Hahn et al (1992) Nature 359:736; Waggoner et al. (1996) Hum. Pathol 27:494). The concept is to treat each cell as a "well" that has spatial and temporal information on the activities of the labeled constituents.
The types of biochemical and molecular information now accessible through fluorescence-based reagents applied to cells include ion concentrations, membrane potential, specific translocations, enzyme activities, gene expression, as well as the presence, amounts and patterns of metabolites, proteins, lipids, carbohydrates, and nucleic acid sequences (DeBiasio et al., (1996) Mol. Biol. Cell. 7:1259;Giuliano et al., (1995) Ann. Rev. Biophys. Biomol. Struct. 24:405; Heim and Tsien, (1996) Curr. Biol. 6:178).
High-content screens can be performed on either fixed cells, using fluorescently labeled antibodies, biological ligands, and/or nucleic acid hybridization probes, or live cells using multicolor fluorescent indicators and "biosensors." The choice of fixed or live cell screens depends on the specific cell-based assay required.
Fixed cell assays are the simplest, since an array of initially living cells in a microtiter plate format can be treated with various compounds and doses being tested, then the cells can be fixed, labeled with specific reagents, and measured. No environmental control of the cells is required after fixation. Spatial information is acquired, but only at one time point. The availability of thousands of antibodies, ligands and nucleic acid hybridization probes that can be applied to cells makes this an attractive approach for many types of cell-based screens. The fixation and labeling steps can be automated, allowing efficient processing of assays.
Live cell assays are more sophisticated and powerful, since an array of living cells containing the desired reagents can be screened over time, as well as space. Environmental control of the cells (temperature, humidity, and carbon dioxide) is required during measurement, since the physiological health of the cells must be maintained for multiple fluorescence measurements over time. There is a growing list of fluorescent physiological indicators and "biosensors" that can report changes in biochemical and molecular activities within cells (Giuliano et al., (1995) Ann. Rev. Biophys. Biomol Struct. 24:405; Hahn et al., (1993) In Fluorescent and Luminescent Probes for Biological Activity. W.T. Mason, (ed.), pp. 349-359, Academic Press, San Diego).
The availability and use of fluorescence-based reagents has helped to advance the development of both fixed and live cell high-content screens. Advances in instrumentation to automatically extract multicolor, high-content information has recently made it possible to develop HCS into an automated tool. An article by Taylor, et al. (American Scientist 80 (1992), p. 322-335) describes many of these methods and their applications. For example, Proffitt et. al. (Cytometry 24: 204-213 (1996)) describe a semi-automated fluorescence digital imaging system for quantifying relative cell numbers in situ in a variety of tissue culture plate formats, especially 96-well microtiter plates. The system consists of an epifluorescence inverted microscope with a motorized stage, video camera, image intensifier, and a microcomputer with a PC- Vision digitizer. Turbo Pascal software controls the stage and scans the plate taking multiple images per well. The software calculates total fluorescence per well, provides for daily calibration, and configures easily for a variety of tissue culture plate formats. Thresholding of digital images and reagents which fluoresce only when taken up by living cells are used to reduce background fluorescence without removing excess fluorescent reagent.
Scanning confocal microscope imaging (Go et al., (1997) Analytical Biochemistry 247:210-215; Goldman et al., (1995) Experimental Cell Research 221 :311-319) and multiphoton microscope imaging (Denk et al., (1990) Science 248:73; Gratton et al., (1994) Proc. of the Microscopical Society of America, pp. 154- 155) are also well established methods for acquiring high resolution images of microscopic samples. The principle advantage of these optical systems is the very shallow depth of focus, which allows features of limited axial extent to be resolved against the background. For example, it is possible to resolve internal cytoplasmic features of adherent cells from the features on the cell surface. Because scanning multiphoton imaging requires very short duration pulsed laser systems to achieve the high photon flux required, fluorescence lifetimes can also be measured in these systems (Lakowicz et al, (1992) Anal. Biochem. 202:316-330; Gerrittsen et al. (1997), J. of Fluorescence 7:1 1-15)), providing additional capability for different detection modes. Small, reliable and relatively inexpensive laser systems, such as laser diode pumped lasers, are now available to allow multiphoton confocal microscopy to be applied in a fairly routine fashion.
A combination of the biological heterogeneity of cells in populations (Bright, et al, (1989). J. Cell. Physiol. 141:410; Giuliano, (1996) Cell Motil Cytoskel 35:237)) as well as the high spatial and temporal frequency of chemical and molecular information present within cells, makes it impossible to extract high-content information from populations of cells using existing whole microtiter plate readers. No existing high- content screening platform has been designed for multicolor, fluorescence-based screens using cells that are analyzed individually. Similarly, no method is currently available that combines automated fluid delivery to arrays of cells for the purpose of systematically screening compounds for the ability to induce a cellular response that is identified by HCS analysis, especially from cells grown in microtiter plates. Furthermore, no method exists in the art combining high throughput well-by-well measurements to identify "hits" in one assay followed by a second high content cell-by- cell measurement on the same plate of only those wells identified as hits.
The instant invention provides systems, methods, and screens that combine high throughput screening (HTS) and high content screening (HCS) that significantly improve target validation and candidate optimization by combining many cell screening formats with fluorescence-based molecular reagents and computer-based feature extraction, data analysis, and automation, resulting in increased quantity and speed of data collection, shortened cycle times, and, ultimately, faster evaluation of promising drug candidates. The instant invention also provides for miniaturizing the methods, thereby allowing increased throughput, while decreasing the volumes of reagents and test compounds required in each assay.
SUMMARY OF THE INVENTION
In one aspect, the present invention relates to a method for analyzing cells comprising
• providing cells containing fluorescent reporter molecules in an array of locations, • treating the cells in the array of locations with one or more reagents,
• imaging numerous cells in each location with fluorescence optics,
• converting the optical information into digital data,
• utilizing the digital data to determine the distribution, environment or activity of the fluorescently labeled reporter molecules in the cells and the distribution of the cells, and
• interpreting that information in terms of a positive, negative or null effect of the compound being tested on the biological function
In this embodiment, the method rapidly determines the distribution, environment, or activity of fluorescently labeled reporter molecules in cells for the purpose of screening large numbers of compounds for those that specifically affect particular biological functions. The array of locations may be a microtiter plate or a microchip which is a microplate having cells in an array of locations. In a preferred embodiment, the method includes computerized means for acquiring, processing, displaying and storing the data received. In a preferred embodiment, the method further comprises automated fluid delivery to the arrays of cells. In another preferred embodiment, the information obtained from high throughput measurements on the same plate are used to selectively perform high content screening on only a subset of the cell locations on the plate. In another aspect of the present invention, a cell screening system is provided that comprises:
• a high magnification fluorescence optical system having a microscope objective, • an XY stage adapted for holding a plate containing an array of cells and having a means for moving the plate for proper alignment and focusing on the cell arrays;
• a digital camera;
• a light source having optical means for directing excitation light to cell arrays and a means for directing fluorescent light emitted from the cells to the digital camera; and
• a computer means for receiving and processing digital data from the digital camera wherein the computer means includes a digital frame grabber for receiving the images from the camera, a display for user interaction and display of assay results, digital storage media for data storage and archiving, and a means for control, acquisition, processing and display of results.
In a preferred embodiment, the cell screening system further comprises a computer screen operatively associated with the computer for displaying data. In another preferred embodiment, the computer means for receiving and processing digital data from the digital camera stores the data in a bioinformatics data base. In a further preferred embodiment, the cell screening system further comprises a reader that measures a signal from many or all the wells in parallel. In another preferred embodiment, the cell screening system further comprises a mechanical-optical means for changing the magnification of the system, to allow changing modes between high throughput and high content screening. In another preferred embodiment, the cell screening system further comprises a chamber and control system to maintain the temperature, CO2 concentration and humidity surrounding the plate at levels required to keep cells alive. In a further preferred embodiment, the cell screening system utilizes a confocal scanning illumination and detection system. In another aspect of the present invention, a machine readable storage medium comprising a program containing a set of instructions for causing a cell screening system to execute procedures for defining the distribution and activity of specific cellular constituents and processes is provided. In a preferred embodiment, the cell screening system comprises a high magnification fluorescence optical system with a stage adapted for holding cells and a means for moving the stage, a digital camera, a light source for receiving and processing the digital data from the digital camera, and a computer means for receiving and processing the digital data from the digital camera. Preferred embodiments of the machine readable storage medium comprise programs consisting of a set of instructions for causing a cell screening system to execute the procedures set forth in Figures 9, 11, 12, 13, 14 or 15. Another preferred embodiment comprises a program consisting of a set of instructions for causing a cell screening system to execute procedures for detecting the distribution and activity of specific cellular constituents and processes. In most preferred embodiments, the cellular processes include, but are not limited to, nuclear translocation of a protein, cellular hypertrophy, apoptosis, and protease-induced translocation of a protein.
In another preferred embodiment, a variety of automated cell screening methods are provided, including screens to identify compounds that affect transcription factor activity, protein kinase activity, cell morphology, microtubule structure, apoptosis, receptor internalization, and protease-induced translocation of a protein.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows a diagram of the components of the cell-based scanning system. Figure 2 shows a schematic of the microscope subassembly. Figure 3 shows the camera subassembly. Figure 4 illustrates cell scanning system process.
Figure 5 illustrates a user interface showing major functions to guide the user. Figure 6 is a block diagram of the two platform architecture of the Dual Mode System for Cell Based Screening in which one platform uses a telescope lens to read all wells of a microtiter plate and a second platform that uses a higher magnification lens to read individual cells in a well.
Figure 7 is a detail of an optical system for a single platform architecture of the Dual Mode System for Cell Based Screening that uses a moveable 'telescope' lens to read all wells of a microtiter plate and a moveable higher magnification lens to read individual cells in a well. Figure 8 is an illustration of the fluid delivery system for acquiring kinetic data on the
Cell Based Screening System.
Figure 9 is a flow chart of processing step for the cell-based scanning system. Figure 10 A-J illustrates the strategy of the Nuclear Translocation Assay.
Figure 11 is a flow chart defining the processing steps in the Dual Mode System for
Cell Based Screening combining high throughput and high content screening of microtiter plates.
Figure 12 is a flow chart defining the processing steps in the High Throughput mode of the System for Cell Based Screening.
Figure 13 is a flow chart defining the processing steps in the High Content mode of the
System for Cell Based Screening.
Figure 14 is a flow chart defining the processing steps required for acquiring kinetic data in the High Content mode of the System for Cell Based Screening. Figure 15 is a flow chart defining the processing steps performed within a well during the acquisition of kinetic data.
Figure 16 is an example of data from a known inhibitor of translocation.
Figure 17 is an example of data from a known stimulator of translocation. Figure 18 illustrates data presentation on a graphical display.
Figure 19 is an illustration of the data from the High Throughput mode of the System for Cell Based Screening, an example of the data passed to the High Content mode, the data acquired in the high content mode, and the results of the analysis of that data.
Figure 20 shows the measurement of a drug-induced cytoplasm to nuclear translocation.
Figure 21 illustrates a graphical user interface of the measurement shown in Figure 20.
Figure 22 illustrates a graphical user interface, with data presentation, of the measurement shown in Fig. 20.
Figure 23 is a graph representing the kinetic data obtained from the measurements depicted in Fig. 20.
Figure 24 details a high-content screen of drug-induced apoptosis.
DETAILED DESCRIPTION OF THE INVENTION All cited patents, patent applications and other references are hereby incorporated by reference in their entirety.
As used herein, the following terms have the specified meaning: Markers of cellular domains. Luminescent probes that have high affinity for specific cellular constituents including specific organelles or molecules. These probes can either be small luminescent molecules or fluorescently tagged macromolecules used as "labeling reagents", "environmental indicators", or "biosensors."
Labeling reagents. Labeling reagents include, but are not limited to, luminescently labeled macromolecules including fluorescent protein analogs and biosensors, luminescent macromolecular chimeras including those formed with the green fluorescent protein and mutants thereof, luminescently labeled primary or secondary antibodies that react with cellular antigens involved in a physiological response, luminescent stains, dyes, and other small molecules.
Markers of cellular translocations. Luminescently tagged macromolecules or organelles that move from one cell domain to another during some cellular process or physiological response. Translocation markers can either simply report location relative to the markers of cellular domains or they can also be "biosensors" that report some biochemical or molecular activity as well.
Biosensors. Macromolecules consisting of a biological functional domain and a luminescent probe or probes that report the environmental changes that occur either internally or on their surface. A class of luminescently labeled macromolecules designed to sense and report these changes have been termed "fluorescent-protein biosensors". The protein component of the biosensor provides a highly evolved molecular recognition moiety. A fluorescent molecule attached to the protein component in the proximity of an active site transduces environmental changes into fluorescence signals that are detected using a system with an appropriate temporal and spatial resolution such as the cell scanning system of the present invention. Because the modulation of native protein activity within the living cell is reversible, and because fluorescent-protein biosensors can be designed to sense reversible changes in protein activity, these biosensors are essentially reusable.
Disease associated sequences ("DAS"). This term refers to nucleic acid sequences identified by standard techniques, such as primary DNA sequence data, genomic methods such as subtraction hybridization and RADE, and proteomic methods in combination with reverse genetics, as being of drug candidate compounds. The term does not mean that the sequence is only associated with a disease state.
High content screening (HCS) can be used to measure the effects of drugs on complex molecular events such as signal transduction pathways, as well as cell functions including, but not limited to, apoptosis, cell division, cell adhesion, locomotion, exocytosis, and cell-cell communication. Multicolor fluorescence permits multiple targets and cell processes to be assayed in a single screen. Cross-coπelation of cellular responses will yield a wealth of information required for target validation and lead optimization. In one aspect of the present invention, a cell screening system is provided comprising a high magnification fluorescence optical system having a microscope objective, an XY stage adapted for holding a plate with an array of locations for holding cells and having a means for moving the plate to align the locations with the microscope objective and a means for moving the plate in the direction to effect focusing; a digital camera; a light source having optical means for directing excitation light to cells in the aπay of locations and a means for directing fluorescent light emitted from the cells to the digital camera; and a computer means for receiving and processing digital data from the digital camera wherein the computer means includes: a digital frame grabber for receiving the images from the camera, a display for user interaction and display of assay results, digital storage media for data storage and archiving, and means for control, acquisition, processing and display of results.
Figure 1 is a schematic diagram of a prefeπed embodiment of the cell scanning system. An inverted fluorescence microscope is used J_, such as a Zeiss Axiovert inverted fluorescence microscope which uses standard objectives with magnification of 1-1 OOx to the camera, and a white light source (e.g. 100W mercury-arc lamp or 75 W xenon lamp) with power supply 2. There is an XY stage 3 to move the plate 4 in the XY direction over the microscope objective. A Z-axis focus drive 5 moves the objective in the Z direction for focusing. A joystick 6 provides for manual movement of the stage in the XYZ direction. A high resolution digital camera 7 acquires images from each well or location on the plate. There is a camera power supply 8^ an automation controller 9 and a central processing unit 10. The PC H. provides a display 12 and has associated software. The printer 13 provides for printing of a hard copy record. Figure 2 is a schematic of one embodiment of the microscope assembly 1 of the invention, showing in more detail the XY stage 3, Z-axis focus drive 5, joystick 6, light source 2, and automation controller 9. Cables to the computer 15 and microscope 16, respectively, are provided. In addition, Figure 2 shows a 96 well microtiter plate 17 which is moved on the XY stage 3 in the XY direction. Light from the light source 2 passes through the PC controlled shutter 18 to a motorized filter wheel 19 with excitation filters 20. The light passes into filter cube 25 which has a dichroic minor 26 and an emission filter 27. Excitation light reflects off the dichroic minor to the wells in the microtiter plate 17 and fluorescent light 28 passes through the dichroic minor 26 and the emission filter 27 and to the digital camera 7. Figure 3 shows a schematic drawing of a prefeπed camera assembly. The digital camera 7, which contains an automatic shutter for exposure control and a power supply 3_1, receives fluorescent light 28 from the microscope assembly. A digital cable 30 transports digital signals to the computer. The standard optical configurations described above use microscope optics to directly produce an enlarged image of the specimen on the camera sensor in order to capture a high resolution image of the specimen. This optical system is commonly refeπed to as 'wide field' microscopy. Those skilled in the art of microscopy will recognize that a high resolution image of the specimen can be created by a variety of other optical systems, including, but not limited to, standard scanning confocal detection of a focused point or line of illumination scanned over the specimen (Go et al. 1997, supra), and multi-photon scanning confocal microscopy (Denk et al., 1990, supra), both of which can form images on a CCD detector or by synchronous digitization of the analog output of a photomultiplier tube. In screening applications, it is often necessary to use a particular cell line, or primary cell culture, to take advantage of particular features of those cells. Those skilled in the art of cell culture will recognize that some cell lines are contact inhibited, meaning that they will stop growing when they become suπounded by other cells, while other cell lines will continue to grow under those conditions and the cells will literally pile up, forming many layers. An example of such a cell line is the HEK 293 (ATCC CRL-1573) line. An optical system that can acquire images of single cell layers in multilayer preparations is required for use with cell lines that tend to form layers. The large depth of field of wide field microscopes produces an image that is a projection through the many layers of cells, making analysis of subcellular spatial distributions extremely difficult in layer-forming cells. Alternatively, the very shallow depth of field that can be achieved on a confocal microscope, (about one micron), allows discrimination of a single cell layer at high resolution, simplifying the determination of the subcellular spatial distribution. Similarly, confocal imaging is preferable when detection modes such as fluorescence lifetime imaging are required.
The output of a standard confocal imaging attachment for a microscope is a digital image that can be converted to the same format as the images produced by the other cell screening system embodiments described above, and can therefore be processed in exactly the same way as those images. The overall control, acquisition and analysis in this embodiment is essentially the same. The optical configuration of the confocal microscope system, is essentially the same as that described above, except for the illuminator and detectors. Illumination and detection systems required for confocal microscopy have been designed as accessories to be attached to standard microscope optical systems such as that of the present invention (Zeiss, Germany). These alternative optical systems therefore can be easily integrated into the system as described above.
Figure 4 illustrates an alternative embodiment of the invention in which cell aπays are in micro wells 40 on a microplate 4J_, described ion co-pending U.S. Application S/N 08/865,341, incoφorated by reference herein in its entirety. Typically the microplate is 20 mm by 30 mm as compared to a standard 96 well microtiter plate which is 86 mm by 129 mm. The higher density aπay of cells on a microplate allows the microplate to be imaged at a low resolution of a few microns per pixel for high throughput and particular locations on the microplate to be imaged at a higher resolution of less than 0.5 microns per pixel. These two resolution modes help to improve the overall throughput of the system.
The microplate chamber 42 serves as a microfluidic delivery system for the addition of compounds to cells. The microplate 4 . in the microplate chamber 42 is placed in an XY microplate reader 43. Digital data is processed as described above. The small size of this microplate system increases throughput, minimizes reagent volume and allows control of the distribution and placement of cells for fast and precise cell-based analysis. Processed data can be displayed on a PC screen J_l and made part of a bioinformatics data base 44. This data base not only permits storage and retrieval of data obtained through the methods of this invention, but also permits acquisition and storage of external data relating to cells. Figure 5 is a PC display which illustrates the operation of the software.
In an alternative embodiment, a high throughput system (HTS) is directly coupled with the HCS either on the same platform or on two separate platforms connected electronically (e.g. via a local area network). This embodiment of the invention, referred to as a dual mode optical system, has the advantage of increasing the throughput of a HCS by coupling it with a HTS and thereby requiring slower high resolution data acquisition and analysis only on the small subset of wells that show a response in the coupled HTS.
High throughput 'whole plate' reader systems are well known in the art and are commonly used as a component of an HTS system used to screen large numbers of compounds (Beggs (1997), J. of Biomolec. Screening 2:71-78; Macaffrey et al, (1996) J. Biomolec. Screening 1 :187-190). In one embodiment of dual mode cell based screening, a two platform architecture in which high throughput acquisition occurs on one platform and high content acquisition occurs on a second platform is provided (Figure 6). Processing occurs on each platform independently, with results passed over a network interface, or a single controller is used to process the data from both platforms.
As illustrated in Figure 6, an exemplified two platform dual mode optical system consists of two light optical instruments, a high throughput platform 60 and a high content platform 65^ which read fluorescent signals emitted from cells cultured in microtiter plates or microwell aπays on a microplate, and communicate with each other via an electronic connection 64. The high throughput platform 60 analyzes all the wells in the whole plate either in parallel or rapid serial fashion. Those skilled in the art of screening will recognize that there are a many such commercially available high throughput reader systems that could be integrated into a dual mode cell based screening system (Topcount (Packard Instruments, Meriden, CT); Spectramax, Lumiskan (Molecular Devices, Sunnyvale, CA); Fluoroscan (Labsystems, Beverly, MA)). The high content platform 65, as described above, scans from well to well and acquires and analyzes high resolution image data collected from individual cells within a well.
The HTS software, residing on the system's computer 62, controls the high throughput instrument, and results are displayed on the monitor 6J_. The HCS software, residing on it's computer system 67, controls the high content instrument hardware 65, optional devices (e.g. plate loader, environmental chamber, fluid dispenser), analyzes digital image data from the plate, displays results on the monitor 66 and manages data measured in an integrated database. The two systems can also share a single computer, in which case all data would be collected, processed and displayed on that computer, without the need for a local area network to transfer the data. Microtiter plates are transfened from the high throughput system to the high content system 63 either manually or by a robotic plate transfer device, as is well known in the art (Beggs ( 1997), supra; Mcaffrey ( 1996), supra).
In a prefeπed embodiment, the dual mode optical system utilizes a single platform system (Figure 7). It consists of two separate optical modules, an HCS module 203 and an HTS module 209 that can be independently or collectively moved so that only one at a time is used to collect data from the microtiter plate 201. The microtiter plate 201 is mounted in a motorized X,Y stage so it can be positioned for imaging in either HTS or HCS mode. After collecting and analyzing the HTS image data as described below, the HTS optical module 209 is moved out of the optical path and the HCS optical module 203 is moved into place.
The optical module for HTS 209 consists of a projection lens 214, excitation wavelength filter 213 and dichroic minor 210 which are used to illuminate the whole bottom of the plate with a specific wavelength band from a conventional microscope lamp system (not illustrated). The fluorescence emission is collected through the dichroic minor 210 and emission wavelength filter 21 1 by a lens 212 which forms an image on the camera 216 with sensor 215.
