WO2017127556A1 - Méthodes et compositions pour identifier, quantifier, et caractériser des analytes cibles et des fragments de liaison - Google Patents

Méthodes et compositions pour identifier, quantifier, et caractériser des analytes cibles et des fragments de liaison Download PDF

Info

Publication number
WO2017127556A1
WO2017127556A1 PCT/US2017/014151 US2017014151W WO2017127556A1 WO 2017127556 A1 WO2017127556 A1 WO 2017127556A1 US 2017014151 W US2017014151 W US 2017014151W WO 2017127556 A1 WO2017127556 A1 WO 2017127556A1
Authority
WO
WIPO (PCT)
Prior art keywords
polynucleotide
proximity
address
solid support
sequence
Prior art date
Application number
PCT/US2017/014151
Other languages
English (en)
Inventor
Heng Zhu
Jiang Qian
Ignacio Pino
Original Assignee
Cdi Laboratories, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cdi Laboratories, Inc. filed Critical Cdi Laboratories, Inc.
Publication of WO2017127556A1 publication Critical patent/WO2017127556A1/fr

Links

Classifications

    • 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
    • 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
    • C12Q1/6869Methods for sequencing
    • C12Q1/6874Methods for sequencing involving nucleic acid arrays, e.g. sequencing by hybridisation
    • 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
    • G01N33/54306Solid-phase reaction mechanisms
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2458/00Labels used in chemical analysis of biological material
    • G01N2458/10Oligonucleotides as tagging agents for labelling antibodies

Definitions

  • binding moieties e.g., polypeptides, nucleic acids, and small molecules
  • binding moieties e.g., antibodies
  • Diseases can be treated by identifying a binding moiety's target analyte and blocking the binding moiety's recognition of its target analyte.
  • screening to identify binding moieties and their target analytes is an important tool and can be used to combat disease progression.
  • binding moieties such as monoclonal antibodies (mAbs), aptamers, and small molecules
  • mAbs monoclonal antibodies
  • aptamers aptamers
  • small molecules small molecules
  • a multiplex assay that allows for screening hundreds to thousands of binding moieties simultaneously can greatly reduce cost and improve throughput.
  • highly multiplexed screens allow a precise measurement of the specificity of a given analyte, because the counter screens are performed simultaneously.
  • Proximity-probe based detection assays and particularly proximity ligation assays have proved very useful in the specific and sensitive detection of proteins in a number of different applications, e.g. the detection of weakly expressed or low abundance proteins.
  • proximity ligation assays are not without their problems and room for improvement exists, with respect to both the sensitivity and specificity of the assay.
  • proximity probes comprising a binding moiety and a proximity
  • polynucleotide are contacted to a solid support comprising a plurality of target analytes and a plurality of address polynucleotides each barcoded to a target analyte.
  • a binding moiety is bound to a target analyte
  • the proximity polynucleotide is coupled to the address polynucleotide.
  • the coupled products are then amplified and sequenced.
  • the binding moiety's barcode and the address polynucleotide barcode will appear in the same sequence. By counting numbers of the reads for the same sequences, the relative strength of the binding moiety can be determined.
  • binding specificity of the binding moiety can be determined.
  • the solid support can comprise a target analyte.
  • the target analyte is attached to the solid support.
  • the solid support can further comprise an address
  • the address polynucleotide can comprise an affinity tag.
  • the affinity tag can be in association with the solid support.
  • the address polynucleotide can be in proximity to the target analyte.
  • the solid support can be magnetic.
  • the magnetic solid support can comprise magnetite, maghemitite, FePt, SrFe, iron, cobalt, nickel, chromium dioxide, ferrites, or mixtures thereof.
  • the solid support can be
  • the solid support can be coated with a polymer.
  • the polymer can be polyethylene glycol, polymethacrylate,
  • the solid support can be spherical or planar. In some embodiments, the solid support can be spherical particle. In some embodiments, the spherical particle can be a bead. In some embodiments, the solid support can comprise an affinity ligand.
  • the affinity ligand can be an antigen, an antibody, an antibody fragment, glutathione, calmodulin, biotin, streptavidin, streptactin, amylose, or a metal chelate.
  • the metal chelate can be nickel, cobalt, zinc, mercury, cupper or iron chelate.
  • the affinity ligand partially coats a surface of the solid support.
  • the affinity ligand can completely coat a surface of said solid support.
  • the target analyte can be barcoded.
  • the target analyte can be barcoded to an address barcode sequence of the address polynucleotide.
  • the target analyte barcode can be unique.
  • the target analyte can be from a biological sample.
  • target analyte can be an antibody.
  • the target analyte can be a polypeptide.
  • the target analyte can be synthesized.
  • the address polynucleotide can be in proximity to a proximity probe when a proximity probe is bound to the target analyte.
  • the proximity probe can comprise a binding moiety.
  • the proximity probe can be bound to the target analyte.
  • the binding moiety can be bound to the target analyte.
  • the proximity probe can comprise a proximity polynucleotide.
  • the binding moiety can be coupled to the proximity polynucleotide.
  • the binding moiety can be barcoded to the proximity polynucleotide to which it is coupled.
  • the proximity polynucleotide can comprise a proximity barcode.
  • the proximity barcode can identify the binding moiety to which it is coupled.
  • the proximity barcode can be unique.
  • the proximity polynucleotide can further comprise a proximity linker sequence. In some
  • the proximity polynucleotide can further comprise a proximity primer binding sequence. In some embodiments, the proximity polynucleotide can further comprise a proximity spacer sequence. In some embodiments, the proximity polynucleotide can be arranged in an order of a proximity linker sequence, proximity barcode, proximity primer binding sequence, and proximity spacer propagating toward said binding moiety. In some embodiments, the proximity polynucleotide can be arranged in an order of the proximity linker sequence, proximity barcode, proximity primer binding sequence, and proximity spacer from a 5' end of said proximity polynucleotide to a 3' end of said proximity polynucleotide. In some
  • the binding moiety can be a polynucleotide.
