US20020164656A1 - Microarrays and uses therefor - Google Patents

Microarrays and uses therefor Download PDF

Info

Publication number
US20020164656A1
US20020164656A1 US10/035,368 US3536801A US2002164656A1 US 20020164656 A1 US20020164656 A1 US 20020164656A1 US 3536801 A US3536801 A US 3536801A US 2002164656 A1 US2002164656 A1 US 2002164656A1
Authority
US
United States
Prior art keywords
antibodies
antigen
cells
protein
binding
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US10/035,368
Inventor
James Hoeffler
Joseph Fernandez
Marc Nasoff
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US10/035,368 priority Critical patent/US20020164656A1/en
Publication of US20020164656A1 publication Critical patent/US20020164656A1/en
Assigned to BANK OF AMERICA, N.A., AS COLLATERAL AGENT reassignment BANK OF AMERICA, N.A., AS COLLATERAL AGENT SECURITY AGREEMENT Assignors: Life Technologies Corporation
Priority to US12/490,069 priority patent/US8012703B2/en
Priority to US13/173,637 priority patent/US8637264B2/en
Priority to US13/648,169 priority patent/US20130123127A1/en
Assigned to Life Technologies Corporation reassignment Life Technologies Corporation LIEN RELEASE Assignors: BANK OF AMERICA, N.A.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/544Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals the carrier being organic
    • G01N33/548Carbohydrates, e.g. dextran
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6834Enzymatic or biochemical coupling of nucleic acids to a solid phase
    • C12Q1/6837Enzymatic or biochemical coupling of nucleic acids to a solid phase using probe arrays or probe chips
    • 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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • 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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/54393Improving reaction conditions or stability, e.g. by coating or irradiation of surface, by reduction of non-specific binding, by promotion of specific binding
    • 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
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6845Methods of identifying protein-protein interactions in protein mixtures
    • 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
    • G01N33/6854Immunoglobulins
    • 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
    • B01J2219/00608DNA chips
    • 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
    • B01J2219/00612Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports the surface being inorganic
    • 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
    • B01J2219/00623Immobilisation or binding
    • B01J2219/00626Covalent
    • 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
    • B01J2219/00639Making arrays on substantially continuous surfaces the compounds being trapped in or bound to a porous medium
    • B01J2219/00641Making arrays on substantially continuous surfaces the compounds being trapped in or bound to a porous medium the porous medium being continuous, e.g. porous oxide substrates
    • 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