The optical module for HCS 203 consists of a projection lens 208, excitation wavelength filter 207 and dichroic minor 204 which are used to illuminate the back aperture of the microscope objective 202, and thereby the field of that objective, from a standard microscope illumination system (not shown). The fluorescence emission is collected by the microscope objective 202, passes through the dichroic minor 204 and emission wavelength filter 205 and is focused by a tube lens 206 which forms an image on the same camera 216 with sensor 215.
In an alternative embodiment of the present invention, the cell screening system further comprises a fluid delivery device for use with the live cell embodiment of the method of cell screening (see below). Figure 8 exemplifies a fluid delivery device for use with the system of the invention. It consists of a bank of 12 syringe pumps 701 driven by a single motor drive. Each syringe 702 is sized according to the volume to be delivered to each well, typically between 1 and 100 μL. Each syringe is attached via flexible tubing 703 to a similar bank of connectors which accept standard pipette tips
705. The bank of pipette tips are attached to a drive system so they can be lowered and raised relative to the microtiter plate 706 to deliver fluid to each well. The plate is mounted on an X,Y stage, allowing movement relative to the optical system 707 for data collection puφoses. This set-up allows one set of pipette tips, or even a single pipette tip, to deliver reagent to all the wells on the plate. The bank of syringe pumps can be used to deliver fluid to 12 wells simultaneously, or to fewer wells by removing some of the tips.
In another aspect, the present invention provides a method for analyzing cells comprising providing an anay of locations which contain multiple cells wherein the cells contain one or more fluorescent reporter molecules; scanning multiple cells in each of the locations containing cells to obtain fluorescent signals from the fluorescent reporter molecule in the cells; converting the fluorescent signals into digital data; and utilizing the digital data to deteπnine the distribution, environment or activity of the fluorescent reporter molecule within the cells. Cell Arrays
Screening large numbers of compounds for activity with respect to a particular biological function requires preparing anays of cells for parallel handling of cells and reagents. Standard 96 well microtiter plates which are 86 mm by 129 mm, with 6mm diameter wells on a 9mm pitch, are used for compatibility with cunent automated loading and robotic handling systems. The microplate is typically 20 mm by 30 mm, with cell locations that are 100-200 microns in dimension on a pitch of about 500 microns. Methods for making microplates are described in U.S. Patent Application Serial No. 08/865,341, incoφorated by reference herein in its entirety. Microplates may consist of coplanar layers of materials to which cells adhere, patterned with materials to which cells will not adhere, or etched 3-dimensional surfaces of similarly pattered materials. For the puφose of the following discussion, the terms 'well' and 'micro well' refer to a location in an aπay of any construction to which cells adhere and within which the cells are imaged. Microplates may also include fluid delivery channels in the spaces between the wells. The smaller format of a microplate increases the overall efficiency of the system by minimizing the quantities of the reagents, storage and handling during preparation and the overall movement required for the scanning operation. In addition, the whole area of the microplate can be imaged more efficiently, allowing a second mode of operation for the microplate reader as described later in this document.
Fluorescence Reporter Molecules A major component of the new drug discovery paradigm is a continually growing family of fluorescent and luminescent reagents that are used to measure the temporal and spatial distribution, content, and activity of intracellular ions, metabolites, macromolecules, and organelles. Classes of these reagents include labeling reagents that measure the distribution and amount of molecules in living and fixed cells, environmental indicators to report signal transduction events in time and space, and fluorescent protein biosensors to measure target molecular activities within living cells. A multiparameter approach that combines several reagents in a single cell is a powerful new tool for drug discovery. The method of the present invention is based on the high affinity of fluorescent or luminescent molecules for specific cellular components. The affinity for specific components is governed by physical forces such as ionic interactions, covalent bonding (which includes chimeric fusion with protein-based chromophores, fluorophores, and lumiphores), as well as hydrophobic interactions, electrical potential, and, in some cases, simple entrapment within a cellular component. The luminescent probes can be small molecules, labeled macromolecules, or genetically engineered proteins, including, but not limited to green fluorescent protein chimeras.
Those skilled in this art will recognize a wide variety of fluorescent reporter molecules that can be used in the present invention, including, but not limited to, fluorescently labeled biomolecules such as proteins, phospholipids and DNA hybridizing probes. Similarly, fluorescent reagents specifically synthesized with particular chemical properties of binding or association have been used as fluorescent reporter molecules (Barak et al., (1997), J. Biol. Chem. 272:27497-27500; Southwick et al., (1990), Cytometry 1 1 :418-430; Tsien (1989) in Methods in Cell Biology, Vol. 29 Taylor and Wang (eds.), pp. 127-156). Fluorescently labeled antibodies are particularly useful reporter molecules due to their high degree of specificity for attaching to a single molecular target in a mixture of molecules as complex as a cell or tissue.
The luminescent probes can be synthesized within the living cell or can be transported into the cell via several non-mechanical modes including diffusion, facilitated or active transport, signal-sequence-mediated transport, and endocytotic or pinocytotic uptake. Mechanical bulk loading methods, which are well known in the art, can also be used to load luminescent probes into living cells (Barber et al. (1996), Neuroscience Letters 207:17-20; Bright et al. (1996), Cytometry 24:226-233; McNeil (1989) in Methods in Cell Biology, Vol. 29, Taylor and Wang (eds.), pp. 153-173). These methods include electroporation and other mechanical methods such as scrape- loading, bead-loading, impact-loading, syringe-loading, hypertonic and hypotonic loading. Additionally, cells can be genetically engineered to express reporter molecules, such as GFP, coupled to a protein of interest as previously described (Chalfie and Prasher U.S. Patent No. 5,491,084; Cubitt et al. (1995), Trends in Biochemical Science 20:448-455).
Once in the cell, the luminescent probes accumulate at their target domain as a result of specific and high affinity interactions with the target domain or other modes of molecular targeting such as signal-sequence-mediated transport. Fluorescently labeled reporter molecules are useful for determining the location, amount and chemical environment of the reporter. For example, whether the reporter is in a lipophilic membrane environment or in a more aqueous environment can be determined (Giuliano et al. (1995), Ann. Rev. of Biophysics and Biomolecular Structure 24:405-434; Giuliano and Taylor (1995), Methods in Neuroscience 27:1-16). The pH environment of the reporter can be determined (Bright et al. (1989), J. Cell Biology 104:1019-1033; Giuliano et al. (1987), Anal. Biochem. 167:362-371 ; Thomas et al. (1979), Biochemistry 18:2210-2218). It can be determined whether a reporter having a chelating group is bound to an ion, such as Ca++, or not (Bright et al. (1989), In Methods in Cell Biology, Vol. 30, Taylor and Wang (eds.), pp. 157-192; Shimoura et al. (1988), J. of Biochemistry (Tokyo) 251 :405-410; Tsien (1989) In Methods in Cell Biology, Vol. 30, Taylor and Wang (eds.), pp. 127-156).
Furthermore, certain cell types within an organism may contain components that can be specifically labeled that may not occur in other cell types. For example, epithelial cells often contain polarized membrane components. That is, these cells asymmetrically distribute macromolecules along their plasma membrane. Connective or supporting tissue cells often contain granules in which are trapped molecules specific to that cell type (e.g., heparin, histamine, serotonin, etc.). Most muscular tissue cells contain a sarcoplasmic reticulum, a specialized organelle whose function is to regulate the concentration of calcium ions within the cell cytoplasm. Many nervous tissue cells contain secretory granules and vesicles in which are trapped neurohormones or neurotransmitters. Therefore, fluorescent molecules can be designed to label not only specific components within specific cells, but also specific cells within a population of mixed cell types.
Those skilled in the art will recognize a wide variety of ways to measure fluorescence. For example, some fluorescent reporter molecules exhibit a change in excitation or emission spectra, some exhibit resonance energy transfer where one fluorescent reporter loses fluorescence, while a second gains in fluorescence, some exhibit a loss (quenching) or appearance of fluorescence, while some report rotational movements (Giuliano et al. (1995), Ann. Rev. of Biophysics and Biomol Structure 24:405-434; Giuliano et al. (1995), Methods in Neuroscience 27:1-16). Scanning cell arrays
Referring to Figure 9, a prefened embodiment is provided to analyze cells that comprises operator-directed parameters being selected based on the assay being conducted, data acquisition by the cell screening system on the distribution of fluorescent signals within a sample, and interactive data review and analysis. At the start of an automated scan the operator enters information 100 that describes the sample, specifies the filter settings and fluorescent channels to match the biological labels being used and the information sought, and then adjusts the camera settings to match the sample brightness. For flexibility to handle a range of samples, the software allows selection of various parameter settings used to identify nuclei and cytoplasm, and selection of different fluorescent reagents, identification of cells of interest based on moφhology or brightness, and cell numbers to be analyzed per well. These parameters are stored in the system's for easy retrieval for each automated run. The system's interactive cell identification mode simplifies the selection of moφhological parameter limits such as the range of size, shape, and intensity of cells to be analyzed. The user specifies which wells of the plate the system will scan and how many fields or how many cells to analyze in each well. Depending on the setup mode selected by the user at step 101. the system either automatically pre-focuses the region of the plate to be scanned using an autofocus procedure to "find focus" of the plate 102 or the user interactively pre-focuses 103 the scanning region by selecting three "tag" points which define the rectangular area to be scanned. A least-squares fit "focal plane model" is then calculated from these tag points to estimate the focus of each well during an automated scan. The focus of each well is estimated by inteφolating from the focal plane model during a scan.
During an automated scan, the software dynamically displays the scan status, including the number of cells analyzed, the cunent well being analyzed, images of each independent wavelength as they are acquired, and the result of the screen for each well as it is determined. The plate 4 (Figure 1) is scanned in a seφentine style as the software automatically moves the motorized microscope XY stage 3 from well to well and field to field within each well of a 96-well plate. Those skilled in the programming art will recognize how to adapt software for scanning of other microplate formats such as 24, 48, and 384 well plates. The scan pattern of the entire plate as well as the scan pattern of fields within each well are programmed. The system adjusts sample focus with an autofocus procedure 104 (Figure 9) through the Z axis focus drive 5, controls filter selection via a motorized filter wheel 19, and acquires and analyzes images of up to four different colors ("channels" or "wavelengths"). The autofocus procedure is called at a user selected frequency, typically for the first field in each well and then once every 4 to 5 fields within each well. The autofocus procedure calculates the starting Z-axis point by inteφolating from the pre-calculated plane focal model. Starting a programmable distance above or below this set point, the procedure moves the mechanical Z-axis through a number of different positions, acquires an image at each position, and finds the maximum of a calculated focus score that estimates the contrast of each image. The Z position of the image with the maximum focus score determines the best focus for a particular field. Those skilled in the art will recognize this as a variant of automatic focusing methods as described in Harms et al. in Cytometry 5 (1984), 236-243, Groen et al. in Cytometry 6 (1985), 81-91, and Firestone et al. in Cytometry 12 (1991), 195-206.
For image acquisition, the camera's exposure time is separately adjusted for each dye to ensure a high-quality image from each channel. Software procedures can be called, at the user's option, to conect for registration shifts between wavelengths by accounting for linear (X and Y) shifts between wavelengths before making any further measurements. The electronic shutter 18 is controlled so that sample photo-bleaching is kept to a minimum. Background shading and uneven illumination can be coπected by the software using methods known in the art (Bright et al. (1987), J. Cell Biol. 104: 1019-1033).
In one channel, images are acquired of a primary marker 105 (Figure 9) (typically cell nuclei counterstained with DAPI or PI fluorescent dyes) which are segmented ("identified") using an adaptive thresholding procedure. The adaptive thresholding procedure 106 is used to dynamically select the threshold of an image for separating cells from the background. The staining of cells with fluorescent dyes can vary to an unknown degree across cells in a microtiter plate sample as well as within images of a field of cells within each well of a microtiter plate. This variation can occur as a result of sample preparation and/or the dynamic nature of cells. A global threshold is calculated for the complete image to separate the cells from background and account for field to field variation. These global adaptive techniques are variants of those described in the art. (Kittler et al. in Computer Vision, Graphics, and Image Processing 30 (1985), 125-147, Ridler et al. in IEEE Trans. Systems, Man, and Cybernetics (1978), 630-632.) An alternative adaptive thresholding method utilizes local region thresholding in contrast to global image thresholding. Image analysis of local regions leads to better overall segmentation since staining of cell nuclei (as well as other labeled components) can vary across an image. Using this global/local procedure, a reduced resolution image (reduced in size by a factor of 2 to 4) is first globally segmented (using adaptive thresholding) to find regions of interest in the image. These regions then serve as guides to more fully analyze the same regions at full resolution. A more localized threshold is then calculated (again using adaptive thresholding) for each region of interest. The output of the segmentation procedure is a binary image wherein the objects are white and the background is black. This binary image, also called a mask in the art, is used to determine if the field contains objects 107. The mask is labeled with a blob labeling method whereby each object (or blob) has a unique number assigned to it. Moφhological features, such as area and shape, of the blobs are used to differentiate blobs likely to be cells from those that are considered artifacts. The user pre-sets the moφhological selection criteria by either typing in known cell moφhological features or by using the interactive training utility. If objects of interest are found in the field, images are acquired for all other active channels 108, otherwise the stage is advanced to the next field 109 in the cunent well. Each object of interest is located in the image for further analysis 110. The software determines if the object meets the criteria for a valid cell nucleus 111 by measuring its moφhological features (size and shape). For each valid cell, the XYZ stage location is recorded, a small image of the cell is stored, and features are measured 112. The cell scanning method of the present invention can be used to perform many different assays on cellular samples by applying a number of analytical methods simultaneously to measure features at multiple wavelengths. An example of one such assay provides for the following measurements: 1. The total fluorescent intensity within the cell nucleus for colors 1 -4
2. The area of the cell nucleus for color 1 (the primary marker)
3. The shape of the cell nucleus for color 1 is described by three shape features: a) perimeter squared area b) box area ratio c) height width ratio
4. The average fluorescent intensity within the cell nucleus for colors 1-4 (i.e. #1 divided by #2)
5. The total fluorescent intensity of a ring outside the nucleus (see Figure 10) that represents fluorescence of the cell's cytoplasm (cytoplasmic mask) for colors 2-4
6. The area of the cytoplasmic mask
7. The average fluorescent intensity of the cytoplasmic mask for colors 2-4 (i.e. #5 divided by #6) 8. The ratio of the average fluorescent intensity of the cytoplasmic mask to average fluorescent intensity within the cell nucleus for colors 2-4 (i.e. #7 divided by #4)
9. The difference of the average fluorescent intensity of the cytoplasmic mask and the average fluorescent intensity within the cell nucleus for colors 2-4 (i.e. #7 minus #4)
10. The number of fluorescent domains (also call spots, dots, or grains) within the cell nucleus for colors 2-4
Features 1 through 4 are general features of the different cell screening assays of the invention. These steps are commonly used in a variety of image analysis applications and are well known in art (Russ (1992) 77ze Image Processing Handbook,
CRC Press Inc.; Gonzales et al. (1987), Digital Image Processing. Addison- Wesley
Publishing Co. pp. 391-448). Features 5-9 have been developed specifically to provide measurements of a cell's fluorescent molecules within the local cytoplasmic region of the cell and the translocation (i.e. movement) of fluorescent molecules from the cytoplasm to the nucleus. These features (steps 5-9) are used for analyzing cells in microplates for the inhibition of nuclear translocation. For example, inhibition of nuclear translocation of transcription factors provides a novel approach to screening intact cells (detailed examples of other types of screens will be provided below). A specific method measures the amount of probe in the nuclear region (feature 4) versus the local cytoplasmic region (feature 7) of each cell. Quantification of the difference between these two sub-cellular compartments provides a measure of cytoplasm-nuclear translocation (feature 9).
Feature 10 describes a screen used for counting of DNA or RNA probes within the nuclear region in colors 2-4. For example, probes are commercially available for identifying chromosome-specific DNA sequences (Life Technologies, Gaithersburg, MD; Genosys, Woodlands, TX; Biotechnologies, Inc., Richmond, CA; Bio 101, Inc., Vista, CA) Cells are three-dimensional in nature and when examined at a high magnification under a microscope one probe may be in-focus while another may be completely out-of-focus. The cell screening method of the present invention provides for detecting three-dimensional probes in nuclei by acquiring images from multiple focal planes. The software moves the Z-axis motor drive 5 (Figure 1) in small steps where the step distance is user selected to account for a wide range of different nuclear diameters. At each of the focal steps, an image is acquired. The maximum gray-level intensity from each pixel in each image is found and stored in a resulting maximum projection image. The maximum projection image is then used to count the probes. The above method works well in counting probes that are not stacked directly above or below another one. To account for probes stacked on top of each other in the Z- direction, users can select an option to analyze probes in each of the focal planes acquired. In this mode, the scanning system performs the maximum plane projection method as discussed above, detects probe regions of interest in this image, then further analyzes these regions in all the focal plane images.
After measuring cell features 112 (Figure 9), the system checks if there are any unprocessed objects in the cunent field U3. If there are any unprocessed objects, it locates the next object 110 and determines whether it meets the criteria for a valid cell nucleus 11 1, and measures its features. Once all the objects in the cunent field are processed, the system determines whether analysis of the cuπent plate is complete 1 14; if not, it determines the need to find more cells in the cunent well 115. If the need exists, the system advances the XYZ stage to the next field within the cunent well 109 or advances the stage to the next well 1 16 of the plate.
After a plate scan is complete, images and data can be reviewed with the system's image review, data review, and summary review facilities. All images, data, and settings from a scan are archived in the system's database for later review or for interfacing with a network information management system. Data can also be exported to other third-party statistical packages to tabulate results and generate other reports. Users can review the images alone of every cell analyzed by the system with an interactive image review procedure 117. The user can review data on a cell-by-cell basis using a combination of interactive graphs, a data spreadsheet of measured features, and images of all the fluorescence channels of a cell of interest with the interactive cell-by-cell data review procedure 118. Graphical plotting capabilities are provided in which data can be analyzed via interactive graphs such as histograms and scatter plots. Users can review summary data that are accumulated and summarized for all cells within each well of a plate with an interactive well-by-well data review procedure 119. Hard copies of graphs and images can be printed on a wide range of standard printers.
As a final phase of a complete scan, reports can be generated on one or more statistics of the measured features. Users can generate a graphical report of data summarized on a well-by-well basis for the scanned region of the plate using an interactive report generation procedure 120. This report includes a summary of the statistics by well in tabular and graphical format and identification information on the sample. The report window allows the operator to enter comments about the scan for later retrieval. Multiple reports can be generated on many statistics and be printed with the touch of one button. Reports can be previewed for placement and data before being printed.
The above-recited embodiment of the method operates in a single high resolution mode refened to as the high content screening (HCS) mode. The HCS mode provides sufficient spatial resolution within a well (on the order of 1 μm) to define the distribution of material within the well, as well as within individual cells in the well. The high degree of information content accessible in that mode, comes at the expense of speed and complexity of the required signal processing.
In an alternative embodiment, a high throughput system (HTS) is directly coupled with the HCS either on the same platform or on two separate platforms connected electronically (e.g. via a local area network). This embodiment of the invention, refeπed to as a dual mode optical system, has the advantage of increasing the throughput of an HCS by coupling it with an HTS and thereby requiring slower high resolution data acquisition and analysis only on the small subset of wells that show a response in the coupled HTS. High throughput 'whole plate' reader systems are well known in the art and are commonly used as a component of an HTS system used to screen large numbers of compounds (Beggs et al. (1997), supra; McCaffrey et al. (1996), supra ). The HTS of the present invention is carried out on the microtiter plate or microwell anay by reading many or all wells in the plate simultaneously with sufficient resolution to make determinations on a well-by-well basis. That is, calculations are made by averaging the total signal output of many or all the cells or the bulk of the material in each well. Wells that exhibit some defined response in the HTS (the 'hits') are flagged by the system. Then on the same microtiter plate or microwell anay, each well identified as a hit is measured via HCS as described above. Thus, the dual mode process involves:
1. Rapidly measuring numerous wells of a microtiter plate or microwell anay,
2. Inteφreting the data to determine the overall activity of fluorescently labeled reporter molecules in the cells on a well-by-well basis to identify "hits" (wells that exhibit a defined response), 3. Imaging numerous cells in each "hit" well, and
4. Inteφreting the digital image data to determine the distribution, environment or activity of the fluorescently labeled reporter molecules in the individual cells (i.e. intracellular measurements) and the distribution of the cells to test for specific biological functions
In a prefened embodiment of dual mode processing (Figure 11), at the start of a run 301. the operator enters information 302 that describes the plate and its contents, specifies the filter settings and fluorescent channels to match the biological labels being used, the information sought and the camera settings to match the sample brightness. These parameters are stored in the system's database for easy retrieval for each automated run. The microtiter plate or microwell anay is loaded into the cell screening system 303 either manually or automatically by controlling a robotic loading device.
An optional environmental chamber 304 is controlled by the system to maintain the temperature, humidity and CO levels in the air suπounding live cells in the microtiter plate or microwell anay. An optional fluid delivery device 305 (see Figure 8) is controlled by the system to dispense fluids into the wells during the scan.
High throughput processing 306 is first performed on the microtiter plate or microwell anay by acquiring and analyzing the signal from each of the wells in the plate. The processing performed in high throughput mode 307 is illustrated in Figure 12 and described below. Wells that exhibit some selected intensity response in this high throughput mode ("hits") are identified by the system. The system performs a conditional operation 308 that tests for hits. If hits are found, those specific hit wells are further analyzed in high content (micro level) mode 309. The processing performed in high content mode 312 is illustrated in Figure 13. The system then updates 310 the informatics database 31 1 with results of the measurements on the plate. If there are more plates to be analyzed 313 the system loads the next plate 303; otherwise the analysis of the plates terminates 314.
The following discussion describes the high throughput mode illustrated in
Figure 12. The prefened embodiment of the system, the single platfoπn dual mode screening system, will be described. Those skilled in the art will recognize that operationally the dual platform system simply involves moving the plate between two optical systems rather than moving the optics. Once the system has been set up and the plate loaded, the system begins the HTS acquisition and analysis 40L The HTS optical module is selected by controlling a motorized optical positioning device 402 on the dual mode system. In one fluorescence channel, data from a primary marker on the plate is acquired 403 and wells are isolated from the plate background using a masking procedure 404. Images are also acquired in other fluorescence channels being used 405. The region in each image conesponding to each well 406 is measured 407. A feature calculated from the measurements for a particular well is compared with a predefined threshold or intensity response 408, and based on the result the well is either flagged as a "hit" 409 or not. The locations of the wells flagged as hits are recorded for subsequent high content mode processing. If there are wells remaining to be processed 410 the program loops back 406 until all the wells have been processed 411 and the system exits high throughput mode.