  • the polynucleotide can be single stranded.
  • the polynucleotide can be double stranded.
  • the polynucleotide can be RNA.
  • the polynucleotide can be DNA.
  • the proximity polynucleotide can be a 5' overhang region of the binding moiety that is a polynucleotide.
  • the sequence of the binding moiety can comprise a proximity barcode.
  • the proximity polynucleotide can comprise a proximity linker sequence.
  • the binding moiety can comprise a universal 3' region. In some embodiments, the universal 3' region can comprise a proximity primer binding sequence. In some embodiments, the solid support can comprise a primer set comprising a first primer that binds to a primer binding site upstream of said address barcode and a second primer that binds to a 3' region of said binding moiety that is a polynucleotide. In some embodiments, the binding moiety can be a polypeptide. In some embodiments, the binding moiety can be an antibody or fragment thereof. In some embodiments, the address polynucleotide can further comprise an address linker sequence. In some embodiments, the address polynucleotide can further comprise an address primer binding sequence.
  • the address polynucleotide can further comprise an address spacer sequence. In some embodiments, the address polynucleotide can arranged in an order of said address linker sequence, said address barcode, said address primer binding sequence, said address spacer, and said affinity tag propagating toward said solid support. In some
  • the address polynucleotide can be arranged in an order of address linker sequence, address barcode, address primer binding sequence, address spacer, and affinity tag from a 3' end of said address polynucleotide to a 5' end of said address polynucleotide.
  • the affinity tag can comprise an antigen tag, an antibody tag, an antibody fragment tag, a calmodulin tag, a glutathione S-transferase (GST) tag, a histidine (His) tag, a streptavidin tag, an avidin tag, a maltose-binding protein tag, a Flag tag or combinations thereof.
  • the affinity tag can be coupled to the solid support covalently.
  • the affinity tag can be coupled to the solid support non-covalently. In some embodiments, the affinity tag can be coupled to the solid support by a linker. In some embodiments, the target analyte can be coupled to the solid support covalently. In some embodiments, the target analyte can be coupled to the solid support non-covalently. In some embodiments, the target analyte can be coupled to the solid support by a linker. In some embodiments, the target analyte can be coupled to the solid support by an affinity tag. In some embodiments, the affinity tag can be a universal tag. In some embodiments, the affinity ligand can be between the solid support and the target analyte.
  • the target analyte can be coupled to the affinity ligand covalently. In some embodiments, the target analyte can be coupled to the affinity ligand non-covalently. In some embodiments, the affinity ligand can be between the affinity tag and the solid support. In some embodiments, the affinity tag can be coupled to the affinity ligand covalently. In some embodiments, the affinity tag can be coupled to the affinity ligand non-covalently. In some embodiments, the proximity linker sequence can be coupled to the address linker sequence. In some embodiments, an end of the proximity polynucleotide can be adjacent to an end of the address polynucleotide.
  • the end of the proximity polynucleotide adjacent to an end of the address polynucleotide can be a 3' end of said proximity polynucleotide. In some embodiments, the end of the address polynucleotide adjacent to an end of the proximity polynucleotide can be a 5' end of said address polynucleotide. In some embodiments, the proximity polynucleotide can be hybridized to a splint polynucleotide. In some embodiments, the solid support can further comprise an amplified product of a polynucleotide comprising a proximity barcode and an address barcode. In some embodiments, the solid support can comprise a plurality of target analytes.
  • the each target analyte of the plurality can be different.
  • the proximity probe can comprise a plurality of proximity probes.
  • the each proximity probe of the plurality can be different.
  • the solid support can comprise a primer set comprising a first primer that can bind to a primer binding site upstream of the address barcode; and a second primer that can bind to a primer binding site upstream of the proximity barcode.
  • the first primer can comprise a 5' overhang region.
  • the 5' overhang region of the first primer can comprise a first universal sequencing primer binding site.
  • the second primer can comprise a 5' overhang region.
  • the 5' overhang region of the second primer can comprise a second universal sequencing primer binding site.
  • the solid support can comprise a plurality of target analytes or address polynucleotide comprising at least 2, or at least about 5, 10, 100, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, 30,000 or more target analytes or address polynucleotide.
  • the each target analyte or polynucleotide of the plurality can be different.
  • each address polynucleotide of the plurality can comprise a unique address barcode.
  • the method can comprise coupling an address polynucleotide to the solid support.
  • the address polynucleotide can comprise an affinity tag.
  • the affinity tag is in association with the solid support.
  • a target analyte can be coupled to the support.
  • the address polynucleotide can identify the target analyte.
  • the solid support can be magnetic.
  • the magnetic solid support can comprise magnetite, maghemitite, FePt, SrFe, iron, cobalt, nickel, chromium dioxide, ferrites, or mixtures thereof.
  • the solid support can be nonmagnetic.
  • the solid support can be coated with a polymer.
  • the polymer can be polyethylene glycol, polymethacrylate,
  • the solid support can be spherical or planar. In some embodiments, the solid support can be spherical particle. In some embodiments, the spherical particle can be a bead. In some embodiments, the solid support can comprise an affinity ligand.
  • the affinity ligand can be an antigen, an antibody, an antibody fragment, glutathione, calmodulin, biotin, streptavidin, streptactin, amylose, or a metal chelate.
  • the metal chelate can be nickel, cobalt, zinc, mercury, cupper or iron chelate.