Definitions

  • the invention disclosed herein relates to new methods of using microarray technologies.
  • the methods are useful for identifying and characterizing specific antibodies as well as the characterization of different tissues or cells by protein or nucleic acid analysis.
  • Antibodies that bind to specific antigens have been produced by a variety of methods including immunization of animals, fusion of mammalian spleen cells to immortalized cells to produce hybridomas, random peptide generation using phage or bacterial display and constrained peptide libraries. Regardless of how the desired antibody is generated, the methods currently available to identify one with a particular binding specificity are generally laborious and incapable of the simultaneous testing of large numbers of unknowns.
  • One method involves binding the antigen to a porous membrane, such as nitrocellulose, contacting the membrane with a source of test antibodies, then determining whether or not any of the test antibodies has bound to the antigen.
  • a porous membrane such as nitrocellulose
  • This method only allows the testing of one source of test antibodies per piece of porous membrane, making the method both inconvenient and wasteful of materials.
  • Antibody/antigen reactions can also be evaluated in plastic plates, such as 96-well microtiter plates, using methods similar to those described above. This method is likewise limited in the number of samples that can be tested in any one assay, thus requiring many assays to fully evaluate a large number of antibody unknowns.
  • Chang U.S. Pat. No. 4,591,570, issued 5/27/86 describes an array of a limited number of characterized antibodies to known antigens on a glass surface that can be used to bind to specific antigens on the surface of whole cells.
  • the invention disclosed herein comprises methods of using microarrays to simplify analysis and characterization of genes and their function.
  • the methods are used to identify and characterize antibodies having binding affinity for a specific target antigen.
  • This method comprises contacting an array of uncharacterized antibodies bound to a solid surface with at least one target antigen and identifying the antibodies to which the target antigen binds.
  • the method can be performed under a variety of conditions to identify antibodies with a range of binding affinities.
  • a second aspect of the invention comprises a method of determining gene expression at the protein level comprising contacting an array of characterized or uncharacterized antibodies on a solid surface with one or more proteins and identifying the antibodies to which said protein(s) binds. This method can be further used to compare the protein expression in two different populations of cells, such as normal cells and cancer cells or resting cells and stimulated cells. A related embodiment can be used as a tool in the diagnosis of various disorders.
  • a further aspect of the invention comprises a method of determining gene expression at the protein level comprising contacting a microarray of nucleic acid samples derived from a variety of different sources with one or more nucleic acid probes then identifying the sample or samples to which the probe binds.
  • FIGS. 1A, 1B, and 1 C show microarrays of antibodies bound to positively charged nylon, reacted with antigen and detected by non-fluorescent means.
  • FIG. 2 shows a microarray produced using a robotic arraying apparatus. Antigen binding is detected by non-fluorescent means.
  • FIG. 3 shows the ability of the antibody microarrays to evaluate relative binding affinities to a specific antigen.
  • FIG. 4 shows a microarray of polyclonal antibodies in comparison to a microarray of monoclonal antibodies.
  • FIGS. 5A, 5B and 5 C show a microarray of antibodies reacted with a cell lysate under conditions that vary the amount of background binding.
  • the present invention discloses methods of using microarrays to simplify analysis and characterization of genes and their function.
  • the methods are used for identifying and characterizing antibodies having binding specificity to a particular antigen or set of antigens.
  • This method utilizes microarray technology to create ordered matrices of large numbers of uncharacterized antibodies which can then be contacted with antigen under a variety of conditions.
  • the method is rapid and simple to perform and is applicable to the simultaneous screening of very large numbers of antibodies.
  • uncharacterized antibodies are bound to a solid surface in an array format consisting of discrete spots whose spatial location can be easily identified. Each location represents an antibody from a known source, such as a particular hybridoma growing in a well in a 96-well microtiter plate. The space between the antibody spots is treated to minimize non-specific binding to the solid support.
  • the arrayed antibodies are then contacted with an antigen, or a set of antigens, for which specific antibodies are sought.
  • the antigen solution is left in contact with the array for an amount of time sufficient to allow antigen:antibody complexes to form (generally 10 minutes to 2 hours), then the unbound antigen is washed away under suitable conditions. Bound antigen is detected at a particular antibody spot using one of a variety of detection methods, thus identifying the source of an antibody specific for the particular antigen.
  • antibody is used herein in the broadest sense and specifically includes intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies) formed from at least two intact antibodies, and antibody fragments, including single chain antibodies, so long as they exhibit the desired binding properties as described herein.
  • a host animal of any of a number of species such as rabbits, goats, sheep, horse, cow, mice, rats, etc. is immunized by injection with an antigenic preparation which may be derived from cells or microorganisms, or may be recombinantly or synthetically produced.
  • adjuvants well known in the art may be used to enhance the production of antibodies by the immunized host, for example, Freund's adjuvant (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, liposomes, potentially useful human adjuvants such as BCG (Bacille Calmette-Guerin) and Propionibacterium acanes , and the like.
  • BCG Bacille Calmette-Guerin
  • the term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen.
  • Preferred antibodies are mAbs, which may be of any immunoglobulin class including IgG, IgM, IgE, IgA, and any subclass or isotype thereof.
  • monoclonal antibodies are advantageous in that they are synthesized by hybridoma culture, uncontaminated by other immunoglobulins.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al., Nature, 256:495 (1975), or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567, incorporated by reference herein).
  • the “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol. Biol., 222:581-597 (1991), for example.
  • the monoclonal antibodies contemplated for use herein specifically include “chimeric” antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567; Morrison et al., Proc. Natl. Acad. Sci. USA, 81:6851-6855 (1984)).
  • chimeric antibodies immunoglobulins in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous
  • “Humanized” forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab′, F(ab′) 2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementarity-determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity, and capacity.
  • humanized antibodies may comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. These modifications are made to further refine and maximize antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence.
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • the humanized antibody includes a PRIMATIZEDTM antibody wherein the antigen-binding region of the antibody is derived from an antibody produced by immunizing macaque monkeys with the antigen of interest.
  • Antibody fragments comprise a portion of an intact antibody, preferably the antigen binding or variable region of the intact antibody.
  • antibody fragments include Fab, Fab′, F(ab′) 2 , and Fv fragments; diabodies; linear antibodies (Zapata et al. Protein Eng. 8(10):1057-1062 (1995)); single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • Single-chain antibodies are antibody fragments comprising the V H and V L domains of an antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide further comprises a polypeptide linker between the V H and V L domains which enables the sFv to form the desired structure for antigen binding.
  • diabodies refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (V H ) connected to a light-chain variable domain (V L ) in the same polypeptide chain (V H -V L ).
  • V H heavy-chain variable domain
  • V L light-chain variable domain
  • the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites.
  • Diabodies are described more fully in, for example, EP 404,097; WO 93/11161; and Hollinger et al., Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993).
  • the antibodies employed in the invention can be isolated prior to creating a microarray.
  • An “isolated” molecule whether an antibody, antigen or nucleic acid, is one which has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials which would interfere with particular uses for the molecule, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes.
  • a protein will be purified (1) to greater than 95% by weight of protein as determined by the Lowry method, and most preferably more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under reducing or nonreducing conditions using Coomassie blue or, preferably, silver stain.
  • Isolated protein includes the protein in situ within recombinant cells since at least one component of the protein's natural environment will not be present. Ordinarily, however, isolated protein will be prepared by at least one purification step. Unpurified antibodies, such as those found in serum, can also be employed in the present invention.
  • isolated in reference to nucleic acid is meant a polymer of 14, 17, 21 or more contiguous nucleotides, including DNA or RNA that is isolated from a natural source or that is synthesized.
  • the isolated nucleic acid of the present invention is unique in the sense that it is not found in a pure or separated state in nature.
  • Use of the term “isolated” indicates that a naturally occurring sequence has been removed from its normal cellular (i.e., chromosomal) environment. Thus, the sequence may be in a cell-free solution or placed in a different cellular environment.
  • sequence is the only nucleotide sequence present, but that it is essentially free (about 90-95% pure at least) of non-nucleotide material naturally associated with it and thus is meant to be distinguished from isolated chromosomes.
  • One particularly useful method of isolating antibodies is affinity purification.
  • Resins suitable for antibody purification are well known in the art, for example, protein A SEPHAROSETM.
  • a recombinant antibody can be engineered to contain an affinity purification tag to facilitate its purification.
  • Resins suitable for antibody purification are well known in the art, for example, protein A SEPHAROSETM resin.
  • Affinity purification tags are generally peptide sequences that can interact with a binding partner immobilized on a solid support. Synthetic DNA sequences encoding multiple consecutive single amino acids, such as histidine, when fused to the expressed protein, may be used for one-step purification of the recombinant protein by high affinity binding to a resin column, such as nickel SEPHAROSETM resin. An endopeptidase recognition sequence can be engineered between the polyamino acid tag and the protein of interest to allow subsequent removal of the leader peptide by digestion with enterokinase, and other proteases.
  • Sequences encoding peptides such as the chitin binding domain (which binds to chitin), biotin (which binds to avidin and strepavidin), and the like can also be used for facilitating purification of the protein of interest.
  • the affinity purification tag can be separated from the protein of interest by methods well known in the art, including the use of inteins (protein self-splicing elements, Chong, et al, Gene 192:271-281, 1997).
  • the process of isolation can be used to simultaneously normalize yield and isolate the antibody.
  • the samples can be contacted with an amount of resin whose maximum binding capacity is 10 mgs. Thus any antibody greater than this amount will pass through the resin unbound. The maximum bound amount can then be eluted from the resin.
  • Methods for creating microarrays are known in the art including printing on a solid surface using pins (passive pins, quill pins, and the like) or spotting with individual drops of solution. Passive pins draw up enough sample to dispense a single spot. Quill pins draw up enough liquid to dispense multiple spots. Bubble printers use a loop to capture a small volume which is dispensed by pushing a rod through the loop. Microdispensing uses a syringe mechanism to deliver multiple spots of a fixed volume.
  • solid supports can be arrayed using piezoelectric (ink jet) technology, which actively transfers samples to a solid support.
  • Shalon and Brown WO 95/35505, published 12/28/95 which is incorporated herein by reference in its entirety.
  • the method and apparatus described in Shalon and Brown can create an array of up to six hundred spots per square centimeter on a glass slide using a volume of 0.01 to 100 nl per spot. Suitable concentrations of antibody range from about 1 ng/ ⁇ l to about 1 ⁇ g/ ⁇ l.
  • each spot can contain one or more than one distinct antibody.
  • microarrays can be created on a variety of solid surfaces such as plastics (e.g. polycarbonate), complex carbohydrates (e.g. agarose and SEPHAROSETM), acrylic resins (e.g. polyacrylamide and latex beads), and nitrocellulose.
  • plastics e.g. polycarbonate
  • complex carbohydrates e.g. agarose and SEPHAROSETM
  • acrylic resins e.g. polyacrylamide and latex beads
  • nitrocellulose e.g. polyacrylamide and latex beads
  • Preferred solid support materials include glass slides, silicon wafers, and positively charged nylon. Specific examples of suitable solid supports are described in the Examples below.
  • BSA bovine serum albumin
  • the arrays used to identify antigen-specific antibodies are contacted with a solution containing one or more known antigens in order to identify antibodies in the array with binding specificity for the antigen.
  • the antigens are often proteins, although they may also be organic chemical compounds, carbohydrates, nucleic acids, and the like. They may be isolated or semi-isolated, recombinant or naturally occurring.
  • the amount of antigen used can vary from about 1-100 ng/ ⁇ l.
  • the antigen is left in contact with the array for an amount of time sufficient for antibody:antigen complexes to form, should one of the antibodies in the array be specific for the antigen. The amount of time sufficient for this purpose will range from 5 minutes to 24 hours, and will generally be from 0.5 to 2 hours.
  • One antigen of particular interest in the practice of the invention is recombinant protein, either a full-length gene product or a fragment thereof, for example an Expressed Sequence Tag (or EST fragment).
  • EST fragments are relatively short cDNA sequences that have been randomly generated and sequenced, generally as part of an ongoing effort to map an entire genome (Adams, et al, Science 252(5013):1651-1656, 1991). Large numbers of these sequences are available in public databases. The identity of the proteins encoded by the vast majority of these sequences is unknown. The following discussion, although directed to the expression of EST-encoded peptides, is equally applicable to any expressed product of a nucleic acid sequence, including full-length proteins.
  • Prokaryotic hosts are, generally, very efficient and convenient for the production of recombinant proteins and are, therefore, one type of preferred expression system for EST fragments. Prokaryotes most frequently are represented by various strains of E. coli . However, other microbial strains may also be used, including other bacterial strains.
  • plasmid vectors that contain replication sites and control sequences derived from a species compatible with the host may be used.
  • suitable plasmid vectors may include pBR322, pUC 118, pUC 119, and the like;
  • suitable phage or bacteriophage vectors may include ⁇ gt10, ⁇ gt11, and the like;
  • suitable virus vectors may include pMAM-neo, PKRC and the like.
  • the selected vector of the present invention has the capacity to replicate in the selected host cell.
  • Recognized prokaryotic hosts include bacteria such as E. coli and those from genera such as Bacillus, Streptomyces, Pseudomonas, Salmonella, Serratia, and the like. However, under such conditions, the polypeptide will not be glycosylated.
  • the prokaryotic host selected for use herein must be compatible with the replicon and control sequences in the expression plasmid.
  • a functional prokaryotic promoter such as the T7 promoter or RSC promoter.
  • Such promoters may be either constitutive or, more preferably, regulatable (i.e., inducible or derepressible).
  • constitutive promoters include the int promoter of bacteriophage ⁇ , the bla promoter of the ⁇ -lactamase gene sequence of pBR322, the CAT promoter of the chloramphenicol acetyl transferase gene sequence of pPR325, and the like.
  • inducible prokaryotic promoters examples include the major right and left promoters of bacteriophage (P L and P R ), the trp, reca, lacZ, LacI, and gal promoters of E. coli , the ⁇ -amylase (Ulmanen et al., J. Bacteriol. 162:176-182, 1985) and the sigma-28-specific promoters of B.
  • subtilis (Gilman et al., Gene sequence 32:11-20(1984)), the promoters of the bacteriophages of Bacillus (Gryczan, In: The Molecular Biology of the Bacilli, Academic Press, Inc., NY (1982)), Streptomyces promoters (Ward et at., Mol. Gen. Genet. 203:468-478, 1986), and the like.
  • Exemplary prokaryotic promoters are reviewed by Glick (J. Ind. Microbiol. 1:277-282, 1987); Cenatiempo (Biochimie 68:505-516, 1986); and Gottesman (Ann. Rev. Genet. 18:415-442, 1984).
  • ribosome binding site upstream of the gene sequence-encoding sequence.
  • Such ribosome binding sites are disclosed, for example, by Gold et at. (Ann. Rev. Microbiol. 35:365-404, 1981).
  • the selection of control sequences, expression vectors, transformation methods, and the like, are dependent on the type of host cell used to express the gene.
  • Host cells which may be used in the expression systems of the present invention are not strictly limited, provided that they are suitable for use in the expression of the peptide of interest. Suitable hosts may often include eukaryotic cells. Preferred eukaryotic hosts include, for example, yeast, fungi, insect cells, and mammalian cells either in vivo, or in tissue culture. Mammalian cells which may be useful as hosts include HeLa cells, cells of fibroblast origin such as VERO, 3T3 or CHOK1, HEK 293 cells or cells of lymphoid origin (such as 32D cells) and their derivatives. Preferred mammalian host cells include SP2/0 and JS58L, as well as neuroblastoma cell lines such as IMR 332 and PC12 which may provide better capacities for correct post-translational processing.
  • eukaryotic hosts include, for example, yeast, fungi, insect cells, and mammalian cells either in vivo, or in tissue culture.
  • plant cells are also available as hosts, and control sequences compatible with plant cells are available, such as the cauliflower mosaic virus 35S and 19S, nopaline synthase promoter and polyadenylation signal sequences, and the like.
  • Another preferred host is an insect cell, for example the Drosophila larvae. Using insect cells as hosts, the Drosophila alcohol dehydrogenase promoter can be used. Rubin, Science 240:1453-1459, 1988).
  • baculovirus vectors can be engineered to express large amounts of peptide encoded by an EST fragment in insects cells (Jasny, Science 238:1653, 1987); Miller et al., In: Genetic Engineering (1986), Setlow, J. K., et al., eds., Plenum, Vol. 8, pp. 277-297).
  • Any of a series of yeast gene sequence expression systems can be utilized which incorporate promoter and termination elements from the actively expressed gene sequences coding for glycolytic enzymes which are produced in large quantities when yeast are grown in media rich in glucose.
  • Known glycolytic gene sequences can also provide very efficient transcriptional control signals.
  • Yeast provides substantial advantages in that it can also carry out post-translational peptide modifications.
  • Yeast recognizes leader sequences on cloned mammalian gene sequence products and secretes peptides bearing leader sequences (i.e., pre-peptides).
  • leader sequences i.e., pre-peptides
  • transcriptional and translational regulatory sequences may be employed, depending upon the nature of the host.
  • the transcriptional and translational regulatory signals may be derived from viral sources, such as adenovirus, bovine papilloma virus, cytomegalovirus, simian virus, or the like, where the regulatory signals are associated with a particular gene sequence which has a high level of expression.
  • promoters from mammalian expression products such as actin, collagen, myosin, and the like, may be employed.
  • Transcriptional initiation regulatory signals may be selected which allow for repression or activation, so that expression of the gene sequences can be modulated.
  • regulatory signals which are temperature-sensitive so that by varying the temperature, expression can be repressed or initiated, or are subject to chemical (such as metabolite) regulation.
  • eukaryotic regulatory regions Such regions will, in general, include a promoter region sufficient to direct the initiation of RNA synthesis.
  • Preferred eukaryotic promoters include, for example, the promoter of the mouse metallothionein I gene sequence (Hamer et al., J. Mol. Appl. Gen. 1:273-288, 1982); the TK promoter of Herpes virus (McKnight, Cell 31:355-365, 1982); the SV40 early promoter (Benoist et al., Nature (London) 290:304-310, 1981); the yeast gal4 gene sequence promoter (Johnston et al., Proc. Natl.
  • An EST fragment and an operably linked promoter may be introduced into a recipient prokaryotic or eukaryotic cell either as a nonreplicating DNA (or RNA) molecule, which may either be a linear molecule or, more preferably, a closed covalent circular molecule (a plasmid). Since such molecules are incapable of autonomous replication, the expression of the gene may occur through the transient expression of the introduced sequence. Alternatively, permanent or stable expression may occur through the integration of the introduced DNA sequence into the host chromosome.
  • RNA nonreplicating DNA
  • a vector may be employed which is capable of integrating the desired gene sequences into the host cell chromosome.
  • Cells which have stably integrated the introduced DNA into their chromosomes can be selected by also introducing one or more markers which allow for selection of host cells which contain the expression vector.
  • the marker may provide for prototrophy to an auxotrophic host, biocide resistance, e.g., antibiotics, or heavy metals, such as copper, or the like.
  • the selectable marker gene sequence can either be directly linked to the DNA gene sequences to be expressed, or introduced into the same cell by cotransfection.
  • Common selectable marker gene sequences include those for resistance to antibiotics such as ampicillin, tetracycline, kanamycin, bleomycin, streptomycin, hygromycin, neomycin, ZeocinTM, and the like.
  • Selectable auxotrophic gene sequences include, for example, hisD, which allows growth in histidine free media in the presence of histidinol.
  • Additional elements may also be needed for optimal synthesis of single chain binding protein mRNA. These elements may include splice signals, as well as transcription promoters, enhancers, and termination signals. cDNA expression vectors incorporating such elements include those described by Okayama, Mol. Cell. Bio. 3:280, 1983.
  • the recombinant antigen may be produced as a fusion protein.