Following HTS analysis, the system starts the high content mode processing 501 defined in Figure 13. The system selects the HCS optical module 502 by controlling the motorized positioning system. For each "hit" well identified in high throughput mode, the XY stage location of the well is retrieved from memory or disk and the stage is then moved to the selected stage location 503. The autofocus procedure 504 is called for the first field in each hit well and then once every 5 to 8 fields within each well. In one channel, images are acquired of the primary marker 505 (typically cell nuclei counterstained with DAPI, Hoechst or PI fluorescent dye). The images are then segmented (separated into regions of nuclei and non-nuclei) using an adaptive thresholding procedure 506. The output of the segmentation procedure is a binary mask wherein the objects are white and the background is black. This binary image, also called a mask in the art, is used to determine if the field contains objects 507. The mask is labeled with a blob labeling method whereby each object (or blob) has a unique number assigned to it. If objects are found in the field, images are acquired for all other active channels 508. otherwise the stage is advanced to the next field 514 in the cunent well. Each object is located in the image for further analysis 509. Moφhological features, such as area and shape of the objects, are used to select objects likely to be cell nuclei 510, and discard (do no further processing on) those that are considered artifacts. For each valid cell nucleus, the XYZ stage location is recorded, a small image of the cell is stored, and assay specific features are measured 511. The system then performs multiple tests on the cells by applying several analytical methods to measure features at each of several wavelengths. After measuring the cell features, the systems checks if there are any unprocessed objects in the cunent field 512. If there are any unprocessed objects, it locates the next object 509 and determines whether it meets the criteria for a valid cell nucleus 510, and measures its features. After processing all the objects in the cunent field, the system deteremines whether it needs to find more cells or fields in the cunent well 513. If it needs to find more cells or fields in the cunent well it advances the XYZ stage to the next field within the cunent well 515. Otherwise, the system checks whether it has any remaining hit wells to measure 515. If so, it advances to the next hit well 503 and proceeds through another cycle of acquisition and analysis, otherwise the HCS mode is finished 516.
In an alternative embodiment of the present invention, a method of kinetic live cell screening is provided. The previously described embodiments of the invention are used to characterize the spatial distribution of cellular components at a specific point in time, the time of chemical fixation. As such, these embodiments have limited utility for implementing kinetic based screens, due to the sequential nature of the image acquisition, and the amount of time required to read all the wells on a plate. For example, since a plate can require 30 - 60 minutes to read through all the wells, only very slow kinetic processes can be measured by simply preparing a plate of live cells and then reading through all the wells more than once. Faster kinetic processes can be measured by taking multiple readings of each well before proceeding to the next well, but the elapsed time between the first and last well would be too long, and fast kinetic processes would likely be complete before reaching the last well.
The kinetic live cell extension of the invention enables the design and use of screens in which a biological process is characterized by its kinetics instead of, or in addition to, its spatial characteristics. In many cases, a response in live cells can be measured by adding a reagent to a specific well and making multiple measurements on that well with the appropriate timing. This dynamic live cell embodiment of the invention therefore includes apparatus for fluid delivery to individual wells of the system in order to deliver reagents to each well at a specific time in advance of reading the well. This embodiment thereby allows kinetic measurements to be made with temporal resolution of seconds to minutes on each well of the plate. To improve the overall efficiency of the dynamic live cell system, the acquisition control program is modified to allow repetitive data collection from sub-regions of the plate, allowing the system to read other wells between the time points required for an individual well.
Figure 8 describes an example of a fluid delivery device for use with the live cell embodiment of the invention and is described above. This set-up allows one set of pipette tips 705, or even a single pipette tip, to deliver reagent to all the wells on the plate. The bank of syringe pumps 701 can be used to deliver fluid to 12 wells simultaneously, or to fewer wells by removing some of the tips 705. The temporal resolution of the system can therefore be adjusted, without sacrificing data collection efficiency, by changing the number of tips and the scan pattern as follows. Typically, the data collection and analysis from a single well takes about 5 seconds. Moving from well to well and focusing in a well requires about 5 seconds, so the overall cycle time for a well is about 10 seconds. Therefore, if a single pipette tip is used to deliver fluid to a single well, and data is collected repetitively from that well, measurements can be made with about 5 seconds temporal resolution. If 6 pipette tips are used to deliver fluids to 6 wells simultaneously, and the system repetitively scans all 6 wells, each scan will require 60 seconds, thereby establishing the temporal resolution. For slower processes which only require data collection every 8 minutes, fluids can be delivered to one half of the plate, by moving the plate during the fluid delivery phase, and then repetitively scanning that half of the plate. Therefore, by adjusting the size of the sub- region being scanned on the plate, the temporal resolution can be adjusted without having to insert wait times between acquisitions. Because the system is continuously scanning and acquiring data, the overall time to collect a kinetic data set from the plate is then simply the time to perform a single scan of the plate, multiplied by the number of time points required. Typically, 1 time point before addition of compounds and 2 or 3 time points following addition should be sufficient for screening puφoses.
Figure 14 shows the acquisition sequence used for kinetic analysis. The start of processing 801 is configuration of the system, much of which is identical to the standard HCS configuration. In addition, the operator must enter information specific to the kinetic analysis being performed 802, such as the sub-region size, the number of time points required, and the required time increment. A sub-region is a group of wells that will be scanned repetitively in order to accumulate kinetic data. The size of the sub-region is adjusted so that the system can scan a whole sub-region once during a single time increment, thus minimizing wait times. The optimum sub-region size is calculated from the setup parameters, and adjusted if necessary by the operator. The system then moves the plate to the first sub-region 803, and to the first well in that sub- region 804 to acquire the prestimulation (time = 0) time points. The acquisition sequence performed in each well is exactly the same as that required for the specific HCS being run in kinetic mode. Figure 15 details a flow chart for that processing. All of the steps between the start 901 and the return 902 are identical to those described as steps 504 - 5_14 in Figure 13.
After processing each well in a sub-region, the system checks to see if all the wells in the sub-region have been processed 806 (Figure 14), and cycles through all the wells until the whole region has been processed. The system then moves the plate into position for fluid addition, and controls fluidic system delivery of fluids to the entire sub-region 807. This may require multiple additions for sub-regions which span several rows on the plate, with the system moving the plate on the X,Y stage between additions. Once the fluids have been added, the system moves to the first well in the sub-region 808 to begin acquisition of time points. The data is acquired from each well 809 and as before the system cycles through all the wells in the sub-region 810. After each pass through the sub-region, the system checks whether all the time points have been collected 811 and if not, pauses 813 if necessary 812 to stay synchronized with the requested time increment. Otherwise, the system checks for additional sub-regions on the plate 814 and either moves to the next sub-region 803 or finishes 815. Thus, the kinetic analysis mode comprises operator identification of sub-regions of the microtiter plate or microwells to be screened, based on the kinetic response to be investigated, with data acquisitions within a sub-region prior to data acquisition in subsequent sub- regions.
Specific Screens In another aspect of the present invention, cell screening methods and machine readable storage medium comprising a program containing a set of instructions for causing a cell screening system to execute procedures for defining the distribution and activity of specific cellular constituents and processes is provided. In a prefeπed embodiment, the cell screening system comprises a high magnification fluorescence optical system with a stage adapted for holding cells and a means for moving the stage, a digital camera, a light source for receiving and processing the digital data from the digital camera, and a computer means for receiving and processing the digital data from the digital camera. This aspect of the invention comprises programs that instruct the cell screening system to define the distribution and activity of specific cellular constituents and processes, using the luminescent probes, the optical imaging system, and the pattern recognition software of the invention. Prefeπed embodiments of the machine readable storage medium comprise programs consisting of a set of instructions for causing a cell screening system to execute the procedures set forth in Figures 9, 11, 12, 13, 14 or 15. Another prefeπed embodiment comprises a program consisting of a set of instructions for causing a cell screening system to execute procedures for detecting the distribution and activity of specific cellular constituents and processes. In most prefened embodiments, the cellular processes include, but are not limited to, nuclear translocation of a protein, cellular moφhology, apoptosis, receptor internalization, and protease-induced translocation of a protein.
In a prefened embodiment, the cell screening methods are used to identify compounds that modify the various cellular processes. The cells can be contacted with a test compound, and the effect of the test compound on a particular cellular process can be analyzed. Alternatively, the cells can be contacted with a test compound and a known agent that modifies the particular cellular process, to determine whether the test compound can inhibit or enhance the effect of the known agent. Thus, the methods can be used to identify test compounds that increase or decrease a particular cellular response, as well as to identify test compounds that affects the ability of other agents to increase or decrease a particular cellular response.
In another prefeπed embodiment, the locations containing cells are analyzed using the above methods at low resolution in a high throughput mode, and only a subset of the locations containing cells are analyzed in a high content mode to obtain luminescent signals from the luminescently labeled reporter molecules in subcellular compartments of the cells being analyzed.
The following examples are intended for puφoses of illustration only and should not be construed to limit the scope of the invention, as defined in the claims appended hereto.
The various chemical compounds, reagents, dyes, and antibodies that are refened to in the following Examples are commercially available from such sources as
Sigma Chemical (St. Louis, MO), Molecular Probes (Eugene, OR), Aldrich Chemical
Company (Milwaukee, WI), Accurate Chemical Company (Westbury, NY), Jackson
Immunolabs, and Clontech (Palo Alto, CA).
Example 1 Cytoplasm to Nucleus Translocation Screening: a. Transcription Factors
Regulation of transcription of some genes involves activation of a transcription factor in the cytoplasm, resulting in that factor being transported into the nucleus where it can initiate transcription of a particular gene or genes. This change in transcription factor distribution is the basis of a screen for the cell-based screening system to detect compounds that inhibit or induce transcription of a particular gene or group of genes. A general description of the screen is given followed by a specific example.
The distribution of the transcription factor is determined by labeling the nuclei with a DNA specific fluorophore like Hoechst 33423 and the transcription factor with a specific fluorescent antibody. After autofocusing on the Hoechst labeled nuclei, an image of the nuclei is acquired in the cell-based screening system and used to create a mask by one of several optional thresholding methods, as described supra. The moφhological descriptors of the regions defined by the mask are compared with the user defined parameters and valid nuclear masks are identified and used with the following method to extract transcription factor distributions. Each valid nuclear mask is eroded to define a slightly smaller nuclear region. The original nuclear mask is then dilated in two steps to define a ring shaped region around the nucleus, which represents a cytoplasmic region. The average antibody fluorescence in each of these two regions is determined, and the difference between these averages is defined as the NucCyt
Difference. Two examples of determining nuclear translocation are discussed below and illustrated in Figure 10A-J. Figure 10A illustrates an unstimulated cell with its nucleus 200 labeled with a blue fluorophore and a transcription factor in the cytoplasm
201 labeled with a green fluorophore. Figure 10B illustrates the nuclear mask 202 derived by the cell-based screening system. Figure 10C illustrates the cytoplasm 203 of the unstimulated cell imaged at a green wavelength. Figure 10D illustrates the nuclear mask 202 is eroded (reduced) once to define a nuclear sampling region 204 with minimal cytoplasmic distribution. The nucleus boundary 202 is dilated (expanded) several times to form a ring that is 2-3 pixels wide that is used to define the cytoplasmic sampling region 205 for the same cell. Figure 10E further illustrates a side view which shows the nuclear sampling region 204 and the cytoplasmic sampling region 205. Using these two sampling regions, data on nuclear translocation can be automatically analyzed by the cell-based screening system on a cell by cell basis. Figure 10F-J illustrates the strategy for determining nuclear translocation in a stimulated cell. Figure 10F illustrates a stimulated cell with its nucleus 206 labeled with a blue fluorophore and a transcription factor in the cytoplasm 207 labeled with a green fluorophore. The nuclear mask 208 in Figure 10G is derived by the cell based screening system. Figure 10H illustrates the cytoplasm 209 of a stimulated cell imaged at a green wavelength. Figure 101 illustrates the nuclear sampling region 211 and cytoplasmic sampling region 212 of the stimulated cell. Figure 10 J further illustrates a side view which shows the nuclear sampling region 21 1 and the cytoplasmic sampling region 212.
A specific application of this method has been used to validate this method as a screen. A human cell line was plated in 96 well microtiter plates. Some rows of wells were titrated with IL-1, a known inducer of the NF-KB transcription factor. The cells were then fixed and stained by standard methods with a fluorescein labeled antibody to the transcription factor, and Hoechst 33423. The cell-based screening system was used to acquire and analyze images from this plate and the NucCyt Difference was found to be strongly conelated with the amount of agonist added to the wells as illustrated in
Figure 16. In a second experiment, an antagonist to the receptor for IL-1, IL-1RA was
titrated in the presence of IL-1 α, progressively inhibiting the translocation induced by
IL-lα. The NucCyt Difference was found to strongly conelate with this inhibition of
translocation, as illustrated in Figure 17. Additional experiments have shown that the NucCyt Difference, as well as the NucCyt ratio, gives consistent results over a wide range of cell densities and reagent concentrations, and can therefore be routinely used to screen compound libraries for specific nuclear translocation activity. Furthermore, the same method can be used with antibodies to other transcription factors, or GFP-transcription factor chimeras, or fluorescently labeled transcription factors introduced into living or fixed cells, to screen for effects on the regulation of transcription factor activity.
Figure 18 is a representative display on a PC screen of data which was obtained in accordance with Example 1. Graph 1 180 plots the difference between the average antibody fluorescence in the nuclear sampling region and cytoplasmic sampling region, NucCyt Difference verses Well #. Graph 2 181 plots the average fluorescence of the antibody in the nuclear sampling region, NP 1 average, versus the Well #. Graph 3 182 plots the average antibody fluorescence in the cytoplasmic sampling region, LIP1 average, versus Well #. The software permits displaying data from each cell. For example, Figure 18 shows a screen display 183. the nuclear image 184. and the fluorescent antibody image 185 for cell #26.
NucCyt Difference refened to in graph 1 180 of Figure 18 is the difference between the average cytoplasmic probe (fluorescent reporter molecule) intensity and the average nuclear probe (fluorescent reporter molecule) intensity. NP1 average refened to in graph 2 181 of Figure 18 is the average of cytoplasmic probe (fluorescent reporter molecule) intensity within the nuclear sampling region. L1P1 average refened to in graph 3 182 of Figure 18 is the average probe (fluorescent reporter molecule) intensity within the cytoplasmic sampling region. It will be understood by one of skill in the art that this aspect of the invention can be performed using other transcription factors that translocate from the cytoplasm to the nucleus upon activation. In another specific example, activation of the c-fos transcription factor was assessed by defining its spatial position within cells. Activated c-fos is found only within the nucleus, while inactivated c-fos resides within the cytoplasm. 3T3 cells were plated at 5000-10000 cells per well in a Polyfiltronics 96-well plate. The cells were allowed to attach and grow overnight. The cells were rinsed twice with 100 μl serum-free medium, incubated for 24-30 hours in serum-free MEM culture medium, and then stimulated with platelet derived growth factor (PDGF-BB) (Sigma Chemical Co., St. Louis, MO) diluted directly into serum free medium at concentrations ranging from 1-50 ng/ml for an average time of 20 minutes.
Following stimulation, cells were fixed for 20 minutes in 3.7% formaldehyde solution in IX Hanks buffered saline solution (HBSS). After fixation, the cells were washed with HBSS to remove residual fixative, permeabilized for 90 seconds with 0.5% Triton X-100 solution in HBSS, and washed twice with HBSS to remove residual detergent. The cells were then blocked for 15 minutes with a 0.1% solution of BSA in HBSS, and further washed with HBSS prior to addition of diluted primary antibody solution. c-Fos rabbit polyclonal antibody (Calbiochem, PC05) was diluted 1 :50 in
HBSS, and 50 μl of the dilution was applied to each well. Cells were incubated in the presence of primary antibody for one hour at room temperature, and then incubated for one hour at room temperature in a light tight container with goat anti-rabbit secondary antibody conjugated to ALEXA™ 488 (Molecular Probes), diluted 1 :500 from a 100 μg/ml stock in HBSS. Hoechst DNA dye (Molecular Probes) was then added at a 1 :1000 dilution of the manufacturer's stock solution (10 mg/ml). The cells were then washed with HBSS, and the plate was sealed prior to analysis with the cell screening system of the invention. The data from these experiments demonstrated that the methods of the invention could be used to measure transcriptional activation of c-fos by defining its spatial position within cells.
One of skill in the art will recognize that while the following method is applied to detection of c-fos activation, it can be applied to the analysis of any transcription factor that translocates from the cytoplasm to the nucleus upon activation. Examples of such transcription factors include, but are not limited to fos and jun homologs, NF-KB (nuclear factor kappa from B cells), NFAT (nuclear factor of activated T-lymphocytes), and STATs (signal transducer and activator of transcription) factors (For example, see Strehlow, I., and Schindler, C. 1998. J. Biol. Chem. 273:28049-28056; Chow, et al. 1997 Science. 278:1638-1641; Ding et al. 1998 J. Biol. Chem. 273:28897-28905; Baldwin, 1996. Annu Rev Immunol. 14:649-83; Kuo, C.T., and J.M. Leiden. 1999. Annu Rev Immunol. 17:149-87; Rao, et al. 1997. Annu Rev Immunol. 15:707-47; Masuda,et al. 1998. Cell Signal. 10:599-611; Hoey, T., and U. Schindler. 1998. Curr Opin Genet Dev. 8:582-7; Liu, et al. 1998. Curr Opin Immunol. 10:271-8.)
Thus, in this aspect of the invention, indicator cells are treated with test compounds and the distribution of luminescently labeled transcription factor is measured in space and time using a cell screening system, such as the one disclosed above. The luminescently labeled transcription factor may be expressed by or added to the cells either before, together with, or after contacting the cells with a test compound. For example, the transcription factor may be expressed as a luminescently labeled protein chimera by transfected indicator cells. Alternatively, the luminescently labeled transcription factor may be expressed, isolated, and bulk-loaded into the indicator cells as described above, or the transcription factor may be luminescently labeled after isolation. As a further alternative, the transcription factor is expressed by the indicator cell, which is subsequently contacted with a luminescent label, such as an antibody, that detects the transcription factor.
In a further aspect, kits are provided for analyzing transcription factor activation, comprising an antibody that specifically recognizes a transcription factor of interest, and instructions for using the antibody for carrying out the methods described above. In a prefened embodiment, the transcription factor-specific antibody, or a secondary antibody that detects the transcription factor antibody, is luminescently labeled. In further prefeπed embodiments, the kit contains cells that express the transcription factor of interest, and/or the kit contains a compound that is known to modify activation of the transcription factor of interest, including but not limited to platelet derived growth factor (PDGF) and serum, which both modify fos activation; and interleukin l(IL-l) and tumor necrosis factor (TNF), which both modify NF-KB activation.
In another embodiment, the kit comprises a recombinant expression vector comprising a nucleic acid encoding a transcription factor of interest that translocates from the cytoplasm to the nucleus upon activation, and instructions for using the expression vector to identify compounds that modify transcription factor activation in a cell of interest. Alternatively, the kits contain a purified, luminescently labeled transcription factor. In a prefeπed embodiment, the transcription factor is expressed as a fusion protein with a luminescent protein, including but not limited to green fluorescent protein, luceriferase, or mutants or fragments thereof. In various prefened embodiments, the kit further contains cells that are transfected with the expression vector, an antibody or fragment that specifically bind to the transcription factor of interest, and/or a compound that is known to modify activation of the transcription factor of interest (as above).
b. Protein Kinases
The cytoplasm to nucleus screening methods can also be used to analyze the activation of any protein kinase that is present in an inactive state in the cytoplasm and is transported to the nucleus upon activation, or that phosphorylates a substrate that translocates from the cytoplasm to the nucleus upon phosphorylation. Examples of appropriate protein kinases include, but are not limited to extracellular signal-regulated protein kinases (ERKs), c-Jun amino-terminal kinases (JNKs), Fos regulating protein kinases (FRKs), p38 mitogen activated protein kinase (p38MAPK), protein kinase A (PKA), and mitogen activated protein kinase kinases (MAPKKs). (For example, see Hall, et al. 1999. J Biol Chem. 274:376-83; Han, et al. 1995. Biochim. Biophys. Acta. 1265:224-227; Jaaro et al. 1997. Proc. Natl. Acad. Sci. U.S.A. 94:3742-3747; Taylor, et al. 1994. J. Biol Chem. 269:308-318; Zhao, Q., and F. S. Lee. 1999. J Biol Chem. 274:8355-8; Paolilloet al. 1999. J Biol Chem. 274:6546-52; Coso et al. 1995. Cell 81:1137-1146; Tibbies, L.A., and J.R. Woodgett. 1999. Cell Mol Life Sci. 55:1230-54; Schaeffer, H.J., and M.J. Weber. 1999. Mol Cell Biol. 19:2435-44.)
Alternatively, protein kinase activity is assayed by monitoring translocation of a luminescently labeled protein kinase substrate from the cytoplasm to the nucleus after being phosphorylated by the protein kinase of interest. In this embodiment, the substrate is non-phosphorylated and cytoplasmic prior to phosphorylation, and is translocated to the nucleus upon phosphorylation by the protein kinase. There is no requirement that the protein kinase itself translocates from the cytoplasm to the nucleus in this embodiment. Examples of such substrates (and the conesponding protein kinase) include, but are not limited to c-jun (JNK substrate); fos (FRK substrate), and p38 (p38 MAPK substrate).
Thus, in these embodiments, indicator cells are treated with test compounds and the distribution of luminescently labeled protein kinase or protein kinase substrate is measured in space and time using a cell screening system, such as the one disclosed above. The luminescently labeled protein kinase or protein kinase substrate may be expressed by or added to the cells either before, together with, or after contacting the cells with a test compound. For example, the protein kinase or protein kinase substrate may be expressed as a luminescently labeled protein chimera by transfected indicator cells. Alternatively, the luminescently labeled protein kinase or protein kinase substrate may be expressed, isolated, and bulk-loaded into the indicator cells as described above, or the protein kinase or protein kinase substrate may be luminescently labeled after isolation. As a further alternative, the protein kinase or protein kinase substrate is expressed by the indicator cell, which is subsequently contacted with a luminescent label, such as a labeled antibody, that detects the protein kinase or protein kinase substrate.
In a further embodiment, protein kinase activity is assayed by monitoring the phosphorylation state (ie: phosphorylated or not phosphorylated) of a protein kinase substrate. In this embodiment, there is no requirement that either the protein kinase or the protein kinase substrate translocate from the cytoplasm to the nucleus upon activation. In a prefened embodiment, phosphorylation state is monitored by contacting the cells with an antibody that binds only to the phosphorylated form of the protein kinase substrate of interest (For example, as disclosed in U.S. Patent No. 5,599,681). In another prefeπed embodiment, a biosensor of phosphorylation is used. For example, a luminescently labeled protein or fragment thereof can be fused to a protein that has been engineered to contain (a) a phosphorylation site that is recognized by a protein kinase of interest; and (b) a nuclear localization signal that is unmasked by the phosphorylation. Such a biosensor will thus be translocated to the nucleus upon phosphorylation, and its translocation can be used as a measure of protein kinase activation.
In another aspect, kits are provided for analyzing protein kinase activation, comprising a primary antibody that specifically binds to a protein kinase, a protein kinase substrate, or a phosphorylated form of the protein kinase substrate of interest and instructions for using the primary antibody to identify compounds that modify protein kinase activation in a cell of interest. In a prefened embodiment, the primary antibody, or a secondary antibody that detects the primary antibody, is luminescently labeled. In other prefened embodiments, the kit further comprises cells that express the protein kinase of interest, and/or a compound that is known to modify activation of the protein kinase of interest, including but not limited to dibutyryl cAMP (modifies PKA), forskolin (PKA), and anisomycin (p38MAPK).