  • the affinity ligand partially coats a surface of the solid support.
  • the affinity ligand can completely coat a surface of the solid support.
  • the address polynucleotide can be in proximity to a proximity probe when a proximity probe is bound to the target analyte.
  • the target analyte can be barcoded to an address barcode sequence of the address polynucleotide.
  • the address barcode can be unique.
  • the address polynucleotide can be in proximity to a proximity probe when the proximity probe is bound to the target analyte.
  • the method can further comprise binding a proximity probe to the target analyte.
  • the proximity probe can be bound to the target analyte.
  • the binding moiety can be bound to the target analyte.
  • the proximity probe can comprise a proximity polynucleotide.
  • the binding moiety can be coupled to the proximity polynucleotide.
  • the binding moiety can be barcoded to the proximity polynucleotide to which it is coupled.
  • the proximity polynucleotide can comprise a proximity barcode.
  • the proximity barcode can identify the binding moiety to which it is coupled.
  • the proximity barcode can be unique.
  • the proximity polynucleotide can further comprise a proximity linker sequence. In some embodiments, the proximity
  • polynucleotide can further comprise a proximity primer binding sequence.
  • the proximity polynucleotide can further comprise a proximity spacer sequence.
  • the proximity polynucleotide can be arranged in an order of the proximity linker sequence, the proximity barcode, the proximity primer binding sequence, and the proximity spacer propagating toward the binding moiety.
  • the proximity polynucleotide can be arranged in an order of the proximity linker sequence, the proximity barcode, the proximity primer binding sequence, and the proximity spacer from a 5' end of the proximity polynucleotide to a 3' end of the proximity polynucleotide.
  • the binding moiety can be a polynucleotide.
  • the polynucleotide can be single stranded. In some embodiments, the polynucleotide double stranded. In some embodiments, the polynucleotide can be RNA. In some embodiments, the polynucleotide can be DNA. In some embodiments, the proximity polynucleotide can be a 5' overhang region of the binding moiety that is a polynucleotide. In some embodiments, the sequence of the binding moiety that is a polynucleotide can comprise a proximity barcode.
  • the proximity polynucleotide that is a 5' overhang region of the binding moiety that is a polynucleotide can comprise a proximity linker sequence.
  • the binding moiety that is a polynucleotide can comprise a universal 3' region.
  • the universal 3' region can comprise a proximity primer binding sequence.
  • the solid support can comprise a primer set.
  • the primer set can comprise a first primer that can bind to a primer binding site upstream of the address barcode and a second primer that can binds to a 3' region of the binding moiety that is a polynucleotide.
  • the binding moiety can be a polypeptide.
  • the polypeptide can be an antibody or fragment thereof.
  • the address polynucleotide can further comprise an address linker sequence.
  • the address polynucleotide can further comprise an address primer binding sequence. In some embodiments, the address polynucleotide can further comprise an address spacer sequence. In some embodiments, the address polynucleotide can be arranged in an order of the address linker sequence, the address barcode, the address primer binding sequence, the address spacer, and the affinity tag propagating toward the solid support. In some embodiments, the address polynucleotide can be arranged in an order of the address linker sequence, the address barcode, the address primer binding sequence, the address spacer, and the affinity tag from a 3' end of the address polynucleotide to a 5' end of the address polynucleotide.
  • the affinity tag can comprise an antigen tag, an antibody tag, an antibody fragment tag, calmodulin tag, glutathione S-transferase (GST) tag, histidine (His) tag, streptavidin tag, avidin tag, or maltose-binding protein tag, Flag tag.
  • the affinity tag can be coupled to the solid support covalently.
  • the affinity tag can be coupled to the solid support non-covalently.
  • the affinity tag can be coupled to the solid support by a linker.
  • the target analyte can be coupled to the solid support covalently.
  • the target analyte can be coupled to the solid support non-covalently.
  • the target analyte can be coupled to the solid support by a linker.
  • the affinity ligand can be between the solid support and the target analyte.
  • the target analyte can be coupled to the affinity ligand covalently.
  • the target analyte can be coupled to the affinity ligand non-covalently.
  • the affinity ligand can be between the affinity tag and the solid support.
  • the affinity tag can be coupled to the affinity ligand covalently.
  • the affinity tag can be coupled to the affinity ligand non- covalently.
  • the proximity linker sequence can be coupled to the address linker sequence.
  • the end of the proximity polynucleotide can be adjacent to an end of the address polynucleotide. In some embodiments, the end of the proximity polynucleotide adjacent to an end of the address polynucleotide can be a 3' end of the proximity polynucleotide. In some embodiments, the end of the address polynucleotide adjacent to an end of the proximity polynucleotide can be a 5' end of the address polynucleotide. In some embodiments, the proximity polynucleotide can be hybridized to a splint polynucleotide. In some embodiments, the solid support can further comprise an amplified product of a
  • the solid support can comprise a plurality of target analytes. In some embodiments, the each target analyte of the plurality can be different. In some embodiments, the solid support can comprises primer set comprising a first primer that can bind to a primer binding site upstream of the address barcode; and a second primer that can bind to a primer binding site upstream of the proximity barcode. In some embodiments, the first primer can comprise a 5' overhang region. In some embodiments, the 5' overhang region of the first primer can comprise a first universal sequencing primer binding site. In some embodiments, the second primer can comprise a 5' overhang region.
  • the 5' overhang region of the second primer can comprise a second universal sequencing primer binding site.
  • the solid support cam comprise a plurality of target analytes or address polynucleotide comprising at least 2, or at least about 5, 10, 100, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, 30,000 or more target analytes or address polynucleotide.
  • the each target analyte or polynucleotide of the plurality can be different.
  • the each address polynucleotide of the plurality can comprise a unique address barcode.