  • a fusion protein When two protein-coding sequences not normally associated with each other in nature are in the same reading frame the resulting expressed protein is called a “fusion protein” as two distinct proteins have been “fused” together. Fusion proteins have a wide variety of uses. For example, two functional enzymes can be fused to produce a single protein with multiple enzymatic activities or short peptide sequences, such as epitope tags or affinity purification tags (see above), can be fused to a larger protein and serve as aids in purification or as means of identifying the expressed protein by serving as epitopes detectable by specific antibodies.
  • Epitope tags are short peptide sequences that are recognized by epitope-specific antibodies.
  • a fusion protein comprising a recombinant protein and an epitope tag can be simply and easily purified using an antibody bound to a chromatography resin.
  • the presence of the epitope tag furthermore allows the recombinant protein to be detected in subsequent assays, such as Western blots, without having to produce an antibody specific for the recombinant protein itself.
  • Examples of commonly used epitope tags include V5, glutathione-S-transferase (GST), hemagglutinin (HA), the peptide Phe-His-His-Thr-Thr, chitin binding domain, and the like.
  • a fusion protein may be a means by which the recombinant antigen protein can be easily detected.
  • the fusion component can itself be a detectable moiety, such as fluorescent protein (fluorescent green protein, fluorescent yellow protein, and the like), or alternatively can be one member of a specific binding pair (such as biotin and streptavidin, for example) which can be detected by reacting with the other member conjugated to a detectable substance.
  • the introduced nucleic acid molecule can be incorporated into a plasmid or viral vector capable of autonomous replication in the recipient host. Any of a wide variety of vectors may be employed for this purpose. Factors of importance in selecting a particular plasmid or viral vector include: the ease with which recipient cells that contain the vector may be recognized and selected from those recipient cells which do not contain the vector; the number of copies of the vector which are desired in a particular host; and whether it is desirable to be able to “shuttle” the vector between host cells of different species.
  • Suitable prokaryotic vectors include plasmids such as those capable of replication in E. coli (for example, pBR322, ColEl, pSC101, PACYC 184, itVX, pRSET, pBAD (Invitrogen, Carlsbad, Calif.), and the like).
  • E. coli for example, pBR322, ColEl, pSC101, PACYC 184, itVX, pRSET, pBAD (Invitrogen, Carlsbad, Calif.), and the like.
  • Such plasmids are disclosed by Sambrook (cf. “Molecular Cloning: A Laboratory Manual”, second edition, edited by Sambrook, Fritsch, & Maniatis, Cold Spring Harbor Laboratory, (1989)).
  • Bacillus plasmids include pC194, pC221, pT127, and the like, and are disclosed by Gryczan (In: The Molecular Biology of the Bacilli, Academic Press, NY (1982), pp. 307-329).
  • Suitable Streptomyces plasmids include plJlOl (Kendall et al., J. Bacteriol. 169:4177-4183,1987), and Streptomyces bacteriophages such as ⁇ C31 (Chater et al., In: Sixth International Symposium on Actinomycetales Biology, Akademiai Kaido, Budapest, Hungary (1986), pp. 45-54).
  • Pseudomonas plasmids are reviewed by John et al. (Rev. Infect. Dis. 8:693-704, 1986), and Izaki (Jpn. J. Bacteriol. 33:729-742, 1978).
  • Suitable eukaryotic plasmids include, for example, BPV, vaccinia, SV40, 2-micron circle, pCDN3.1 (Invitrogen), and the like, or their derivatives.
  • Such plasmids are well known in the art (Botstein et al., Miami Wntr. Symp. 19:265-274, 1982); Broach, In: “The Molecular Biology of the Yeast Saccharomyces: Life Cycle and Inheritance”, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., p. 445-470 (1981); Broach, Cell 28:203-204, 1982); Dilon et at., J. Clin. Hematol. Oncol. 10:39-48, 1980); Maniatis, In: Cell Biology: A Comprehensive Treatise, Vol. 3, Gene Sequence Expression, Academic Press, NY, pp. 563-608 (1980).
  • the antibody:antigen complexes can be detected using one of several techniques known in the art. Suitable washing conditions are known to those skilled in the art (see, for example, Ausubel, et al, Short Protocols in Molecular Biology, 3rd ed. 1995). Exemplary washing conditions are shown in the examples below.
  • expression vectors can be used that form chimeric fusion peptides as described above.
  • the epitope tagged antigen can be 55464 detected using an antibody specific for the tag sequence. This antibody may be itself detectably labeled or can be detected with a third detectably-labeled antibody.
  • the antigen can be complexed with biotin and detected using detectably-labeled avidin or streptavidin. The antigen itself can also be detectably labeled, such as with a fluorescent dye compound.
  • detectably labeled is intended to encompass antigen directly coupled to a detectable substance, such as a fluorescent dye, and antigen coupled to a member of binding pair, such as biotin/streptavidin, or an epitope tag that can specifically interact with a molecule that can be detected, such as by producing a colored substrate or fluorescence.
  • Substances suitable for detectably labeling proteins include fluorescent dyes such as fluorescein isothiocyanate (FITC), fluorescein, rhodamine, tetramethyl-rhodamine-5-(and 6)-isothiocyanate (TRITC), Texas red, cyanine dyes (Cy3 and Cy5, for example), and the like; and enzymes that react with colorimetric substrates such as horseradish peroxidase.
  • fluorescent dyes is generally preferred in the practice of the invention as they can be detected at very low amounts.
  • each antigen can be labeled with a distinct fluorescent compound for simultaneous detection. Labeled spots on the array are detected using a fluorimeter, the presence of a signal indicating an antigen bound to a specific antibody.
  • Antigen-containing reaction solutions can contain varying degrees of salt or be conducted at varying pH levels.
  • the binding reaction can be carried out at varying temperatures.
  • Each set of conditions will identify antibodies with different affinity for the antigen.
  • antibodies that bind at pH 2 may have utility under highly acidic conditions such as those that exist in the stomach.
  • antibodies that bind at temperatures near boiling may be useful in studying thermophilic organisms.
  • pH conditions will range from 2-10 (most preferably around pH 8), temperatures from 0° C.-100° C. and salt conditions from 1 ⁇ M to 5 M (in the case of NaCl).
  • Affinity constants are a measure of the interaction between a particular ligand and its cognate receptor.
  • the “binding affinity” or the measure of the strength of association between a particular antibody:antigen interaction is generally measured by affinity constants for the equilibrium concentrations of associated and dissociated configurations of the antibody and its antigen.
  • Antibody fragments will generally have affinities in the range of about 10 ⁇ 6 M to 10 ⁇ 7 M.
  • microarrays of uncharacterized antibodies are used to compare the protein expression profiles of cells. For example, comparisons can be made between a population of cells from one tissue, such as arterial endothelial cells, and a second tissue, such as venous endothelial cells or from cells derived from a particular tissue but from different species. Comparisons can be made between normal cells and cells from the same tissue type that originate from an individual with a pathogenic disorder. For example, comparisons can be made between normal cells and cancer cells. Comparisons can additionally be made between cells in a resting state and cells in an activated state, for example, resting T-cells and activated T-cells.
  • the disclosed arrays are useful for evaluating the expression of proteins by pathogens, such as, for example, bacteria, parasites, viruses, and the like.
  • a solution (such as a lysate) made from the pathogen which represents all proteins expressed by the pathogen can be used to contact an antibody array to identify antibodies recognizing pathogen-expressed proteins. These antibodies have utility as diagnostic agents as well as potential therapeutics.
  • Cellular lysates can be used as “antigens” as described above and reacted with two identical microarrays. Antibodies reactive in one array but not the other would indicate the presence of a differentially expressed protein. This antibody is then useful for the subsequent isolation and identification of those proteins that are different in two populations of cells. In the case of normal and cancer cells, for example, one may be able to identify proteins expressed in the cancer cell that contribute to its malignant state.
  • microarrays can be composed of previously characterized antibodies. These microarrays have a variety of uses, one of which is cell profiling.
  • an array can be composed of antibodies that recognize a set of antigens known to be present in activated T-cells but not in resting T-cells. A population of T-cells can then be lysed and the lysate contacted with the array to determine if the population has the profile of activated or resting T-cells.
  • Microarrays and the methods disclosed herein can be used in methods of diagnosing particular disorders. For example, a collection of antibodies specific for a range of antigens associated with one or more disorders can be arrayed and contacted with a bodily fluid containing antigens whose presence, or absence, would indicate a particular disorder.
  • the advantage of using a microarray over a conventional immunoassay is the ability to include a population of antibodies diagnostic for a variety of disorders on a single surface, significantly reducing time, costs and materials needed to effect a diagnosis.
  • a single microarray assay could be used to make a specific diagnosis, thus allowing the patient to be properly treated.
  • Patients suffering from stroke or brain infarcts release several proteins into cerebrospinal fluid, examples of which are neuron specific enolyse (NSE) from neuronal cells and S-100 from glial cells and astrocytes.
  • NSE neuron specific enolyse
  • Such proteins are not released in conditions that may have similar symptoms, such as drug reactions, making proper diagnosis more difficult.
  • a diagnostic array could readily detect these and other proteins in the CSF, leading to a rapid clinical diagnosis and treatment.
  • microarrays are employed to characterize protein expression patterns using nucleic acid samples. Briefly, nucleic acid molecules from a whole cell or tissue are applied to a solid support using a microarray format. The arrayed nucleic acid samples are then contacted with a nucleic acid probe specific for a gene encoding a known protein. The probe solution is left in contact with the array for an amount of time sufficient to allow sample:probe complexes to form, then the unbound probe is washed away under suitable conditions (see, for example, Ausubel, et al, Short Protocols in Molecular Biology, 3rd ed. 1995 and the examples below). Bound probe is detected at one or more nucleic acid sample spots using one of a variety of detection methods.
  • the microarray can be constructed from nucleic acid samples isolated from a single tissue type but from a large number of species, with each spot representing a particular species. Thus in a single assay format one can determine the evolutionary development of the protein represented by the probe.
  • the microarray can be constructed of multiple tissue types from a single species, or from different developmental stages of a single species (or multiple species) thus simply and efficiently determining tissue expression of the protein represented by the probe.
  • a microarray can be constructed with arrayed samples representing all the developmental stages of Drosophila, a well known organism the study of which has led to a greater understanding of mammalian physiology and development.
  • the nucleic acid sample can be messenger ribonucleic acid (mRNA) or can be complementary deoxyribonucleic acid (cDNA), including EST fragments.
  • Methods for extracting and isolating nucleic acids from cells are well known in the art (for example phenol extraction/ethanol precipitation, ammonium acetate precipitation, cesium chloride gradients, and the like), as are methods for generating cDNA (see, for example, “Molecular Cloning: A Laboratory Manual,” second edition, edited by Sambrook, Fritsch, & Maniatis, Cold Spring Harbor Laboratory, 1989; and Ausubel, et al, Short Protocols in Molecular Biology, 3rd ed. 1995, both of which are incorporated by reference herein).
  • Microarrays of these nucleic acids are created using the methods described above. Techniques for coupling nucleic acids to solid supports used to construct microarrays are well known in the art, including the poly-L-lysine and phenylboronic acid methods described in the Examples below.
  • the nucleic acid probes used in the invention methods can be designed based on the sequence of a gene encoding a known protein or can be an EST fragment, as described above.
  • One skilled in the art can readily design such probes based on the known sequence using methods of computer alignment and sequence analysis known in the art (e.g., “Molecular Cloning: A Laboratory Manual”, second edition, edited by Sambrook, Fritsch, & Maniatis, Cold Spring Harbor Laboratory, 1989; Ausubel, et al, Short Protocols in Molecular Biology, 3rd ed. 1995).
  • the probe can comprise any number of nucleotides but will preferably be not fewer than 10 nucleotides and preferably not more than about 300 nucleotides in length.
  • the probes of the invention can be labeled by standard labeling techniques such as with a radiolabel, enzyme label, fluorescent label, biotin-avidin label, chemiluminescent label, and the like. After hybridization, the probes may be detected using known methods. Preferred labels are fluorescent labels, as described above.
  • the nucleic acid probes of the present invention include RNA as well as DNA probes and nucleic acids modified in the sugar, phosphate or even the base portion as long as the probe still retains the ability to specifically hybridize under conditions as disclosed herein. Such probes are generated using techniques known in the art.
  • hybridize refers to a method of interacting a nucleic acid sequence with a DNA or RNA molecule in solution or on a solid support, such as cellulose or nitrocellulose. If a nucleic acid sequence binds to the DNA or RNA molecule with sufficiently high affinity, it is said to “hybridize” to the DNA or RNA molecule.
  • the strength of the interaction between the probing sequence and its target can be assessed by varying the stringency of the hybridization conditions. Various low to high stringency hybridization conditions may be used depending upon the specificity and selectivity desired. Stringency is controlled by varying salt or denaturant concentrations. Examples of hybridization conditions are shown in the Examples below.
  • microarrays can be composed of randomly generated polynucleotides (DNA or RNA) and contacted with proteins to identify unique binding pairs.
  • Polynucleotides are now known to bind to proteins and may have potential as diagnostics and therapeutics (see, for example, Allen, et al, Virology 209(2):327-336, 1995; Binkley, et al, Nucleic Acids Res. 23(16):3198-3205, 1995). Polynucleotides can be evaluated in very large numbers using the methods disclosed herein thus increasing the likelihood of identifying a useful binder.
  • Glass slides are prepared as follows: NaOH (50 g) is dissolved in 150 ml of double distilled water (ddH 2 O), then 200 ml of 95% EtOH is added while stirring. If the solution becomes cloudy, ddH 2 O is added until it becomes clear. Approximately 30 glass slides (Gold Seal, Cat. No. 3010) are soaked in the NaOH/EtOH solution for 2 hours, shaking. The slides are then rinsed three times with ddH 2 O. The slides are next soaked in a poly-L-lysine solution (70 ml poly-L-lysine (Sigma Cat. No. 8920) to 280 ddH 2 O) for 1 hour. Excess liquid is removed by spinning the slides in a rack on a microtiter plate carrier at 500 rpm. The slides are dried at 40° C. for 5 minutes, then stored in a closed box for at least 2 weeks prior to use.
  • ddH 2 O double distilled water
  • EtOH 95% EtOH
  • a cDNA microarray is prepared as follows: Total MRNA is isolated from tissue (for example, nerve cells) of a variety of species representative of different classes of organisms such as Drosophila, nematode, salmon, clam, chicken, mouse, dog, goat, spider monkey, chimpanzee, human, and the like, by the FastTrac method (Stratagene, La Jolla, Calif.) or other common methods. mRNA is also obtained from a variety of unicellular organisms such as E. coli , yeast, B. subtilis , mycoplasma and the like.
  • Eukaryotic mRNA is enriched from total RNA using oligo(dT) cellulose (Ausubel, et al, Short Protocols in Molecular Biology, 3rd ed. 1995, pgs 4-11-4-12). Equivalent amounts (for example, 1 ⁇ g) of mRNA from each source are placed in a separate well of one or more 96 well microtiter plates and precipitated with cold EtOH. The precipitate is rinsed with 70% EtOH and allowed to dry.
  • the dried mRNA is resuspended in 3 ⁇ SSC (sodium chloride/sodium citrate—20X solution is 3 M NaCl (175g/L0 0.3 M trisodium citrate 2H 2 O (88g/L adjusted to pH 7.0 with 1 M HCl) then spotted onto a previously prepared glass slide using an array device (for example, Shalon and Brown (WO 95/35505, published 12/28/95)).
  • the prepared array can be kept for a long period of time before probing, however, if the slides are to be kept for long periods of time, stability is increased by converting each mRNA sample into cDNA using techniques known in the art, such as PCR.
  • the array is rehydrated by suspending the slide over a dish of ddH 2 O (50° C.) for approximately one minute.
  • the slide is quickly (approximately 3 seconds) dried by placing it on a surface heated to 100° C. (mRNA side up).
  • the mRNA is crosslinked to the poly-L-lysine coating of the slide using ultraviolet radiation using a StratalinkerTM UV device according to the manufacturer's instructions (Stratagene) set at 60 milliJoules.
  • the slides are next soaked in a solution of 5 grams of succinic anhydride (Aldrich Cat. No. 23,969-0) dissolved in 315 ml of N-methyl-pyrrilidinone (Aldrich Cat. No. 32,863-4) plus 35 mls of 0.2 M sodium borate (brought to pH 8.0 with NaOH) for 15 minutes with shaking.
  • the slide is then transferred to a 95° C. water bath for 2 minutes followed by 95% EtOH for 1 minute. Excess liquid is removed from the slides by spinning a rack of slides on a microtiter plate carrier at 500 rpm.
  • a probe sequence of a known protein is labeled using standard protocols, for example by using a CyDyeTM Nick Translation kit (Amersham).
  • the labeled probe (approximately 1 ⁇ g/ml) is resuspended in 4 ⁇ SSC (10 ⁇ l) to which is added 0.2 ⁇ l 10% sodium dodecyl sulfate (SDS).
  • SSC sodium dodecyl sulfate
  • the probe is boiled for 2 minutes, then cooled for 10 seconds and transferred to the array by pipette.
  • the array is covered by a 22 mm ⁇ 22 mm cover slip, and the slide is placed in a humid hybridization chamber and submerged into a hot water bath ( ⁇ 75° C.).
  • the slide is left in the bath for 10-24 hours, then the cover slip is removed and the slide rinsed in 0.2 ⁇ SSC with 0.1% SDS several times. Excess wash buffer is removed by centrifugation on a microtiter plate carrier as described above. The slide is scanned using a spectrofluorometer, such as the ScanArray 3000 (General Scanning Inc., Watertown, Mass.). For probes labeled with Cy5, for example, fluorescence is measured at 670 nm. Localization of spots on the array to which the probe hybridizes indicates that the species represented by the spot expresses a protein similar or identical to the probe protein.
  • the slides are rinsed with isopropyl alcohol, then sonicated in isopropyl alcohol for several minutes. Sonication should remove any white silane residue on the slides. If the residue remains, the slides should be discarded. After sonication, the slides are left to cure/dry for at least 24 hours before use.
  • the cured slides are next soaked in a linker solution overnight at room temperature.
  • the linker solution is made by dissolving 115 mg of 9Y SA(OCH 2 CN)-X-COOH (Prolix, Bothell, Wash.) in 1 ml dimethylformamide (DMF) plus 60 ⁇ l DIEA, then adding 60 mg TSTU (Sigma) and leaving for 15 minutes at room temperature. This stock is diluted in 270 ml of isopropyl alcohol plus 270 ⁇ l DIEA before using.
  • the slides are removed from the linker solution and soaked in 1 M NH 2 OH, 1 mM EDTA, 0.1 M NaHCO 3 (pH 10) for 4 hours at room temperature. This solution is removed, the slides are extensively washed with water then let air dry at room temperature. The slides can be stored at room temperature away from light before using to make arrays.
  • Aldehyde, nitrocellulose, polystyrene and Surmodics slides were purchased from various outside vendors (aldehyde Slides—Cel Associates, Inc., Houston, Tex.; nitrocellulose Slides —Molecular Probes, Inc., Eugene, Oreg.; polystyrene Slides—Nunc, Inc., Naperville, Ill.; Surmodics Slides—Surmodics, Inc., Eden Prairie, Minn.). Surmodics slides have an undisclosed polymer on the glass surface which forms a covalent linkage with proteins under the appropriate conditions (described by the manufacturer).
  • D1 is a creatine kinase fusion protein isolated from a human fetal heart cDNA library and cloned into the pBAD-Thio-His-TOPO vector (Invitrogen, Carlsbad, Calif.) to create a Thioredoxin-V5-His-creatine kinase fusion protein.
  • D1 was biotinylated using the EZ-LinkTM Sulfo-NHS-LC Biotinylation Kit (Pierce, Rockford, Ill.) used according to the manufacturer's instructions).
  • the filters were washed 5 times with PBST, dried, and developed by immersion in ECL chemiluminescent substrate (ECL—Amersham, Arlington Heights, Ill.) or the chromogenic substrate BCIP/NBT (Sigma Chemicals, St. Louis, Mo.). Filters developed with ECL were exposed to Kodak chemiluminescent film for 1 to 10 seconds.
  • ECL ECL chemiluminescent substrate
  • BCIP/NBT Sigma Chemicals, St. Louis, Mo.
  • Anti-His, anti-V5, anti-FOS, anti-PLC-gamma, 25ClDG, and anti-VEGF (vascular endothelial growth factor) antibodies were arrayed on a nitrocellulose slide and reacted with biotinylated D1 protein as previously described. Binding was detected with streptavidin-Cy3 as described above. The anti-V5 antibodies spots showed red, the anti-His spots showed green, while the negative controls were undetectable (see FIG. 3). When viewed in a black and white drawing, relative increase in binding affinity is visualized by an increase of white in a given area. The color of the spots generally indicates a higher amount of fluorescently labeled antigen present, and thus indicates relative binding affinity between antibody and antigen. Colors, in descending order from highest to lowest affinity, are white, red, yellow, green, and blue. Using this technique, multiple antibodies can be tested for their affinity to a single antigen.
  • polyclonal antibodies were arrayed by hand on a nitrocellulose slide, three polyclonal antibodies (anti-E12 (unpurified rabbit polyclonal sera to a His-V5-thioredoxin-thymidine kinase fusion protein), anti-lexA (lexA repressor protein), and anti-GFP(Green fluorescent protein)) and three monoclonal antibodies (anti-V5, anti-His and anti-GalU (a mammalian transcription factor).
  • the slide was blocked with PBST and 3% milk for 1 hour at room temperature, and incubated with the E12-biotin conjugate, prepared according to the protocol used for D1 protein.
  • the slides were incubated with streptavidin-CY3 conjugate (Amersham, Arlington Heights, Ill.) for 1 hour at room temperature, washed 5 times with PBST and dried by centrifugation prior to scanning on the Scan Array 3000.
  • binding was detected with both the antigen specific polyclonal antibody (anti-E12) and the antigen specific monoclonal antibodies (anti-His, anti-V5) and not with any of the negative control antibodies.
  • CHO cells expressing high levels of beta-galactosidase were grown to confluency in a T-175 flask. (Hams media with Pen/Strep, and L-glutamine plus 10% FCS, at 37° C. with 5% CO 2 ) Cells were harvested using Trypsin/EDTA. NP40 extracts were prepared by pelleting the cells (10 7 cells), washing once in PBS and resuspending in 5% NP40. Cell debris was removed by centrifugation. Soluble protein was biotinylated using a Pierce biotinylation kit according to the manufacturer's instructions.