Alternatively, the kits comprise an expression vector encoding a protein kinase or a protein kinase substrate of interest that translocates from the cytoplasm to the nucleus upon activation and instructions for using the expression vector to identify compounds that modify protein kinase activation in a cell of interest. Alternatively, the kits contain a purified, luminescently labeled protein kinase or protein kinase substrate. In a prefeπed embodiment, the protein kinase or protein kinase substrate of interest is expressed as a fusion protein with a luminescent protein. In further prefeπed embodiments, the kit further comprises cells that are transfected with the expression vector, an antibody or fragment thereof that specifically binds to the protein kinase or protein kinase substrate of interest, and/or a compound that is known to modify activation of the protein kinase of interest, (as above)
In another aspect, the present invention comprises a machine readable storage medium comprising a program containing a set of instructions for causing a cell screening system to execute the methods disclosed for analyzing transcription factor or protein kinase activation, wherein the cell screening system comprises an optical system with a stage adapted for holding a plate containing cells, a digital camera, a means for directing fluorescence or luminescence emitted from the cells to the digital camera, and a computer means for receiving and processing the digital data from the digital camera.
Example 2 Automated Screen for Compounds that Modify Cellular Morphology
Changes in cell size are associated with a number of cellular conditions, such as hypertrophy, cell attachment and spreading, differentiation, growth and division, necrotic and programmed cell death, cell motility, moφhogenesis, tube formation, and colony formation. For example, cellular hypertrophy has been associated with a cascade of alterations in gene expression and can be characterized in cell culture by an alteration in cell size, that is clearly visible in adherent cells growing on a coverslip.
Cell size can also be measured to determine the attachment and spreading of adherent cells. Cell spreading is the result of selective binding of cell surface receptors to substrate ligands and subsequent activation of signaling pathways to the cytoskeleton. Cell attachment and spreading to substrate molecules is an important step for the metastasis of cancer cells, leukocyte activation during the inflammatory response, keratinocyte movement during wound healing, and endothelial cell movement during angiogenesis. Compounds that affect these surface receptors, signaling pathways, or the cytoskeleton will affect cell spreading and can be screened by measuring cell size.
Total cellular area can be monitored by labeling the entire cell body or the cell cytoplasm using cytoskeletal markers, cytosolic volume markers, or cell surface markers, in conjunction with a DNA label. Examples of such labels (many available from Molecular Probes (Eugene, Oregon) and Sigma Chemical Co. (St. Louis, Missouri)) include the following:
CELL SIZE AND AREA MARKERS
Cytoskeletal Markers
ALEXA'M 488 phalloidin (Molecular Probes, Oregon)
Tubulin-green fluorescent protein chimeras
Cytokeratin-green fluorescent protein chimeras
Antibodies to cytoskeletal proteins
Cytosolic Volume Markers
Green fluorescent proteins
Chloromethylfluorescein diacetate (CMFDA)
Calcein green
BCECF/AM ester
Rhodamine dextran
Cell Surface Markers for Lipid, Protein, or Oligosaccharide
• Dihexadecyl tetramethylindocarbocyanine perchlorate (DilC 16) lipid dyes Triethylammonium propyl dibutylamino styryl pyridinium (FM 4-64, FM 1-43) lipid dyes
MITOTRACKER™~Green FM
Lectins to oligosaccarides such as fluorescein concanavalin A or wheat germ agglutinin
SYPRO Red non-specific protein markers
Antibodies to various surface proteins such as epidermal growth factor
Biotin labeling of surface proteins followed by fluorescent strepavidin labeleing
Protocols for cell staining with these various agents are well known to those skilled in the art. Cells are stained live or after fixation and the cell area can be measured. For example, live cells stained with DiIC16 have homogeneously labeled plasma membranes, and the projected cross-sectional area of the cell is uniformly discriminated from background by fluorescence intensity of the dye. Live cells stained with cytosolic stains such as CMFDA produce a fluorescence intensity that is proportional to cell thickness. Although cell labeling is dimmer in thin regions of the cell, total cell area can be discriminated from background. Fixed cells can be stained with cytoskeletal markers such as ALEXA™ 488 phalloidin that label polymerized actin. Phalloidin does not homogeneously stain the cytoplasm, but still permits discrimination of the total cell area from background.
Cellular hypertrophy A screen to analyze cellular hypertrophy is implemented using the following strategy. Primary rat myocytes can be cultured in 96 well plates, treated with various compounds and then fixed and labeled with a fluorescent marker for the cell membrane or cytoplasm, or cytoskeleton, such as an antibody to a cell surface marker or a fluorescent marker for the cytoskeleton like rhodamine-phalloidin, in combination with a DNA label like Hoechst. After focusing on the Hoechst labeled nuclei, two images are acquired, one of the Hoechst labeled nuclei and one of the fluorescent cytoplasm image. The nuclei are identified by thresholding to create a mask and then comparing the moφhological descriptors of the mask with a set of user defined descriptor values. Each non-nucleus image (or "cytoplasmic image") is then processed separately. The original cytoplasm image can be thresholded, creating a cytoplasmic mask image. Local regions containing cells are defined around the nuclei. The limits of the cells in those regions are then defined by a local dynamic threshold operation on the same region in the fluorescent antibody image. A sequence of erosions and dilations is used to separate slightly touching cells and a second set of moφhological descriptors is used to identify single cells. The area of the individual cells is tabulated in order to define the distribution of cell sizes for comparison with size data from normal and hypertrophic cells.
Responses from entire 96-well plates (measured as average cytoplasmic area/cell) were analyzed by the above methods, and the results demonstrated that the assay will perform the same on a well-to-well, plate-to-plate, and day-to-day basis
(below a 15% cov for maximum signal). The data showed very good coπelation for each day, and that there was no variability due to well position in the plate.
The following totals can be computed for the field. The aggregate whole nucleus area is the number of nonzero pixels in the nuclear mask. The average whole nucleus area is the aggregate whole nucleus area divided by the total number of nuclei. For each cytoplasm image several values can be computed. These are the total cytoplasmic area, which is the count of nonzero pixels in the cytoplasmic mask. The aggregate cytoplasm intensity is the sum of the intensities of all pixels in the cytoplasmic mask. The cytoplasmic area per nucleus is the total cytoplasmic area divided by the total nucleus count. The cytoplasmic intensity per nucleus is the aggregate cytoplasm intensity divided by the total nucleus count. The average cytoplasm intensity is the aggregate cytoplasm intensity divided by the cytoplasm area.
The cytoplasm nucleus ratio is the total cytoplasm area divided by the total nucleus area.
Additionally, one or more fluorescent antibodies to other cellular proteins, such as the major muscle proteins actin or myosin, can be included. Images of these additional labeled proteins can be acquired and stored with the above images, for later review, to identify anomalies in the distribution and moφhology of these proteins in hypertrophic cells. This example of a multi-parametric screen allows for simultaneous analysis of cellular hypertrophy and changes in actin or myosin distribution.
One of skill in the art will recognize that while the example analyzes myocyte hypertrophy, the methods can be applied to analyzing hypertrophy, or general moφhological changes in any cell type.
Cell morphology assays for prostate carcinoma
Cell spreading is a measure of the response of cell surface receptors to substrate attachment ligands. Spreading is proportional to the ligand concentration or to the concentration of compounds that reduce receptor-ligand function. One example of selective cell-substrate attachment is prostate carcinoma cell adhesion to the extracellular matrix protein collagen. Prostate carcinoma cells metastasize to bone via selective adhesion to collagen.
Compounds that interfere with metastasis of prostate carcinoma cells were screened as follows. PC3 human prostate carcinoma cells were cultured in media with appropriate stimulants and are passaged to collagen coated 96 well plates. Ligand concentration can be varied or inhibitors of cell spreading can be added to the wells. Examples of compounds that can affect spreading are receptor antagonists such as integrin- or proteoglycan-blocking antibodies, signaling inhibitors including phosphatidyl inositol-3 kinase inhibitors, and cytoskeletal inhibitors such as cytochalasin D. After two hours, cells were fixed and stained with ALEXA™ 488 phalloidin (Molecular Probes) and Hoechst 33342 as per the protocol for cellular hypertrophy. The size of cells under these various conditions, as measured by cytoplasmic staining, can be distinguished above background levels. The number of cells per field is determined by measuring the number of nuclei stained with the Hoechst DNA dye. The area per cell is found by dividing the cytoplasmic area (phalloidin image) by the cell number (Hoechst image). The size of cells is proportional to the ligand-receptor function. Since the area is determined by ligand concentration and by the resultant function of the cell, drug efficacy, as well as drug potency, can be determined by this cell-based assay. Other measurements can be made as discussed above for cellular hypertrophy.
The methods for analyzing cellular moφhology can be used in a combined high throughput-high content screen. In one example, the high throughput mode scans the whole well for an increase in fluorescent phalloidin intensity. A threshold is set above which both nuclei (Hoechst) and cells (phalloidin) are measured in a high content mode. In another example, an environmental biosensor (examples include, but are not limited to, those biosensors that are sensitive to calcium and pH changes) is added to the cells, and the cells are contacted with a compound. The cells are scanned in a high throughput mode, and those wells that exceed a pre-determined threshold for luminescence of the biosensor are scanned in a high content mode.
In a further aspect, kits are provided for analyzing cellular moφhology, comprising a luminescent compound that can be used to specifically label the cell cytoplasm, membrane, or cytoskeleton (such as those described above), and instructions for using the luminescent compound to identify test stimuli that induce or inhibit changes in cellular moφhology according to the above methods. In a prefened embodiment, the kit further comprises a luminescent marker for cell nuclei. In a further prefened embodiment, the kit comprises at least one compound that is known to modify cellular moφhology, including, but not limited to integrin- or proteoglycan- blocking antibodies, signaling inhibitors including phosphatidyl inositol-3 kinase inhibitors, and cytoskeletal inhibitors such as cytochalasin D.
In another aspect, the present invention comprises a machine readable storage medium comprising a program containing a set of instructions for causing a cell screening system to execute the disclosed methods for analyzing cellular moφhology, wherein the cell screening system comprises an optical system with a stage adapted for holding a plate containing cells, a digital camera, a means for directing fluorescence or luminescence emitted from the cells to the digital camera, and a computer means for receiving and processing the digital data from the digital camera.
Example 3 Dual Mode High Throughput and High-Content Screen
The following example is a screen for activation of a G-protein coupled receptor
(GPCR) as detected by the translocation of the GPCR from the plasma membrane to a proximal nuclear location. This example illustrates how a high throughput screen can be coupled with a high-content screen in the dual mode System for Cell Based Screening.
G-protein coupled receptors are a large class of 7 trans-membrane domain cell surface receptors. Ligands for these receptors stimulate a cascade of secondary signals in the cell, which may include, but are not limited to, Ca++ transients, cyclic AMP production, inositol triphosphate (IP ) production and phosphorylation. Each of these signals are rapid, occuring in a matter of seconds to minutes, but are also generic. For example, many different GPCRs produce a secondary Ca++ signal when activated. Stimulation of a GPCR also results in the transport of that GPCR from the cell surface membrane to an internal, proximal nuclear compartment. This intemalization is a much more receptor-specific indicator of activation of a particular receptor than are the secondary signals described above.
Figure 19 illustrates a dual mode screen for activation of a GPCR. Cells carrying a stable chimera of the GPCR with a blue fluorescent protein (BFP) would be loaded with the acetoxymethyl ester form of Fluo-3, a cell permeable calcium indicator (green fluorescence) that is trapped in living cells by the hydrolysis of the esters. They would then be deposited into the wells of a microtiter plate 601. The wells would then be treated with an anay of test compounds using a fluid delivery system, and a short sequence of Fluo-3 images of the whole microtiter plate would be acquired and analyzed for wells exhibiting a calcium response (i.e., high throughput mode). The images would appear like the illustration of the microtiter plate 601 in Figure 19. A small number of wells, such as wells C4 and E9 in the illustration, would fluoresce more brightly due to the Ca++ released upon stimulation of the receptors. The locations of wells containing compounds that induced a response 602, would then be transfened to the HCS program and the optics switched for detailed cell by cell analysis of the blue fluorescence for evidence of GPCR translocation to the perinuclear region. The bottom of Figure 19 illustrates the two possible outcomes of the analysis of the high resolution cell data. The camera images a sub-region 604 of the well area 603, producing images of the fluorescent cells 605. In well C4, the uniform distribution of the fluorescence in the cells indicates that the receptor has not internalized, implying that the Ca++ response seen was the result of the stimulation of some other signalling system in the cell. The cells in well E9 606 on the other hand, clearly indicate a concentration of the receptor in the perinuclear region clearly indicating the full activation of the receptor. Because only a few hit wells have to be analyzed with high resolution, the overall throughput of the dual mode system can be quite high, comparable to the high throughput system alone.
Example 4 Kinetic High Content Screen The following is an example of a screen to measure the kinetics of intemalization of a receptor. As described above, the stimulation of a GPCR, results in the intemalization of the receptor, with a time course of about 15 min. Simply detecting the endpoint as internalized or not, may not be sufficient for defining the potency of a compound as a GPCR agonist or antagonist. However, 3 time points at 5 min intervals would provide information not only about potency during the time course of measurement, but would also allow extrapolation of the data to much longer time periods. To perform this assay, the sub-region would be defined as two rows, the sampling interval as 5 minutes and the total number of time points 3. The system would then start by scanning two rows, and then adding reagent to the two rows, establishing the time=0 reference. After reagent addition, the system would again scan the two row sub-region acquiring the first time point data. Since this process would take about 250 seconds, including scanning back to the beginning of the sub-region, the system would wait 50 seconds to begin acquisition of the second time point. Two more cycles would produce the three time points and the system would move on to the second 2 row sub-region. The final two 2-row sub-regions would be scanned to finish all the wells on the plate, resulting in four time points for each well over the whole plate. Although the time points for the wells would be offset slightly relative to time=0, the spacing of the time points would be very close to the required 5 minutes, and the actual acquisition times and results recorded with much greater precision than in a fixed-cell screen.
Example 5 High-content screen of human glucocorticoid receptor translocation
One class of HCS involves the drug-induced dynamic redistribution of intracellular constituents. The human glucocorticoid receptor (hGR), a single "sensor" in the complex environmental response machinery of the cell, binds steroid molecules that have diffused into the cell. The ligand-receptor complex translocates to the nucleus where transcriptional activation occurs (Htun et al., Proc. Natl. Acad. Sci. 93:4845, 1996). In general, hormone receptors are excellent drug targets because their activity lies at the apex of key intracellular signaling pathways. Therefore, a high-content screen of hGR translocation has distinct advantage over in vitro ligand-receptor binding assays. The availability of up to two more channels of fluorescence in the cell screening system of the present invention permits the screen to contain two additional parameters in parallel, such as other receptors, other distinct targets or other cellular processes.
Plasmid construct. A eukaryotic expression plasmid containing a coding sequence for a green fluorescent protein - human glucocorticoid receptor (GFP-hGR) chimera was prepared using GFP mutants (Palm et al., Nat. Struct. Biol. 4:361 (1997). The construct was used to transfect a human cervical carcinoma cell line (HeLa).
Cell preparation and transfection. HeLa cells (ATCC CCL-2) were trypsinized and plated using DMEM containing 5% charcoal/dextran-treated fetal bovine serum (FBS) (HyClone) and 1% penicillin-streptomycin (C-DMEM) 12-24 hours prior to transfection and incubated at 37°C and 5% CO . Transfections were performed by calcium phosphate co-precipitation (Graham and Van der Eb, Virology 52:456, 1973; Sambrook et al., (1989). Molecular Cloning: A Laboratory Manual, Second ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 1989) or with Lipofectamine (Life Technologies, Gaithersburg, MD). For the calcium phosphate transfections, the medium was replaced, prior to transfection, with DMEM containing 5% charcoal/dextran-treated FBS. Cells were incubated with the calcium phosphate-DΝA precipitate for 4-5 hours at 37°C and 5% CO2, washed 3-4 times with DMEM to remove the precipitate, followed by the addition of C-DMEM.
Lipofectamine transfections were performed in serum-free DMEM without antibiotics according to the manufacturer's instructions (Life Technologies, Gaithersburg, MD). Following a 2-3 hour incubation with the DΝA-liposome complexes, the medium was removed and replaced with C-DMEM. All transfected cells in 96-well microtiter plates were incubated at 33°C and 5% CO2 for 24-48 hours prior to drug treatment. Experiments were performed with the receptor expressed transiently in HeLa cells.
Dexamethasone induction of GFP-hGR translocation. To obtain receptor- ligand translocation kinetic data, nuclei of transfected cells were first labeled with 5 μg/ml Hoechst 33342 (Molecular Probes) in C-DMEM for 20 minutes at 33°C and 5% CO2. Cells were washed once in Hank's Balanced Salt Solution (HBSS) followed by the addition of 100 nM dexamethasone in HBSS with 1% charcoal/dextran-treated FBS. To obtain fixed time point dexamethasone titration data, transfected HeLa cells
were first washed with DMEM and then incubated at 33°C and 5% CO2 for 1 h in the presence of 0 - 1000 nM dexamethasone in DMEM containing 1% charcoal/dextran- treated FBS. Cells were analyzed live or they were rinsed with HBSS, fixed for 15 min with 3.7% formaldehyde in HBSS, stained with Hoechst 33342, and washed before analysis. The intracellular GFP-hGR fluorescence signal was not diminished by this fixation procedure. Image acquisition and analysis. Kinetic data were collected by acquiring fluorescence image pairs (GFP-hGR and Hoechst 33342-labeled nuclei) from fields of living cells at 1 min intervals for 30 min after the addition of dexamethasone. Likewise, image pairs were obtained from each well of the fixed time point screening plates 1 h after the addition of dexamethasone. In both cases, the image pairs obtained at each time point were used to define nuclear and cytoplasmic regions in each cell. Translocation of GFP-hGR was calculated by dividing the integrated fluorescence intensity of GFP-hGR in the nucleus by the integrated fluorescence intensity of the chimera in the cytoplasm or as a nuclear-cytoplasmic difference of GFP fluorescence. In the fixed time point screen this translocation ratio was calculated from data obtained from at least 200 cells at each concentration of dexamethasone tested. Drug-induced translocation of GFP-hGR from the cytoplasm to the nucleus was therefore conelated with an increase in the translocation ratio.
Results. Figure 20 schematically displays the drag-induced cytoplasm 253 to nucleus 252 translocation of the human glucocorticoid receptor. The upper pair of schematic diagrams depicts the localization of GFP-hGR within the cell before 250 (A) and after 251 (B) stimulation with dexamethasone. Under these experimental conditions, the drug induces a large portion of the cytoplasmic GFP-hGR to translocate into the nucleus. This redistribution is quantified by determining the integrated intensities ratio of the cytoplasmic and nuclear fluorescence in treated 255 and untreated 254 cells. The lower pair of fluorescence micrographs show the dynamic redistribution of GFP-hGR in a single cell, before 254 and after 255 treatment. The HCS is performed on wells containing hundreds to thousands of transfected cells and the translocation is quantified for each cell in the field exhibiting GFP fluorescence. Although the use of a stably transfected cell line would yield the most consistently labeled cells, the heterogeneous levels of GFP-hGR expression induced by transient transfection did not interfere with analysis by the cell screening system of the present invention.
To execute the screen, the cell screening system scans each well of the plate, images a population of cells in each, and analyzes cells individually. Here, two channels of fluorescence are used to define the cytoplasmic and nuclear distribution of the GFP-hGR within each cell. Depicted in Figure 21 is the graphical user interface of the cell screening system near the end of a GFP-hGR screen. The user interface depicts the parallel data collection and analysis capability of the system. The windows labeled "Nucleus" 261 and "GFP-hGR" 262 show the pair of fluorescence images being obtained and analyzed in a single field. The window labeled "Color Overlay" 260 is formed by pseudocoloring the above images and merging them so the user can immediately identify cellular changes. Within the "Stored Object Regions" window 265, an image containing each analyzed cell and its neighbors is presented as it is archived. Furthermore, as the HCS data are being collected, they are analyzed, in this case for GFP-hGR translocation, and translated into an immediate "hit" response. The
96 well plate depicted in the lower window of the screen 267 shows which wells have met a set of user-defined screening criteria. For example, a white-colored well 269 indicates that the drag-induced translocation has exceeded a predetermined threshold value of 50%. On the other hand, a black-colored well 270 indicates that the drag being tested induced less than 10% translocation. Gray-colored wells 268 indicate "hits" where the translocation value fell between 10% and 50%. Row "E" on the 96 well plate being analyzed 266 shows a titration with a drag known to activate GFP-hGR translocation, dexamethasone. This example screen used only two fluorescence channels. Two additional channels (Channels 3 263 and 4 264) are available for parallel analysis of other specific targets, cell processes, or cytotoxicity to create multiple parameter screens.
There is a link between the image database and the information database that is a powerful tool during the validation process of new screens. At the completion of a screen, the user has total access to image and calculated data (Figure 22). The comprehensive data analysis package of the cell screening system allows the user to examine HCS data at multiple levels. Images 276 and detailed data in a spread sheet 279 for individual cells can be viewed separately, or summary data can be plotted. For example, the calculated results of a single parameter for each cell in a 96 well plate are shown in the panel labeled Graph 1 275. By selecting a single point in the graph, the user can display the entire data set for a particular cell that is recalled from an existing database. Shown here are the image pair 276 and detailed fluorescence and moφhometric data from a single cell (Cell #118, gray line 277). The large graphical insert 278 shows the results of dexamethasone concentration on the translocation of GFP-hGR. Each point is the average of data from at least 200 cells. The calculated EC50 for dexamethasone in this assay is 2 nM.
A powerful aspect of HCS with the cell screening system is the capability of kinetic measurements using multicolor fluorescence and moφhometric parameters in living cells. Temporal and spatial measurements can be made on single cells within a population of cells in a field. Figure 23 shows kinetic data for the dexamethasone- induced translocation of GFP-hGR in several cells within a single field. Human HeLa cells transfected with GFP-hGR were treated with 100 nM dexamethasone and the translocation of GFP-hGR was measured over time in a population of single cells. The graph shows the response of transfected cells 285, 286, 287, and 288 and non- transfected cells 289. These data also illustrate the ability to analyze cells with different expression levels.
Example 6 High-content screen of drug-induced apoptosis
Apoptosis is a complex cellular program that involves myriad molecular events and pathways. To understand the mechanisms of drug action on this process, it is essential to measure as many of these events within cells as possible with temporal and spatial resolution. Therefore, an apoptosis screen that requires little cell sample preparation yet provides an automated readout of several apoptosis-related parameters would be ideal. A cell-based assay designed for the cell screening system has been used to simultaneously quantify several of the moφhological, organellar, and macromolecular hallmarks of paclitaxel -induced apoptosis. Cell preparation. The cells chosen for this study were mouse connective tissue fibroblasts (L-929; ATCC CCL-1) and a highly invasive glioblastoma cell line (SNB- 19; ATCC CRL-2219) (Welch et al, In Vitro Cell. Dev. Biol 31:610, 1995). The day before treatment with an apoptosis inducing drug, 3500 cells were placed into each well
of a 96-well plate and incubated overnight at 37°C in a humidified 5% CO2 atmosphere. The following day, the culture medium was removed from each well and replaced with fresh medium containing various concentrations of paclitaxel (0 - 50 μM) from a 20 mM stock made in DMSO. The maximal concentration of DMSO used in these experiments was 0.25%. The cells were then incubated for 26 h as above. At the end of the paclitaxel treatment period, each well received fresh medium containing 750 nM MitoTracker Red (Molecular Probes; Eugene, OR) and 3 μg/ml Hoechst 33342 DNA-binding dye (Molecular Probes) and was incubated as above for 20 min. Each well on the plate was then washed with HBSS and fixed with 3.7% formaldehyde in HBSS for 15 min at room temperature. The formaldehyde was washed out with HBSS and the cells were permeabilized for 90 s with 0.5% (v/v) Triton X-100, washed with HBSS, incubated with 2 U ml"1 Bodipy FL phallacidin (Molecular Probes) for 30 min, and washed with HBSS. The wells on the plate were then filled with 200 μl HBSS,
sealed, and the plate stored at 4°C if necessary. The fluorescence signals from plates
stored this way were stable for at least two weeks after preparation. As in the nuclear translocation assay, fluorescence reagents can be designed to convert this assay into a live cell high-content screen.
Image acquisition and analysis on the ArrayScan System. The fluorescence intensity of intracellular MitoTracker Red, Hoechst 33342, and Bodipy FL phallacidin was measured with the cell screening system as described supra. Moφhometric data from each pair of images obtained from each well was also obtained to detect each object in the image field (e.g., cells and nuclei), and to calculate its size, shape, and integrated intensity.