  • the methods described herein can be used to identify a biomarker of a disease or condition.
  • the methods can further comprise the step of identify a molecule capable of binding said binding moiety.
  • the molecule can be a drug or treatment.
  • the binding moiety can be contacted with the molecule.
  • the molecule can be identified by detecting a reduced amplified coupling product of the proximity probe in the discrete address region and the address polynucleotide in the discrete address region, when compared to a binding moiety not contacted with said molecule.
  • the method can comprise contacting to a solid support a proximity probe.
  • the solid support can comprises a discrete address region comprising a first discrete location comprising an address polynucleotide, and a second discrete location comprising a target analyte.
  • the address polynucleotide can comprise an affinity tag.
  • the affinity tag can be in association with the solid support.
  • the address polynucleotide can be barcoded to the target analyte.
  • the solid support can be magnetic.
  • the magnetic solid support can comprise magnetite, maghemitite, FePt, SrFe, iron, cobalt, nickel, chromium dioxide, ferrites, or mixtures thereof.
  • the solid support can be nonmagnetic.
  • the solid support can be coated with a polymer.
  • the polymer can be polyethylene glycol, polymethacrylate, polymethylmethacrylate, polyethylenimine, polyvinyl alcohol, polyvinyl acetate, polystyrene, polyglutaraldehyde, polyacrylamide, agarose, chitosan, alginate or a combination thereof.
  • the solid support can be spherical or planar.
  • the solid support can be spherical particle.
  • the spherical particle can be a bead.
  • the solid support can comprise an affinity ligand.
  • the affinity ligand can be an antigen, an antibody, an antibody fragment, glutathione, calmodulin, biotin, streptavidin, streptactin, amylose, or a metal chelate.
  • the metal chelate can be nickel, cobalt, zinc, mercury, cupper or iron chelate.
  • the affinity ligand partially coats a surface of the solid support.
  • the affinity ligand can completely coat a surface of the solid support.
  • the address polynucleotide can be in proximity to a proximity probe when a proximity probe is bound to the target analyte.
  • the target analyte can be barcoded to an address barcode sequence of the address polynucleotide.
  • the address barcode can be unique.
  • the address polynucleotide can be in proximity to a proximity probe when the proximity probe is bound to the target analyte.
  • the method can further comprise binding a proximity probe to the target analyte.
  • the proximity probe can be bound to the target analyte.
  • the binding moiety can be bound to the target analyte.
  • the proximity probe can comprise a proximity polynucleotide.
  • the proximity probe can comprise a proximity polynucleotide coupled to a binding moiety.
  • the method can further comprise coupling the proximity probe in the discrete address region to the address polynucleotide in the discrete address region.
  • the method can further comprise amplifying a coupled product.
  • the method can further comprise detecting the coupled product or an amplified product thereof.
  • the solid support can comprise a plurality of discrete address regions.
  • the first and second discrete locations can be in proximity.
  • the binding moiety can be coupled to the proximity polynucleotide.
  • the binding moiety can be barcoded to the proximity polynucleotide to which it is coupled.
  • the proximity polynucleotide can comprise a proximity barcode.
  • the proximity barcode can identify the binding moiety to which it is coupled.
  • the proximity barcode can be unique.
  • the proximity polynucleotide can further comprise a proximity linker sequence.
  • the proximity polynucleotide can further comprise a proximity primer binding sequence.
  • the proximity polynucleotide can further comprise a proximity spacer sequence. In some embodiments, the proximity
  • the polynucleotide can be arranged in an order of the proximity linker sequence, the proximity barcode, the proximity primer binding sequence, and the proximity spacer propagating toward the binding moiety.
  • the proximity polynucleotide can be arranged in an order of the proximity linker sequence, the proximity barcode, the proximity primer binding sequence, and the proximity spacer from a 5' end of the proximity polynucleotide to a 3' end of the proximity polynucleotide.
  • the binding moiety can be a
  • the polynucleotide can be single stranded. In some embodiments, the polynucleotide double stranded. In some embodiments, the polynucleotide can be RNA. In some embodiments, the polynucleotide can be DNA. In some embodiments, the proximity polynucleotide can be a 5' overhang region of the binding moiety that is a
  • sequence of the binding moiety that is a
  • polynucleotide can comprise a proximity barcode.
  • the proximity polynucleotide that is a 5' overhang region of the binding moiety that is a polynucleotide can comprise a proximity linker sequence.
  • the binding moiety that is a polynucleotide can comprise a universal 3' region.
  • the universal 3' region can comprise a proximity primer binding sequence.
  • the solid support can comprise a primer set.
  • the primer set can comprise a first primer that can bind to a primer binding site upstream of the address barcode and a second primer that can binds to a 3' region of the binding moiety that is a polynucleotide.
  • the binding moiety can be a polypeptide.
  • the polypeptide can be an antibody or fragment thereof.
  • the address polynucleotide can further comprise an address linker sequence.
  • the address polynucleotide can further comprise an address primer binding sequence.
  • the address polynucleotide can further comprise an address spacer sequence.
  • the address polynucleotide can be arranged in an order of the address linker sequence, the address barcode, the address primer binding sequence, the address spacer, and the affinity tag propagating toward the solid support.
  • the address polynucleotide can be arranged in an order of the address linker sequence, the address barcode, the address primer binding sequence, the address spacer, and the affinity tag from a 3 ' end of the address polynucleotide to a 5' end of the address polynucleotide.
  • the affinity tag can comprise an antigen tag, an antibody tag, an antibody fragment tag, calmodulin tag, glutathione S -transferase (GST) tag, histidine (His) tag, streptavidin tag, avidin tag, or maltose- binding protein tag, Flag tag.