Abstract

Methods of using microarrays to simplify analysis and characterization of genes and their function are provided. Such methods can be used to identify and characterize antibodies having binding affinity for a specific target antigen. A method of determining gene expression at the protein level by contacting an array of characterized or uncharacterized antibodies on a solid surface with one or more proteins and identifying the antibodies to which said protein(s) binds also is provided. This method can be used to compare the protein expression in two different populations of cells, such as normal cells and cancer cells or resting cells and stimulated cells. In addition, a method of determining gene expression at the protein level by contacting a microarray of nucleic acid samples derived from a variety of different sources with one or more nucleic acid probes then identifying the sample or samples to which the probe binds is provided.

Description

  • This application is a divisional application of U.S. Ser. No. 09/245,615, filed Feb. 4, 1999, which claims the benefit of priority under 35 U.S.C. § 119 of U.S. Ser. No. 60/073,605, filed Feb. 4, 1998 (now abandoned), the entire contents of each of which is incorporated herein by reference.[0001]
  • FIELD OF THE INVENTION
  • The invention disclosed herein relates to new methods of using microarray technologies. The methods are useful for identifying and characterizing specific antibodies as well as the characterization of different tissues or cells by protein or nucleic acid analysis. [0002]
  • BACKGROUND OF THE INVENTION
  • Recent breakthroughs in nucleic acid sequencing technology have made possible the sequencing of entire genomes from a variety of organisms, including humans. The potential benefits of a complete genome sequence are many, ranging from applications in medicine to a greater understanding of evolutionary processes. These benefits cannot be fully realized, however, without an understanding of how and where these newly sequenced genes function. [0003]
  • Traditionally, functional understanding started with recognizing an activity, isolating a protein associated with that activity, then isolating the gene, or genes, encoding that protein. The isolated protein was also used to generate antibody reagents. Specific antibodies and fragments of the isolated gene were both employed to study tissue expression and function. [0004]
  • Several methods have been used to study protein expression patterns including in situ hybridization studies of tissue sections and northern blots. These methods are both time consuming and require relatively large amounts of material to perform successfully. [0005]
  • Antibodies that bind to specific antigens have been produced by a variety of methods including immunization of animals, fusion of mammalian spleen cells to immortalized cells to produce hybridomas, random peptide generation using phage or bacterial display and constrained peptide libraries. Regardless of how the desired antibody is generated, the methods currently available to identify one with a particular binding specificity are generally laborious and incapable of the simultaneous testing of large numbers of unknowns. [0006]
  • One method involves binding the antigen to a porous membrane, such as nitrocellulose, contacting the membrane with a source of test antibodies, then determining whether or not any of the test antibodies has bound to the antigen. This method only allows the testing of one source of test antibodies per piece of porous membrane, making the method both inconvenient and wasteful of materials. [0007]
  • Antibody/antigen reactions can also be evaluated in plastic plates, such as 96-well microtiter plates, using methods similar to those described above. This method is likewise limited in the number of samples that can be tested in any one assay, thus requiring many assays to fully evaluate a large number of antibody unknowns. Chang (U.S. Pat. No. 4,591,570, issued 5/27/86) describes an array of a limited number of characterized antibodies to known antigens on a glass surface that can be used to bind to specific antigens on the surface of whole cells. [0008]
  • Recently new technologies have arisen that allow the creation of microarrays containing thousands or millions of different elements. Such array technology has been applied mainly to forming arrays of individual nucleic acids (see, for example, Marshall and Hodgson, Nature Biotech. 16:27-31, 1998; Ramsay, Nature Biotech. 16:40-44, 1998), in particular short oligonucleotides synthesized in situ. [0009]
  • Methods are needed to simply and rapidly screen very large numbers of uncharacterized antibodies for those specific for a given antigen as well as for the characterization of tissues and cells by nucleic acid and/or protein analysis. The invention described herein addresses that need. [0010]
  • BRIEF DESCRIPTION OF THE INVENTION
  • The invention disclosed herein comprises methods of using microarrays to simplify analysis and characterization of genes and their function. In one aspect of the invention the methods are used to identify and characterize antibodies having binding affinity for a specific target antigen. This method comprises contacting an array of uncharacterized antibodies bound to a solid surface with at least one target antigen and identifying the antibodies to which the target antigen binds. The method can be performed under a variety of conditions to identify antibodies with a range of binding affinities. [0011]
  • A second aspect of the invention comprises a method of determining gene expression at the protein level comprising contacting an array of characterized or uncharacterized antibodies on a solid surface with one or more proteins and identifying the antibodies to which said protein(s) binds. This method can be further used to compare the protein expression in two different populations of cells, such as normal cells and cancer cells or resting cells and stimulated cells. A related embodiment can be used as a tool in the diagnosis of various disorders. [0012]
  • A further aspect of the invention comprises a method of determining gene expression at the protein level comprising contacting a microarray of nucleic acid samples derived from a variety of different sources with one or more nucleic acid probes then identifying the sample or samples to which the probe binds.[0013]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIGS. 1A, 1B, and [0014] 1C show microarrays of antibodies bound to positively charged nylon, reacted with antigen and detected by non-fluorescent means.
  • FIG. 2 shows a microarray produced using a robotic arraying apparatus. Antigen binding is detected by non-fluorescent means. [0015]
  • FIG. 3 shows the ability of the antibody microarrays to evaluate relative binding affinities to a specific antigen. [0016]
  • FIG. 4 shows a microarray of polyclonal antibodies in comparison to a microarray of monoclonal antibodies. [0017]
  • FIGS. 5A, 5B and [0018] 5C show a microarray of antibodies reacted with a cell lysate under conditions that vary the amount of background binding.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention discloses methods of using microarrays to simplify analysis and characterization of genes and their function. In a first aspect of the invention the methods are used for identifying and characterizing antibodies having binding specificity to a particular antigen or set of antigens. This method utilizes microarray technology to create ordered matrices of large numbers of uncharacterized antibodies which can then be contacted with antigen under a variety of conditions. The method is rapid and simple to perform and is applicable to the simultaneous screening of very large numbers of antibodies. [0019]
  • Briefly, uncharacterized antibodies are bound to a solid surface in an array format consisting of discrete spots whose spatial location can be easily identified. Each location represents an antibody from a known source, such as a particular hybridoma growing in a well in a 96-well microtiter plate. The space between the antibody spots is treated to minimize non-specific binding to the solid support. The arrayed antibodies are then contacted with an antigen, or a set of antigens, for which specific antibodies are sought. The antigen solution is left in contact with the array for an amount of time sufficient to allow antigen:antibody complexes to form (generally 10 minutes to 2 hours), then the unbound antigen is washed away under suitable conditions. Bound antigen is detected at a particular antibody spot using one of a variety of detection methods, thus identifying the source of an antibody specific for the particular antigen. [0020]
  • The term “antibody” is used herein in the broadest sense and specifically includes intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies) formed from at least two intact antibodies, and antibody fragments, including single chain antibodies, so long as they exhibit the desired binding properties as described herein. [0021]
  • Various procedures well-known in the art may be used for the production of polyclonal antibodies to an epitope or antigen of interest. A host animal of any of a number of species, such as rabbits, goats, sheep, horse, cow, mice, rats, etc. is immunized by injection with an antigenic preparation which may be derived from cells or microorganisms, or may be recombinantly or synthetically produced. Various adjuvants well known in the art may be used to enhance the production of antibodies by the immunized host, for example, Freund's adjuvant (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, liposomes, potentially useful human adjuvants such as BCG (Bacille Calmette-Guerin) and [0022] Propionibacterium acanes, and the like.
  • The term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. Preferred antibodies are mAbs, which may be of any immunoglobulin class including IgG, IgM, IgE, IgA, and any subclass or isotype thereof. [0023]
  • In addition to their specificity, monoclonal antibodies are advantageous in that they are synthesized by hybridoma culture, uncontaminated by other immunoglobulins. The modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al., [0024] Nature, 256:495 (1975), or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567, incorporated by reference herein). The “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol. Biol., 222:581-597 (1991), for example.
  • The monoclonal antibodies contemplated for use herein specifically include “chimeric” antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567; Morrison et al., [0025] Proc. Natl. Acad. Sci. USA, 81:6851-6855 (1984)).
  • “Humanized” forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab′, F(ab′)[0026] 2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementarity-determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity, and capacity. In some instances, Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, humanized antibodies may comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. These modifications are made to further refine and maximize antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. For further details, see Jones et al., Nature, 321:522-525 (1986); Reichmann et al., Nature, 332:323-329 (1988); and Presta, Curr. Op. Struct. Biol., 2:593-596 (1992). The humanized antibody includes a PRIMATIZED™ antibody wherein the antigen-binding region of the antibody is derived from an antibody produced by immunizing macaque monkeys with the antigen of interest.
  • “Antibody fragments” comprise a portion of an intact antibody, preferably the antigen binding or variable region of the intact antibody. Examples of antibody fragments include Fab, Fab′, F(ab′)[0027] 2, and Fv fragments; diabodies; linear antibodies (Zapata et al. Protein Eng. 8(10):1057-1062 (1995)); single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • Particularly preferred in the practice of the invention are single-chain antibodies. “Single-chain” or “sFv” antibodies are antibody fragments comprising the V[0028] H and VLdomains of an antibody, wherein these domains are present in a single polypeptide chain. Preferably, the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the sFv to form the desired structure for antigen binding. For a review of sFvs see Pluckthun in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269-315 (1994).
  • Large quantities of single chain antibodies with uncharacterized randomized binding specificity can be produced using a number of methodologies known in the art. Recombinant antibody libraries can be created in filamentous phage particles (Daniels and Lane, Methods 9(3):494-507, 1996; Reichmann and Weill, Biochemistry 32(34):8848-8855; Rader and Barbas, Curr Opin Biotechnol 9(4):503-508, 1997; Iba and Kurosawa, Immunol Cell Biol 75(2):217-221, 1997, WO 90/05144, WO 92/01047, WO 92/20791, WO 93/19172, GB 9722131.8, GB9810228.8 and GB 9810223.9, all of which are incorporated by reference herein in their entirety), for example, or similarly in yeast, bacteria, and the like. Other methods for creating random libraries of sFvs include various solid state synthesis methods. [0029]
  • The term “diabodies” refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (V[0030] H) connected to a light-chain variable domain (VL) in the same polypeptide chain (VH-VL). By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites. Diabodies are described more fully in, for example, EP 404,097; WO 93/11161; and Hollinger et al., Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993).
  • The antibodies employed in the invention can be isolated prior to creating a microarray. An “isolated” molecule, whether an antibody, antigen or nucleic acid, is one which has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials which would interfere with particular uses for the molecule, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes. In preferred embodiments, a protein will be purified (1) to greater than 95% by weight of protein as determined by the Lowry method, and most preferably more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under reducing or nonreducing conditions using Coomassie blue or, preferably, silver stain. Isolated protein includes the protein in situ within recombinant cells since at least one component of the protein's natural environment will not be present. Ordinarily, however, isolated protein will be prepared by at least one purification step. Unpurified antibodies, such as those found in serum, can also be employed in the present invention. [0031]
  • By “isolated” in reference to nucleic acid is meant a polymer of 14, 17, 21 or more contiguous nucleotides, including DNA or RNA that is isolated from a natural source or that is synthesized. The isolated nucleic acid of the present invention is unique in the sense that it is not found in a pure or separated state in nature. Use of the term “isolated” indicates that a naturally occurring sequence has been removed from its normal cellular (i.e., chromosomal) environment. Thus, the sequence may be in a cell-free solution or placed in a different cellular environment. The term does not imply that the sequence is the only nucleotide sequence present, but that it is essentially free (about 90-95% pure at least) of non-nucleotide material naturally associated with it and thus is meant to be distinguished from isolated chromosomes. [0032]
  • One particularly useful method of isolating antibodies, such as single chain antibodies from a cell extract, is affinity purification. Resins suitable for antibody purification are well known in the art, for example, protein A SEPHAROSE™. A recombinant antibody can be engineered to contain an affinity purification tag to facilitate its purification. Resins suitable for antibody purification are well known in the art, for example, protein A SEPHAROSE™ resin. [0033]
  • Affinity purification tags are generally peptide sequences that can interact with a binding partner immobilized on a solid support. Synthetic DNA sequences encoding multiple consecutive single amino acids, such as histidine, when fused to the expressed protein, may be used for one-step purification of the recombinant protein by high affinity binding to a resin column, such as nickel SEPHAROSE™ resin. An endopeptidase recognition sequence can be engineered between the polyamino acid tag and the protein of interest to allow subsequent removal of the leader peptide by digestion with enterokinase, and other proteases. Sequences encoding peptides such as the chitin binding domain (which binds to chitin), biotin (which binds to avidin and strepavidin), and the like can also be used for facilitating purification of the protein of interest. The affinity purification tag can be separated from the protein of interest by methods well known in the art, including the use of inteins (protein self-splicing elements, Chong, et al, Gene 192:271-281, 1997). [0034]
  • By using an amount of resin with binding sites sufficient for only a small portion of the antibody present in the unpurified mixture, the process of isolation can be used to simultaneously normalize yield and isolate the antibody. For example, although each sample will contain a different and unknown amount of antibody protein, the samples can be contacted with an amount of resin whose maximum binding capacity is 10 mgs. Thus any antibody greater than this amount will pass through the resin unbound. The maximum bound amount can then be eluted from the resin. [0035]
  • Methods for creating microarrays are known in the art including printing on a solid surface using pins (passive pins, quill pins, and the like) or spotting with individual drops of solution. Passive pins draw up enough sample to dispense a single spot. Quill pins draw up enough liquid to dispense multiple spots. Bubble printers use a loop to capture a small volume which is dispensed by pushing a rod through the loop. Microdispensing uses a syringe mechanism to deliver multiple spots of a fixed volume. In addition, solid supports, can be arrayed using piezoelectric (ink jet) technology, which actively transfers samples to a solid support. [0036]
  • One method is described in Shalon and Brown (WO 95/35505, published 12/28/95) which is incorporated herein by reference in its entirety. The method and apparatus described in Shalon and Brown can create an array of up to six hundred spots per square centimeter on a glass slide using a volume of 0.01 to 100 nl per spot. Suitable concentrations of antibody range from about 1 ng/μl to about 1 μg/μl. In the present invention, each spot can contain one or more than one distinct antibody. [0037]
  • Other methods of creating arrays are known in the art, including photolithographic printing (Pease, et al, [0038] Proc. Natl. Acad. Sci. USA, 91(11):5022-5026, 1994) and in situ synthesis. While known in situ synthesis methods are less useful for synthesizing polypeptides long enough to be antibodies, they can be used to make polypeptides up to 50 amino acids in length, which can serve as binding proteins as described below.
  • The microarrays can be created on a variety of solid surfaces such as plastics (e.g. polycarbonate), complex carbohydrates (e.g. agarose and SEPHAROSE™), acrylic resins (e.g. polyacrylamide and latex beads), and nitrocellulose. Preferred solid support materials include glass slides, silicon wafers, and positively charged nylon. Specific examples of suitable solid supports are described in the Examples below. [0039]
  • Methods for covalent attachment of antibodies to a solid support are known in the art. Examples of such methods are found in Bhatia, et al, Anal. Biochem. 178(2):408-413, 1989; Ahluwalia, et al, Biosens. Bioelectron. 7(3):207-214, 1992; Jonsson, et al, Biochem. J. 227(2):373-378, 1985; and Freij-Larsson, et al, Biomaterials 17(22):2199-2207, 1996, all of which are incorporated by reference herein in their entirety. Proteins may additionally be attached to a solid support using methods described in the examples below. [0040]
  • Methods of reducing non-specific binding to a solid surface are well known in the art and include washing the arrayed solid surface with bovine serum albumin (BSA), reconstituted non-fat milk, salmon sperm DNA, porcine heparin, and the like (see Ausubel, et al., Short Protocols in Molecular Biology, 3rd ed. 1995). [0041]
  • The arrays used to identify antigen-specific antibodies are contacted with a solution containing one or more known antigens in order to identify antibodies in the array with binding specificity for the antigen. The antigens are often proteins, although they may also be organic chemical compounds, carbohydrates, nucleic acids, and the like. They may be isolated or semi-isolated, recombinant or naturally occurring. The amount of antigen used can vary from about 1-100 ng/μl. The antigen is left in contact with the array for an amount of time sufficient for antibody:antigen complexes to form, should one of the antibodies in the array be specific for the antigen. The amount of time sufficient for this purpose will range from 5 minutes to 24 hours, and will generally be from 0.5 to 2 hours. [0042]
  • One antigen of particular interest in the practice of the invention is recombinant protein, either a full-length gene product or a fragment thereof, for example an Expressed Sequence Tag (or EST fragment). EST fragments are relatively short cDNA sequences that have been randomly generated and sequenced, generally as part of an ongoing effort to map an entire genome (Adams, et al, Science 252(5013):1651-1656, 1991). Large numbers of these sequences are available in public databases. The identity of the proteins encoded by the vast majority of these sequences is unknown. The following discussion, although directed to the expression of EST-encoded peptides, is equally applicable to any expressed product of a nucleic acid sequence, including full-length proteins. [0043]
  • Techniques are available in the art by which cells can be genetically engineered to express the peptide encoded by a given EST fragment. The methods of the invention can then be used to identify antibodies specific for the peptide. These antibodies are then useful as reagents that can be employed in purification and identification of the full-length protein, and in other experimental procedures designed to elucidate the protein's location and function. [0044]
  • Prokaryotic hosts are, generally, very efficient and convenient for the production of recombinant proteins and are, therefore, one type of preferred expression system for EST fragments. Prokaryotes most frequently are represented by various strains of [0045] E. coli. However, other microbial strains may also be used, including other bacterial strains.
  • In prokaryotic systems, plasmid vectors that contain replication sites and control sequences derived from a species compatible with the host may be used. Examples of suitable plasmid vectors may include pBR322, pUC 118, pUC 119, and the like; suitable phage or bacteriophage vectors may include λgt10, λgt11, and the like; and suitable virus vectors may include pMAM-neo, PKRC and the like. Preferably, the selected vector of the present invention has the capacity to replicate in the selected host cell. [0046]
  • Recognized prokaryotic hosts include bacteria such as [0047] E. coli and those from genera such as Bacillus, Streptomyces, Pseudomonas, Salmonella, Serratia, and the like. However, under such conditions, the polypeptide will not be glycosylated. The prokaryotic host selected for use herein must be compatible with the replicon and control sequences in the expression plasmid.
  • To express an EST fragment in a prokaryotic cell, it is necessary to operably link the gene sequence to a functional prokaryotic promoter such as the T7 promoter or RSC promoter. Such promoters may be either constitutive or, more preferably, regulatable (i.e., inducible or derepressible). Examples of constitutive promoters include the int promoter of bacteriophage λ, the bla promoter of the β-lactamase gene sequence of pBR322, the CAT promoter of the chloramphenicol acetyl transferase gene sequence of pPR325, and the like. Examples of inducible prokaryotic promoters include the major right and left promoters of bacteriophage (P[0048] L and PR), the trp, reca, lacZ, LacI, and gal promoters of E. coli, the α-amylase (Ulmanen et al., J. Bacteriol. 162:176-182, 1985) and the sigma-28-specific promoters of B. subtilis (Gilman et al., Gene sequence 32:11-20(1984)), the promoters of the bacteriophages of Bacillus (Gryczan, In: The Molecular Biology of the Bacilli, Academic Press, Inc., NY (1982)), Streptomyces promoters (Ward et at., Mol. Gen. Genet. 203:468-478, 1986), and the like. Exemplary prokaryotic promoters are reviewed by Glick (J. Ind. Microbiol. 1:277-282, 1987); Cenatiempo (Biochimie 68:505-516, 1986); and Gottesman (Ann. Rev. Genet. 18:415-442, 1984).
  • Proper expression in a prokaryotic cell also requires the presence of a ribosome binding site upstream of the gene sequence-encoding sequence. Such ribosome binding sites are disclosed, for example, by Gold et at. (Ann. Rev. Microbiol. 35:365-404, 1981). The selection of control sequences, expression vectors, transformation methods, and the like, are dependent on the type of host cell used to express the gene. [0049]
  • Host cells which may be used in the expression systems of the present invention are not strictly limited, provided that they are suitable for use in the expression of the peptide of interest. Suitable hosts may often include eukaryotic cells. Preferred eukaryotic hosts include, for example, yeast, fungi, insect cells, and mammalian cells either in vivo, or in tissue culture. Mammalian cells which may be useful as hosts include HeLa cells, cells of fibroblast origin such as VERO, 3T3 or CHOK1, HEK 293 cells or cells of lymphoid origin (such as 32D cells) and their derivatives. Preferred mammalian host cells include SP2/0 and JS58L, as well as neuroblastoma cell lines such as IMR 332 and PC12 which may provide better capacities for correct post-translational processing. [0050]
  • In addition, plant cells are also available as hosts, and control sequences compatible with plant cells are available, such as the cauliflower mosaic virus 35S and 19S, nopaline synthase promoter and polyadenylation signal sequences, and the like. Another preferred host is an insect cell, for example the Drosophila larvae. Using insect cells as hosts, the Drosophila alcohol dehydrogenase promoter can be used. Rubin, Science 240:1453-1459, 1988). Alternatively, baculovirus vectors can be engineered to express large amounts of peptide encoded by an EST fragment in insects cells (Jasny, Science 238:1653, 1987); Miller et al., In: Genetic Engineering (1986), Setlow, J. K., et al., eds., Plenum, Vol. 8, pp. 277-297). [0051]
  • Any of a series of yeast gene sequence expression systems can be utilized which incorporate promoter and termination elements from the actively expressed gene sequences coding for glycolytic enzymes which are produced in large quantities when yeast are grown in media rich in glucose. Known glycolytic gene sequences can also provide very efficient transcriptional control signals. Yeast provides substantial advantages in that it can also carry out post-translational peptide modifications. A number of recombinant DNA strategies exist which utilize strong promoter sequences and high copy number of plasmids which can be utilized for production of the desired proteins in yeast. Yeast recognizes leader sequences on cloned mammalian gene sequence products and secretes peptides bearing leader sequences (i.e., pre-peptides). For a mammalian host, several possible vector systems are available for the expression of and EST fragment [0052]
  • A wide variety of transcriptional and translational regulatory sequences may be employed, depending upon the nature of the host. The transcriptional and translational regulatory signals may be derived from viral sources, such as adenovirus, bovine papilloma virus, cytomegalovirus, simian virus, or the like, where the regulatory signals are associated with a particular gene sequence which has a high level of expression. Alternatively, promoters from mammalian expression products, such as actin, collagen, myosin, and the like, may be employed. Transcriptional initiation regulatory signals may be selected which allow for repression or activation, so that expression of the gene sequences can be modulated. Of interest are regulatory signals which are temperature-sensitive so that by varying the temperature, expression can be repressed or initiated, or are subject to chemical (such as metabolite) regulation. [0053]
  • Expression of an EST fragment in eukaryotic hosts involves the use of eukaryotic regulatory regions. Such regions will, in general, include a promoter region sufficient to direct the initiation of RNA synthesis. Preferred eukaryotic promoters include, for example, the promoter of the mouse metallothionein I gene sequence (Hamer et al., J. Mol. Appl. Gen. 1:273-288, 1982); the TK promoter of Herpes virus (McKnight, Cell 31:355-365, 1982); the SV40 early promoter (Benoist et al., Nature (London) 290:304-310, 1981); the yeast gal4 gene sequence promoter (Johnston et al., Proc. Natl. Acad. Sci. (USA) 79:6971-6975, 1982); Silver et al., Proc. Natl. Acad. Sci. (USA) 81:5951-5955, 1984), the CMV promoter, the EF-1 promoter, and the like. [0054]
  • An EST fragment and an operably linked promoter may be introduced into a recipient prokaryotic or eukaryotic cell either as a nonreplicating DNA (or RNA) molecule, which may either be a linear molecule or, more preferably, a closed covalent circular molecule (a plasmid). Since such molecules are incapable of autonomous replication, the expression of the gene may occur through the transient expression of the introduced sequence. Alternatively, permanent or stable expression may occur through the integration of the introduced DNA sequence into the host chromosome. [0055]
  • A vector may be employed which is capable of integrating the desired gene sequences into the host cell chromosome. Cells which have stably integrated the introduced DNA into their chromosomes can be selected by also introducing one or more markers which allow for selection of host cells which contain the expression vector. The marker may provide for prototrophy to an auxotrophic host, biocide resistance, e.g., antibiotics, or heavy metals, such as copper, or the like. The selectable marker gene sequence can either be directly linked to the DNA gene sequences to be expressed, or introduced into the same cell by cotransfection. Common selectable marker gene sequences include those for resistance to antibiotics such as ampicillin, tetracycline, kanamycin, bleomycin, streptomycin, hygromycin, neomycin, Zeocin™, and the like. Selectable auxotrophic gene sequences include, for example, hisD, which allows growth in histidine free media in the presence of histidinol. [0056]
  • Additional elements may also be needed for optimal synthesis of single chain binding protein mRNA. These elements may include splice signals, as well as transcription promoters, enhancers, and termination signals. cDNA expression vectors incorporating such elements include those described by Okayama, Mol. Cell. Bio. 3:280, 1983. [0057]
  • The recombinant antigen may be produced as a fusion protein. When two protein-coding sequences not normally associated with each other in nature are in the same reading frame the resulting expressed protein is called a “fusion protein” as two distinct proteins have been “fused” together. Fusion proteins have a wide variety of uses. For example, two functional enzymes can be fused to produce a single protein with multiple enzymatic activities or short peptide sequences, such as epitope tags or affinity purification tags (see above), can be fused to a larger protein and serve as aids in purification or as means of identifying the expressed protein by serving as epitopes detectable by specific antibodies. [0058]
  • Epitope tags are short peptide sequences that are recognized by epitope-specific antibodies. A fusion protein comprising a recombinant protein and an epitope tag can be simply and easily purified using an antibody bound to a chromatography resin. The presence of the epitope tag furthermore allows the recombinant protein to be detected in subsequent assays, such as Western blots, without having to produce an antibody specific for the recombinant protein itself. Examples of commonly used epitope tags include V5, glutathione-S-transferase (GST), hemagglutinin (HA), the peptide Phe-His-His-Thr-Thr, chitin binding domain, and the like. [0059]