Calculations and output. A total of 50-250 cells were measured per image field. For each field of cells, the following calculations were performed: (1) The average nuclear area (μm2) was calculated by dividing the total nuclear area in a field by the number of nuclei detected. (2) The average nuclear perimeter (μm) was calculated by dividing the sum of the perimeters of all nuclei in a field by the number of nuclei detected in that field. Highly convoluted apoptotic nuclei had the largest nuclear perimeter values. (3) The average nuclear brightness was calculated by dividing the integrated intensity of the entire field of nuclei by the number of nuclei in that field.
An increase in nuclear brightness was conelated with increased DNA content. (4) The average cellular brightness was calculated by dividing the integrated intensity of an entire field of cells stained with MitoTracker dye by the number of nuclei in that field. Because the amount of MitoTracker dye that accumulates within the mitochondria is proportional to the mitochondrial potential, an increase in the average cell brightness is consistent with an increase in mitochondrial potential. (5) The average cellular brightness was also calculated by dividing the integrated intensity of an entire field of cells stained with Bodipy FL phallacidin dye by the number of nuclei in that field. Because the phallotoxins bind with high affinity to the polymerized form of actin, the amount of Bodipy FL phallacidin dye that accumulates within the cell is proportional to actin polymerization state. An increase in the average cell brightness is consistent with an increase in actin polymerization. Results. Figure 24 (top panels) shows the changes paclitaxel induced in the nuclear moφhology of L-929 cells. Increasing amounts of paclitaxel caused nuclei to enlarge and fragment 293, a hallmark of apoptosis. Quantitative analysis of these and other images obtained by the cell screening system is presented in the same figure. Each parameter measured showed that the L-929 cells 296 were less sensitive to low concentrations of paclitaxel than were SNB-19 cells 297. At higher concentrations though, the L-929 cells showed a response for each parameter measured. The multiparameter approach of this assay is useful in dissecting the mechanisms of drag action. For example, the area, brightness, and fragmentation of the nucleus 298 and actin polymerization values 294 reached a maximum value when SNB-19 cells were treated with 10 nM paclitaxel (Figure 24; top and bottom graphs). However, mitochondrial potential 295 was minimal at the same concentration of paclitaxel (Figure 24; middle graph). The fact that all the parameters measured approached control levels at increasing paclitaxel concentrations (>10 nM) suggests that SNB-19 cells have low affinity drag metabolic or clearance pathways that are compensatory at sufficiently high levels of the drag. Contrasting the drag sensitivity of SNB-19 cells 297. L-929 showed a different response to paclitaxel 296. These fibroblastic cells showed a maximal response in many parameters at 5 μM paclitaxel, a 500-fold higher dose than SNB-19 cells. Furthermore, the L-929 cells did not show a shaφ decrease in mitochondrial potential 295 at any of the paclitaxel concentrations tested. This result is consistent with the presence of unique apoptosis pathways between a normal and cancer cell line. Therefore, these results indicate that a relatively simple fluorescence labeling protocol can be coupled with the cell screening system of the present invention to produce a high-content screen of key events involved in programmed cell death.
Example 7. Protease induced translocation of a signaling enzyme containing a disease-associated sequence from cytoplasm to nucleus.
Plasmid construct. A eukaryotic expression plasmid containing a coding sequence for a green fluorescent protein - caspase (Cohen (1997), Biochemical J. 326:1-16; Liang et al. (1997), J. ofMolec. Biol. 274:291-302) chimera is prepared using GFP mutants. The construct is used to transfect eukaryotic cells.
Cell preparation and transfection. Cells are trypsinized and plated 24 h prior to transfection and incubated at 37°C and 5% CO2. Transfections are performed by methods including, but not limited to calcium phosphate coprecipitation or lipofection. Cells are incubated with the calcium phosphate-DNA precipitate for 4-5 hours at 37°C and 5% CO2, washed 3-4 times with DMEM to remove the precipitate, followed by the addition of C-DMEM. Lipofectamine transfections are performed in serum-free DMEM without antibiotics according to the manufacturer's instructions. Following a 2-3 hour incubation with the DNA-liposome complexes, the medium is removed and replaced with C-DMEM.
Apopototic induction of Caspase-GFP translocation. To obtain Caspase-GFP translocation kinetic data, nuclei of transfected cells are first labeled with 5 μg/ml Hoechst 33342 (Molecular Probes) in C-DMEM for 20 minutes at 37°C and 5% CO2. Cells are washed once in Hank's Balanced Salt Solution (HBSS) followed by the addition of compounds that induce apoptosis. These compounds include, but are not limited to paclitaxel, staurosporine, ceramide, and tumor necrosis factor. To obtain fixed time point titration data, transfected cells are first washed with DMEM and then
incubated at 37°C and 5% CO2 for 1 h in the presence of 0 - 1000 nM compound in
DMEM. Cells are analyzed live or they are rinsed with HBSS, fixed for 15 min with 3.7%o formaldehyde in HBSS, stained with Hoechst 33342, and washed before analysis.
Image acquisition and analysis. Kinetic data are collected by acquiring fluorescence image pairs (Caspase-GFP and Hoechst 33342-labeled nuclei) from fields of living cells at 1 min intervals for 30 min after the addition of compound. Likewise, image pairs are obtained from each well of the fixed time point screening plates 1 h after the addition of compound. In both cases, the image pairs obtained at each time point are used to define nuclear and cytoplasmic regions in each cell. Translocation of Caspase-GFP is calculated by dividing the integrated fluorescence intensity of Caspase- GFP in the nucleus by the integrated fluorescence intensity of the chimera in the cytoplasm or as a nuclear-cytoplasmic difference of GFP fluorescence. In the fixed time point screen this translocation ratio is calculated from data obtained from at least 200 cells at each concentration of compound tested. Drag-induced translocation of Caspase-GFP from the cytoplasm to the nucleus is therefore conelated with an increase in the translocation ratio. Molecular interaction libraries including, but not limited to those comprising putative activators or inhibitors of apoptosis-activated enzymes are use to screen the indicator cell lines and identify a specific ligand for the DAS, and a pathway activated by compound activity.
Example 8. Identification of novel steroid receptors from DAS Two sources of material and/or information are required to make use of this embodiment, which allows assessment of the function of an uncharacterized gene. First, disease associated sequence bank(s) containing cDNA sequences suitable for transfection into mammalian cells can be used. Because every RADE or differential expression experiment generates up to several hundred sequences, it is possible to generate an ample supply of DAS. Second, information from primary sequence database searches can be used to place DAS into broad categories, including, but not limited to, those that contain signal sequences, seven trans-membrane motifs, conserved protease active site domains, or other identifiable motifs. Based on the information acquired from these sources, method types and indicator cell lines to be transfected are selected. A large number of motifs are already well characterized and encoded in the linear sequences contained within the large number genes in existing genomic databases.
In one embodiment, the following steps are taken:
1) Information from the DAS identification experiment (including database searches) is used as the basis for selecting the relevant biological processes, (for example, look at the DAS from a tumor line for cell cycle modulation, apoptosis, metastatic proteases, etc.)
2) Sorting of DNA sequences or DAS by identifiable motifs (ie. signal sequences, 7- transmembrane domains, conserved protease active site domains, etc.)
This initial grouping will determine fluorescent tagging strategies, host cell lines, indicator cell lines, and banks of bioactive molecules to be screened, as described supra.
3) Using well established molecular biology methods, ligate DAS into an expression vector designed for this puφose. Generalized expression vectors contain promoters, enhancers, and terminators for which to deliver target sequences to the cell for transient expression. Such vectors may also contain antibody tagging sequences, direct association sequences, chromophore fusion sequences like GFP, etc. to facilitate detection when expressed by the host. 4) Transiently transfect cells with DAS containing vectors using standard transfection protocols including: calcium phosphate co-precipitation, liposome mediated, DEAE dextran mediated, polycationic mediated, viral mediated, or electroporation, and plate into microtiter plates or microwell aπays. Alternatively, transfection can be done directly in the microtiter plate itself.
5) Carry out the cell screening methods as described supra.
In this embodiment, DAS shown to possess a motif(s) suggestive of transcriptional activation potential (for example, DNA binding domain, amino terminal modulating domain, hinge region, or carboxy terminal ligand binding domain) are utilized to identify novel steroid receptors.
Defining the fluorescent tags for this experiment involves identification of the nucleus through staining, and tagging the DAS by creating a GFP chimera via insertion of DAS into an expression vector, proximally fused to the gene encoding GFP. Alternatively, a single chain antibody fragment with high affinity to some portion of the expressed DAS could be constructed using technology available in the art (Cambridge Antibody Technologies) and linked to a fluorophore (FITC) to tag the putative transcriptional activator/receptor in the cells. This alternative would provide an external tag requiring no DNA transfection and therefore would be useful if distribution data were to be gathered from the original primary cultures used to generate the DAS.
Plasmid construct. A eukaryotic expression plasmid containing a coding sequence for a green fluorescent protein - DAS chimera is prepared using GFP mutants. The construct is used to transfect HeLa cells. The plasmid, when transfected into the host cell, produces a GFP fused to the DAS protein product, designated GFP- DASpp. Cell preparation and transfection. HeLa cells are trypsinized and plated using DMEM containing 5% charcoal/dextran-treated fetal bovine serum (FBS) (Hyclone) and 1%) penicillin-streptomycin (C-DMEM) 12-24 hours prior to transfection and incubated at 37°C and 5% CO . Transfections are performed by calcium phosphate coprecipitation or with Lipofectamine (Life Technologies). For the calcium phosphate transfections, the medium is replaced, prior to transfection, with DMEM containing 5% charcoal/dextran-treated FBS. Cells are incubated with the calcium phosphate-DNA precipitate for 4-5 hours at 37°C and 5% CO2, and washed 3-4 times with DMEM to remove the precipitate, followed by the addition of C-DMEM. Lipofectamine transfections are performed in seram-free DMEM without antibiotics according to the manufacturer's instructions. Following a 2-3 hour incubation with the DNA-liposome complexes, the medium is removed and replaced with C-DMEM. All transfected cells in 96-well microtiter plates are incubated at 33°C and 5% CO2 for 24-48 hours prior to drug treatment. Experiments are performed with the receptor expressed transiently in HeLa cells.
Localization of expressed GFP-DASpp inside cells. To obtain cellular distribution data, nuclei of transfected cells are first labeled with 5 μg/ml Hoechst 33342 (Molecular Probes) in C-DMEM for 20 minutes at 33°C and 5% CO . Cells are washed once in Hank's Balanced Salt Solution (HBSS). The cells are analyzed live or they are rinsed with HBSS, fixed for 15 min with 3.7% formaldehyde in HBSS, stained with Hoechst 33342, and washed before analysis.
In a prefeπed embodiment, image acquisition and analysis are performed using the cell screening system of the present invention. The intracellular GFP-DASpp fluorescence signal is collected by acquiring fluorescence image pairs (GFP-DASpp and Hoechst 33342-labeled nuclei) from field cells. The image pairs obtained at each time point are used to define nuclear and cytoplasmic regions in each cell. Data demonstrating dispersed signal in the cytoplasm would be consistent with known steroid receptors that are DNA transcriptional activators. Screening for induction of GFP-DASpp translocation. Using the above construct, confirmed for appropriate expression of the GFP-DASpp, as an indicator cell line, a screen of various ligands is performed using a series of steroid type ligands including, but not limited to: estrogen, progesterone, retinoids, growth factors, androgens, and many other steroid and steroid based molecules. Image acquisition and analysis are performed using the cell screening system of the invention. The intracellular GFP-DASpp fluorescence signal is collected by acquiring fluorescence image pairs (GFP-DASpp and Hoechst 33342-labeled nuclei) from fields cells. The image pairs obtained at each time point are used to define nuclear and cytoplasmic regions in each cell. Translocation of GFP-DASpp is calculated by dividing the integrated fluorescence intensity of GFP-DASpp in the nucleus by the integrated fluorescence intensity of the chimera in the cytoplasm or as a nuclear-cytoplasmic difference of GFP fluorescence. A translocation from the cytoplasm into the nucleus indicates a ligand binding activation of the DASpp thus identifying the potential receptor class and action. Combining this data with other data obtained in a similar fashion using known inhibitors and modifiers of steroid receptors, would either validate the DASpp as a target, or more data would be generated from various sources.
Example 9 Additional Screens
Translocation between the plasma membrane and the cytoplasm: Profilactin complex dissociation and binding of profilin to the plasma membrane. In one embodiment, a fluorescent protein biosensor of profilin membrane binding is prepared by labeling purified profilin (Federov et al.(1994), J. Molec. Biol. 241 :480-482; Lanbrechts et al. (1995), Eur. J. Biochem. 230:281-286) with a probe possessing a fluorescence lifetime in the range of 2-300 ns. The labeled profilin is introduced into living indicator cells using bulk loading methodology and the indicator cells are treated with test compounds. Fluorescence anisotropy imaging microscopy (Gough and Taylor (1993), J. Cell Biol. 121:1095-1107) is used to measure test- compound dependent movement of the fluorescent derivative of profilin between the cytoplasm and membrane for a period of time after treatment ranging from 0.1 s to 10 h.
Rho-RhoGDI complex translocation to the membrane. In another embodiment, indicator cells are treated with test compounds and then fixed, washed, and permeabilized. The indicator cell plasma membrane, cytoplasm, and nucleus are all labeled with distinctly colored markers followed by immunolocalization of Rho protein (Self et al. (1995), Methods in Enzymology 256:3-10; Tanaka et al. (1995), Methods in Enzymology 256:41-49) with antibodies labeled with a fourth color. Each of the four labels is imaged separately using the cell screening system, and the images used to calculate the amount of inhibition or activation of translocation effected by the test compound. To do this calculation, the images of the probes used to mark the plasma membrane and cytoplasm are used to mask the image of the immunological probe marking the location of intracellular Rho protein. The integrated brightness per unit area under each mask is used to form a translocation quotient by dividing the plasma membrane integrated brightness/area by the cytoplasmic integrated brightness/area. By comparing the translocation quotient values from control and experimental wells, the percent translocation is calculated for each potential lead compound.
β-Arrestin translocation to the plasma membrane upon G-protein receptor activation.
In another embodiment of a cytoplasm to membrane translocation high-content
screen, the translocation of β-anestin protein from the cytoplasm to the plasma
membrane is measured in response to cell treatment. To measure the translocation, living indicator cells containing luminescent domain markers are treated with test
compounds and the movement of the β-aπestin marker is measured in time and space
using the cell screening system of the present invention. In a prefened embodiment, the indicator cells contain luminescent markers consisting of a green fluorescent protein
β-anestin (GFP-β-aπestin) protein chimera (Barak et al. (1997), J. Biol. Chem.
272:27497-27500; Daaka et al. (1998), J. Biol. Chem. 273:685-688) that is expressed by the indicator cells through the use of transient or stable cell transfection and other reporters used to mark cytoplasmic and membrane domains. When the indicator cells are in the resting state, the domain marker molecules partition predominately in the plasma membrane or in the cytoplasm. In the high-content screen, these markers are used to delineate the cell cytoplasm and plasma membrane in distinct channels of fluorescence. When the indicator cells are treated with a test compound, the dynamic
redistribution of the GFP-β-anestin is recorded as a series of images over a time scale
ranging from 0.1 s to 10 h. In a prefened embodiment, the time scale is 1 h. Each
image is analyzed by a method that quantifies the movement of the GFP-β-aπestin
protein chimera between the plasma membrane and the cytoplasm. To do this calculation, the images of the probes used to mark the plasma membrane and cytoplasm
are used to mask the image of the GFP-β-aπestin probe marking the location of
intracellular GFP-β-anestin protein. The integrated brightness per unit area under each
mask is used to form a translocation quotient by dividing the plasma membrane integrated brightness/area by the cytoplasmic integrated brightness/area. By comparing the translocation quotient values from control and experimental wells, the percent translocation is calculated for each potential lead compound. The output of the high- content screen relates quantitative data describing the magnitude of the translocation within a large number of individual cells that have been treated with test compounds of interest.
Translocation between the endoplasmic reticulum and the Golgi:
In one embodiment of an endoplasmic reticulum to Golgi translocation high- content screen, the translocation of a VSVG protein from the ts045 mutant strain of vesicular stomatitis virus (Ellenberg et al. (1997), J. Cell Biol 138:1193-1206; Presley et al. (1997) Nature 389:81-85) from the endoplasmic reticulum to the Golgi domain is measured in response to cell treatment. To measure the translocation, indicator cells containing luminescent reporters are treated with test compounds and the movement of the reporters is measured in space and time using the cell screening system of the present invention. The indicator cells contain luminescent reporters consisting of a GFP-VSVG protein chimera that is expressed by the indicator cell through the use of transient or stable cell transfection and other domain markers used to measure the localization of the endoplasmic reticulum and Golgi domains. When the indicator cells are in their resting state at 40°C, the GFP-VSVG protein chimera molecules are partitioned predominately in the endoplasmic reticulum. In this high-content screen, domain markers of distinct colors used to delineate the endoplasmic reticulum and the Golgi domains in distinct channels of fluorescence. When the indicator cells are treated with a test compound and the temperature is simultaneously lowered to 32°C, the dynamic redistribution of the GFP-VSVG protein chimera is recorded as a series of images over a time scale ranging from 0.1 s to 10 h. Each image is analyzed by a method that quantifies the movement of the GFP-VSVG protein chimera between the endoplasmic reticulum and the Golgi domains. To do this calculation, the images of the probes used to mark the endoplasmic reticulum and the Golgi domains are used to mask the image of the GFP-VSVG probe marking the location of intracellular GFP- VSVG protein. The integrated brightness per unit area under each mask is used to form a translocation quotient by dividing the endoplasmic reticulum integrated brightness/area by the Golgi integrated brightness/area. By comparing the translocation quotient values from control and experimental wells, the percent translocation is calculated for each potential lead compound. The output of the high-content screen relates quantitative data describing the magnitude of the translocation within a large number of individual cells that have been treated with test compounds of interest at final concentrations ranging from IO"12 M to 103 M for a period ranging from 1 min to 10 h.
Induction and inhibition of or ganellar function: Intracellular microtubule stability.
In another aspect of the invention, an automated method for identifying compounds that modify microtubule structure is provided. In this embodiment, indicator cells are treated with test compounds and the distribution of luminescent microtubule-labeling molecules is measured in space and time using a cell screening system, such as the one disclosed above. The luminescent microtubule-labeling molecules may be expressed by or added to the cells either before, together with, or after contacting the cells with a test compound. In one embodiment of this aspect of the invention, living cells express a luminescently labeled protein biosensor of microtubule dynamics, comprising a protein that labels microtubules fused to a luminescent protein. Appropriate microtubule-
labeling proteins for this aspect of the invention include, but are not limited to α and β
tubulin isoforms, and MAP4. Prefened embodiments of the luminescent protein include, but are not limited to green fluorescent protein (GFP) and GFP mutants. In a prefeπed embodiment, the method involves transfecting cells with a microtubule labeling luminescent protein, wherein the microtubule labeling protein can be, but is
not limited to, α-tubulin, β-tubulin, or microtubule-associated protein 4 (MAP4). The
approach outlined here enables those skilled in the art to make live cell measurements to determine the effect of lead compounds on tubulin activity and microtubule stability in vivo.
In a most prefened embodiment, MAP4 is fused to a modified version of the Aequorea victoria green fluorescent protein (GFP). A DNA construct has been made which consists of a fusion between the EGFP coding sequence (available from Clontech) and the coding sequence for mouse MAP4. (Olson et al., (1995), J. Cell Biol. 130(3): 639-650). MAP4 is a ubiquitous microtubule-associated protein that is known to interact with microtubules in inteφhase as well as mitotic cells (Olmsted and Murofushi, (1993), MAP4. In "Guidebook to the Cytoskeleton and Motor Proteins."
Oxford University Press. T. Kreis and R. Vale, eds.) Its localization, then, can serve as an indicator of the localization, organization, and integrity of microtubules in living (or fixed) cells at all stages of the cell cycle for cell-based HCS assays. While MAP2 and tau (microtubule associated proteins expressed specifically in neuronal cells) have been used to form GFP chimeras (Kaech et al, (1996) Neuron. 17: 1189-1199; Hall et al, (1997), Proc. Nat. Acad. Sci. 94: 4733-4738) their restricted cell type distribution and the tendency of these proteins to bundle microtubules when overexpressed make these proteins less desirable as molecular reagents for analysis in live cells originating from varied tissues and organs. Moderate overexpression of GFP-MAP4 does not disrupt microtubule function or integrity (Olson et al., 1995). Similar constructs can be made
using β-tubulin or α-tubulin via standard techniques in the art. These chimeras will
provide a means to observe and analyze microtubule activity in living cells during all stages of the cell cycle.
In another embodiment, the luminescently labeled protein biosensor of microtubule dynamics is expressed, isolated, and added to the cells to be analyzed via bulk loading techniques, such as microinjection, scrape loading, and impact-mediated loading. In this embodiment, there is not an issue of overexpression within the cell,
and thus α and β tubulin isoforms, MAP4, MAP2 and/or tau can all be used.
In a further embodiment, the protein biosensor is expressed by the cell, and the cell is subsequently contacted with a luminescent label, such as a labeled antibody, that detects the protein biosensor, endogenous levels of a protein antigen, or both. In this
embodiment, a luminescent label that detects α and β tubulin isoforms, MAP4, MAP2
and/or tau, can be used. A variety of GFP mutants are available, all of which would be effective in this invention, including, but not limited to, GFP mutants which are commercially available ■ (Clontech, California).
The MAP4 construct has been introduced into several mammalian cell lines (BHK-21, Swiss 3T3, HeLa, HEK 293, LLCPK) and the organization and localization of tubulin has been visualized in live cells by virtue of the GFP fluorescence as an indicator of MAP4 localization. The construct can be expressed transiently or stable cell lines can be prepared by standard methods. Stable HeLa cell lines expressing the EGFP-MAP4 chimera have been obtained, indicating that expression of the chimera is not toxic and does not interfere with mitosis.
Possible selectable markers for establishment and maintenance of stable cell lines include, but are not limited to the neomycin resistance gene, hygromycin resistance gene, zeocin resistance gene, puromycin resistance gene, bleomycin resistance gene, and blastacidin resistance gene. The utility of this method for the monitoring of microtubule assembly, disassembly, and reanangement has been demonstrated by treatment of transiently and stably transfected cells with microtubule drags such as paclitaxel, nocodazole, vincristine, or vinblastine.
The present method provides high-content and combined high throughput-high content cell-based screens for anti-microtubule drags, particularly as one parameter in a multi-parametric cancer target screen. The EGFP-MAP4 construct used herein can also be used as one of the components of a high-content screen that measures multiple signaling pathways or physiological events. In a prefened embodiment, a combined high throughput and high content screen is employed, wherein multiple cells in each of the locations containing cells are analyzed in a high throughput mode, and only a subset of the locations containing cells are analyzed in a high content mode. The high throughput screen can be any screen that would be useful to identify those locations containing cells that should be further analyzed, including, but not limited to, identifying locations with increased luminescence intensity, those exhibiting expression of a reporter gene, those undergoing calcium changes, and those undergoing pH changes.
In addition to drag screening applications, the present invention may be applied to clinical diagnostics, the detection of chemical and biological warfare weapons, and the basic research market since fundamental cell processes, such as cell division and motility, are highly dependent upon microtubule dynamics.
Image Acquisition and Analysis
Image data can be obtained from either fixed or living indicator cells. To extract moφhometric data from each of the images obtained the following method of analysis is used:
1. Threshold each nucleus and cytoplasmic image to produce a mask that has value = 0 for each pixel outside a nucleus or cell boundary.
2. Overlay the mask on the original image, detect each object in the field (i.e., nucleus or cell), and calculate its size, shape, and integrated intensity.
3. Overlay the whole cell mask obtained above on the conesponding luminescent microtubule image and apply one or more of the following set of classifiers to determine the micrtotubule moφhology and the effect of drugs on microtubule moφhology. Microtubule moφhology is defined using a set of classifiers to quantify aspects of microtubule shape, size, aggregation state, and polymerization state. These classifiers can be based on approaches that include co-occunence matrices, texture measurements, spectral methods, structural methods, wavelet transforms, statistical methods, or combinations thereof. Examples of such classifiers are as follows:
1. A classifier to quantify microtubule length and width using edge detection methods such as that discussed in Kolega et al. ((1993). Biolmaging 1 :136- 150), which discloses a non-automated method to determine edge strength in individual cells), to calculate the total edge strength within each cell. To normalize for cell size, the total edge strength can be divided by the cell area to give a "microtubule moφhology" value. Large microtubule moφhology values are associated with strong edge strength values and are therefore maximal in cells containing distinct microtubule structures. Likewise, small microtubule moφhology values are associated with weak edge strength and are minimal in cells with depolymerized microtubules. The physiological range of microtubule moφhology values is set by treating cells with either the microtubule stabilizing drug paclitaxel (10 μM) or the microtubule depolymerizing drag nocodazole (10 μg/ml).
2. A classifier to quantify microtubule aggregation into punctate spots or foci using methodology from the receptor intemalization methods discussed supra.
3. A classifier to quantify microtubule depolymerization using a measure of image texture. 4. A classifier to quantify apparent interconnectivity, or branching (or both), of the microtubules.
5. Measurement of the kinetics of microtubule reorganization using the above classifiers on a time series of images of cells treated with test compounds.
In a further aspect, kits are provided for analyzing microtubule stability, comprising an expression vector comprising a nucleic acid that encodes a microtubule labeling protein and instructions for using the expression vector for carrying out the methods described above. In a prefened embodiment, the expression vector further comprises a nucleic acid that encodes a luminescent protein, wherein the microtubule binding protein and the luminescent protein thereof are expressed as a fusion protein. Alternatively, the kit may contain an antibody that specifically binds to the microtubule-labeling protein. In a further embodiment, the kit includes cells that express the microtubule labeling protein. In a prefened embodiment, the cells are transfected with the expression vector. In another prefened embodiment, the kits further contain a compound that is known to disrapt microtubule structure, including but not limited to curacin, nocodazole, vincristine, or vinblastine. In another prefened embodiment, the kits further comprise a compound that is known to stabilize microtubule structure, including but not limited to taxol (paclitaxel), and discodermolide. In another aspect, the present invention comprises a machine readable storage medium comprising a program containing a set of instructions for causing a cell screening system to execute the disclosed methods for analyzing microtubule stability, wherein the cell screening system comprises an optical system with a stage adapted for holding a plate containing cells, a digital camera, a means for directing fluorescence or luminescence emitted from the cells to the digital camera, and a computer means for receiving and processing the digital data from the digital camera.
High-content screens involving the functional localization of macromolecules
Within this class of high-content screen, the functional localization of macromolecules in response to external stimuli is measured within living cells.
Glycolytic enzyme activity regulation. In a prefened embodiment of a cellular enzyme activity high-content screen, the activity of key glycolytic regulatory enzymes are measured in treated cells. To measure enzyme activity, indicator cells containing luminescent labeling reagents are treated with test compounds and the activity of the reporters is measured in space and time using cell screening system of the present invention.
In one embodiment, the reporter of intracellular enzyme activity is fructose-6- phosphate, 2-kinase/fructose-2,6-bisphosphatase (PFK-2), a regulatory enzyme whose phosphorylation state indicates intracellular carbohydrate anabolism or catabolism (Deprez et al. (1997) J. Biol. Chem. 272:17269-17275; Kealer et al. (1996) FEBS Letters 395:225-227; Lee et al. (1996), Biochemistiγ 35:6010-6019). The indicator cells contain luminescent reporters consisting of a fluorescent protein biosensor of PFK-2 phosphorylation. The fluorescent protein biosensor is constructed by introducing an environmentally sensitive fluorescent dye near to the known phosphorylation site of the enzyme (Deprez et al. (1997), supra; Giuliano et al. (1995), supra). The dye can be of the ketocyanine class (Kessler and Wolfbeis (1991), Spectrochimica Acta 47A:187-192 ) or any class that contains a protein reactive moiety and a fluorochrome whose excitation or emission spectrum is sensitive to solution polarity. The fluorescent protein biosensor is introduced into the indicator cells using bulk loading methodology.
Living indicator cells are treated with test compounds, at final concentrations ranging from IO"12 M to IO"3 M for times ranging from 0.1 s to 10 h. In a prefened embodiment, ratio image data are obtained from living treated indicator cells by collecting a spectral pair of fluorescence images at each time point. To extract moφhometric data from each time point, a ratio is made between each pair of images by numerically dividing the two spectral images at each time point, pixel by pixel. Each pixel value is then used to calculate the fractional phosphorylation of PFK-2. At small fractional values of phosphorylation, PFK-2 stimulates carbohydrate catabolism. At high fractional values of phosphorylation, PFK-2 stimulates carbohydrate anabolism.
Protein kinase A activity and localization of sub units. In another embodiment of a high-content screen, both the domain localization and activity of protein kinase A (PKA) within indicator cells are measured in response to treatment with test compounds.
The indicator cells contain luminescent reporters including a fluorescent protein biosensor of PKA activation. The fluorescent protein biosensor is constructed by introducing an environmentally sensitive fluorescent dye into the catalytic subunit of PKA near the site known to interact with the regulatory subunit of PKA (Harootunian et al. (1993), Mol. Biol. of the Cell 4:993-1002; Johnson et al. (1996), Cell 85: 149-158; Giuliano et al. (1995), supra). The dye can be of the ketocyanine class (Kessler, and Wolfbeis (1991), Spectrochimica Acta 47A:187-192) or any class that contains a protein reactive moiety and a fluorochrome whose excitation or emission spectrum is sensitive to solution polarity. The fluorescent protein biosensor of PKA activation is introduced into the indicator cells using bulk loading methodology.
In one embodiment, living indicator cells are treated with test compounds, at final concentrations ranging from IO"12 M to 10"3 M for times ranging from 0.1 s to 10 h. In a prefened embodiment, ratio image data are obtained from living treated indicator cells. To extract biosensor data from each time point, a ratio is made between each pair of images, and each pixel value is then used to calculate the fractional activation of PKA (e.g., separation of the catalytic and regulatory subunits after cAMP binding). At high fractional values of activity, PFK-2 stimulates biochemical cascades within the living cell.
To measure the translocation of the catalytic subunit of PKA, indicator cells containing luminescent reporters are treated with test compounds and the movement of the reporters is measured in space and time using the cell screening system. The indicator cells contain luminescent reporters consisting of domain markers used to measure the localization of the cytoplasmic and nuclear domains. When the indicator cells are treated with a test compounds, the dynamic redistribution of a PKA fluorescent protein biosensor is recorded intracellularly as a series of images over a time scale ranging from 0.1 s to 10 h. Each image is analyzed by a method that quantifies the movement of the PKA between the cytoplasmic and nuclear domains. To do this calculation, the images of the probes used to mark the cytoplasmic and nuclear domains are used to mask the image of the PKA fluorescent protein biosensor. The integrated brightness per unit area under each mask is used to form a translocation quotient by dividing the cytoplasmic integrated brightness/area by the nuclear integrated brightness/area. By comparing the translocation quotient values from control and experimental wells, the percent translocation is calculated for each potential lead compound. The output of the high-content screen relates quantitative data describing the magnitude of the translocation within a large number of individual cells that have been treated with test compound in the concentration range of 10" M to 10" M. High-content screens involving the induction or inhibition of gene expression RNA-based fluorescent biosensors
Cytoskeletal protein transcription and message localization. Regulation of the general classes of cell physiological responses including cell-substrate adhesion, cell-cell adhesion, signal transduction, cell-cycle events, intermediary and signaling molecule metabolism, cell locomotion, cell-cell communication, and cell death can involve the alteration of gene expression. High-content screens can also be designed to measure this class of physiological response.
In one embodiment, the reporter of intracellular gene expression is an oligonucleotide that can hybridize with the target mRNA and alter its fluorescence signal. In a prefeπed embodiment, the oligonucleotide is a molecular beacon (Tyagi and Kramer (1996) Nat. Biotechnol. 14:303-308), a luminescence-based reagent whose fluorescence signal is dependent on intermolecular and intramolecular interactions.
The fluorescent biosensor is constructed by introducing a fluorescence energy transfer pair of fluorescent dyes such that there is one at each end (5' and 3') of the reagent.
The dyes can be of any class that contains a protein reactive moiety and fluorochromes whose excitation and emission spectra overlap sufficiently to provide fluorescence energy transfer between the dyes in the resting state, including, but not limited to, fluorescein and rhodamine (Molecular Probes, Inc.). In a prefened embodiment, a
portion of the message coding for β-actin (Kislauskis et al. (1994), J. Cell Biol.
127:441-451; McCann et al. (1997), Proc. Natl. Acad. Sci. 94:5679-5684; Sutoh
(1982), Biochemistry 21:3654-3661) is inserted into the loop region of a haiφin-shaped oligonucleotide with the ends tethered together due to intramolecular hybridization. At each end of the biosensor a fluorescence donor (fluorescein) and a fluorescence acceptor (rhodamine) are covalently bound. In the tethered state, the fluorescence energy transfer is maximal and therefore indicative of an unhybridized molecule.
When hybridized with the mRNA coding for β-actin, the tether is broken and energy
transfer is lost. The complete fluorescent biosensor is introduced into the indicator cells using bulk loading methodology.
In one embodiment, living indicator cells are treated with test compounds, at final concentrations ranging from IO"12 M to IO"3 M for times ranging from 0.1 s to 10 h. In a prefened embodiment, ratio image data are obtained from living treated indicator cells. To extract moφhometric data from each time point, a ratio is made between each pair of images, and each pixel value is then used to calculate the fractional hybridization of the labeled nucleotide. At small fractional values of
hybridization little expression of β-actin is indicated. At high fractional values of
hybridization, maximal expression of β-actin is indicated. Furthermore, the distribution
of hybridized molecules within the cytoplasm of the indicator cells is also a measure of the physiological response of the indicator cells.
Cell surface binding of a ligand
Labeled insulin binding to its cell surface receptor in living cells. Cells whose plasma membrane domain has been labeled with a labeling reagent of a particular color are incubated with a solution containing insulin molecules (Lee et al.
(1997), Biochemistry 36:2701-2708; Martinez-Zaguilan et al. (1996), Am. J. Physiol.
270:C1438-C1446) that are labeled with a luminescent probe of a different color for an appropriate time under the appropriate conditions. After incubation, unbound insulin molecules are washed away, the cells fixed and the distribution and concentration of the insulin on the plasma membrane is measured. To do this, the cell membrane image is used as a mask for the insulin image. The integrated intensity from the masked insulin image is compared to a set of images containing known amounts of labeled insulin. The amount of insulin bound to the cell is determined from the standards and used in conjunction with the total concentration of insulin incubated with the cell to calculate a dissociation constant or insulin to its cell surface receptor.
Labeling of cellular compartments Whole cell labeling Whole cell labeling is accomplished by labeling cellular components such that dynamics of cell shape and motility of the cell can be measured over time by analyzing fluorescence images of cells.
In one embodiment, small reactive fluorescent molecules are introduced into living cells. These membrane-permeant molecules both diffuse through and react with protein components in the plasma membrane. Dye molecules react with intracellular molecules to both increase the fluorescence signal emitted from each molecule and to entrap the fluorescent dye within living cells. These molecules include reactive chloromethyl derivatives of aminocoumarins, hydroxycoumarins, eosin diacetate, fluorescein diacetate, some Bodipy dye derivatives, and tetramethylrhodamine. The reactivity of these dyes toward macromolecules includes free primary amino groups and free sulfhydryl groups.
In another embodiment, the cell surface is labeled by allowing the cell to interact with fluorescently labeled antibodies or lectins (Sigma Chemical Company, St. Louis, MO) that react specifically with molecules on the cell surface. Cell surface protein chimeras expressed by the cell of interest that contain a green fluorescent protein, or mutant thereof, component can also be used to fluorescently label the entire cell surface. Once the entire cell is labeled, images of the entire cell or cell aπay can become a parameter in high content screens, involving the measurement of cell shape, motility, size, and growth and division.
Plasma membrane labeling
In one embodiment, labeling the whole plasma membrane employs some of the same methodology described above for labeling the entire cells. Luminescent molecules that label the entire cell surface act to delineate the plasma membrane.
In a second embodiment subdomains of the plasma membrane, the extracellular surface, the lipid bilayer, and the intracellular surface can be labeled separately and used as components of high content screens. In the first embodiment, the extracellular surface is labeled using a brief treatment with a reactive fluorescent molecule such as the succinimidyl ester or iodoacetamde derivatives of fluorescent dyes such as the fluoresceins, rhodamines, cyanines, and Bodipys.
In a third embodiment, the extracellular surface is labeled using fluorescently labeled macromolecules with a high affinity for cell surface molecules. These include fluorescently labeled lectins such as the fluorescein, rhodamine, and cyanine derivatives of lectins derived from jack bean (Con A), red kidney bean
(erythroagglutinin PHA-E), or wheat germ.
In a fourth embodiment, fluorescently labeled antibodies with a high affinity for cell surface components are used to label the extracellular region of the plasma membrane. Extracellular regions of cell surface receptors and ion channels are examples of proteins that can be labeled with antibodies.
In a fifth embodiment, the lipid bilayer of the plasma membrane is labeled with fluorescent molecules. These molecules include fluorescent dyes attached to long chain hydrophobic molecules that interact strongly with the hydrophobic region in the center of the plasma membrane lipid bilayer. Examples of these dyes include the PKH series of dyes (U.S. 4,783,401, 4,762701, and 4,859,584; available commercially from Sigma Chemical Company, St. Loius, MO), fluorescent phospholipids such as nitrobenzoxadiazole glycerophosphoethanolamine and fluorescein-derivatized dihexadecanoylglycerophosphoetha-nolamine, fluorescent fatty acids such as 5-butyl- 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene-3-nonanoic acid and 1 -pyrenedecanoic acid (Molecular Probes, Inc.), fluorescent sterols including cholesteryl 4,4-difluoro-5,7- dimethyl-4-bora-3a,4a-diaza-s-indacene-3-dodecanoate and cholesteryl 1- pyrenehexanoate, and fluorescently labeled proteins that interact specifically with lipid bilayer components such as the fluorescein derivative of annexin V (Caltag Antibody Co, Burlingame, CA).
In another embodiment, the intracellular component of the plasma membrane is labeled with fluorescent molecules. Examples of these molecules are the intracellular components of the trimeric G-protein receptor, adenylyl cyclase, and ionic transport proteins. These molecules can be labeled as a result of tight binding to a fluorescently labeled specific antibody or by the incoφoration of a fluorescent protein chimera that is comprised of a membrane-associated protein and the green fluorescent protein, and mutants thereof. Endosome fluorescence labeling
In one embodiment, ligands that are transported into cells by receptor-mediated endocytosis are used to trace the dynamics of endosomal organelles. Examples of labeled ligands include Bodipy FL-labeled low density lipoprotein complexes, tetramethylrhodamine transferrin analogs, and fluorescently labeled epidermal growth factor (Molecular Probes, Inc.)
In a second embodiment, fluorescently labeled primary or secondary antibodies (Sigma Chemical Co. St. Louis, MO; Molecular Probes, Inc. Eugene, OR; Caltag Antibody Co.) that specifically label endosomal ligands are used to mark the endosomal compartment in cells.
In a third embodiment, endosomes are fluorescently labeled in cells expressing protein chimeras formed by fusing a green fluorescent protein, or mutants thereof, with a receptor whose intemalization labels endosomes. Chimeras of the EGF, transferrin, and low density lipoprotein receptors are examples of these molecules.
Lysosome labeling
In one embodiment, membrane permeant lysosome-specific luminescent reagents are used to label the lysosomal compartment of living and fixed cells. These reagents include the luminescent molecules neutral red, N-(3-((2,4- dinitrophenyl)amino)propyl)-N-(3-aminopropyl)methylamine, and the LysoTracker probes which report intralysosomal pH as well as the dynamic distribution of lysosomes (Molecular Probes, Inc.)
In a second embodiment, antibodies against lysosomal antigens (Sigma Chemical Co.; Molecular Probes, Inc.; Caltag Antibody Co.) are used to label lysosomal components that are localized in specific lysosomal domains. Examples of these components are the degradative enzymes involved in cholesterol ester hydrolysis, membrane protein proteases, and nucleases as well as the ATP-driven lysosomal proton pump. In a third embodiment, protein chimeras consisting of a lysosomal protein genetically fused to an intrinsically luminescent protein such as the green fluorescent protein, or mutants thereof, are used to label the lysosomal domain. Examples of these components are the degradative enzymes involved in cholesterol ester hydrolysis, membrane protein proteases, and nucleases as well as the ATP-driven lysosomal proton pump.
Cytoplasmic fluorescence labeling
In one embodiment, cell permeant fluorescent dyes (Molecular Probes, Inc.) with a reactive group are reacted with living cells. Reactive dyes including monobromobimane, 5-chloromethylfluorescein diacetate, carboxy fluorescein diacetate succinimidyl ester, and chloromethyl tetramethylrhodamine are examples of cell permeant fluorescent dyes that are used for long term labeling of the cytoplasm of cells.
In a second embodiment, polar tracer molecules such as Lucifer yellow and cascade blue-based fluorescent dyes (Molecular Probes, Inc.) are introduced into cells using bulk loading methods and are also used for cytoplasmic labeling.
In a third embodiment, antibodies against cytoplasmic components (Sigma Chemical Co.; Molecular Probes, Inc.; Caltag Antibody Co.) are used to fluorescently label the cytoplasm. Examples of cytoplasmic antigens are many of the enzymes involved in intermediary metabolism. Enolase, phosphofractokinase, and acetyl-CoA dehydrogenase are examples of uniformly distributed cytoplasmic antigens.
In a fourth embodiment, protein chimeras consisting of a cytoplasmic protein genetically fused to an intrinsically luminescent protein such as the green fluorescent protein, or mutants thereof, are used to label the cytoplasm. Fluorescent chimeras of uniformly distributed proteins are used to label the entire cytoplasmic domain. Examples of these proteins are many of the proteins involved in intermediary metabolism and include enolase, lactate dehydrogenase, and hexokinase.
In a fifth embodiment, antibodies against cytoplasmic antigens (Sigma Chemical Co.; Molecular Probes, Inc.; Caltag Antibody Co.) are used to label cytoplasmic components that are localized in specific cytoplasmic sub-domains. Examples of these components are the cytoskeletal proteins actin, tubulin, and cytokeratin. A population of these proteins within cells is assembled into discrete structures, which in this case, are fibrous. Fluorescence labeling of these proteins with antibody-based reagents therefore labels a specific sub-domain of the cytoplasm.
In a sixth embodiment, non-antibody-based fluorescently labeled molecules that interact strongly with cytoplasmic proteins are used to label specific cytoplasmic components. One example is a fluorescent analog of the enzyme DNAse I (Molecular Probes, Inc.) Fluorescent analogs of this enzyme bind tightly and specifically to cytoplasmic actin, thus labeling a sub-domain of the cytoplasm. In another example, fluorescent analogs of the mushroom toxin phalloidin or the drag paclitaxel (Molecular Probes, Inc.) are used to label components of the actin- and microtubule-cytoskeletons, respectively. In a seventh embodiment, protein chimeras consisting of a cytoplasmic protein genetically fused to an intrinsically luminescent protein such as the green fluorescent protein, or mutants thereof, are used to label specific domains of the cytoplasm. Fluorescent chimeras of highly localized proteins are used to label cytoplasmic sub- domains. Examples of these proteins are many of the proteins involved in regulating the cytoskeleton. They include the structural proteins actin, tubulin, and cytokeratin as
well as the regulatory proteins microtubule associated protein 4 and α-actinin.
Nuclear labeling In one embodiment, membrane permeant nuclei c-acid-specific luminescent reagents (Molecular Probes, Inc.) are used to label the nucleus of living and fixed cells.
These reagents include cyanine-based dyes (e.g., TOTO®, YOYO®, and BOBO™),
phenanthidines and acridines (e.g., ethidium bromide, propidium iodide, and acridine orange), indoles and imidazoles (e.g., Hoechst 33258, Hoechst 33342, and 4', 6- diamidino-2-phenylindole), and other similar reagents (e.g., 7-aminoactinomycin D, hydroxystilbamidine, and the psoralens).
In a second embodiment, antibodies against nuclear antigens (Sigma Chemical Co.; Molecular Probes, Inc.; Caltag Antibody Co.) are used to label nuclear components that are localized in specific nuclear domains. Examples of these components are the macromolecules involved in maintaining DNA structure and function. DNA, RNA, histones, DNA polymerase, RNA polymerase, lamins, and nuclear variants of cytoplasmic proteins such as actin are examples of nuclear antigens.
In a third embodiment, protein chimeras consisting of a nuclear protein genetically fused to an intrinsically luminescent protein such as the green fluorescent protein, or mutants thereof, are used to label the nuclear domain. Examples of these proteins are many of the proteins involved in maintaining DNA stracture and function. Histones, DNA polymerase, RNA polymerase, lamins, and nuclear variants of cytoplasmic proteins such as actin are examples of nuclear proteins.
Mitochondrial labeling
In one embodiment, membrane permeant mitochondrial-specific luminescent reagents (Molecular Probes, Inc.) are used to label the mitochondria of living and fixed cells. These reagents include rhodamine 123, tetramethyl rosamine, JC-1, and the MitoTracker reactive dyes.
In a second embodiment, antibodies against mitochondrial antigens (Sigma Chemical Co.; Molecular Probes, Inc.; Caltag Antibody Co.) are used to label mitochondrial components that are localized in specific mitochondrial domains. Examples of these components are the macromolecules involved in maintaining mitochondrial DNA stracture and function. DNA, RNA, histones, DNA polymerase, RNA polymerase, and mitochondrial variants of cytoplasmic macromolecules such as mitochondrial tRNA and rRNA are examples mitochondrial antigens. Other examples of mitochondrial antigens are the components of the oxidative phosphorylation system found in the mitochondria (e.g., cytochrome c, cytochrome c oxidase, and succinate dehydrogenase).
In a third embodiment, protein chimeras consisting of a mitochondrial protein genetically fused to an intrinsically luminescent protein such as the green fluorescent protein, or mutants thereof, are used to label the mitochondrial domain. Examples of these components are the macromolecules involved in maintaining mitochondrial DNA structure and function. Examples include histones, DNA polymerase, RNA polymerase, and the components of the oxidative phosphorylation system found in the mitochondria (e.g., cytochrome c, cytochrome c oxidase, and succinate dehydrogenase). Endoplasmic reticulum labeling
In one embodiment, membrane permeant endoplasmic reticulum-specific luminescent reagents (Molecular Probes, Inc.) are used to label the endoplasmic reticulum of living and fixed cells. These reagents include short chain carbocyanine dyes (e.g., DiOC and DiOC3), long chain carbocyanine dyes (e.g., DiICi6 and DiICι ), and luminescently labeled lectins such as concanavalin A.
In a second embodiment, antibodies against endoplasmic reticulum antigens
(Sigma Chemical Co.; Molecular Probes, Inc.; Caltag Antibody Co.) are used to label endoplasmic reticulum components that are localized in specific endoplasmic reticulum domains. Examples of these components are the macromolecules involved in the fatty acid elongation systems, glucose-6-phosphatase, and HMG CoA-reductase.
In a third embodiment, protein chimeras consisting of a endoplasmic reticulum protein genetically fused to an intrinsically luminescent protein such as the green fluorescent protein, or mutants thereof, are used to label the endoplasmic reticulum domain. Examples of these components are the macromolecules involved in the fatty acid elongation systems, glucose-6-phosphatase, and HMG CoA-reductase.
Golgi labeling
In one embodiment, membrane permeant Golgi-specific luminescent reagents (Molecular Probes, Inc.) are used to label the Golgi of living and fixed cells. These reagents include luminescently labeled macromolecules such as wheat germ agglutinin and Brefeldin A as well as luminescently labeled ceramide.
In a second embodiment, antibodies against Golgi antigens (Sigma Chemical
Co.; Molecular Probes, Inc.; Caltag Antibody Co.) are used to label Golgi components that are localized in specific Golgi domains. Examples of these components are N- acetylglucosamine phosphotransferase, Golgi-specific phosphodiesterase, and mannose-6-phosphate receptor protein.
In a third embodiment, protein chimeras consisting of a Golgi protein genetically fused to an intrinsically luminescent protein such as the green fluorescent protein, or mutants thereof, are used to label the Golgi domain. Examples of these components are N-acetylglucosamine phosphotransferase, Golgi-specific phosphodiesterase, and mannose-6-phosphate receptor protein.
While many of the examples presented involve the measurement of single cellular processes, this is again is intended for puφoses of illustration only. Multiple parameter high-content screens can be produced by combining several single parameter screens into a multiparameter high-content screen or by adding cellular parameters to any existing high-content screen. Furthermore, while each example is described as being based on either live or fixed cells, each high-content screen can be designed to be used with both live and fixed cells. Those skilled in the art will recognize a wide variety of distinct screens that can be developed based on the disclosure provided herein. There is a large and growing list of known biochemical and molecular processes in cells that involve translocations or reorganizations of specific components within cells. The signaling pathway from the cell surface to target sites within the cell involves the translocation of plasma membrane-associated proteins to the cytoplasm. For example, it is known that one of the src family of protein tyrosine kinases, pp60c-src (Walker et al (1993), J. Biol Chem. 268:19552-19558) translocates from the plasma membrane to the cytoplasm upon stimulation of fibroblasts with platelet-derived growth factor (PDGF). Additionally, the targets for screening can themselves be converted into fluorescence- based reagents that report molecular changes including ligand-binding and post- translocational modifications.