  • the affinity tag can be coupled to the solid support covalently.
  • the affinity tag can be coupled to the solid support non-covalently. In some embodiments, the affinity tag can be coupled to the solid support by a linker. In some embodiments, the target analyte can be coupled to the solid support covalently. In some embodiments, the target analyte can be coupled to the solid support non-covalently. In some embodiments, the target analyte can be coupled to the solid support by a linker. In some embodiments, the affinity ligand can be between the solid support and the target analyte. In some embodiments, the target analyte can be coupled to the affinity ligand covalently. In some embodiments, the target analyte can be coupled to the affinity ligand non-covalently.
  • the affinity ligand can be between the affinity tag and the solid support.
  • the affinity tag can be coupled to the affinity ligand covalently.
  • the affinity tag can be coupled to the affinity ligand non-covalently.
  • the proximity linker sequence can be coupled to the address linker sequence.
  • the end of the proximity polynucleotide can be adjacent to an end of the address polynucleotide.
  • the end of the proximity polynucleotide adjacent to an end of the address polynucleotide can be a 3' end of the proximity polynucleotide.
  • the end of the address polynucleotide adjacent to an end of the proximity polynucleotide can be a 5' end of the address polynucleotide.
  • the proximity polynucleotide can be hybridized to a splint polynucleotide.
  • the solid support can further comprise an amplified product of a polynucleotide comprising a proximity barcode and an address barcode.
  • the solid support can comprise a plurality of target analytes. In some embodiments, the each target analyte of the plurality can be different.
  • the solid support can comprises primer set comprising a first primer that can bind to a primer binding site upstream of the address barcode; and a second primer that can bind to a primer binding site upstream of the proximity barcode.
  • the first primer can comprise a 5' overhang region.
  • the 5' overhang region of the first primer can comprise a first universal sequencing primer binding site.
  • the second primer can comprise a 5' overhang region.
  • the 5' overhang region of the second primer can comprise a second universal sequencing primer binding site.
  • the solid support cam comprise a plurality of target analytes or address polynucleotide comprising at least 2, or at least about 5, 10, 100, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, 30,000 or more target analytes or address polynucleotide.
  • the each target analyte or polynucleotide of the plurality can be different.
  • the each address polynucleotide of the plurality can comprise a unique address barcode.
  • the methods described herein can be used to identify a biomarker of a disease or condition.
  • the methods can further comprise the step of identify a molecule capable of binding said binding moiety.
  • the molecule can be a drug or treatment.
  • the binding moiety can be contacted with the molecule.
  • the molecule can be identified by detecting a reduced amplified coupling product of the proximity probe in the discrete address region and the address polynucleotide in the discrete address region, when compared to a binding moiety not contacted with said molecule.
  • the method can comprise forming a complex between a target analyte and a proximity probe.
  • the target analyte can be coupled to a solid support.
  • the solid support can comprise a discrete address region.
  • the discrete address region can comprise a first discrete location comprising an address polynucleotide, and a second discrete location comprising the target analyte.
  • the address polynucleotide comprises an affinity tag.
  • the affinity tag can be in association with the solid support.
  • the target analyte does not base pair with the address polynucleotide.
  • the solid support can be magnetic.
  • the magnetic solid support can comprise magnetite, maghemitite, FePt, SrFe, iron, cobalt, nickel, chromium dioxide, ferrites, or mixtures thereof.
  • the solid support can be
  • the solid support can be coated with a polymer.
  • the polymer can be polyethylene glycol, polymethacrylate,
  • the solid support can be spherical or planar. In some embodiments, the solid support can be spherical particle. In some embodiments, the spherical particle can be a bead. In some embodiments, the solid support can comprise an affinity ligand.
  • the affinity ligand can be an antigen, an antibody, an antibody fragment, glutathione, calmodulin, biotin, streptavidin, streptactin, amylose, or a metal chelate.
  • the metal chelate can be nickel, cobalt, zinc, mercury, cupper or iron chelate.
  • the affinity ligand partially coats a surface of the solid support.
  • the affinity ligand can completely coat a surface of the solid support.
  • the address polynucleotide can be in proximity to a proximity probe when a proximity probe is bound to the target analyte.
  • the target analyte can be barcoded to an address barcode sequence of the address polynucleotide.
  • the address barcode can be unique.
  • the address polynucleotide can be in proximity to a proximity probe when the proximity probe is bound to the target analyte.
  • the method can further comprise binding a proximity probe to the target analyte.
  • the proximity probe can be bound to the target analyte.
  • the binding moiety can be bound to the target analyte.
  • the proximity probe can comprise a proximity polynucleotide.
  • the proximity probe can comprise a proximity polynucleotide coupled to a binding moiety.
  • the method can further comprise coupling the proximity probe in the discrete address region to the address polynucleotide in the discrete address region.
  • the method can further comprise amplifying a coupled product.
  • the method can further comprise detecting the coupled product or an amplified product thereof.
  • the solid support can comprise a plurality of discrete address regions.
  • the first and second discrete locations can be in proximity.
  • the binding moiety can be coupled to the proximity polynucleotide.
  • the binding moiety can be barcoded to the proximity polynucleotide to which it is coupled.
  • the proximity polynucleotide can comprise a proximity barcode.
  • the proximity barcode can identify the binding moiety to which it is coupled.
  • the proximity barcode can be unique.
  • the proximity polynucleotide can further comprise a proximity linker sequence.
  • the proximity polynucleotide can further comprise a proximity primer binding sequence.
  • the proximity polynucleotide can further comprise a proximity spacer sequence.