  • A fusion protein may be a means by which the recombinant antigen protein can be easily detected. For example, the fusion component can itself be a detectable moiety, such as fluorescent protein (fluorescent green protein, fluorescent yellow protein, and the like), or alternatively can be one member of a specific binding pair (such as biotin and streptavidin, for example) which can be detected by reacting with the other member conjugated to a detectable substance. [0060]
  • The foregoing elements can be combined to produce vectors suitable for use in the methods of the invention. Those of skill in the art would be able to select and combine the elements suitable for use in their particular system. [0061]
  • The introduced nucleic acid molecule can be incorporated into a plasmid or viral vector capable of autonomous replication in the recipient host. Any of a wide variety of vectors may be employed for this purpose. Factors of importance in selecting a particular plasmid or viral vector include: the ease with which recipient cells that contain the vector may be recognized and selected from those recipient cells which do not contain the vector; the number of copies of the vector which are desired in a particular host; and whether it is desirable to be able to “shuttle” the vector between host cells of different species. [0062]
  • Suitable prokaryotic vectors include plasmids such as those capable of replication in [0063] E. coli (for example, pBR322, ColEl, pSC101, PACYC 184, itVX, pRSET, pBAD (Invitrogen, Carlsbad, Calif.), and the like). Such plasmids are disclosed by Sambrook (cf. “Molecular Cloning: A Laboratory Manual”, second edition, edited by Sambrook, Fritsch, & Maniatis, Cold Spring Harbor Laboratory, (1989)). Bacillus plasmids include pC194, pC221, pT127, and the like, and are disclosed by Gryczan (In: The Molecular Biology of the Bacilli, Academic Press, NY (1982), pp. 307-329). Suitable Streptomyces plasmids include plJlOl (Kendall et al., J. Bacteriol. 169:4177-4183,1987), and Streptomyces bacteriophages such as φC31 (Chater et al., In: Sixth International Symposium on Actinomycetales Biology, Akademiai Kaido, Budapest, Hungary (1986), pp. 45-54). Pseudomonas plasmids are reviewed by John et al. (Rev. Infect. Dis. 8:693-704, 1986), and Izaki (Jpn. J. Bacteriol. 33:729-742, 1978).
  • Suitable eukaryotic plasmids include, for example, BPV, vaccinia, SV40, 2-micron circle, pCDN3.1 (Invitrogen), and the like, or their derivatives. Such plasmids are well known in the art (Botstein et al., Miami Wntr. Symp. 19:265-274, 1982); Broach, In: “The Molecular Biology of the Yeast Saccharomyces: Life Cycle and Inheritance”, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., p. 445-470 (1981); Broach, Cell 28:203-204, 1982); Dilon et at., J. Clin. Hematol. Oncol. 10:39-48, 1980); Maniatis, In: Cell Biology: A Comprehensive Treatise, Vol. 3, Gene Sequence Expression, Academic Press, NY, pp. 563-608 (1980). [0064]
  • Once antibody:antigen complexes have been formed and unbound antigen washed away under suitable conditions, the antibody:antigen complexes can be detected using one of several techniques known in the art. Suitable washing conditions are known to those skilled in the art (see, for example, Ausubel, et al, Short Protocols in Molecular Biology, 3rd ed. 1995). Exemplary washing conditions are shown in the examples below. [0065]
  • For detection in the case of recombinant antigens, expression vectors can be used that form chimeric fusion peptides as described above. The epitope tagged antigen can be 55464 detected using an antibody specific for the tag sequence. This antibody may be itself detectably labeled or can be detected with a third detectably-labeled antibody. Alternatively, the antigen can be complexed with biotin and detected using detectably-labeled avidin or streptavidin. The antigen itself can also be detectably labeled, such as with a fluorescent dye compound. [0066]
  • The term “detectably labeled” as used herein is intended to encompass antigen directly coupled to a detectable substance, such as a fluorescent dye, and antigen coupled to a member of binding pair, such as biotin/streptavidin, or an epitope tag that can specifically interact with a molecule that can be detected, such as by producing a colored substrate or fluorescence. [0067]
  • Substances suitable for detectably labeling proteins include fluorescent dyes such as fluorescein isothiocyanate (FITC), fluorescein, rhodamine, tetramethyl-rhodamine-5-(and 6)-isothiocyanate (TRITC), Texas red, cyanine dyes (Cy3 and Cy5, for example), and the like; and enzymes that react with colorimetric substrates such as horseradish peroxidase. The use of fluorescent dyes is generally preferred in the practice of the invention as they can be detected at very low amounts. Furthermore, in the case where multiple antigens are reacted with a single array, each antigen can be labeled with a distinct fluorescent compound for simultaneous detection. Labeled spots on the array are detected using a fluorimeter, the presence of a signal indicating an antigen bound to a specific antibody. [0068]
  • The formation of antibody:antigen complexes can be performed under a variety of conditions to identify antibodies with varying binding characteristics. Antigen-containing reaction solutions can contain varying degrees of salt or be conducted at varying pH levels. In addition, the binding reaction can be carried out at varying temperatures. Each set of conditions will identify antibodies with different affinity for the antigen. For example, antibodies that bind at [0069] pH 2 may have utility under highly acidic conditions such as those that exist in the stomach. Similarly, antibodies that bind at temperatures near boiling may be useful in studying thermophilic organisms. In general pH conditions will range from 2-10 (most preferably around pH 8), temperatures from 0° C.-100° C. and salt conditions from 1 μM to 5 M (in the case of NaCl).
  • Affinity constants are a measure of the interaction between a particular ligand and its cognate receptor. The “binding affinity” or the measure of the strength of association between a particular antibody:antigen interaction is generally measured by affinity constants for the equilibrium concentrations of associated and dissociated configurations of the antibody and its antigen. Preferably the binding of the antigen should occur at an affinity of about k[0070] a=10−6M or greater to be useful for the present invention, with greater than about 10−7M being more preferable, and most preferably between about 10−8M and about 10−11M. Antibody fragments will generally have affinities in the range of about 10−6M to 10−7M.
  • In another embodiment of the invention, microarrays of uncharacterized antibodies are used to compare the protein expression profiles of cells. For example, comparisons can be made between a population of cells from one tissue, such as arterial endothelial cells, and a second tissue, such as venous endothelial cells or from cells derived from a particular tissue but from different species. Comparisons can be made between normal cells and cells from the same tissue type that originate from an individual with a pathogenic disorder. For example, comparisons can be made between normal cells and cancer cells. Comparisons can additionally be made between cells in a resting state and cells in an activated state, for example, resting T-cells and activated T-cells. [0071]
  • In another example, the disclosed arrays are useful for evaluating the expression of proteins by pathogens, such as, for example, bacteria, parasites, viruses, and the like. A solution (such as a lysate) made from the pathogen which represents all proteins expressed by the pathogen can be used to contact an antibody array to identify antibodies recognizing pathogen-expressed proteins. These antibodies have utility as diagnostic agents as well as potential therapeutics. [0072]
  • Cellular lysates can be used as “antigens” as described above and reacted with two identical microarrays. Antibodies reactive in one array but not the other would indicate the presence of a differentially expressed protein. This antibody is then useful for the subsequent isolation and identification of those proteins that are different in two populations of cells. In the case of normal and cancer cells, for example, one may be able to identify proteins expressed in the cancer cell that contribute to its malignant state. [0073]
  • In a further aspect of the invention, microarrays can be composed of previously characterized antibodies. These microarrays have a variety of uses, one of which is cell profiling. For example, an array can be composed of antibodies that recognize a set of antigens known to be present in activated T-cells but not in resting T-cells. A population of T-cells can then be lysed and the lysate contacted with the array to determine if the population has the profile of activated or resting T-cells. [0074]
  • Microarrays and the methods disclosed herein can be used in methods of diagnosing particular disorders. For example, a collection of antibodies specific for a range of antigens associated with one or more disorders can be arrayed and contacted with a bodily fluid containing antigens whose presence, or absence, would indicate a particular disorder. The advantage of using a microarray over a conventional immunoassay is the ability to include a population of antibodies diagnostic for a variety of disorders on a single surface, significantly reducing time, costs and materials needed to effect a diagnosis. [0075]
  • For example, if a patient presents with symptoms that are characteristic of several distinct disorders which can be distinguished on the basis of the presence or absence of one or more proteins, a single microarray assay could be used to make a specific diagnosis, thus allowing the patient to be properly treated. Patients suffering from stroke or brain infarcts release several proteins into cerebrospinal fluid, examples of which are neuron specific enolyse (NSE) from neuronal cells and S-100 from glial cells and astrocytes. Such proteins are not released in conditions that may have similar symptoms, such as drug reactions, making proper diagnosis more difficult. A diagnostic array could readily detect these and other proteins in the CSF, leading to a rapid clinical diagnosis and treatment. [0076]
  • In another aspect of the invention microarrays are employed to characterize protein expression patterns using nucleic acid samples. Briefly, nucleic acid molecules from a whole cell or tissue are applied to a solid support using a microarray format. The arrayed nucleic acid samples are then contacted with a nucleic acid probe specific for a gene encoding a known protein. The probe solution is left in contact with the array for an amount of time sufficient to allow sample:probe complexes to form, then the unbound probe is washed away under suitable conditions (see, for example, Ausubel, et al, Short Protocols in Molecular Biology, 3rd ed. 1995 and the examples below). Bound probe is detected at one or more nucleic acid sample spots using one of a variety of detection methods. [0077]
  • This aspect of the invention has a variety of uses. For example, the microarray can be constructed from nucleic acid samples isolated from a single tissue type but from a large number of species, with each spot representing a particular species. Thus in a single assay format one can determine the evolutionary development of the protein represented by the probe. Similarly, the microarray can be constructed of multiple tissue types from a single species, or from different developmental stages of a single species (or multiple species) thus simply and efficiently determining tissue expression of the protein represented by the probe. For example, a microarray can be constructed with arrayed samples representing all the developmental stages of Drosophila, a well known organism the study of which has led to a greater understanding of mammalian physiology and development. [0078]
  • The nucleic acid sample can be messenger ribonucleic acid (mRNA) or can be complementary deoxyribonucleic acid (cDNA), including EST fragments. Methods for extracting and isolating nucleic acids from cells are well known in the art (for example phenol extraction/ethanol precipitation, ammonium acetate precipitation, cesium chloride gradients, and the like), as are methods for generating cDNA (see, for example, “Molecular Cloning: A Laboratory Manual,” second edition, edited by Sambrook, Fritsch, & Maniatis, Cold Spring Harbor Laboratory, 1989; and Ausubel, et al, Short Protocols in Molecular Biology, 3rd ed. 1995, both of which are incorporated by reference herein). Microarrays of these nucleic acids are created using the methods described above. Techniques for coupling nucleic acids to solid supports used to construct microarrays are well known in the art, including the poly-L-lysine and phenylboronic acid methods described in the Examples below. [0079]
  • The nucleic acid probes used in the invention methods can be designed based on the sequence of a gene encoding a known protein or can be an EST fragment, as described above. One skilled in the art can readily design such probes based on the known sequence using methods of computer alignment and sequence analysis known in the art (e.g., “Molecular Cloning: A Laboratory Manual”, second edition, edited by Sambrook, Fritsch, & Maniatis, Cold Spring Harbor Laboratory, 1989; Ausubel, et al, Short Protocols in Molecular Biology, 3rd ed. 1995). The probe can comprise any number of nucleotides but will preferably be not fewer than 10 nucleotides and preferably not more than about 300 nucleotides in length. [0080]
  • The probes of the invention can be labeled by standard labeling techniques such as with a radiolabel, enzyme label, fluorescent label, biotin-avidin label, chemiluminescent label, and the like. After hybridization, the probes may be detected using known methods. Preferred labels are fluorescent labels, as described above. [0081]
  • The nucleic acid probes of the present invention include RNA as well as DNA probes and nucleic acids modified in the sugar, phosphate or even the base portion as long as the probe still retains the ability to specifically hybridize under conditions as disclosed herein. Such probes are generated using techniques known in the art. [0082]
  • The term “hybridize” as used herein refers to a method of interacting a nucleic acid sequence with a DNA or RNA molecule in solution or on a solid support, such as cellulose or nitrocellulose. If a nucleic acid sequence binds to the DNA or RNA molecule with sufficiently high affinity, it is said to “hybridize” to the DNA or RNA molecule. The strength of the interaction between the probing sequence and its target can be assessed by varying the stringency of the hybridization conditions. Various low to high stringency hybridization conditions may be used depending upon the specificity and selectivity desired. Stringency is controlled by varying salt or denaturant concentrations. Examples of hybridization conditions are shown in the Examples below. Those skilled in the art readily recognize how such conditions can be varied to vary specificity and selectivity. For example, under highly stringent hybridization conditions only highly complementary nucleic acid sequences hybridize. Preferably, such conditions prevent hybridization of nucleic acids having even one or two mismatches out of 20 contiguous nucleotides. [0083]
  • In a further aspect of the invention, microarrays can be composed of randomly generated polynucleotides (DNA or RNA) and contacted with proteins to identify unique binding pairs. Polynucleotides are now known to bind to proteins and may have potential as diagnostics and therapeutics (see, for example, Allen, et al, Virology 209(2):327-336, 1995; Binkley, et al, Nucleic Acids Res. 23(16):3198-3205, 1995). Polynucleotides can be evaluated in very large numbers using the methods disclosed herein thus increasing the likelihood of identifying a useful binder. [0084]
  • The invention will now be described in greater detail by reference to the following non-limiting examples. [0085]
  • EXAMPLES Example I-Nucleic Acid Microarrays
  • The following procedures are conducted at room temperature and using double distilled water unless otherwise noted. These methods are applicable to arrays of polypeptides or polynucleic acids. [0086]
  • Glass slides are prepared as follows: NaOH (50 g) is dissolved in 150 ml of double distilled water (ddH[0087] 2O), then 200 ml of 95% EtOH is added while stirring. If the solution becomes cloudy, ddH2O is added until it becomes clear. Approximately 30 glass slides (Gold Seal, Cat. No. 3010) are soaked in the NaOH/EtOH solution for 2 hours, shaking. The slides are then rinsed three times with ddH2O. The slides are next soaked in a poly-L-lysine solution (70 ml poly-L-lysine (Sigma Cat. No. 8920) to 280 ddH2O) for 1 hour. Excess liquid is removed by spinning the slides in a rack on a microtiter plate carrier at 500 rpm. The slides are dried at 40° C. for 5 minutes, then stored in a closed box for at least 2 weeks prior to use.
  • A cDNA microarray is prepared as follows: Total MRNA is isolated from tissue (for example, nerve cells) of a variety of species representative of different classes of organisms such as Drosophila, nematode, salmon, clam, chicken, mouse, dog, goat, spider monkey, chimpanzee, human, and the like, by the FastTrac method (Stratagene, La Jolla, Calif.) or other common methods. mRNA is also obtained from a variety of unicellular organisms such as [0088] E. coli, yeast, B. subtilis, mycoplasma and the like. Eukaryotic mRNA is enriched from total RNA using oligo(dT) cellulose (Ausubel, et al, Short Protocols in Molecular Biology, 3rd ed. 1995, pgs 4-11-4-12). Equivalent amounts (for example, 1 μg) of mRNA from each source are placed in a separate well of one or more 96 well microtiter plates and precipitated with cold EtOH. The precipitate is rinsed with 70% EtOH and allowed to dry.
  • The dried mRNA is resuspended in 3× SSC (sodium chloride/sodium citrate—20X solution is 3 M NaCl (175g/L0 0.3 M trisodium citrate 2H[0089] 2O (88g/L adjusted to pH 7.0 with 1 M HCl) then spotted onto a previously prepared glass slide using an array device (for example, Shalon and Brown (WO 95/35505, published 12/28/95)). The prepared array can be kept for a long period of time before probing, however, if the slides are to be kept for long periods of time, stability is increased by converting each mRNA sample into cDNA using techniques known in the art, such as PCR.
  • The array is rehydrated by suspending the slide over a dish of ddH[0090] 2O (50° C.) for approximately one minute. The slide is quickly (approximately 3 seconds) dried by placing it on a surface heated to 100° C. (mRNA side up). The mRNA is crosslinked to the poly-L-lysine coating of the slide using ultraviolet radiation using a Stratalinker™ UV device according to the manufacturer's instructions (Stratagene) set at 60 milliJoules.
  • The slides are next soaked in a solution of 5 grams of succinic anhydride (Aldrich Cat. No. 23,969-0) dissolved in 315 ml of N-methyl-pyrrilidinone (Aldrich Cat. No. 32,863-4) plus 35 mls of 0.2 M sodium borate (brought to pH 8.0 with NaOH) for 15 minutes with shaking. The slide is then transferred to a 95° C. water bath for 2 minutes followed by 95% EtOH for 1 minute. Excess liquid is removed from the slides by spinning a rack of slides on a microtiter plate carrier at 500 rpm. [0091]
  • A probe sequence of a known protein (for example, human nerve growth factor, GenBank Accession No. E03589) is labeled using standard protocols, for example by using a CyDye™ Nick Translation kit (Amersham). The labeled probe (approximately 1 μg/ml) is resuspended in 4× SSC (10 μl) to which is added 0.2 μl 10% sodium dodecyl sulfate (SDS). The probe is boiled for 2 minutes, then cooled for 10 seconds and transferred to the array by pipette. The array is covered by a 22 mm×22 mm cover slip, and the slide is placed in a humid hybridization chamber and submerged into a hot water bath (≧75° C.). [0092]
  • The slide is left in the bath for 10-24 hours, then the cover slip is removed and the slide rinsed in 0.2× SSC with 0.1% SDS several times. Excess wash buffer is removed by centrifugation on a microtiter plate carrier as described above. The slide is scanned using a spectrofluorometer, such as the ScanArray 3000 (General Scanning Inc., Watertown, Mass.). For probes labeled with Cy5, for example, fluorescence is measured at 670 nm. Localization of spots on the array to which the probe hybridizes indicates that the species represented by the spot expresses a protein similar or identical to the probe protein. [0093]
  • The procedure outlined below is an alternative method for binding arrayed molecules to a solid support, using an SA(OCH[0094] 2CN)-X-NHS linkage (see, for example, U.S. Pat. No. 5,594,111, issued 1/14/97; U.S. Pat. No. 5,648,470, issued May 15, 1997; U.S. Pat. No. 5,623,055, issued Apr. 22, 1997; all of which are incorporated by reference herein).
  • Glass slides (Fisher Catalog No. 12-544-4) are soaked in an acid bath (1 hour in 0.1 M HCl), then washed with water and dried at room temperature. The slides should not be aggressively dried, such as in an oven. The slides are next soaked in a silane solution overnight at room temperature (5% APTES (3-aminopropyl-triethoxysilane, Aldrich 28,177-8), 0.3% DIEA (Sigma) v/v in EtOH). The slides may be sonicated for 10-15 minutes right after being placed in the APTES solution. [0095]
  • The slides are rinsed with isopropyl alcohol, then sonicated in isopropyl alcohol for several minutes. Sonication should remove any white silane residue on the slides. If the residue remains, the slides should be discarded. After sonication, the slides are left to cure/dry for at least 24 hours before use. [0096]
  • The cured slides are next soaked in a linker solution overnight at room temperature. The linker solution is made by dissolving 115 mg of 9Y SA(OCH[0097] 2CN)-X-COOH (Prolix, Bothell, Wash.) in 1 ml dimethylformamide (DMF) plus 60 μl DIEA, then adding 60 mg TSTU (Sigma) and leaving for 15 minutes at room temperature. This stock is diluted in 270 ml of isopropyl alcohol plus 270 μl DIEA before using.
  • The slides are removed from the linker solution and soaked in 1 M NH[0098] 2OH, 1 mM EDTA, 0.1 M NaHCO3 (pH 10) for 4 hours at room temperature. This solution is removed, the slides are extensively washed with water then let air dry at room temperature. The slides can be stored at room temperature away from light before using to make arrays.
  • Example II Determination of Optimal Concentrations of Antibody and Antigen
  • Various concentrations (1 μg/μl, 100 ng/μl, 10 ng/μl, 1 ng/μl) of total mouse IgG or a mouse monoclonal anti-PLC-gamma were spotted on aldehyde slides (Cel Associates, Inc., Houston, Tex.), which allow non-covalent attachment of proteins. Using a manual [0099] 8 pin hand arrayer the slides were blocked for 1 hour with PBST (phosphate buffered saline and 0.10% Tween 20), and 3% milk protein. The slides were subsequently washed three times, 15 minutes each, in PBST. Duplicate slides were incubated with 50 μl of goat anti-mouse IgG antibody (GAMG) conjugated with CY3 or CY5 fluorescent dye compounds (Amersham, Arlington Heights, Ill.) at 10 μg/ml or 1 μg/ml. Slides were then washed for 15 minutes in PBST three additional times and dried by centrifugation prior to scanning. Binding was detected as shown in Table 1 below.
  • Example III Comparison of Solid Supports
  • Serial dilutions (1 μg/ml, 100 ng/ml, 10 ng/ml, 1 ng/ml) of mouse IgG or PLC-gamma were hand arrayed onto aldehyde, polystyrene, nitrocellulose and Surmodics slides. Aldehyde, nitrocellulose, polystyrene and Surmodics slides were purchased from various outside vendors (aldehyde Slides—Cel Associates, Inc., Houston, Tex.; nitrocellulose Slides —Molecular Probes, Inc., Eugene, Oreg.; polystyrene Slides—Nunc, Inc., Naperville, Ill.; Surmodics Slides—Surmodics, Inc., Eden Prairie, Minn.). Surmodics slides have an undisclosed polymer on the glass surface which forms a covalent linkage with proteins under the appropriate conditions (described by the manufacturer). [0100]
  • Following hand arraying of the antibodies (approximately 20-30 nanoliters per spot), the nitrocellulose, aldehyde, and polystyrene slides were immediately blocked for 1 hour with PBST and 3% milk, washed 3 times with PBST, and hybridized with 50 μl of GAMG-CY3 for 30 minutes. Surmodics slides were incubated overnight in a moist salt chamber as recommended by the manufacturer. The following day, the Surmodics slides were processed as described above. Following hybridization all of the various slides were washed 3 times in PB ST, dried and scanned using a Scan Array 3000 fluorescent scanner. [0101]
    TABLE 1
    Detection
    Antibody Conc. Antigen Conc. Level
    PLC-gamma 1 μg/μl GAMG-CY3 10 μg/ml +++
    100 ng/μl 10 μg/ml +++
    10 ng/μl 10 μg/ml +
    1 ng/μl 10 μg/ml
    mouse IgG 1 μg/μl GAMG-CY3 10 μg/ml +++
    100 ng/μl 10 μg/ml +++
    10 ng/μl 10 μg/ml +
    1 ng/μl 10 μg/ml
    PLC-gamma 1 μg/μl GAMG-CY3 1 μg/ml +
    100 ng/μl 1 μg/ml +
    10 ng/μl 1 μg/ml
    1 ng/μl 1 μg/ml
    mouse IgG 1 ng/μl GAMG-CY3 1 μg/ml +
    100 ng/μl 1 μg/ml +
    10 ng/μl 1 μg/ml
    1 ng/μl 1 μg/ml
    PLC-gamma 1 μg/gl GAMG-CY5 1 μg/ml +++
    100 ng/μl 1 μg/ml +++
    10 ng/μl 1 μg/ml +
    1 ng/μl 1 μg/ml
    mouse IgG 1 ng/μl GAMG-CY5 1 μg/ml +++
    100 ng/μl 1 μg/ml +++
    10 ng/μl 1 μg/ml +
    1 ng/μl 1 μg/ml
    PLC-gamma 1 μg/μl GAMG-CY5 1 μg/ml +
    100 ng/μl 1 μg/ml +
    10 ng/μl 1 μg/ml
    1 ng/μl 1 μg/ml
    mouse IgG 1 μg/μl GAMG-CY5 1 μg/ml +
    100 ng/μl 1 μg/ml +
    10 ng/μl 1 μg/ml
    1 ng/μl 1 μg/ml
  • All of the slides tested allowed for the detection of antigen:antibody binding at higher concentrations of antibody. The Aldehyde and Nitrocellulose treated slides were the most efficient at binding antibody, and antibody:antigen interaction could be detected at 1 ng/μl. [0102]
  • Example IV Detection of Binding Using Non-Fluorescent Methods
  • Positively charged nylon filters (Zeta Probe Membranes, BioRad Laboratories, Hercules, Calif.) were hand arrayed using 1 μl of anti-His, anti-V5, anti-thioredoxin (anti-Thio), anti-FOS, anti-PLC-gamma and anti-CREB antibodies (Invitrogen, Carlsbad, Calif.; all antibodies were approximately 1 mg/ml). Filters were blocked for 1 hour with PBST and 3% milk, washed three times with PBST, and incubated with 1 μg/ml biotinylated D1 protein for three hours at room temperature. D1 is a creatine kinase fusion protein isolated from a human fetal heart cDNA library and cloned into the pBAD-Thio-His-TOPO vector (Invitrogen, Carlsbad, Calif.) to create a Thioredoxin-V5-His-creatine kinase fusion protein. D1 was biotinylated using the EZ-Link™ Sulfo-NHS-LC Biotinylation Kit (Pierce, Rockford, Ill.) used according to the manufacturer's instructions). [0103]
  • Following three additional washes with the same buffer, filters were treated with streptavidin/alkaline phosphatase conjugate or streptavidin/horseradish peroxidase conjugate (Boehringer Mannheim, GmbH Germany) for 1 hour at room temperature. [0104]
  • The filters were washed 5 times with PBST, dried, and developed by immersion in ECL chemiluminescent substrate (ECL—Amersham, Arlington Heights, Ill.) or the chromogenic substrate BCIP/NBT (Sigma Chemicals, St. Louis, Mo.). Filters developed with ECL were exposed to Kodak chemiluminescent film for 1 to 10 seconds. [0105]
  • The results are shown in FIGS. 1A, 1B, and [0106] 1C. In all cases, only the antibodies specific for epitopes on the fusion protein antigen were detectable, and only in the arrayed spots, showing that the system has both good signal to noise ratio and specificity.
  • The experiment was repeated using an array created with an automated arrayer. Antibodies (1 mg/ml) were spotted using an automated 96 pin microarrayer developed at Invitrogen. Fifteen negative control antibodies (assorted mouse monoclonals) were arrayed along with the three positive control antibodies (anti-His, anti-Thio, anti-V5). Filters were treated as described above using the alkaline phosphatase conjugate and the chromogenic substrate BCIP/NBT. [0107]
  • As can been seen in FIG. 2, binding and detection of antibody:antigen complexes was highly specific and sensitive. [0108]
  • Example V Evaluation of Antibody Affinity
  • Anti-His, anti-V5, anti-FOS, anti-PLC-gamma, 25ClDG, and anti-VEGF (vascular endothelial growth factor) antibodies were arrayed on a nitrocellulose slide and reacted with biotinylated D1 protein as previously described. Binding was detected with streptavidin-Cy3 as described above. The anti-V5 antibodies spots showed red, the anti-His spots showed green, while the negative controls were undetectable (see FIG. 3). When viewed in a black and white drawing, relative increase in binding affinity is visualized by an increase of white in a given area. The color of the spots generally indicates a higher amount of fluorescently labeled antigen present, and thus indicates relative binding affinity between antibody and antigen. Colors, in descending order from highest to lowest affinity, are white, red, yellow, green, and blue. Using this technique, multiple antibodies can be tested for their affinity to a single antigen. [0109]
  • Example VI Polyclonal Antibody Microarrays
  • To demonstrate specific binding to polyclonal antibodies, six antibodies were arrayed by hand on a nitrocellulose slide, three polyclonal antibodies (anti-E12 (unpurified rabbit polyclonal sera to a His-V5-thioredoxin-thymidine kinase fusion protein), anti-lexA (lexA repressor protein), and anti-GFP(Green fluorescent protein)) and three monoclonal antibodies (anti-V5, anti-His and anti-GalU (a mammalian transcription factor). The slide was blocked with PBST and 3% milk for 1 hour at room temperature, and incubated with the E12-biotin conjugate, prepared according to the protocol used for D1 protein. Following extensive washing with PBST, the slides were incubated with streptavidin-CY3 conjugate (Amersham, Arlington Heights, Ill.) for 1 hour at room temperature, washed 5 times with PBST and dried by centrifugation prior to scanning on the Scan Array 3000. [0110]
  • As can be seen in FIG. 4, binding was detected with both the antigen specific polyclonal antibody (anti-E12) and the antigen specific monoclonal antibodies (anti-His, anti-V5) and not with any of the negative control antibodies. [0111]
  • Example VII Microarray Analysis of Labeled Cell Lysate
  • A series of experiments were conducted to determine if a microarray of antibodies could specifically detect antigens in a cell lysate. [0112]
  • CHO cells expressing high levels of beta-galactosidase were grown to confluency in a T-175 flask. (Hams media with Pen/Strep, and L-glutamine plus 10% FCS, at 37° C. with 5% CO[0113] 2) Cells were harvested using Trypsin/EDTA. NP40 extracts were prepared by pelleting the cells (107 cells), washing once in PBS and resuspending in 5% NP40. Cell debris was removed by centrifugation. Soluble protein was biotinylated using a Pierce biotinylation kit according to the manufacturer's instructions.
  • Nitrocellulose slides (see above) containing arrayed monoclonal antibodies (anti-beta-gal, anti-His, anti-Thio, anti-V5, anti-FOS, anti-PLC-gamma, anti-VEGF and 25C10G (an anti-CREB antibody) were blocked, washed, hybridized and developed with streptavidin-CY3 as described in Example VI supra. As can be seen in FIG. 5A, beta-galactosidase binding was seen, however, some non-specific binding was detected as well. [0114]
  • The experiment was repeated, except that after centrifugation of the extract, soluble protein was dialyzed overnight against 50 mM phosphate buffer at 4° C. prior to biotinylation. As can be seen in Figure [0115] 5B, much of the non-specific binding seen in the previous experiment was eliminated.
  • In the next experiment dialyzed extract containing the biotinylated soluble proteins was adjusted to 10% glycerol to reduce non-specific hydrophobic interactions. Furthermore, the sodium chloride concentration was adjusted to 0.2 M NaCl to increase specific ionic interactions. All other conditions remained identical. As can be seen in FIG. 5C, all non-specific binding was eliminated using this protocol. [0116]