Claims

We claim:
1. An -automated method for identifying compounds that modify transcription factor activation comprising: a) providing an aπay of locations containing multiple cells to be treated with a test compound, wherein the cells possess a luminescently labeled transcription factor, wherein the luminescently labeled transcription factor translocates from the cytoplasm to the nucleus upon activation, and wherein the luminescently labeled transcription factor is present in the cells either before, together with, or after treatment with the test compound; b) contacting the cells with the test compound c) scanning multiple cells in each of the locations containing cells to obtain luminescent signals from the luminescently labeled transcription factor in subcellular compartments of the cells being analyzed, wherein the subcellular compartments comprise the cell nucleus and the cell cytoplasm; d) converting the luminescent signals into digital data; and e) utilizing the digital data to automatically make measurements, wherein the measurements are used to automatically calculate changes in the distribution of the luminescently labeled transcription factor within or between the cell nucleus and the cell cytoplasm of the cells being analyzed, and wherein the change in distribution conelates to modification of transcription factor activity induced by the test compound.
2. The method of claim 1 further comprising scanning multiple cells in each of the locations containing cells in a high throughput mode, and selectively scanning only a subset of the locations containing cells in a high content mode to obtain luminescent signals from the luminescently labeled transcription factor in subcellular compartments of the cells-being analyzed.
3. The method of claim 1 wherein the measurements comprise determining one or more of the following: a) a total or average luminescence intensity of the luminescently labeled transcription factor on or in the cell nucleus; b) a total or average luminescence intensity of the luminescently labeled transcription factor outside the cell nucleus, representing the cell cytoplasm; c) an area of the cell nucleus; or d) an area of the cytoplasm.
4. The method of claim 3 wherein the calculated changes comprise one or more of the following: a) changes in the total or average luminescence intensity of the luminescently labeled transcription factor on or in the cell nucleus of the cells being analyzed; b) changes in the total or average luminescence intensity of the luminescently labeled transcription factor in the cell cytoplasm of the cells being analyzed; c) changes in the ratio of the total luminescence intensity or average luminescence intensity of the cytoplasm to total luminescence intensity or average luminescence intensity on or in the nucleus; or d) changes in the difference of the total luminescence intensity or average luminescence intensity of the cytoplasm and the total luminescence intensity or average luminescence intensity on or in the nucleus.
5. The method of claim 1 wherein the transcription factor of interest is luminescently labeled by contacting the cell with a luminescently labeled antibody or antibody fragment.
6. The method of claim 1 where the transcription factor of interest is selected from the group consisting of c-fos and fos homologs, c-jun and c-jun homologs, NF-KB, NFAT, and STATs.
7. A kit for identifying compounds that modify transcription factor activation comprising: a) a primary antibody that specifically binds to a transcription factor of interest; and b) instructions for using the primary antibody to identify compounds that modify transcription factor activation in a cell of interest, according to the method of claim 1.
8. The kit of claim 7 wherein the primary antibody is luminescently labeled.
9. The kit of claim 7 further comprising a secondary antibody that can detect the primary antibody.
10. The-kit of claim 9 wherein the secondary antibody is luminescently labeled.
11. The kit of claim 7 further comprising at least one of the following: a) cells that express the transcription factor of interest; or b) a compound that is known to modify activation of the transcription factor of interest.
12. The kit of claim 7 where the transcription factor is selected from the group consisting of c-fos and fos homologs, c-jun and c-jun homologs, NF-KB, NFAT, and
STATs.
13. A kit for identifying compounds that modify transcription factor activation comprising: a) an expression vector comprising a nucleic acid encoding a transcription factor of interest that translocates from the cytoplasm to the nucleus upon activation; and b) instructions for using the expression vector to identify compounds that modify transcription factor activation in a cell of interest, according to the method of claim 1.
14. The kit of claim 13 wherein the expression vector further comprises a nucleic acid encoding a luminescent protein, wherein the luminescent protein and the transcription factor of interest are expressed as a fusion protein.
15. The kit of claim 13 further comprising at least one of the following: a) _ an antibody or fragment thereof that specifically binds to the transcription factor of interest; b) cells that are transfected with the expression vector; or c) at least one compound that is known to modify activation of the transcription factor of interest.
16. The kit of claim 13 where the transcription factor is selected from the group consisting of c-fos and fos homologs, c-jun and c-jun homologs, NF-KB, NFAT, and STATs.
17. A kit for identifying compounds that modify transcription factor activation comprising: a) an isolated protein comprising a luminescently labeled transcription factor; and b) instructions for using the luminescently labeled transcription factor to identify compounds that modify transcription factor activation in a cell of interest, according to the method of claim 1.
18. The kit of claim 17 further comprising at least one compound that is known to modify activation of the transcription factor of interest.
19. The kit of claim 17 where the transcription factor is selected from the group consisting of c-fos and fos homologs, c-jun and c-jun homologs, NF-KB, NFAT, and STATs.
20. An automated method for identifying compounds that modify protein kinase activation comprising: a) providing an anay of locations containing multiple cells to be treated with a test compound, wherein the cells possess a luminescently labeled protein kinase or protein kinase substrate, wherein the luminescently labeled protein kinase or protein kinase substrate translocates from the cytoplasm to the nucleus upon activation of the protein kinase, and wherein the luminescently labeled protein kinase or protein kinase substrate is present in the cells either before, together with, or after treatment with the test compound; b) contacting the cells with the test compound c) scanning multiple cells in each of the locations containing cells to obtain luminescent signals from the luminescently labeled protein kinase or protein kinase substrate, in subcellular compartments of the cells being analyzed, wherein the subcellular compartments comprise the cell nucleus and the cell cytoplasm; d) converting the luminescent signals into digital data; and e) utilizing the digital data to automatically make measurements, wherein the measurements are used to automatically calculate changes in the distribution of the luminescently labeled protein kinase or protein kinase substrate within or between the cell nucleus and the cell cytoplasm of the cells being analyzed, and wherein the change in distribution conelates to modification of protein kinase activity induced by the test compound.
21. The method of claim 20 further comprising scanning multiple cells in each of the locations containing cells in a high throughput mode, and selectively scanning only a subset of the locations containing cells in a high content mode to obtain luminescent signals from the luminescently labeled protein kinase or protein kinase substrate in subcellular compartments of the cells being analyzed.
22. The method of claim 20 wherein the measurements comprise determining one or more of the following: a) an total or average luminescence intensity of the luminescently labeled protein kinase or protein kinase substrate on or in the cell nucleus; b) an total or average luminescence intensity of the luminescently labeled protein kinase or protein kinase substrate outside the cell nucleus, representing the cell cytoplasm; c) an area of the cell nucleus; or d) an area of the cytoplasm.
23. The method of claim 22 wherein the calculated changes comprise one or more of the following: a) changes in the total or average luminescence intensity of the luminescently labeled protein kinase or protein kinase substrate on or in the cell nucleus of the cells being analyzed; b) changes in the total or average luminescence intensity of the luminescently labeled protein kinase or protein kinase substrate in the cell cytoplasm of the cells being analyzed; c) changes in the ratio of the total luminescence intensity or average luminescence intensity of the cytoplasm to total luminescence intensity or average luminescence intensity on or in the nucleus; or d) changes in the difference of the total luminescence intensity or average luminescence intensity of the cytoplasm and the total luminescence intensity or average luminescence intensity on or in the nucleus.
24. The method of claim 20 wherein the protein kinase or protein kinase substrate of interest is luminescently labeled by contacting the cell with a luminescently labeled antibody or antibody fragment.
25. The method of claim 20 where the protein kinase of interest is selected from the group consisting of extracellular signal-regulated protein kinases (ERKs), c-Jun amino- terminal kinases (JNKs), Fos regulating protein kinases (FRKs), p38 mitogen activated protein kinase (p38MAPK); protein kinase A (PKA), and mitogen activated protein kinase kinases (MAPKKs).
26. A kit for identifying compounds that modify protein kinase activation comprising: a) a primary antibody that specifically binds to a protein kinase or a protein kinase substrate of interest; and b) instructions for using the antibody to identify compounds that modify protein kinase activation in a cell of interest, according to the method of claim 20.
27. The kit of claim 26 wherein the primary antibody is luminescently labeled.
I ll
28. The kit of claim 26 further comprising a secondary antibody that can detect the primary antibody.
29. The kit of claim 28 wherein the secondary antibody is luminescently labeled.
30. The kit of claim 26 further comprising at least one of the following: a) cells that express the protein kinase or protein kinase substrate of interest; or b) a compound that is known to modify activation of the protein kinase of interest.
31. The kit of claim 26 where the protein kinase is selected from the group consisting of extracellular signal-regulated protein kinases (ERKs), c-Jun amino- terminal kinases (JNKs), Fos regulating protein kinases (FRKs), p38 mitogen activated protein kinase (p38MAPK); protein kinase A (PKA), and mitogen activated protein kinase kinases (MAPKKs).
32. A kit for identifying compounds that modify protein kinase activation comprising: a) an expression vector comprising a nucleic acid encoding a protein kinase or a protein kinase substrate of interest that translocates from the cytoplasm to the nucleus upon activation; and b) instructions for using the expression vector to identify compounds that modify protein kinase activation in a cell of interest, according to the method of claim 20.
33. The kit of claim 32 wherein the expression vector further comprises a nucleic acid encoding a luminescent protein, wherein the luminescent protein and the protein kinase or protein kinase substrate of interest are expressed as a fusion protein.
34. The kit of claim 32 further comprising at least one of the following: a) cells that are transfected with the expression vector; b) an antibody or fragment thereof that specifically binds to the protein kinase or protein kinase substrate of interest; or c) a compound that is known to modify activation of the protein kinase of interest.
35. The kit of claim 32 where the protein kinase is selected from the group consisting of extracellular signal-regulated protein kinases (ERKs), c-Jun amino- terminal kinases (JNKs), Fos regulating protein kinases (FRKs), p38 mitogen activated protein kinase (p38MAPK); protein kinase A (PKA), and mitogen activated protein kinase kinases (MAPKKs).
36. A kit for identifying compounds that modify protein kinase activation comprising: a) an isolated protein comprising a luminescently labeled protein kinase or luminescently labeled protein kinase substrate; and b) instructions for using the luminescently labeled protein kinase or luminescently labeled protein kinase substrate, according to the method of claim 20.
37. The kit of claim 36 further comprising a compound that is known to modify activation of the protein kinase.
38. The kit of claim 36 where the protein kinase is selected from the group consisting of extracellular signal-regulated protein kinases (ERKs), c-Jun amino- terminal kinases (JNKs), Fos regulating protein kinases (FRKs), p38 mitogen activated protein kinase (p38MAPK); protein kinase A (PKA), and mitogen activated protein kinase kinases (MAPKKs).
39. A machine readable storage medium comprising a program containing a set of instructions for causing a cell screening system to execute the method of claim 1 wherein the cell screening system comprises an optical system with a stage adapted for holding a plate containing cells, a digital camera, a means for directing fluorescence or luminescence emitted from the cells to the digital camera, and a computer means for receiving and processing the digital data from the digital camera.
40. A machine readable storage medium comprising a program containing a set of instructions for causing a cell screening system to execute the method of claim 20 wherein the cell screening system comprises an optical system with a stage adapted for holding a plate containing cells, a digital camera, a means for directing fluorescence or luminescence emitted from the cells to the digital camera, and a computer means for receiving and processing the digital data from the digital camera.
41. An automated method for analyzing compounds that modify cellular moφhology comprising: a) providing an aπay of locations which contain multiple cells that are to be contacted with a test compound, wherein the cells possess one or more luminescent reporter molecules, and wherein the one or more luminescent reporter molecules can be expressed by the cells, or added to the cells either before, together with, or after the cells are contacted with the test compound; b) contacting the cells with the test compound; c) scanning multiple cells in each of the locations containing cells to obtain luminescent signals from the luminescent reporter molecules in subcellular compartments of the cells, wherein the subcellular compartments comprise the cell nucleus, cell cytoplasm, cell membrane, and cell cytoskeleton; d) converting the luminescent signals into digital data; and e) utilizing the digital data to automatically make measurements, wherein the measurements are used to automatically calculate changes in the distribution of the luminescent reporter molecules within or between the subcellular compartments, and wherein the calculated changes coπelate with changes in cellular moφhology induced by the test compound.
42 The method of claim 41 comprising scanning multiple cells in each of the locations containing cells in a high throughput mode, and selectively scanning only a subset of the locations containing cells in a high content mode to obtain luminescent signals from the luminescently labeled reporter molecules in subcellular compartments of the cells being analyzed.
43. The method of claim 41 further comprising at least one of the following: a. acquiring an image of the cell nuclei; b. acquiring an image of the cell cytoplasm; c. acquiring an image of the cell membrane; or d. acquiring an image of the cell cytoskeleton.
44. The method of claim 43 wherein the measurements comprise one or more of the following: a. an aggregate whole nucleus area; b. an average whole nucleus area; c. a total cytoplasmic area; d. an aggregate cytoplasmic intensity; e. a cytoplasmic area per cell nucleus; f. a cytoplasmic intensity per cell nucleus; g. an average cell cytoplasm intensity; h. a cell cytoplasm-cell nucleus ratio; i. a measure of actin cytoskeleton stracture; or j. an average cell area.
45. The method of claim 44 wherein the calculated changes comprise one or more of the following: a. changes in the aggregate whole nucleus area; b. changes in the average whole nucleus area; c. changes in the total cytoplasmic area; d. changes in the aggregate cytoplasmic intensity; e. changes in the cytoplasmic area per cell nucleus; f. changes in the cytoplasmic intensity per cell nucleus; g. changes in the average cell cytoplasm intensity; h. changes in the cell cytoplasm-cell nucleus ratio; i. changes in the measure of actin cytoskeleton stracture; or j. changes in the average cell area.
46. The method of claim 41 wherein sub-regions of the aπay of locations containing multiple cells are sampled multiple times at intervals to provide kinetic measurement of changes in cellular moφhology.
47. The method of claim 41 wherein the cells possess a luminescent reporter molecule selected from the group consisting of cytoskeletal markers, cytosolic volume markers, and cell surface markers.
48. The method of claim 47 wherein the luminescent reporter molecule selectively detects actin filaments.
49. A kit for identifying stimuli that modify cellular moφhology, comprising: a. a luminescent compound that specifically labels the cell membrane, cytoplasm, or cytoskeleton; and b. instructions for using the luminescent compound to identify test compounds that modify cellular moφhology according to the method of claim 41.
50. The kit of claim 49 further comprising at least one of the following: a) a luminescent compound that specifically labels the cell nucleus; or b) at least one compound that is known to modify cellular moφhology.
51. A machine readable storage medium comprising a program containing a set of instructions for causing a cell screening system to execute the method of claim 41 wherein the cell screening system comprises an optical system with a stage adapted for holding a plate containing cells, a digital camera, a means for directing fluorescence or luminescence emitted from the cells to the digital camera, and a computer means for receiving and processing the digital data from the digital camera.
52. An automated method for identifying compounds that modify microtubule structure comprising: a) providing an aπay of locations containing multiple cells to be treated with a test compound, wherein the cells possess a luminescently labeled microtubule- labeling molecule that is expressed by or added to the cells either before, together with, or after contacting the cells with the test compound; b) contacting the cells with the test compound; c) scanning multiple cells in each of the locations containing cells to obtain luminescent signals from the luminescently labeled microtubule-labeling molecule in the cell cytoplasm or the cell cytoskeleton; d) converting the luminescent signals into digital data; and e) utilizing the digital data to automatically make measurements, wherein the measurements are used to automatically calculate changes in the distribution of the luminescently labeled microtubule-labeling molecule within the cell cytoplasm or cell cytoskeleton, and wherein the calculated changes in the distribution of the luminescently labeled microtubule-labeling molecule within the cell cytoplasm or the cell cytoskeleton conelate with changes in microtubule stracture induced by the test compound.
53. The method of claim 52 further comprising scanning multiple cells in each of the locations containing cells in a high throughput mode, and selectively scanning only a subset of the locations containing cells in a high content mode to obtain luminescent signals from the luminescently labeled microtubule-labeling molecule in subcellular compartments of the cells being analyzed.
54. The method of claim 52 wherein the luminescently labeled microtubule-labeling
molecule is selected from the group consisting of α and β tubulin isoforms, MAP4, and
antibodies that specifically recognize a protein selected from the group consisting of α
and β tubulin isoforms, MAP 4, MAP 2, and tau.
55. The method of claim 52 wherein the measurements comprise determining at least one of the following: a. edge strength; b. cell area; c. cell size; d. cell shape; e. integrated luminescence intensity; and f. cell microtubule moφhology; g. punctate distribution of the cell microtubule cytoskeleton; h. interconnectivity or branching of the cell microtubule cytoskeleton; i. microtubule length or width; j. microtubule aggregation; k. microtubule depolymerization; or
1. microtubule reorganization.
56. The method of claim 55 wherein the calculated changes comprise determining at least one of the following: a. changes in edge strength; b. changes in cell area; c. changes in cell size; d. changes in cell shape; e. changes in integrated luminescence intensity; f. changes in cell microtubule moφhology; g. changes in punctate distribution of the cell microtubule cytoskeleton; h. changes in interconnectivity or branching of the cell microtubule cytoskeleton; i. changes in microtubule length or width; j. " changes in microtubule aggregation; k. changes in microtubule depolymerization; or
1. changes in microtubule reorganization.
57. The method of claim 52 wherein sub-regions of the aπay of locations containing multiple cells are sampled multiple times at intervals to provide kinetic measurement of changes in microtubule structure.
58. A kit for automated analysis of microtubule stracture comprising: a. an expression vector comprising a nucleic acid that encodes a microtubule binding protein; and b. instructions for using the expression vector for carrying out the method of claim 52.
59. The kit of claim 58 wherein the expression vector further comprises a nucleic acid that encodes a luminescent protein, wherein the microtubule binding protein and the luminescent protein thereof are expressed as a fusion protein.
60. The kit of claim 58 further comprising at least one of the following: a) cells that are transfected with the expression vector; or b) a compound that is known to modify microtubule stracture.
61. A kit for automated analysis of microtubule stracture comprising: a. a primary antibody that specifically binds to a microtubule-labeling molecule of interest; and b. instructions for using the expression vector for carrying out the method of claim 52.
62. The kit of claim 61 wherein the primary antibody is luminescently labeled.
63. The kit of claim 61 further comprising a secondary antibody that can detect the primary antibody.
64. The kit of claim 63 wherein the secondary antibody is luminescently labeled.
65. The kit of claim 61 further comprising at least one of the following: a) cells that express the microtubule labeling molecule of interest; or b) a compound that is known to modify microtubule stracture.
66. The kit of claim 61 where the microtubule labeling molecule is selected from
the group consisting of α and β tubulin isoforms, MAP 4, MAP 2, and tau.
67. A kit for automated analysis of microtubule structure comprising: a. an isolated protein comprising a luminescently labeled microtubule- labeling molecule; and and b. instructions for using the luminescently labeled microtubule-labeling molecule for carrying out the method of claim 52.
68. The kit of claim 67 wherein the luminescently labeled microtubule-labeling molecule is selected from the group consisting of α and β tubulin isoforms, MAP 4,
MAP 2, and tau.
69. A machine readable storage medium comprising a program containing a set of instructions for causing a cell screening system to execute the method of claim 52 wherein the cell screening system comprises an optical system with a stage adapted for holding a plate containing cells, a digital camera, a means for directing fluorescence or luminescence emitted from the cells to the digital camera, and a computer means for receiving and processing the digital data from the digital camera.
PCT/US1999/021561 1998-09-18 1999-09-17 A system for cell-based screening WO2000017643A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
AU60485/99A AU6048599A (en) 1998-09-18 1999-09-17 A system for cell-based screening
CA002344567A CA2344567A1 (en) 1998-09-18 1999-09-17 A system for cell-based screening
EP99969495A EP1114320A2 (en) 1998-09-18 1999-09-17 A system for cell-based screening
JP2000571253A JP2002525603A (en) 1998-09-18 1999-09-17 System for cell-based screening

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10097398P 1998-09-18 1998-09-18
US60/100,973 1998-09-18

Publications (4)

Publication Number Publication Date
WO2000017643A2 true WO2000017643A2 (en) 2000-03-30
WO2000017643A8 WO2000017643A8 (en) 2000-06-22
WO2000017643A9 WO2000017643A9 (en) 2000-08-17
WO2000017643A3 WO2000017643A3 (en) 2000-10-12

Family

ID=22282473

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/021561 WO2000017643A2 (en) 1998-09-18 1999-09-17 A system for cell-based screening

Country Status (5)

Country Link
EP (1) EP1114320A2 (en)
JP (1) JP2002525603A (en)
AU (1) AU6048599A (en)
CA (1) CA2344567A1 (en)
WO (1) WO2000017643A2 (en)