  • the proximity polynucleotide can be arranged in an order of the proximity linker sequence, the proximity barcode, the proximity primer binding sequence, and the proximity spacer propagating toward the binding moiety. In some embodiments, the proximity polynucleotide can be arranged in an order of the proximity linker sequence, the proximity barcode, the proximity primer binding sequence, and the proximity spacer from a 5' end of the proximity polynucleotide to a 3' end of the proximity polynucleotide.
  • the binding moiety can be a polynucleotide. In some embodiments, the polynucleotide can be single stranded. In some embodiments, the polynucleotide double stranded.
  • the polynucleotide can be RNA. In some embodiments, the polynucleotide can be DNA. In some embodiments, the proximity polynucleotide can be a 5' overhang region of the binding moiety that is a polynucleotide. In some embodiments, the sequence of the binding moiety that is a polynucleotide can comprise a proximity barcode. In some embodiments, the proximity polynucleotide that is a 5' overhang region of the binding moiety that is a polynucleotide can comprise a proximity linker sequence. In some embodiments,
  • the binding moiety that is a polynucleotide can comprise a universal 3' region.
  • the universal 3' region can comprise a proximity primer binding sequence.
  • the solid support can comprise a primer set.
  • the primer set can comprise a first primer that can bind to a primer binding site upstream of the address barcode and a second primer that can binds to a 3' region of the binding moiety that is a polynucleotide.
  • the binding moiety can be a polypeptide.
  • the polypeptide can be an antibody or fragment thereof.
  • the address polynucleotide can further comprise an address linker sequence.
  • the address polynucleotide can further comprise an address primer binding sequence. In some embodiments, the address polynucleotide can further comprise an address spacer sequence. In some embodiments, the address polynucleotide can be arranged in an order of the address linker sequence, the address barcode, the address primer binding sequence, the address spacer, and the affinity tag propagating toward the solid support. In some embodiments, the address polynucleotide can be arranged in an order of the address linker sequence, the address barcode, the address primer binding sequence, the address spacer, and the affinity tag from a 3' end of the address polynucleotide to a 5' end of the address polynucleotide.
  • the affinity tag can comprise an antigen tag, an antibody tag, an antibody fragment tag, calmodulin tag, glutathione S-transferase (GST) tag, histidine (His) tag, streptavidin tag, avidin tag, or maltose-binding protein tag, Flag tag.
  • the affinity tag can be coupled to the solid support covalently.
  • the affinity tag can be coupled to the solid support non-covalently.
  • the affinity tag can be coupled to the solid support by a linker.
  • the target analyte can be coupled to the solid support covalently.
  • the target analyte can be coupled to the solid support non-covalently.
  • the target analyte can be coupled to the solid support by a linker.
  • the affinity ligand can be between the solid support and the target analyte.
  • the target analyte can be coupled to the affinity ligand covalently.
  • the target analyte can be coupled to the affinity ligand non-covalently.
  • the affinity ligand can be between the affinity tag and the solid support.
  • the affinity tag can be coupled to the affinity ligand covalently.
  • the affinity tag can be coupled to the affinity ligand non- covalently.
  • the proximity linker sequence can be coupled to the address linker sequence.
  • the end of the proximity polynucleotide can be adjacent to an end of the address polynucleotide. In some embodiments, the end of the proximity polynucleotide adjacent to an end of the address polynucleotide can be a 3' end of the proximity polynucleotide. In some embodiments, the end of the address polynucleotide adjacent to an end of the proximity polynucleotide can be a 5' end of the address polynucleotide. In some embodiments, the proximity polynucleotide can be hybridized to a splint polynucleotide. In some embodiments, the solid support can further comprise an amplified product of a
  • the solid support can comprise a plurality of target analytes. In some embodiments, the each target analyte of the plurality can be different. In some embodiments, the solid support can comprises primer set comprising a first primer that can bind to a primer binding site upstream of the address barcode; and a second primer that can bind to a primer binding site upstream of the proximity barcode. In some embodiments, the first primer can comprise a 5' overhang region. In some embodiments, the 5' overhang region of the first primer can comprise a first universal sequencing primer binding site. In some embodiments, the second primer can comprise a 5' overhang region.
  • the 5' overhang region of the second primer can comprise a second universal sequencing primer binding site.
  • the solid support cam comprise a plurality of target analytes or address polynucleotide comprising at least 2, or at least about 5, 10, 100, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, 30,000 or more target analytes or address polynucleotide.
  • the each target analyte or polynucleotide of the plurality can be different.
  • the each address polynucleotide of the plurality can comprise a unique address barcode.
  • the methods described herein can be used to identify a biomarker of a disease or condition.
  • the methods can further comprise the step of identify a molecule capable of binding said binding moiety.
  • the molecule can be a drug or treatment.
  • the binding moiety can be contacted with the molecule.
  • the molecule can be identified by detecting a reduced amplified coupling product of the proximity probe in the discrete address region and the address polynucleotide in the discrete address region, when compared to a binding moiety not contacted with said molecule.
  • FIG. 1A depicts a schematic of the various compositions used in the methods described herein.
  • An array comprising a solid support is depicted, wherein an address polynucleotide comprising an address barcode is in proximity to a target analyte which is bound / complexed to a binding moiety comprising a proximity polynucleotide comprising a proximity barcode, wherein the target analyte is coupled to the solid support and the address polynucleotide is coupled to the solid support.
  • FIG. IB depicts a schematic of the various compositions used in the methods described herein to determine protein-target analyte interactions, such as protein-protein interactions (pDAPPL).
  • pDAPPL protein-protein interactions
  • FIG. 1C depicts a schematic of the various compositions used in the methods described herein to determine antibody-target analyte interactions, such as antibody-protein interactions (aDAPPL).