Claims (50)

That which is claimed is:
1. A method of identifying antibodies having binding affinity for an antigen, said method comprising:
(a) contacting an array of uncharacterized antibodies on a solid surface with at least one antigen; and
(b) identifying the antibodies to which the antigen binds.
2. A method according to claim 1 wherein the antigen is a protein.
3. A method according to claim 1 wherein the antigen is an intact cell.
4. A method according to claim 1 wherein the antigen is a cell lysate.
5. A method according to claim 2 wherein the protein is recombinant.
6. A method according to claim 5 wherein the protein is full-length.
7. A method according to claim 5 wherein the protein is a protein fragment.
8. A method according to claim 7 wherein the protein fragment is encoded by an EST fragment.
9. A method according to claim 1 wherein the antibodies are monoclonal antibodies.
10. A method according to claim 1 wherein the antibodies are polyclonal antibodies.
11. A method according to claim 1 wherein the antibodies are antibody fragments.
12. A method according to claim 11 wherein the antibody fragments are single chain antibodies.
13. A method according to claim 1 wherein the antibodies are recombinant antibodies.
14. A method according to claim 1 wherein the antigen is detectably labeled.
15. A method according to claim 14 wherein the detectable label is a fluorescent moiety, avidin, streptavidin, or biotin.
16. A method according to claim 1 wherein the antigen is a fusion protein comprised of an epitope tag or a fluorescent protein.
17. A method according to claim 1 wherein the binding affinity of said antibody for said antigen is determined by iterative washing of said solid surface with a suitable diluent and detecting when antigen is no longer released therefrom.
18. A method of comparing protein expression in two or more populations of cells, said method comprising:
(a) contacting an array of antibodies on a solid surface with a cell lysate of a first cell population, generating a first binding pattern;
(b) contacting a duplicate array of antibodies on a solid surface with a cell lysate of a second cell population, generating a second binding pattern; and
(c) comparing the binding pattern of the first cell lysate with the binding pattern of the second cell lysate.
19. A method according to claim 18 wherein the antibodies are uncharacterized antibodies.
20. A method according to claim 18 wherein the antibodies are recombinant antibodies.
21. A method according to claim 18 wherein the first cell lysate is derived from normal cells and the second cell lysate is derived from abnormal cells.
22. A method according to claim 21 wherein the abnormal cells are cancer cells.
23. A method according to claim 18 wherein the first cell lysate is derived from normal cells in a resting state and the second cell lysate is derived from normal cells in a stimulated state.
24. A method according to claim 18 wherein the difference between the first and second set of cells is the presence of a different detectable label.
25. A method for determining the effect of varying binding conditions on the binding affinity of antibodies to a specific antigen, said method comprising:
(a) contacting an array of antibodies on a solid surface with at least one antigen under a first set of binding conditions, generating a first binding pattern;
(b) contacting a duplicate array with the antigen under a second set of binding conditions, generating a second binding pattern;
(c) comparing the first and second binding patterns.
26. A method according to claim 21 wherein said varying binding conditions comprise varying pH, temperature, salt concentration, and/or duration.
27. A method for characterizing a cell, based on the pattern of protein expression produced thereby, said method comprising:
(a) contacting an array of antibodies on a solid surface with a cell lysate; and
(b) identifying the profile of antibodies to which components of the lysate binds.
28. A method of diagnosing a disorder, said method comprising:
(a) contacting an array of antibodies specific for one or more antigens characteristic of a disorder with a biological sample obtained from a subject under conditions suitable for the formation of an antigen:antibody complex, wherein the presence of the antigens in the biological sample would be indicative of the disorder;
and
(b) detecting the formation of any antibody: antigen complexes.
29. A method according to claim 28 wherein the biological sample is cerebral spinal fluid, blood, plasma, urine, feces, saliva, tears, or extracted tissue.
30. A method according to claim 29 wherein the disorder is stroke, cerebral hemorrhage, myocardial infarction, peripheral blood clots, diabetes, cancer, Alzheimer's disease, and sepsis.
31. A kit comprising:
(a) an array of uncharacterized antibodies on a solid surface; and
(b) instructions for using the array.
32. A kit according to claim 31 wherein the instructions are for identifying antibodies to a specific antigen, comparing protein expression in two or more populations of cells, characterizing a cell based on the pattern of protein expression produced thereby, or determining the effect of varying binding conditions on the binding affinity of the antibodies.
33. A kit according to claim 31 wherein the antibodies are monoclonal antibodies, polyclonal antibodies or antibody fragments.
34. A kit according to claim 33 wherein the antibody fragments are single chain antibodies.
35. A kit according to claim 31 wherein the antibodies are recombinant antibodies.
36. A kit according to claim 31 further comprising reagents for detecting an antigen and instructions for use thereof.
37. A kit comprising:
(a) an array of antibodies on a solid surface; and
(b) instructions for using the array; wherein the instructions are for diagnosing a disorder, characterizing a cell based on the pattern of protein expression produced thereby, or comparing protein expression in two or more populations of cells.
38. A kit according to claim 37 further comprising reagents for detecting an antigen and instructions for use thereof.
39. A kit according to claim 37 wherein the antibodies are recombinant antibodies.
40. A kit according to claim 37 wherein the antibodies are single chain antibodies.
41. A method of comparing protein expression patterns, said method comprising:
(a) contacting a microarray of nucleic acid samples derived from different sources with one or more nucleic acid probes and
(b) identifying the sample or samples to which the probe(s) binds.
42. A method according to claim 41 wherein the microarray comprises nucleic acid samples derived from a single tissue type but from different species.
43. A method according to claim 41 wherein the microarray comprises nucleic acid samples derived from a single species but from different tissue types.
44. A method according to claim 41 wherein the microarray comprises nucleic acid samples derived from the same tissue type at different developmental stages.
45. A method according to claim 41 wherein the nucleic acid samples are comprised of mRNA or cDNA.
46. A method according to claim 41 wherein the probe is detectably labeled.
47. A method according to claim 46 wherein the detectable label is a fluorescent label.
48. A method according to claim 18 wherein the first and second cell lysates are derived from cells from a single tissue type but from different species.
49. A method according to claim 18 wherein the first and second cell lysates are derived from cells from a single species but from different tissue types.
50. A method according to claim 18 wherein the first and second cell lysates are derived from cells from the same tissue type at different developmental stages.
US10/035,368 1998-02-04 2001-10-26 Microarrays and uses therefor Abandoned US20020164656A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US10/035,368 US20020164656A1 (en) 1998-02-04 2001-10-26 Microarrays and uses therefor
US12/490,069 US8012703B2 (en) 1998-02-04 2009-06-23 Microarrays and uses therefor
US13/173,637 US8637264B2 (en) 1998-02-04 2011-06-30 Microarrays and uses therefor
US13/648,169 US20130123127A1 (en) 1998-02-04 2012-10-09 Microarrays and uses thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US7360598P 1998-02-04 1998-02-04
US09/245,615 US7794946B1 (en) 1998-02-04 1999-02-04 Microarray and uses therefor
US10/035,368 US20020164656A1 (en) 1998-02-04 2001-10-26 Microarrays and uses therefor

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/245,615 Division US7794946B1 (en) 1998-02-04 1999-02-04 Microarray and uses therefor

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/490,069 Continuation US8012703B2 (en) 1998-02-04 2009-06-23 Microarrays and uses therefor

Publications (1)

Publication Number Publication Date
US20020164656A1 true US20020164656A1 (en) 2002-11-07

Family

ID=22114700

Family Applications (5)

Application Number Title Priority Date Filing Date
US09/245,615 Expired - Fee Related US7794946B1 (en) 1998-02-04 1999-02-04 Microarray and uses therefor
US10/035,368 Abandoned US20020164656A1 (en) 1998-02-04 2001-10-26 Microarrays and uses therefor
US12/490,069 Expired - Fee Related US8012703B2 (en) 1998-02-04 2009-06-23 Microarrays and uses therefor
US13/173,637 Expired - Fee Related US8637264B2 (en) 1998-02-04 2011-06-30 Microarrays and uses therefor
US13/648,169 Abandoned US20130123127A1 (en) 1998-02-04 2012-10-09 Microarrays and uses thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/245,615 Expired - Fee Related US7794946B1 (en) 1998-02-04 1999-02-04 Microarray and uses therefor

Family Applications After (3)

Application Number Title Priority Date Filing Date
US12/490,069 Expired - Fee Related US8012703B2 (en) 1998-02-04 2009-06-23 Microarrays and uses therefor
US13/173,637 Expired - Fee Related US8637264B2 (en) 1998-02-04 2011-06-30 Microarrays and uses therefor
US13/648,169 Abandoned US20130123127A1 (en) 1998-02-04 2012-10-09 Microarrays and uses thereof

Country Status (8)

Country Link
US (5) US7794946B1 (en)
EP (3) EP2189791A3 (en)
JP (1) JP2002502977A (en)
AT (1) ATE393912T1 (en)
AU (1) AU2583899A (en)
CA (1) CA2318175A1 (en)
DE (1) DE69938623T2 (en)
WO (1) WO1999040434A1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20010039014A1 (en) * 2000-01-11 2001-11-08 Maxygen, Inc. Integrated systems and methods for diversity generation and screening
EP1583845A2 (en) * 2003-01-02 2005-10-12 Bioforce Nanosciences, Inc. Method and apparatus for molecular analysis in small sample volumes
US20050260683A1 (en) * 2002-10-23 2005-11-24 University Of Hawaii Methods for diagnosing and treating pre-term labor
US20060292559A1 (en) * 2005-06-23 2006-12-28 Beckman Coulter, Inc. Cell-based microarrays, and methods for their preparation and use
US20080070235A1 (en) * 2003-04-02 2008-03-20 Sirs-Lab Gmbh Method for Recognizing Acute Generalized Inflammatory Conditions (Sirs), Sepsis, Sepsis-Like Conditions and Systemic Infections
US20090075831A1 (en) * 2004-10-13 2009-03-19 Stefan Russwurm Method for differentiating between the non-infectious and infectious causes of multiple organ failure
US7687437B2 (en) 2001-07-13 2010-03-30 Nanosphere, Inc. Method for immobilizing molecules onto surfaces
US20100086909A1 (en) * 2004-03-30 2010-04-08 Sirs-Lab Gmbh Method for the prediction of individual disease course in sepsis
US7794946B1 (en) 1998-02-04 2010-09-14 Life Technologies Corporation Microarray and uses therefor
US20120015347A1 (en) * 2010-07-16 2012-01-19 Anupam Singhal Methods for Assaying Cellular Binding Interactions
US8399383B2 (en) 2000-05-04 2013-03-19 Yale University Protein chips for high throughput screening of protein activity
DE10315031B4 (en) * 2003-04-02 2014-02-13 Analytik Jena Ag Method for detecting sepsis and / or sepsis-like conditions
US20230347021A1 (en) * 2021-10-25 2023-11-02 Singular Genomics Systems, Inc. Manipulating and detecting biological samples

Families Citing this family (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2319828A1 (en) * 1998-01-29 1999-08-05 Miller, Samuel High density arrays for proteome analysis and methods and compositions therefor
US6780582B1 (en) 1998-07-14 2004-08-24 Zyomyx, Inc. Arrays of protein-capture agents and methods of use thereof
US6406921B1 (en) 1998-07-14 2002-06-18 Zyomyx, Incorporated Protein arrays for high-throughput screening
US6682942B1 (en) 1998-07-14 2004-01-27 Zyomyx, Inc. Microdevices for screening biomolecules
US6576478B1 (en) 1998-07-14 2003-06-10 Zyomyx, Inc. Microdevices for high-throughput screening of biomolecules
CA2350111A1 (en) 1998-11-16 2000-05-25 Genway Biotech, Inc. Generation of antibodies using polynucleotide vaccination in avian species
NZ525421A (en) 1998-12-23 2004-10-29 Univ Sydney Method of treating cancer by determining the presence of the disease condition using an assay to determine the binding pattern of immobilised immunoglobulins
JP3942431B2 (en) * 1999-08-31 2007-07-11 三菱化学株式会社 Protein-molecule interaction analysis method
JP2003511696A (en) * 1999-10-13 2003-03-25 インサイト・ゲノミックス・インコーポレイテッド Multiple cytokine analysis
JP2003516526A (en) 1999-11-28 2003-05-13 ラ ホヤ ファーマシューティカル カンパニー Methods for treating lupus based on antibody affinity and screening methods and compositions for use thereof
GB9928789D0 (en) * 1999-12-03 2000-02-02 Medical Res Council Naive screening method
US9335328B2 (en) * 2000-02-03 2016-05-10 Instant Medical Diagnostics, Llc Method and apparatus for signal transduction pathway profiling
CA2399189A1 (en) * 2000-02-22 2001-08-30 Genospectra, Inc. Microarray fabrication techniques and apparatus
JP2003529056A (en) 2000-02-22 2003-09-30 ジェノスペクトラ,インコーポレイティド Microarray fabrication technology and equipment
DE10023130B4 (en) 2000-05-11 2007-10-18 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Hypha specific factors from Candida albicans
WO2002008765A2 (en) * 2000-07-26 2002-01-31 Stanford University Basal cell markers in breast cancer and uses thereof
WO2002014866A2 (en) * 2000-08-11 2002-02-21 Qianjin Hu Methods and universal monoclonal antibody array
AUPR005600A0 (en) * 2000-09-12 2000-10-05 University Of Sydney, The Diagnostic assay
WO2002025288A2 (en) 2000-09-22 2002-03-28 Clontech Laboratories Inc. Highly sensitive proteomic analysis methods and kits and systems for practicing the same
US20020137104A1 (en) * 2000-11-27 2002-09-26 Antonio Cosma High throughput determination of antigen expression
EP1270740A1 (en) * 2001-06-29 2003-01-02 SIRS-Lab GmbH Biochip and its use for determining inflammation
KR20030011982A (en) * 2001-07-25 2003-02-12 주식회사 인투젠 Target gene for diagnosis of gastric cancer and development of anticancer drugs identified by cDNA microarray analysis and solid support for microarray analysis arrayed the same
US20030228709A1 (en) * 2002-03-25 2003-12-11 Kozlowski Roland Zbignieiw Arrays
AU2003282832B2 (en) * 2002-10-15 2009-09-10 Abmetrix, Inc. Sets of digital antibodies directed against short epitopes, and methods using same
WO2007024877A2 (en) 2005-08-22 2007-03-01 Cornell Research Foundation, Inc. Compositions and methods for analyzing protein interactions
AU2010221418B2 (en) 2009-03-02 2015-06-04 Dignity Health Diagnostic devices and methods of use
CN107574157A (en) 2009-03-16 2018-01-12 盘古生物制药有限公司 Include the composition and method of the Histidyl-tRNA-synthetase splicing variants with non-classical bioactivity
US20100310576A1 (en) 2009-03-31 2010-12-09 Adams Ryan A COMPOSITIONS AND METHODS COMPRISING ASPARTYL-tRNA SYNTHETASES HAVING NON-CANONICAL BIOLOGICAL ACTIVITIES
WO2011139714A2 (en) 2010-04-26 2011-11-10 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of cysteinyl-trna synthetase
CN103096911B (en) 2010-04-27 2018-05-29 Atyr 医药公司 Treatment relevant with the protein fragments of Isoleucyl-tRNA synthetase, diagnosis and the innovation of antibody compositions are found
JP6008837B2 (en) 2010-04-28 2016-10-19 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of alanyl tRNA synthetase
US9598734B2 (en) 2010-04-29 2017-03-21 Medical Prognosis Institute A/S Methods and devices for predicting treatment efficacy
CN103118693B (en) 2010-04-29 2017-05-03 Atyr 医药公司 Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of valyl tRNA synthetases
EP2563912B1 (en) 2010-04-29 2018-09-05 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of asparaginyl trna synthetases
WO2011139986A2 (en) 2010-05-03 2011-11-10 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of arginyl-trna synthetases
US9034321B2 (en) 2010-05-03 2015-05-19 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-alpha-tRNA synthetases
CA2797978C (en) 2010-05-03 2019-12-03 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of methionyl-trna synthetases
CA2798139C (en) 2010-05-04 2019-09-24 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of p38 multi-trna synthetase complex
JP6396656B2 (en) 2010-05-14 2018-09-26 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of phenylalanyl βtRNA synthetase
JP6027965B2 (en) 2010-05-17 2016-11-16 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of leucyl-tRNA synthetase
EP2575856B1 (en) 2010-05-27 2017-08-16 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutaminyl-trna synthetases
US8962560B2 (en) 2010-06-01 2015-02-24 Atyr Pharma Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of Lysyl-tRNA synthetases
JP6116479B2 (en) 2010-07-12 2017-04-19 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of glycyl-tRNA synthetase
CN103108650A (en) 2010-08-25 2013-05-15 Atyr医药公司 Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of tyrosyl-trna synthetases
CA2836836A1 (en) 2011-06-01 2012-12-06 Medical Prognosis Institute A/S Methods and devices for prognosis of cancer relapse
ES1076494Y (en) * 2012-02-15 2012-06-12 Duart Andoni Monforte DEVICE FOR DRINKING ELABORATION
JP6170077B2 (en) 2012-02-16 2017-07-26 エータイアー ファーマ, インコーポレイテッド Histidyl tRNA synthetase for treating autoimmune and inflammatory diseases
DE102012020496A1 (en) 2012-10-18 2014-04-24 Charité - Universitätsmedizin Berlin Invitro diagnosis and/or early detection of neurofibromatosis type 1 and/or neurofibromatosis type 1-associated tumor from sample of a patient for treating the patient with the tumor, comprises detecting protein or its fragment in sample
US10260103B2 (en) 2012-11-27 2019-04-16 Pontificia Universidad Catolica De Chile Compositions and methods for diagnosing thyroid tumors
WO2014195032A1 (en) 2013-06-07 2014-12-11 Medical Prognosis Institute A/S Methods and devices for predicting treatment efficacy of fulvestrant in cancer patients
DK3169815T3 (en) 2014-07-15 2021-02-15 Ontario Institute For Cancer Res METHODS AND DEVICES FOR PREDICTING THE EFFICIENCY OF ANTHRACYCLINE TREATMENT
CN108398554B (en) * 2018-03-07 2020-08-07 深圳市伯劳特生物制品有限公司 anti-TORCH-IgM antibody spectrum chip, preparation method thereof and TORCH detection kit
CN108398564B (en) * 2018-03-07 2020-08-07 深圳市伯劳特生物制品有限公司 anti-TORCH-IgG type antibody spectrum chip, preparation method thereof and TORCH detection kit
CN113238039B (en) * 2018-03-07 2023-03-31 深圳市伯劳特生物制品有限公司 Detection kit

Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4591570A (en) * 1983-02-02 1986-05-27 Centocor, Inc. Matrix of antibody-coated spots for determination of antigens
US4994373A (en) * 1983-01-27 1991-02-19 Enzo Biochem, Inc. Method and structures employing chemically-labelled polynucleotide probes
US5252743A (en) * 1989-11-13 1993-10-12 Affymax Technologies N.V. Spatially-addressable immobilization of anti-ligands on surfaces
US5283283A (en) * 1991-08-02 1994-02-01 The Goodyear Tire & Rubber Company High modulus blend of poly(α-amino acid) in rubbery elastomer
US5384263A (en) * 1987-10-13 1995-01-24 Terrapin Technologies, Inc. Method to produce immunodiagnostic reagents
US5436134A (en) * 1993-04-13 1995-07-25 Molecular Probes, Inc. Cyclic-substituted unsymmetrical cyanine dyes
US5532142A (en) * 1993-02-12 1996-07-02 Board Of Regents, The University Of Texas System Method of isolation and purification of fusion polypeptides
US5538897A (en) * 1994-03-14 1996-07-23 University Of Washington Use of mass spectrometry fragmentation patterns of peptides to identify amino acid sequences in databases
US5674712A (en) * 1993-03-10 1997-10-07 Eniricerche S.P.A. Recombinant vector and use thereof for exocellular preparation of antibodies in single molecule form from Bacillus subtilis
US5807522A (en) * 1994-06-17 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods for fabricating microarrays of biological samples
US6309820B1 (en) * 1995-04-07 2001-10-30 University Of North Carolina At Chapel Hill Polypeptides having a functional domain of interest and methods of identifying and using same
US6635311B1 (en) * 1999-01-07 2003-10-21 Northwestern University Methods utilizing scanning probe microscope tips and products therefor or products thereby
US6899137B2 (en) * 1999-06-28 2005-05-31 California Institute Of Technology Microfabricated elastomeric valve and pump systems
US20050182242A1 (en) * 2001-05-11 2005-08-18 Michael Snyder Global analysis of protein activities using proteome chips
US7132251B1 (en) * 2000-05-04 2006-11-07 Procognia Ltd Method and composition for analyzing a carbohydrate polymer

Family Cites Families (185)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4071409A (en) 1976-05-20 1978-01-31 Corning Glass Works Immobilization of proteins on inorganic support materials
US4281061A (en) * 1979-07-27 1981-07-28 Syva Company Double antibody for enhanced sensitivity in immunoassay
US4722896A (en) 1981-01-26 1988-02-02 The Beth Israel Hospital Association Method for affinity purification of hybridoma antibodies
US4444879A (en) * 1981-01-29 1984-04-24 Science Research Center, Inc. Immunoassay with article having support film and immunological counterpart of analyte
NZ199722A (en) 1981-02-25 1985-12-13 Genentech Inc Dna transfer vector for expression of exogenous polypeptide in yeast;transformed yeast strain
DE3269567D1 (en) 1981-04-29 1986-04-10 Ciba Geigy Ag New devices and kits for immunological analysis
US4483929A (en) * 1982-05-03 1984-11-20 Liposome Technology Incorporated Liposomes with glycolipid-linked antibodies
US4690715A (en) 1982-06-18 1987-09-01 American Telephone And Telegraph Company, At&T Bell Laboratories Modification of the properties of metals
US4514508A (en) * 1982-07-06 1985-04-30 Biond Inc. Assaying for a multiplicity of antigens or antibodies with a detection compound
US4973493A (en) 1982-09-29 1990-11-27 Bio-Metric Systems, Inc. Method of improving the biocompatibility of solid surfaces
US4865998A (en) * 1983-03-11 1989-09-12 Sloan-Kettering Institute For Cancer Research Monoclonal antibodies to human lung cancers and method
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
GB8314523D0 (en) 1983-05-25 1983-06-29 Lowe C R Diagnostic device
US5096807A (en) 1985-03-06 1992-03-17 Murex Corporation Imaging immunoassay detection system with background compensation and its use
WO1986006170A1 (en) * 1985-04-10 1986-10-23 Immunicon Corporation Direct homogeneous assay
US5866363A (en) 1985-08-28 1999-02-02 Pieczenik; George Method and means for sorting and identifying biological information
US4894146A (en) 1986-01-27 1990-01-16 University Of Utah Thin channel split flow process and apparatus for particle fractionation
US4802951A (en) 1986-03-07 1989-02-07 Trustees Of Boston University Method for parallel fabrication of nanometer scale multi-device structures
US4728591A (en) 1986-03-07 1988-03-01 Trustees Of Boston University Self-assembled nanometer lithographic masks and templates and method for parallel fabrication of nanometer scale multi-device structures
US5643948A (en) 1986-06-11 1997-07-01 Procyon Pharmaceuticals, Inc. Protein kinase C modulators. K.
US4859538A (en) 1986-11-20 1989-08-22 Ribi Hans O Novel lipid-protein compositions and articles and methods for their preparation
US5079600A (en) 1987-03-06 1992-01-07 Schnur Joel M High resolution patterning on solid substrates
JPH056800Y2 (en) * 1987-04-27 1993-02-22
US5154808A (en) 1987-06-26 1992-10-13 Fuji Photo Film Co., Ltd. Functional organic thin film and process for producing the same
US4987032A (en) 1987-06-26 1991-01-22 Fuji Photo Film Co., Ltd. Functional organic thin film and method of manufacture thereof
US4880750A (en) 1987-07-09 1989-11-14 Miragen, Inc. Individual-specific antibody identification methods
GB8803000D0 (en) 1988-02-10 1988-03-09 Ekins Roger Philip Determination of ambient concentrations of several analytes
US5807755A (en) 1987-08-06 1998-09-15 Multilyte Limited Determination of ambient concentrations of several analytes
US5300425A (en) 1987-10-13 1994-04-05 Terrapin Technologies, Inc. Method to produce immunodiagnostic reagents
CA1339069C (en) * 1987-11-09 1997-07-29 Henry Lee Niman Polypeptide-induced monoclonal receptors to protein ligand
US6692751B1 (en) 1988-05-06 2004-02-17 New York Blood Center Methods and systems for producing recombinant viral antigens
US4908112A (en) 1988-06-16 1990-03-13 E. I. Du Pont De Nemours & Co. Silicon semiconductor wafer for analyzing micronic biological samples
US5075077A (en) 1988-08-02 1991-12-24 Abbott Laboratories Test card for performing assays
US5281540A (en) 1988-08-02 1994-01-25 Abbott Laboratories Test array for performing assays
SE462454B (en) 1988-11-10 1990-06-25 Pharmacia Ab METHOD FOR USE IN BIOSENSORS
EP0368684B2 (en) 1988-11-11 2004-09-29 Medical Research Council Cloning immunoglobulin variable domain sequences.
IT1229691B (en) 1989-04-21 1991-09-06 Eniricerche Spa SENSOR WITH ANTIGEN CHEMICALLY LINKED TO A SEMICONDUCTIVE DEVICE.
US5143854A (en) 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
US6346413B1 (en) 1989-06-07 2002-02-12 Affymetrix, Inc. Polymer arrays
US5424186A (en) 1989-06-07 1995-06-13 Affymax Technologies N.V. Very large scale immobilized polymer synthesis
US5800992A (en) 1989-06-07 1998-09-01 Fodor; Stephen P.A. Method of detecting nucleic acids
US6919211B1 (en) 1989-06-07 2005-07-19 Affymetrix, Inc. Polypeptide arrays
US6040138A (en) * 1995-09-15 2000-03-21 Affymetrix, Inc. Expression monitoring by hybridization to high density oligonucleotide arrays
US6416952B1 (en) 1989-06-07 2002-07-09 Affymetrix, Inc. Photolithographic and other means for manufacturing arrays
US5744101A (en) 1989-06-07 1998-04-28 Affymax Technologies N.V. Photolabile nucleoside protecting groups
US5491097A (en) * 1989-06-15 1996-02-13 Biocircuits Corporation Analyte detection with multilayered bioelectronic conductivity sensors
DE3920358A1 (en) 1989-06-22 1991-01-17 Behringwerke Ag BISPECIFIC AND OLIGO-SPECIFIC, MONO- AND OLIGOVALENT ANTI-BODY CONSTRUCTS, THEIR PRODUCTION AND USE
DE3939973A1 (en) 1989-12-02 1991-06-06 Basf Ag TWO-DIMENSIONAL CRYSTALLIZED MACROMOLECULAR LAYERS
US5283173A (en) 1990-01-24 1994-02-01 The Research Foundation Of State University Of New York System to detect protein-protein interactions
US5858188A (en) 1990-02-28 1999-01-12 Aclara Biosciences, Inc. Acrylic microchannels and their use in electrophoretic applications
SE470347B (en) 1990-05-10 1994-01-31 Pharmacia Lkb Biotech Microstructure for fluid flow systems and process for manufacturing such a system
JPH06508511A (en) 1990-07-10 1994-09-29 ケンブリッジ アンティボディー テクノロジー リミティド Method for producing specific binding pair members
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5665582A (en) 1990-10-29 1997-09-09 Dekalb Genetics Corp. Isolation of biological materials
US5639620A (en) 1990-10-31 1997-06-17 Coulter Corporation Polymeric particles having a biodegradable gelatin or aminodextran coating and process for making same
WO1992008788A1 (en) 1990-11-19 1992-05-29 The Board Of Trustees Of The University Of Illinois Mutant orientable proteins and coated substrates
US5294369A (en) 1990-12-05 1994-03-15 Akzo N.V. Ligand gold bonding
DE69132905T2 (en) 1990-12-06 2002-08-01 Affymetrix Inc N D Ges D Staat Detection of nucleic acid sequences
WO1992013629A1 (en) * 1991-01-31 1992-08-20 Wayne State University A method for analyzing an organic sample
US5763170A (en) 1991-04-16 1998-06-09 Amersham International Plc Method for forming an array of biological particles
JP3097866B2 (en) 1991-10-15 2000-10-10 マルティライト リミティド Binding assays using labeling reagents
US5605662A (en) * 1993-11-01 1997-02-25 Nanogen, Inc. Active programmable electronic devices for molecular biological analysis and diagnostics
US6051380A (en) 1993-11-01 2000-04-18 Nanogen, Inc. Methods and procedures for molecular biological analysis and diagnostics
US5846708A (en) 1991-11-19 1998-12-08 Massachusetts Institiute Of Technology Optical and electrical methods and apparatus for molecule detection
IL103674A0 (en) 1991-11-19 1993-04-04 Houston Advanced Res Center Method and apparatus for molecule detection
US6943034B1 (en) 1991-11-22 2005-09-13 Affymetrix, Inc. Combinatorial strategies for polymer synthesis
US5412087A (en) 1992-04-24 1995-05-02 Affymax Technologies N.V. Spatially-addressable immobilization of oligonucleotides and other biological polymers on surfaces
WO1993009668A1 (en) 1991-11-22 1993-05-27 Affymax Technology N.V. Combinatorial strategies for polymer synthesis
US5384261A (en) 1991-11-22 1995-01-24 Affymax Technologies N.V. Very large scale immobilized polymer synthesis using mechanically directed flow paths
DK1136556T3 (en) 1991-11-25 2005-10-03 Enzon Inc Process for the preparation of multivalent antigen-binding proteins
DK1024191T3 (en) 1991-12-02 2008-12-08 Medical Res Council Preparation of autoantibodies displayed on phage surfaces from antibody segment libraries
DE59108591D1 (en) 1991-12-06 1997-04-10 Ciba Geigy Ag Electrophoretic separation device and electrophoretic separation process
JP3507073B2 (en) 1992-03-24 2004-03-15 ケンブリッジ アンティボディー テクノロジー リミティド Methods for producing members of a specific binding pair
CA2064683A1 (en) 1992-03-26 1993-09-27 Krishna Mohan Rao Kallury Formation of thermostable enzymes with extra-ordinary heat tolerance by immobilization on phospholipid matrices
US6100099A (en) 1994-09-06 2000-08-08 Abbott Laboratories Test strip having a diagonal array of capture spots
DE69305160T2 (en) 1992-04-22 1997-04-30 Ecole Polytech LIPID MEMBRANES FOR BIOSENSOR APPLICATIONS
US6107059A (en) 1992-04-29 2000-08-22 Affymax Technologies N.V. Peptide library and screening method
US5637469A (en) 1992-05-01 1997-06-10 Trustees Of The University Of Pennsylvania Methods and apparatus for the detection of an analyte utilizing mesoscale flow systems
US5726026A (en) 1992-05-01 1998-03-10 Trustees Of The University Of Pennsylvania Mesoscale sample preparation device and systems for determination and processing of analytes
US5304487A (en) 1992-05-01 1994-04-19 Trustees Of The University Of Pennsylvania Fluid handling in mesoscale analytical devices
US5348886A (en) 1992-09-04 1994-09-20 Monsanto Company Method of producing recombinant eukaryotic viruses in bacteria
DE4237113B4 (en) 1992-11-03 2006-10-12 "Iba Gmbh" Peptides and their fusion proteins, expression vector and method of producing a fusion protein
CA2108705A1 (en) 1992-11-06 1994-05-07 Richard Barner Biologically recognizing layers on new ti02 waveguide for biosensors
US5472881A (en) 1992-11-12 1995-12-05 University Of Utah Research Foundation Thiol labeling of DNA for attachment to gold surfaces
US5677196A (en) 1993-05-18 1997-10-14 University Of Utah Research Foundation Apparatus and methods for multi-analyte homogeneous fluoro-immunoassays
US5512492A (en) 1993-05-18 1996-04-30 University Of Utah Research Foundation Waveguide immunosensor with coating chemistry providing enhanced sensitivity
CA2512290C (en) 1993-05-28 2010-02-02 Baylor College Of Medicine Method and apparatus for desorption and ionization of analytes
US5861242A (en) 1993-06-25 1999-01-19 Affymetrix, Inc. Array of nucleic acid probes on biological chips for diagnosis of HIV and methods of using the same
US5837832A (en) 1993-06-25 1998-11-17 Affymetrix, Inc. Arrays of nucleic acid probes on biological chips
US5441876A (en) 1993-07-30 1995-08-15 The United States Of America As Represented By The Secretary Of The Navy Process for the preparation of headgroup-modified phospholipids using phosphatidylhydroxyalkanols as intermediates
RU2041262C1 (en) 1993-08-11 1995-08-09 Институт молекулярной биологии им.В.А.Энгельгардта РАН Method for immobilization of water soluble bioorganic compounds on capillary-porous carrier
US5776748A (en) 1993-10-04 1998-07-07 President And Fellows Of Harvard College Method of formation of microstamped patterns on plates for adhesion of cells and other biological materials, devices and uses therefor
US5512131A (en) 1993-10-04 1996-04-30 President And Fellows Of Harvard College Formation of microstamped patterns on surfaces and derivative articles
ATE424927T1 (en) 1993-10-28 2009-03-15 Houston Advanced Res Ct MICROFABRICATED POROUS FLOW DEVICE FOR DISCRETE DETERMINATION OF BONDING REACTIONS
US5429807A (en) 1993-10-28 1995-07-04 Beckman Instruments, Inc. Method and apparatus for creating biopolymer arrays on a solid support surface
US5429708A (en) 1993-12-22 1995-07-04 The Board Of Trustees Of The Leland Stanford Junior University Molecular layers covalently bonded to silicon surfaces
GB9326451D0 (en) 1993-12-24 1994-02-23 Multilyte Ltd Binding assay
GB9326450D0 (en) * 1993-12-24 1994-02-23 Multilyte Ltd Binding assay
DE4435728A1 (en) 1994-01-19 1995-07-20 Boehringer Mannheim Gmbh Biotin silane compounds and binding matrix containing these compounds
ES2179853T3 (en) 1994-01-19 2003-02-01 Roche Diagnostics Gmbh BIOTIN-SILANOS COMPOUNDS AND FIXING MATRIX CONTAINING THESE COMPOUNDS.
US5623055A (en) 1994-01-28 1997-04-22 Prolinx, Inc. Phenylboronic acid complexes derived from aminosalicylic acid for bioconjugate preparation
US5594151A (en) 1994-01-28 1997-01-14 Prolinx, Inc. Phenylboronic acid complexing reagents derived from aminosalicylic acid
US5594111A (en) 1994-01-28 1997-01-14 Prolinx, Inc. Phenylboronic acid complexes for bioconjugate preparation
CA2179309A1 (en) 1994-02-09 1995-08-17 Ted J. Hanagan Diagnostic flow cell device
US6335160B1 (en) 1995-02-17 2002-01-01 Maxygen, Inc. Methods and compositions for polypeptide engineering
GB9404709D0 (en) 1994-03-11 1994-04-27 Multilyte Ltd Binding assay
US5514501A (en) 1994-06-07 1996-05-07 The United States Of America As Represented By The Secretary Of Commerce Process for UV-photopatterning of thiolate monolayers self-assembled on gold, silver and other substrates
US6287850B1 (en) 1995-06-07 2001-09-11 Affymetrix, Inc. Bioarray chip reaction apparatus and its manufacture
EP0695941B1 (en) 1994-06-08 2002-07-31 Affymetrix, Inc. Method and apparatus for packaging a chip
US5498545A (en) * 1994-07-21 1996-03-12 Vestal; Marvin L. Mass spectrometer system and method for matrix-assisted laser desorption measurements
SE9403245D0 (en) 1994-09-26 1994-09-26 Pharmacia Biosensor Ab Improvements relating to bilayer lipid membranes
US5620850A (en) 1994-09-26 1997-04-15 President And Fellows Of Harvard College Molecular recognition at surfaces derivatized with self-assembled monolayers
US6121048A (en) 1994-10-18 2000-09-19 Zaffaroni; Alejandro C. Method of conducting a plurality of reactions
US5571410A (en) 1994-10-19 1996-11-05 Hewlett Packard Company Fully integrated miniaturized planar liquid sample handling and analysis device
US5603351A (en) 1995-06-07 1997-02-18 David Sarnoff Research Center, Inc. Method and system for inhibiting cross-contamination in fluids of combinatorial chemistry device
US5585069A (en) 1994-11-10 1996-12-17 David Sarnoff Research Center, Inc. Partitioned microelectronic and fluidic device array for clinical diagnostics and chemical synthesis
US5688642A (en) 1994-12-01 1997-11-18 The United States Of America As Represented By The Secretary Of The Navy Selective attachment of nucleic acid molecules to patterned self-assembled surfaces
US5622826A (en) 1994-12-22 1997-04-22 Houston Advanced Research Center Method for immobilization of molecules on platinum solid support surfaces
US5814565A (en) 1995-02-23 1998-09-29 University Of Utah Research Foundation Integrated optic waveguide immunosensor
DE19507166C1 (en) 1995-03-01 1996-04-18 Deutsches Krebsforsch Antibodies specific for fusion polypeptide(s) contg. a histidine component
US5629213A (en) 1995-03-03 1997-05-13 Kornguth; Steven E. Analytical biosensor
JPH11503005A (en) 1995-03-08 1999-03-23 アクゾ ノーベル ナムローゼ フェンノートシャップ Surface-modified affinity separation membrane
US6140045A (en) 1995-03-10 2000-10-31 Meso Scale Technologies Multi-array, multi-specific electrochemiluminescence testing
BR9607193B1 (en) 1995-03-10 2009-01-13 multispecific multispecific electrochemical test.
KR19990008052A (en) * 1995-04-25 1999-01-25 마이클 피 노바 Remotely programmable matrix with storage device and uses thereof
US5624711A (en) 1995-04-27 1997-04-29 Affymax Technologies, N.V. Derivatization of solid supports and methods for oligomer synthesis
US5827659A (en) * 1995-05-19 1998-10-27 Perseptive Biosystems, Inc. Methods and apparatus for sequencing polymers using mass spectrometry
US5625184A (en) * 1995-05-19 1997-04-29 Perseptive Biosystems, Inc. Time-of-flight mass spectrometry analysis of biomolecules
US5700642A (en) 1995-05-22 1997-12-23 Sri International Oligonucleotide sizing using immobilized cleavable primers
US5776674A (en) 1995-06-05 1998-07-07 Seq, Ltd Chemical biochemical and biological processing in thin films
US5545531A (en) 1995-06-07 1996-08-13 Affymax Technologies N.V. Methods for making a device for concurrently processing multiple biological chip assays
US5840300A (en) 1995-09-11 1998-11-24 Trustees Of The University Of Pennsylvania Methods and compositions comprising single chain recombinant antibodies
DE19543232A1 (en) 1995-11-07 1997-05-15 Knoell Hans Forschung Ev Production of matrix-bound miniaturised combinatorial polymer and oligomer library
US5763263A (en) 1995-11-27 1998-06-09 Dehlinger; Peter J. Method and apparatus for producing position addressable combinatorial libraries
WO1997021094A1 (en) 1995-12-01 1997-06-12 Innogenetics N.V. Impedimetric detection system and method of production thereof
DE19548152A1 (en) 1995-12-22 1997-06-26 Boehringer Mannheim Gmbh Process for covering a surface with a film of an oligoethylene glycol derivative
US5837196A (en) * 1996-01-26 1998-11-17 The Regents Of The University Of California High density array fabrication and readout method for a fiber optic biosensor
US5942443A (en) 1996-06-28 1999-08-24 Caliper Technologies Corporation High throughput screening assay systems in microscale fluidic devices
US5925552A (en) 1996-04-25 1999-07-20 Medtronic, Inc. Method for attachment of biomolecules to medical devices surfaces
US5731152A (en) 1996-05-13 1998-03-24 Motorola, Inc. Methods and systems for biological reagent placement
US6103479A (en) 1996-05-30 2000-08-15 Cellomics, Inc. Miniaturized cell array methods and apparatus for cell-based screening
JPH1025299A (en) 1996-07-12 1998-01-27 Nec Corp Aligned peptide, and detection of bound/interaction site in protein using the same
US5827658A (en) 1996-08-09 1998-10-27 The United States Of America As Reprsented By The Department Of Health And Human Services Isolation of amplified genes via cDNA subtractive hybridization
US20030017149A1 (en) 1996-10-10 2003-01-23 Hoeffler James P. Single chain monoclonal antibody fusion reagents that regulate transcription in vivo
GB9624927D0 (en) 1996-11-29 1997-01-15 Oxford Glycosciences Uk Ltd Gels and their use
US5905024A (en) 1996-12-17 1999-05-18 University Of Chicago Method for performing site-specific affinity fractionation for use in DNA sequencing
AU6646398A (en) 1996-12-31 1998-07-31 Genometrix Incorporated Multiplexed molecular analysis apparatus and method
NZ516848A (en) 1997-06-20 2004-03-26 Ciphergen Biosystems Inc Retentate chromatography apparatus with applications in biology and medicine
US5981734A (en) 1997-07-17 1999-11-09 University Of Chicago Methods for immobilizing nucleic acids on a gel substrate
JP4313861B2 (en) 1997-08-01 2009-08-12 キヤノン株式会社 Manufacturing method of probe array
US6960457B1 (en) 1997-09-04 2005-11-01 Stanford University Reversible immobilization of arginine-tagged moieties on a silicate surface
US5922617A (en) 1997-11-12 1999-07-13 Functional Genetics, Inc. Rapid screening assay methods and devices
US6232066B1 (en) 1997-12-19 2001-05-15 Neogen, Inc. High throughput assay system
US6087102A (en) 1998-01-07 2000-07-11 Clontech Laboratories, Inc. Polymeric arrays and methods for their use in binding assays
CA2319828A1 (en) 1998-01-29 1999-08-05 Miller, Samuel High density arrays for proteome analysis and methods and compositions therefor
US7794946B1 (en) 1998-02-04 2010-09-14 Life Technologies Corporation Microarray and uses therefor
US6087103A (en) 1998-03-04 2000-07-11 Lifespan Biosciences, Inc. Tagged ligand arrays for identifying target-ligand interactions
US6001607A (en) 1998-04-13 1999-12-14 Incyte Pharmaceuticals, Inc. Human growth-associated methyltransfeases
US6350369B1 (en) 1998-04-14 2002-02-26 California Institute Of Technology Method and system for determining analyte activity
EP1073770B1 (en) 1998-04-30 2004-11-17 Max-Planck-Gesellschaft Zur Förderung Der Wissenschaften E.V. Novel method for the identification of clones conferring a desired biological property from an expression library
US6406921B1 (en) 1998-07-14 2002-06-18 Zyomyx, Incorporated Protein arrays for high-throughput screening
US6682942B1 (en) 1998-07-14 2004-01-27 Zyomyx, Inc. Microdevices for screening biomolecules
US6780582B1 (en) 1998-07-14 2004-08-24 Zyomyx, Inc. Arrays of protein-capture agents and methods of use thereof
US6576478B1 (en) 1998-07-14 2003-06-10 Zyomyx, Inc. Microdevices for high-throughput screening of biomolecules
US20020119579A1 (en) 1998-07-14 2002-08-29 Peter Wagner Arrays devices and methods of use thereof
US6897073B2 (en) 1998-07-14 2005-05-24 Zyomyx, Inc. Non-specific binding resistant protein arrays and methods for making the same
US6197599B1 (en) 1998-07-30 2001-03-06 Guorong Chin Method to detect proteins
US6190908B1 (en) 1998-08-12 2001-02-20 The Scripps Research Institute Modulation of polypeptide display on modified filamentous phage
FR2792285B1 (en) 1999-04-16 2001-06-08 Onera (Off Nat Aerospatiale) AERODYNAMIC SURFACE OF AIRCRAFT WITH LEAK EDGE DEFLECTOR
US7107329B1 (en) 1999-05-21 2006-09-12 Lucent Technologies Inc. In networks of interconnected router nodes for routing data traffic, a method of and system for imperceptibly upgrading router node software and the like without traffic interruption
KR100379411B1 (en) 1999-06-28 2003-04-10 엘지전자 주식회사 biochip and method for patterning and measuring biomaterial of the same
CA2392753A1 (en) 1999-12-10 2001-06-14 Invitrogen Corporation Use of multiple recombination sites with unique specificity in recombinational cloning
US6406840B1 (en) 1999-12-17 2002-06-18 Biomosaic Systems, Inc. Cell arrays and the uses thereof
EP2202001A3 (en) 2000-02-23 2011-05-18 Zyomyx, Inc. Microfluidic devices and methods
AU2001259512B2 (en) 2000-05-04 2007-03-01 Yale University High density protein arrays for screening of protein activity
US6531283B1 (en) 2000-06-20 2003-03-11 Molecular Staging, Inc. Protein expression profiling
DE10039643A1 (en) 2000-08-14 2002-02-28 Max Planck Gesellschaft Functionalized perylene tetracarboxylic acid diimides
US7094568B2 (en) 2000-08-17 2006-08-22 Sense Proteomic Ltd. Method for producing proteins tagged at the N- or C-terminus
WO2002025288A2 (en) 2000-09-22 2002-03-28 Clontech Laboratories Inc. Highly sensitive proteomic analysis methods and kits and systems for practicing the same
US6699665B1 (en) 2000-11-08 2004-03-02 Surface Logix, Inc. Multiple array system for integrating bioarrays
WO2003043487A2 (en) 2001-11-19 2003-05-30 Protometrix, Inc. Method of using a non-antibody protein to detect and measure an analyte
US20050233473A1 (en) 2002-08-16 2005-10-20 Zyomyx, Inc. Methods and reagents for surface functionalization
GB0300150D0 (en) 2003-01-04 2003-02-05 Banks J & Co Ltd Lockable releasable safety catch
US20040248323A1 (en) 2003-06-09 2004-12-09 Protometrix, Inc. Methods for conducting assays for enzyme activity on protein microarrays
US20050026215A1 (en) 2003-07-17 2005-02-03 Predki Paul F. Method for the prediction of an epitope