Cited By (65)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000070528A2 (en) * 1999-05-14 2000-11-23 Cytokinetics, Inc. Method and apparatus for predictive cellular bioinformatics
WO2000079241A2 (en) * 1999-06-21 2000-12-28 Cellomics, Inc. A system for cell-based screening
WO2001011341A2 (en) * 1999-08-05 2001-02-15 Cellomics, Inc. A system for cell-based screening
WO2001096597A2 (en) * 2000-06-13 2001-12-20 Glaxo Group Limited High throughput screening of compounds for biological activity
WO2002067182A2 (en) * 2001-02-20 2002-08-29 Cytokinetics, Inc. Characterizing biological stimuli by response curves
WO2002073200A1 (en) 2001-03-12 2002-09-19 Cellomics, Inc. Methods to increase the capacity of high content cell-based screening assays
US6599694B2 (en) 2000-12-18 2003-07-29 Cytokinetics, Inc. Method of characterizing potential therapeutics by determining cell-cell interactions
EP1333099A1 (en) * 2000-11-06 2003-08-06 Ajinomoto Co., Inc. Method of searching for gene encoding nuclear transport protein
WO2003071270A2 (en) * 2002-02-19 2003-08-28 University Of Bristol Screening method
US6614452B1 (en) 1999-11-15 2003-09-02 Xenogen Corporation Graphical user interface for in-vivo imaging
US6615141B1 (en) 1999-05-14 2003-09-02 Cytokinetics, Inc. Database system for predictive cellular bioinformatics
US6651008B1 (en) 1999-05-14 2003-11-18 Cytokinetics, Inc. Database system including computer code for predictive cellular bioinformatics
US6683735B2 (en) 2000-11-17 2004-01-27 Universal Imaging Corporation Rapidly changing dichroic beamsplitter in epifluorescent microscopes
US6716588B2 (en) 1999-12-09 2004-04-06 Cellomics, Inc. System for cell-based screening
US6759206B1 (en) 1997-02-27 2004-07-06 Cellomics, Inc. System for cell-based screening
US6775567B2 (en) 2000-02-25 2004-08-10 Xenogen Corporation Imaging apparatus
EP1517134A1 (en) * 2003-09-22 2005-03-23 Yokogawa Electric Corporation Drug screening method and device using a Nipkow confocal scanner
US6875578B2 (en) 1997-02-27 2005-04-05 Cellomics, Inc. System for cell-based screening
WO2005031322A1 (en) * 2003-09-26 2005-04-07 Nikon Corporation Environment holding apparatus and environment control type analyzer
US6919919B2 (en) 2002-02-06 2005-07-19 Xenogen Corporation Light calibration device for use in low level light imaging systems
US6956961B2 (en) 2001-02-20 2005-10-18 Cytokinetics, Inc. Extracting shape information contained in cell images
US6986993B1 (en) 1999-08-05 2006-01-17 Cellomics, Inc. System for cell-based screening
US6999607B2 (en) 2001-02-20 2006-02-14 Cytokinetics, Inc. Method and apparatus for automated cellular bioinformatics
US7113217B2 (en) 2001-07-13 2006-09-26 Xenogen Corporation Multi-view imaging apparatus
US7116354B2 (en) 2001-06-20 2006-10-03 Xenogen Corporation Absolute intensity determination for a light source in low level light imaging systems
US7151847B2 (en) 2001-02-20 2006-12-19 Cytokinetics, Inc. Image analysis of the golgi complex
US7177024B2 (en) 2002-02-22 2007-02-13 Xenogen Corporation Bottom fluorescence illumination assembly for an imaging apparatus
US7218764B2 (en) 2000-12-04 2007-05-15 Cytokinetics, Inc. Ploidy classification method
US7235353B2 (en) 2003-07-18 2007-06-26 Cytokinetics, Inc. Predicting hepatotoxicity using cell based assays
US7246012B2 (en) 2003-07-18 2007-07-17 Cytokinetics, Inc. Characterizing biological stimuli by response curves
US7250298B2 (en) 2004-04-07 2007-07-31 The University Of Chicago Monomeric red fluorescent proteins
WO2007085923A2 (en) 2006-01-25 2007-08-02 Evrogen, Ip Novel fluorescent proteins and methods for using same
US7298415B2 (en) 2001-07-13 2007-11-20 Xenogen Corporation Structured light imaging apparatus
US7323318B2 (en) 2004-07-15 2008-01-29 Cytokinetics, Inc. Assay for distinguishing live and dead cells
US7331341B2 (en) 2002-02-20 2008-02-19 Xenogen Corporation Living specimen induction chamber
US7390458B2 (en) 2000-10-13 2008-06-24 Irm Llc High throughput processing system and method of using
US7403812B2 (en) 2001-05-17 2008-07-22 Xenogen Corporation Method and apparatus for determining target depth, brightness and size within a body region
US7417131B2 (en) 2005-11-04 2008-08-26 Evrogen Joint Stock Company Modified green fluorescent proteins and methods for using same
US7474398B2 (en) 2002-02-22 2009-01-06 Xenogen Corporation Illumination system for an imaging apparatus with low profile output device
US7474399B2 (en) 2002-02-22 2009-01-06 Xenogen Corporation Dual illumination system for an imaging apparatus and method
US7496277B2 (en) 2003-06-02 2009-02-24 Disney Enterprises, Inc. System and method of programmatic window control for consumer video players
US7565247B1 (en) 1997-02-27 2009-07-21 Cellomics, Inc. Method for acquisition, storage, and retrieval of cell screening data on a computer system
US7581191B2 (en) 1999-11-15 2009-08-25 Xenogen Corporation Graphical user interface for 3-D in-vivo imaging
EP2100958A1 (en) 2002-11-12 2009-09-16 Zakrytoe Aktsionernoe Obschestvo 'Evrogen' Fluorescent proteins and chromoproteins from non-aequorea hydrozoa species and methods for using same
US7629573B2 (en) 2002-02-06 2009-12-08 Xenogen Corporation Tissue phantom calibration device for low level light imaging systems
US7649185B2 (en) 2002-02-06 2010-01-19 Xenogen Corporation Fluorescent phantom device
US7678893B2 (en) 2002-12-26 2010-03-16 Zakrytoe Aktsionernoe Obschestvo “Evrogen” Fluorescent proteins from Copepoda species and methods for using same
US7817840B2 (en) 2003-07-18 2010-10-19 Cytokinetics, Inc. Predicting hepatotoxicity using cell based assays
US7838287B2 (en) 2001-01-25 2010-11-23 Evolva Sa Library of a collection of cells
US7853411B2 (en) 1997-02-27 2010-12-14 Cellomics, Inc. System for cell-based screening
EP2308974A2 (en) 1999-10-14 2011-04-13 Clontech Laboratories Inc. Anthozoa derived chromo/fluoroproteins and methods for using the same
US8008459B2 (en) 2001-01-25 2011-08-30 Evolva Sa Concatemers of differentially expressed multiple genes
US8044996B2 (en) 2005-05-11 2011-10-25 Xenogen Corporation Surface construction using combined photographic and structured light information
US8050868B2 (en) 2001-03-26 2011-11-01 Cellomics, Inc. Methods for determining the organization of a cellular component of interest
US8114615B2 (en) 2006-05-17 2012-02-14 Cernostics, Inc. Method for automated tissue analysis
US20120300194A1 (en) * 2007-02-13 2012-11-29 Bti Holdings, Inc. Universal multidetection system for microplates
US8563703B2 (en) 2006-01-25 2013-10-22 Evrogen IP Joint Stock Company Fluorescent proteins and methods for using same
US8664471B2 (en) 2001-12-19 2014-03-04 The University Of Chicago Rapidly maturing fluorescent proteins and methods for using the same
US8680235B2 (en) 2006-09-22 2014-03-25 Stowers Institute For Medical Research Branchiostoma derived fluorescent proteins
US8679749B2 (en) 2007-11-01 2014-03-25 The University Of Chicago Red fluorescent proteins with enhanced bacterial expression, increased brightness and reduced aggregation
JPWO2013094307A1 (en) * 2011-12-22 2015-04-27 孝広 落谷 Exosome analysis method, reagent for exosome analysis, and exosome analyzer
US10018631B2 (en) 2011-03-17 2018-07-10 Cernostics, Inc. Systems and compositions for diagnosing Barrett's esophagus and methods of using the same
CN110993030A (en) * 2019-11-21 2020-04-10 复旦大学 Method and system for monitoring RNA (ribonucleic acid) nuclear flux in single living cell
US10775308B2 (en) 2006-08-24 2020-09-15 Xenogen Corporation Apparatus and methods for determining optical tissue properties
US11730370B2 (en) 2006-08-24 2023-08-22 Xenogen Corporation Spectral unmixing for in-vivo imaging

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6135795B2 (en) * 2015-04-22 2017-05-31 小野薬品工業株式会社 Lead compound extraction method, drug discovery target selection method, scatter diagram generation device, data visualization method and visualization device
US20170241911A1 (en) * 2016-02-22 2017-08-24 Miltenyi Biotec Gmbh Automated analysis tool for biological specimens

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997020931A1 (en) * 1995-12-08 1997-06-12 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Method and compositions for monitoring dna binding molecules in living cells
WO1998038490A1 (en) * 1997-02-27 1998-09-03 Cellomics, Inc. A system for cell-based screening

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997020931A1 (en) * 1995-12-08 1997-06-12 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Method and compositions for monitoring dna binding molecules in living cells
WO1998038490A1 (en) * 1997-02-27 1998-09-03 Cellomics, Inc. A system for cell-based screening

Cited By (109)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7853411B2 (en) 1997-02-27 2010-12-14 Cellomics, Inc. System for cell-based screening
US7565247B1 (en) 1997-02-27 2009-07-21 Cellomics, Inc. Method for acquisition, storage, and retrieval of cell screening data on a computer system
US6875578B2 (en) 1997-02-27 2005-04-05 Cellomics, Inc. System for cell-based screening
US6759206B1 (en) 1997-02-27 2004-07-06 Cellomics, Inc. System for cell-based screening
WO2000070528A2 (en) * 1999-05-14 2000-11-23 Cytokinetics, Inc. Method and apparatus for predictive cellular bioinformatics
US6615141B1 (en) 1999-05-14 2003-09-02 Cytokinetics, Inc. Database system for predictive cellular bioinformatics
US6651008B1 (en) 1999-05-14 2003-11-18 Cytokinetics, Inc. Database system including computer code for predictive cellular bioinformatics
US6631331B1 (en) 1999-05-14 2003-10-07 Cytokinetics, Inc. Database system for predictive cellular bioinformatics
US6738716B1 (en) 1999-05-14 2004-05-18 Cytokinetics, Inc. Database system for predictive cellular bioinformatics
US6743576B1 (en) 1999-05-14 2004-06-01 Cytokinetics, Inc. Database system for predictive cellular bioinformatics
WO2000070528A3 (en) * 1999-05-14 2001-03-08 Cytokinetics Inc Method and apparatus for predictive cellular bioinformatics
WO2000079241A2 (en) * 1999-06-21 2000-12-28 Cellomics, Inc. A system for cell-based screening
WO2000079241A3 (en) * 1999-06-21 2002-05-10 Cellomics Inc A system for cell-based screening
WO2001011341A2 (en) * 1999-08-05 2001-02-15 Cellomics, Inc. A system for cell-based screening
WO2001011341A3 (en) * 1999-08-05 2001-07-05 Cellomics Inc A system for cell-based screening
US8062856B2 (en) 1999-08-05 2011-11-22 Cellomics, Inc. System for cell-based screening
US6986993B1 (en) 1999-08-05 2006-01-17 Cellomics, Inc. System for cell-based screening
EP2308974A2 (en) 1999-10-14 2011-04-13 Clontech Laboratories Inc. Anthozoa derived chromo/fluoroproteins and methods for using the same
US6614452B1 (en) 1999-11-15 2003-09-02 Xenogen Corporation Graphical user interface for in-vivo imaging
US7299420B2 (en) 1999-11-15 2007-11-20 Xenogen Corporation Graphical user interface for in-vivo imaging
US8734342B2 (en) 1999-11-15 2014-05-27 Xenogen Corporation Graphical user interface for in-vivo imaging
US7581191B2 (en) 1999-11-15 2009-08-25 Xenogen Corporation Graphical user interface for 3-D in-vivo imaging
US7765487B2 (en) 1999-11-15 2010-07-27 Xenogen Corporation Graphical user interface for in-vivo imaging
US6716588B2 (en) 1999-12-09 2004-04-06 Cellomics, Inc. System for cell-based screening
US6775567B2 (en) 2000-02-25 2004-08-10 Xenogen Corporation Imaging apparatus
US7383078B2 (en) 2000-02-25 2008-06-03 Xenogen Corporation Imaging apparatus with heating element
WO2001096597A2 (en) * 2000-06-13 2001-12-20 Glaxo Group Limited High throughput screening of compounds for biological activity
WO2001096597A3 (en) * 2000-06-13 2003-09-12 Glaxo Group Ltd High throughput screening of compounds for biological activity
JP2004503249A (en) * 2000-06-13 2004-02-05 グラクソ グループ リミテッド High-throughput method for screening candidate compounds for biological activity
US7390458B2 (en) 2000-10-13 2008-06-24 Irm Llc High throughput processing system and method of using
US7195869B2 (en) 2000-11-06 2007-03-27 Ajinomoto Co., Inc. Method of searching for gene encoding nuclear transport protein
EP1333099A1 (en) * 2000-11-06 2003-08-06 Ajinomoto Co., Inc. Method of searching for gene encoding nuclear transport protein
EP1333099B1 (en) * 2000-11-06 2008-10-29 Ajinomoto Co., Inc. Method of searching for gene encoding nuclear transport protein
US6683735B2 (en) 2000-11-17 2004-01-27 Universal Imaging Corporation Rapidly changing dichroic beamsplitter in epifluorescent microscopes
US6927903B2 (en) 2000-11-17 2005-08-09 Universal Imaging Corporation Rapidly changing dichroic beamsplitter
US7218764B2 (en) 2000-12-04 2007-05-15 Cytokinetics, Inc. Ploidy classification method
US6599694B2 (en) 2000-12-18 2003-07-29 Cytokinetics, Inc. Method of characterizing potential therapeutics by determining cell-cell interactions
US8008459B2 (en) 2001-01-25 2011-08-30 Evolva Sa Concatemers of differentially expressed multiple genes
US7838287B2 (en) 2001-01-25 2010-11-23 Evolva Sa Library of a collection of cells
GB2389120B (en) * 2001-02-20 2005-01-05 Cytokinetics Inc Characterizing biological stimuli by response curves
US6999607B2 (en) 2001-02-20 2006-02-14 Cytokinetics, Inc. Method and apparatus for automated cellular bioinformatics
US7151847B2 (en) 2001-02-20 2006-12-19 Cytokinetics, Inc. Image analysis of the golgi complex
WO2002067182A2 (en) * 2001-02-20 2002-08-29 Cytokinetics, Inc. Characterizing biological stimuli by response curves
US7657076B2 (en) 2001-02-20 2010-02-02 Cytokinetics, Inc. Characterizing biological stimuli by response curves
US6956961B2 (en) 2001-02-20 2005-10-18 Cytokinetics, Inc. Extracting shape information contained in cell images
US7269278B2 (en) 2001-02-20 2007-09-11 Cytokinetics, Inc. Extracting shape information contained in cell images
GB2389120A (en) * 2001-02-20 2003-12-03 Cytokinetics Inc Characterizing Biological Stimuli by Response Curves
WO2002067182A3 (en) * 2001-02-20 2003-07-03 Cytokinetics Inc Characterizing biological stimuli by response curves
WO2002073200A1 (en) 2001-03-12 2002-09-19 Cellomics, Inc. Methods to increase the capacity of high content cell-based screening assays
US8050868B2 (en) 2001-03-26 2011-11-01 Cellomics, Inc. Methods for determining the organization of a cellular component of interest
US7403812B2 (en) 2001-05-17 2008-07-22 Xenogen Corporation Method and apparatus for determining target depth, brightness and size within a body region
US7663664B2 (en) 2001-06-20 2010-02-16 Xenogen Corporation Absolute intensity determination for a fluorescent light source in low level light imaging systems
US7116354B2 (en) 2001-06-20 2006-10-03 Xenogen Corporation Absolute intensity determination for a light source in low level light imaging systems
US7113217B2 (en) 2001-07-13 2006-09-26 Xenogen Corporation Multi-view imaging apparatus
US7298415B2 (en) 2001-07-13 2007-11-20 Xenogen Corporation Structured light imaging apparatus
US8279334B2 (en) 2001-07-13 2012-10-02 Xenogen Corporation Structured light imaging apparatus
US7595838B2 (en) 2001-07-13 2009-09-29 Xenogen Corporation Multi-view imaging apparatus
US7589786B2 (en) 2001-07-13 2009-09-15 Xenogen Corporation Multi-view imaging of a sample in a box
US8664471B2 (en) 2001-12-19 2014-03-04 The University Of Chicago Rapidly maturing fluorescent proteins and methods for using the same
US6919919B2 (en) 2002-02-06 2005-07-19 Xenogen Corporation Light calibration device for use in low level light imaging systems
US7649185B2 (en) 2002-02-06 2010-01-19 Xenogen Corporation Fluorescent phantom device
US7629573B2 (en) 2002-02-06 2009-12-08 Xenogen Corporation Tissue phantom calibration device for low level light imaging systems
WO2003071270A2 (en) * 2002-02-19 2003-08-28 University Of Bristol Screening method
WO2003071270A3 (en) * 2002-02-19 2004-02-26 Univ Bristol Screening method
US7461652B2 (en) 2002-02-20 2008-12-09 Xenogen Corporation Imaging system with anesthesia system
US7503323B2 (en) 2002-02-20 2009-03-17 Xenogen Corporation Multiple output anesthesia system
US7331341B2 (en) 2002-02-20 2008-02-19 Xenogen Corporation Living specimen induction chamber
US7464707B2 (en) 2002-02-20 2008-12-16 Xenogen Corporation Anesthesia delivery device for use in a light imaging system
US7806082B2 (en) 2002-02-20 2010-10-05 Xenogen Corporation Living specimen induction chamber
US7474399B2 (en) 2002-02-22 2009-01-06 Xenogen Corporation Dual illumination system for an imaging apparatus and method
US7177024B2 (en) 2002-02-22 2007-02-13 Xenogen Corporation Bottom fluorescence illumination assembly for an imaging apparatus
US7474398B2 (en) 2002-02-22 2009-01-06 Xenogen Corporation Illumination system for an imaging apparatus with low profile output device
US7466418B2 (en) 2002-02-22 2008-12-16 Xenogen Corporation Fluorescence illumination assembly for an imaging apparatus and method
EP2100958A1 (en) 2002-11-12 2009-09-16 Zakrytoe Aktsionernoe Obschestvo 'Evrogen' Fluorescent proteins and chromoproteins from non-aequorea hydrozoa species and methods for using same
US7678893B2 (en) 2002-12-26 2010-03-16 Zakrytoe Aktsionernoe Obschestvo “Evrogen” Fluorescent proteins from Copepoda species and methods for using same
US7496277B2 (en) 2003-06-02 2009-02-24 Disney Enterprises, Inc. System and method of programmatic window control for consumer video players
US7235353B2 (en) 2003-07-18 2007-06-26 Cytokinetics, Inc. Predicting hepatotoxicity using cell based assays
US7246012B2 (en) 2003-07-18 2007-07-17 Cytokinetics, Inc. Characterizing biological stimuli by response curves
US7817840B2 (en) 2003-07-18 2010-10-19 Cytokinetics, Inc. Predicting hepatotoxicity using cell based assays
EP1517134A1 (en) * 2003-09-22 2005-03-23 Yokogawa Electric Corporation Drug screening method and device using a Nipkow confocal scanner
JPWO2005031322A1 (en) * 2003-09-26 2007-11-15 株式会社ニコン Environmental maintenance device and environmental control analyzer
WO2005031322A1 (en) * 2003-09-26 2005-04-07 Nikon Corporation Environment holding apparatus and environment control type analyzer
US8066962B2 (en) 2003-09-26 2011-11-29 Nikon Corporation Environment holding apparatus and environment control type analyzer
US7910714B2 (en) 2004-04-07 2011-03-22 The University Of Chicago Monomeric red fluorescent proteins
US7250298B2 (en) 2004-04-07 2007-07-31 The University Of Chicago Monomeric red fluorescent proteins
US7671185B2 (en) 2004-04-07 2010-03-02 The University Of Chicago Monomeric red fluorescent proteins
US7323318B2 (en) 2004-07-15 2008-01-29 Cytokinetics, Inc. Assay for distinguishing live and dead cells
US8044996B2 (en) 2005-05-11 2011-10-25 Xenogen Corporation Surface construction using combined photographic and structured light information
US7417131B2 (en) 2005-11-04 2008-08-26 Evrogen Joint Stock Company Modified green fluorescent proteins and methods for using same
US7605230B2 (en) 2005-11-04 2009-10-20 Evrogen Joint Stock Company Modified green fluorescent proteins and methods for using same
US8563703B2 (en) 2006-01-25 2013-10-22 Evrogen IP Joint Stock Company Fluorescent proteins and methods for using same
US8138320B2 (en) 2006-01-25 2012-03-20 Evrogen IP Joint Stock Company Fluorescent proteins and methods for using same
WO2007085923A2 (en) 2006-01-25 2007-08-02 Evrogen, Ip Novel fluorescent proteins and methods for using same
US7638615B1 (en) 2006-01-25 2009-12-29 Evrogen IP Joint Stock Company Fluorescent proteins and methods for using same
US8114615B2 (en) 2006-05-17 2012-02-14 Cernostics, Inc. Method for automated tissue analysis
US8597899B2 (en) 2006-05-17 2013-12-03 Cernostics, Inc. Method for automated tissue analysis
US11730370B2 (en) 2006-08-24 2023-08-22 Xenogen Corporation Spectral unmixing for in-vivo imaging
US10775308B2 (en) 2006-08-24 2020-09-15 Xenogen Corporation Apparatus and methods for determining optical tissue properties
US8680235B2 (en) 2006-09-22 2014-03-25 Stowers Institute For Medical Research Branchiostoma derived fluorescent proteins
US10072982B2 (en) 2007-02-13 2018-09-11 Biotek Instruments, Inc. Universal multidetection system for microplates
US9557217B2 (en) * 2007-02-13 2017-01-31 Bti Holdings, Inc. Universal multidetection system for microplates
US20120300194A1 (en) * 2007-02-13 2012-11-29 Bti Holdings, Inc. Universal multidetection system for microplates
US8679749B2 (en) 2007-11-01 2014-03-25 The University Of Chicago Red fluorescent proteins with enhanced bacterial expression, increased brightness and reduced aggregation
US10018631B2 (en) 2011-03-17 2018-07-10 Cernostics, Inc. Systems and compositions for diagnosing Barrett's esophagus and methods of using the same
EP2801822A4 (en) * 2011-12-22 2015-07-01 Exosome analysis method, reagent for use in analysis of exosome, and exosome analysis apparatus
JPWO2013094307A1 (en) * 2011-12-22 2015-04-27 孝広 落谷 Exosome analysis method, reagent for exosome analysis, and exosome analyzer
US11391741B2 (en) 2011-12-22 2022-07-19 Theoria Science Inc. Method of exosome analysis, reagent for exosome analysis, and analyzer for exosome
CN110993030A (en) * 2019-11-21 2020-04-10 复旦大学 Method and system for monitoring RNA (ribonucleic acid) nuclear flux in single living cell
CN110993030B (en) * 2019-11-21 2023-05-02 复旦大学 Method and system for monitoring nuclear flux of RNA in single living cell

Also Published As

Publication number Publication date
JP2002525603A (en) 2002-08-13
WO2000017643A8 (en) 2000-06-22
WO2000017643A3 (en) 2000-10-12
EP1114320A2 (en) 2001-07-11
CA2344567A1 (en) 2000-03-30
WO2000017643A9 (en) 2000-08-17
AU6048599A (en) 2000-04-10

Similar Documents

Publication Publication Date Title
EP1203214B1 (en) Optical system analysis of cells
US8062856B2 (en) System for cell-based screening
US6671624B1 (en) Machine readable storage media for detecting distribution of macromolecules between nucleus and cytoplasm in cells
US6727071B1 (en) System for cell-based screening
US6716588B2 (en) System for cell-based screening
US7117098B1 (en) Machine-readable storage medium for analyzing distribution of macromolecules between the cell membrane and the cell cytoplasm
WO2000017643A2 (en) A system for cell-based screening
US20040063162A1 (en) System for cell-based screening
AU759180B2 (en) A system for cell-based screening
US7853411B2 (en) System for cell-based screening
WO1998038490A9 (en) A system for cell-based screening
WO2000003246A2 (en) A system for cell-based screening
WO2000050872A9 (en) A system for cell-based screening
WO2001011341A2 (en) A system for cell-based screening
EP1185863A2 (en) Optical system analysis of cells
WO2000079241A2 (en) A system for cell-based screening
EP1439384B1 (en) A system for screening biological cells
AU773088B2 (en) A system for cell-based screening
MXPA01002684A (en) A system for cell-based screening

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AU CA JP MX NZ US

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: C1

Designated state(s): AU CA JP MX NZ US

AL Designated countries for regional patents

Kind code of ref document: C1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE

CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i

Free format text: DELETE "(63)"

AK Designated states

Kind code of ref document: C2

Designated state(s): AU CA JP MX NZ US

AL Designated countries for regional patents

Kind code of ref document: C2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE

COP Corrected version of pamphlet

Free format text: PAGES 1/24-24/24, DRAWINGS, REPLACED BY NEW PAGES 1/24-24/24; DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE

AK Designated states

Kind code of ref document: A3

Designated state(s): AU CA JP MX NZ US

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE

WWE Wipo information: entry into national phase

Ref document number: 510368

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: PA/a/2001/002684

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 1999969495

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2344567

Country of ref document: CA

Ref country code: CA

Ref document number: 2344567

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 60485/99

Country of ref document: AU

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2000 571253

Kind code of ref document: A

Format of ref document f/p: F

WWP Wipo information: published in national office

Ref document number: 1999969495

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1999969495

Country of ref document: EP