  • aDAPPL antibody-protein interactions
  • FIG. ID depicts a schematic of the various compositions used in the methods described herein to determine DNA-target analyte interactions, such as DNA-transcription factor (TF) interactions, using a proximity probe containing a proximity barcode.
  • TF DNA-transcription factor
  • FIG. IE depicts a schematic of the various compositions used in the methods described herein to determine DNA-target analyte interactions, such as DNA-TF interactions, where the proximity barcode is a sequence of the DNA.
  • FIG. IF depicts a schematic of the various compositions used in the methods described herein to determine RNA- target analyte interactions (rDAPPL), such as RNA-RNA-binding protein (RBP) interactions, using a proximity probe containing a proximity barcode.
  • rDAPPL RNA-target analyte interactions
  • RBP RNA-RNA-binding protein
  • FIG. 1G depicts a schematic of the various compositions used in the methods described herein to determine RNA- target analyte interactions (rDAPPL) such as RNA-RBP interactions, where the proximity barcode is a sequence of the RNA.
  • rDAPPL RNA-target analyte interactions
  • FIG. 1H depicts a schematic of the various compositions used in the methods described herein to determine aptamer-target analyte interactions using a proximity probe containing a proximity barcode.
  • FIG. II depicts a schematic of the various compositions used in the methods described herein to determine aptamer-target analyte interactions where the proximity barcode is a sequence of the aptamer.
  • FIG. 1 J depicts a schematic of the various compositions used in the methods described herein to determine small molecule-target analyte interactions.
  • FIG. IK depicts a schematic of the various compositions used in the methods described herein. An array comprising a solid support is depicted, wherein an address polynucleotide comprising an address barcode is in proximity to a target analyte which is bound / complexed to a binding moiety comprising a proximity polynucleotide comprising a proximity barcode, wherein the target analyte is coupled to the solid support via a linker and the address
  • polynucleotide is coupled to the solid support.
  • FIG. 1L depicts a schematic of the various compositions used in the methods described herein.
  • An array comprising a solid support is depicted, wherein an address polynucleotide comprising an address barcode is in proximity to a target analyte which is bound / complexed to a binding moiety comprising a proximity polynucleotide comprising a proximity barcode, wherein the target analyte is coupled to the solid support via an antibody linker and the address polynucleotide is attached to the solid support.
  • FIG. 1M depicts a schematic of the various compositions used in the methods described herein.
  • An array comprising a solid support is depicted, wherein an address polynucleotide comprising an address barcode is in proximity to a target macrocycle/small molecule which is bound/complexed to a binding moiety comprising a proximity polynucleotide comprising a proximity barcode, wherein the target macrocycle/small molecule is coupled to the solid support and the address polynucleotide is attached to the solid support.
  • FIG. 2A depicts a schematic of the oligonucleotides of the compositions used in the methods described herein where the proximity probe contains a proximity barcode.
  • FIG. 2B depicts a schematic of the oligonucleotides of the compositions used in the methods described herein where the binding moiety is a polynucleotide, such as a DNA, RNA, or aptamer that contains a proximity barcode.
  • the binding moiety is a polynucleotide, such as a DNA, RNA, or aptamer that contains a proximity barcode.
  • FIG. 3A depicts a schematic of a protein array, a magnification of a portion of the array, and a magnification of one well of the portion of the array.
  • FIG. 3B depicts a schematic of a virion array comprising the depicted exemplary membrane proteins within virion particles.
  • FIG. 4A depicts a schematic of the ligation of the methods described herein.
  • FIG. 4B depicts a schematic of the amplification and sequencing steps of the methods described herein.
  • FIG. 5A depicts a solid support with an address polynucleotide comprising an address barcode in proximity to a target analyte which is bound / complexed to a binding moiety comprising a proximity polynucleotide comprising a proximity barcode, wherein the target analyte is attached to the solid support and the address polynucleotide is attached to the solid support.
  • FIG. 5A depicts a solid support with an address polynucleotide comprising an address barcode in proximity to a target analyte which is bound / complexed to a binding moiety comprising a proximity polynucleotide comprising a proximity barcode, wherein the target analyte is attached to the solid support and the address polynucleotide is attached to the solid support.
  • 5B depicts a solid support, with an address polynucleotide comprising an address barcode in proximity to a target analyte which is bound / complexed to a binding moiety comprising a proximity polynucleotide which comprises a proximity barcode, wherein the target analyte is attached to the solid support and the address polynucleotide is attached to the solid support, wherein the proximity polynucleotide and the address polynucleotide are ligated.
  • FIG. 6 depicts a schematic of the step of library preparation for sequencing of the ligated products produced by the methods described herein.
  • FIG. 7A depicts a schematic of the methods described herein to determine aptamer- protein interactions.
  • FIG. 7B depicts a schematic of the methods described herein to determine DNA-protein interactions.
  • FIG. 7C depicts a schematic of the methods described herein to determine DNA-protein interactions.
  • FIG. 8A depicts a schematic of the methods described herein to apply previously identified aptamers that specifically bind to a target protein to enable proteome-wide detection of protein abundance inside a cell or tissue (WB-omix). Briefly, after a pool of the aptamers is mixed with biotinylated lysates, streptavidin beads are added to the mixture and washed. In the example shown, each of the aptamers in the pool is known to specifically interact with a particular protein Bound aptamers are then PCR amplified. The number of sequence reads for a particular aptamer sequence can be used to determine the approximate level or relative abundance of a particular protein.
  • FIG. 8B depicts a schematic of a bead coupled via an avidin-biotin linker to a complex of a target analyte bound to an aptamer containing a pair of adapter sequences for PCR amplification.
  • FIG. 8C depicts a schematic of a complex of a target analyte bound to an aptamer containing a pair of adapter sequences and coupled to a bead via hybridization with the adapter sequences.