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4994373A (en) * 1983-01-27 1991-02-19 Enzo Biochem, Inc. Method and structures employing chemically-labelled polynucleotide probes
US4591570A (en) * 1983-02-02 1986-05-27 Centocor, Inc. Matrix of antibody-coated spots for determination of antigens
US5384263A (en) * 1987-10-13 1995-01-24 Terrapin Technologies, Inc. Method to produce immunodiagnostic reagents
US5252743A (en) * 1989-11-13 1993-10-12 Affymax Technologies N.V. Spatially-addressable immobilization of anti-ligands on surfaces
US5283283A (en) * 1991-08-02 1994-02-01 The Goodyear Tire & Rubber Company High modulus blend of poly(α-amino acid) in rubbery elastomer
US5532142A (en) * 1993-02-12 1996-07-02 Board Of Regents, The University Of Texas System Method of isolation and purification of fusion polypeptides
US5674712A (en) * 1993-03-10 1997-10-07 Eniricerche S.P.A. Recombinant vector and use thereof for exocellular preparation of antibodies in single molecule form from Bacillus subtilis
US5436134A (en) * 1993-04-13 1995-07-25 Molecular Probes, Inc. Cyclic-substituted unsymmetrical cyanine dyes
US5538897A (en) * 1994-03-14 1996-07-23 University Of Washington Use of mass spectrometry fragmentation patterns of peptides to identify amino acid sequences in databases
US5807522A (en) * 1994-06-17 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods for fabricating microarrays of biological samples
US6309820B1 (en) * 1995-04-07 2001-10-30 University Of North Carolina At Chapel Hill Polypeptides having a functional domain of interest and methods of identifying and using same
US6635311B1 (en) * 1999-01-07 2003-10-21 Northwestern University Methods utilizing scanning probe microscope tips and products therefor or products thereby
US6899137B2 (en) * 1999-06-28 2005-05-31 California Institute Of Technology Microfabricated elastomeric valve and pump systems
US7132251B1 (en) * 2000-05-04 2006-11-07 Procognia Ltd Method and composition for analyzing a carbohydrate polymer
US20050182242A1 (en) * 2001-05-11 2005-08-18 Michael Snyder Global analysis of protein activities using proteome chips

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8637264B2 (en) 1998-02-04 2014-01-28 Life Technologies Corporation Microarrays and uses therefor
US8012703B2 (en) 1998-02-04 2011-09-06 Life Technologies Corporation Microarrays and uses therefor
US7794946B1 (en) 1998-02-04 2010-09-14 Life Technologies Corporation Microarray and uses therefor
US7462469B2 (en) 2000-01-11 2008-12-09 Maxygen, Inc. Integrated system for diversity generation and screening
US20030054384A1 (en) * 2000-01-11 2003-03-20 Maxygen, Inc. Integrated systems and methods for diversity generation and screening
US20010039014A1 (en) * 2000-01-11 2001-11-08 Maxygen, Inc. Integrated systems and methods for diversity generation and screening
US8014961B2 (en) 2000-01-11 2011-09-06 Codexis Mayflower Holdings, Llc Integrated systems and methods for diversity generation and screening
US20080015116A1 (en) * 2000-01-11 2008-01-17 Maxygen, Inc. Integrated Systems and Methods for Diversity Generation and Screening
US8399383B2 (en) 2000-05-04 2013-03-19 Yale University Protein chips for high throughput screening of protein activity
US7687437B2 (en) 2001-07-13 2010-03-30 Nanosphere, Inc. Method for immobilizing molecules onto surfaces
US7524636B2 (en) * 2002-10-23 2009-04-28 University Of Hawaii Methods for diagnosing labor
US20050260683A1 (en) * 2002-10-23 2005-11-24 University Of Hawaii Methods for diagnosing and treating pre-term labor
EP1583845A4 (en) * 2003-01-02 2006-06-28 Bioforce Nanosciences Inc Method and apparatus for molecular analysis in small sample volumes
EP1583845A2 (en) * 2003-01-02 2005-10-12 Bioforce Nanosciences, Inc. Method and apparatus for molecular analysis in small sample volumes
US20060035234A1 (en) * 2003-01-02 2006-02-16 Eric Henderson Method and apparatus for molecular analysis in small sample volumes
DE10315031B4 (en) * 2003-04-02 2014-02-13 Analytik Jena Ag Method for detecting sepsis and / or sepsis-like conditions
US20080070235A1 (en) * 2003-04-02 2008-03-20 Sirs-Lab Gmbh Method for Recognizing Acute Generalized Inflammatory Conditions (Sirs), Sepsis, Sepsis-Like Conditions and Systemic Infections
US20100086909A1 (en) * 2004-03-30 2010-04-08 Sirs-Lab Gmbh Method for the prediction of individual disease course in sepsis
US8476200B2 (en) 2004-10-13 2013-07-02 Sirs-Lab Gmbh Method for differentiating between the non-infectious and infectious causes of multiple organ failure
US20090075831A1 (en) * 2004-10-13 2009-03-19 Stefan Russwurm Method for differentiating between the non-infectious and infectious causes of multiple organ failure
US20060292559A1 (en) * 2005-06-23 2006-12-28 Beckman Coulter, Inc. Cell-based microarrays, and methods for their preparation and use
US10578618B2 (en) 2010-07-16 2020-03-03 The University Of British Columbia Methods for assaying cellular binding interactions
US10718768B1 (en) 2010-07-16 2020-07-21 The University Of British Columbia Methods for assaying cellular binding interactions
US10107812B2 (en) 2010-07-16 2018-10-23 The University Of British Columbia Methods for assaying cellular binding interactions
US10274494B2 (en) 2010-07-16 2019-04-30 The University Of British Columbia Methods for assaying cellular binding interactions
US10466241B2 (en) 2010-07-16 2019-11-05 The University Of British Columbia Methods for assaying cellular binding interactions
US20120015347A1 (en) * 2010-07-16 2012-01-19 Anupam Singhal Methods for Assaying Cellular Binding Interactions
US10697962B1 (en) 2010-07-16 2020-06-30 The University Of British Columbia Methods for assaying cellular binding interactions
US9188593B2 (en) * 2010-07-16 2015-11-17 The University Of British Columbia Methods for assaying cellular binding interactions
US10746737B2 (en) 2010-07-16 2020-08-18 The University Of British Columbia Methods for assaying cellular binding interactions
US10753933B2 (en) 2010-07-16 2020-08-25 The University Of British Columbia Methods for assaying cellular binding interactions
US10775378B1 (en) 2010-07-16 2020-09-15 The University Of British Columbia Methods for assaying cellular binding interactions
US10775377B1 (en) 2010-07-16 2020-09-15 The University Of British Columbia Methods for assaying cellular binding interactions
US10775376B2 (en) 2010-07-16 2020-09-15 The University Of British Columbia Methods for assaying cellular binding interactions
US10794910B1 (en) 2010-07-16 2020-10-06 The University Of British Columbia Methods for assaying cellular binding interactions
US20230347021A1 (en) * 2021-10-25 2023-11-02 Singular Genomics Systems, Inc. Manipulating and detecting biological samples

Also Published As

Publication number Publication date
EP1060395A1 (en) 2000-12-20
CA2318175A1 (en) 1999-08-12
ATE393912T1 (en) 2008-05-15
DE69938623T2 (en) 2009-05-28
JP2002502977A (en) 2002-01-29
US8637264B2 (en) 2014-01-28
EP1060395B1 (en) 2008-04-30
AU2583899A (en) 1999-08-23
DE69938623D1 (en) 2008-06-12
US8012703B2 (en) 2011-09-06
WO1999040434A1 (en) 1999-08-12
US20120004131A1 (en) 2012-01-05
EP2189791A3 (en) 2011-03-09
EP1965213A2 (en) 2008-09-03
US20100022407A1 (en) 2010-01-28
US20130123127A1 (en) 2013-05-16
EP1060395A4 (en) 2005-02-09
EP1965213A3 (en) 2009-07-15
EP2189791A2 (en) 2010-05-26
US7794946B1 (en) 2010-09-14

Similar Documents

Publication Publication Date Title
US8637264B2 (en) Microarrays and uses therefor
US8969009B2 (en) Identification of discriminant proteins through antibody profiling, methods and apparatus for identifying an individual
US20060003381A1 (en) Methods for assembling protein microarrays
US20080139405A1 (en) Colloidal compositions for solid phase biomolecular analytical, preparative and identification systems
WO2001014425A1 (en) Multipurpose diagnostic systems using protein chips
AU2005250325A1 (en) Methods and apparatus for detection of viral infection
US9410965B2 (en) Identification of discriminant proteins through antibody profiling, methods and apparatus for identifying an individual
WO2011153464A2 (en) In situ oriented immobilization of proteins on a support
WO2007130549A1 (en) Monoclonal antibody microarray
JP2003527605A (en) Methods for making and using biological material microarrays
Sompuram et al. A water-stable protected isocyanate glass array substrate
AU2003252937A1 (en) Microarrays and Uses Therefor
AU2007200935A1 (en) Microarrays and uses therefor
EP1258731A2 (en) Reactive solid support for DNA fragment detection
US20010053520A1 (en) Methods of making and using microarrays of biological materials
US20030082560A1 (en) Method of making interactive protein arrays
US9714948B2 (en) Method for high-throughput protein detection with two antibody microarrays
RU2206574C2 (en) Polypeptide comprising antigenic site of mycobacterium tuberculosis soluble antigen, method (variants) and diagnostic set (variants) for detection of mycobacterium tuberculosis infection
WO2004094638A1 (en) Vasculitis antigen and method of diagnosing vasculitis
US20140274758A1 (en) Antibody profiling, methods and apparatus for identifying an individual or source of a biological material
US20040152130A1 (en) Method for determining secondary modifications of molecules using arrays

Legal Events

Date Code Title Description
AS Assignment

Owner name: BANK OF AMERICA, N.A., AS COLLATERAL AGENT, WASHIN

Free format text: SECURITY AGREEMENT;ASSIGNOR:LIFE TECHNOLOGIES CORPORATION;REEL/FRAME:021975/0467

Effective date: 20081121

Owner name: BANK OF AMERICA, N.A., AS COLLATERAL AGENT,WASHING

Free format text: SECURITY AGREEMENT;ASSIGNOR:LIFE TECHNOLOGIES CORPORATION;REEL/FRAME:021975/0467

Effective date: 20081121

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: LIFE TECHNOLOGIES CORPORATION, CALIFORNIA

Free format text: LIEN RELEASE;ASSIGNOR:BANK OF AMERICA, N.A.;REEL/FRAME:030182/0461

Effective date: 20100528