  • FIG. 8D depicts a schematic of a bead coupled to a plurality of complexes each containing a target analyte bound to an aptamer containing a unique sequence corresponding to the target analyte to which the aptamer is bound.
  • FIG. 9A depicts a schematic of the methods described herein to apply previously identified aptamers that specifically bind to a target protein to determine protein-protein interactions by testing multiple combinations using lysates from cells (e.g., a cell line) and/or tissue (PPI-omix).
  • lysates from cells (e.g., a cell line) and/or tissue (PPI-omix).
  • PPI-omix tissue
  • a mixture of DNA/RNA aptamers that each recognizes a unique human protein is utilized to examine all possible combinations of protein-protein interactions inside a cell or tissue.
  • FIG. 9B depicts a schematic of a bead coupled to one target analyte of a protein complex, comprised of either two different proteins (i.e., a heterodimer) or the same proteins (i.e., a homodimer), wherein each target analyte is bound to an aptamer and the two aptamers are ligated via a splint polynucleotide hybridized to the adapter sequences of each aptamer.
  • a heterodimer i.e., a heterodimer
  • FIG. 9C depicts a schematic of a bead coupled via hybridization to a protein complex, wherein each target analyte is bound to an aptamer and the two aptamers are ligated via a splint polynucleotide hybridized to the adapter sequences of each aptamer.
  • each target analyte is bound to an aptamer and the two aptamers are ligated via a splint polynucleotide hybridized to the adapter sequences of each aptamer.
  • FIG. 9D depicts a schematic of a bead coupled to a plurality of protein complexes, wherein each target analyte of a complex is bound to an aptamer and the two aptamers of each complex are ligated via a splint polynucleotide hybridized to the adapter sequences of each aptamer.
  • a heterodimer When two different aptamers are found in the same sequence read based on the sequencing results, it indicates formation of a heterodimer. When the same aptamers are found on the same read sequence based on the sequencing results, it indicates formation of a homodimer.
  • FIG. 10 depicts a schematic of the methods described herein to perform screens for disease-specific aptamers.
  • FIG. 11 depicts a schematic exemplary methods and uses of dimeric and multimeric aptamer scaffolds.
  • FIG. 12A depicts a schematic of the methods described herein to perform aptamer screens against human transmembrane proteins on virion arrays (VirD arrays). Briefly, a library of DNA or RNA aptamers containing ends with fixed sequences are incubated on an array of virus particles (i.e., virions) containing transmembrane proteins of interest and bound aptamers are then recovered and amplified. Asymmetric amplification of the amplified products is then performed to regenerate the aptamers. In the case of using an RNA aptamer library, in vitro transcription is then performed to regenerate the RNA aptamers. This process is repeated for 4 cycles.
  • virus particles i.e., virions
  • bound aptamers are ligated to address polynucleotides, amplified, and sequenced to identify transmembrane proteins recognized by the aptamers. This process is repeated for a 6 th and 7 th cycle. Sequencing data from cycles 5, 6, and 7 can be compared to identify high affinity aptamers specific to transmembrane proteins.
  • FIG. 12B depicts a schematic of the methods described herein to perform peptide ligand screens for human G-protein coupled receptors (GPCRs) on VirD arrays.
  • GPCRs G-protein coupled receptors
  • FIG. 13 depicts a graph of the number of transcription factor binding motifs as a function of the number of binding proteins.
  • FIG. 14 depicts a schematic for DNA probe preparation.
  • FIG. 15A depicts a schematic of the methods described herein using a transcription factor array and a library of DNA probes with potential transcription factor binding motifs.
  • FIG. 15B depicts unmethylated and methylated DNA probes used in the methods described herein for determining binding of proteins to methylated and unmethylated DNA binding sites.
  • FIG. 16 depicts an agarose gel of three polynucleotide libraries used for determining binding of proteins to methylated and unmethylated DNA binding sites.
  • FIG. 17 depicts a schematic of the methods described herein using a transcription factor array and a library of methylated DNA probes with potential protein binding motifs.
  • FIG. 18 depicts a heat map generated from the methods in FIG. 17 used to determine the transcription factors that bind to the probes.
  • FIG. 19A depicts a schematic of the methods described herein using a transcription factor array and a library of methylated DNA probes with potential protein binding motifs.
  • FIG. 19B depicts a schematic of the methods described herein using a transcription factor array and a library of methylated DNA probes with potential protein binding motifs.
  • hTF human transcription factor.
  • FIG. 20 depicts agarose gels demonstrating detection of ligated DNA after performing protein-DNA binding assays on arrays to detect interactions between a methylated motif and a human TF, such as those depicted in FIG. 19A and FIG. 19B, and amplification by PCR with the indicated number of PCR cycles. Controls are depicted on the left. The right panel shows that MPG2 captures a methylated motif M62.
  • FIG. 21 depicts Sanger sequencing results confirming the expected sequences of the ligated products after performing protein-DNA binding assays on arrays to detect interactions between a methylated motif and a human TF, such as those depicted in FIG. 20.
  • FIG. 22 depicts agarose gels demonstrating detection of ligated DNA after performing protein-DNA binding assays on arrays with two different probe sets and the indicated methylated polynucleotide probe libraries.
  • FIG. 23 depicts an agarose gel of input DNA probes loaded on the gel at the indicated concentrations. Ligated products were analyzed by gel electrophoresis. Competitor (double stranded DNA) was analyzed separately.
  • FIG. 24 depicts agarose gels of PCR amplified ligated products at the indicated number of PCR cycles.
  • FIG. 25A depicts a schematic of the methods described herein to determine RNA- protein interactions (rDAPPL).
  • RBP RNA binding protein.
  • FIG. 25B depicts an agarose gel of PCR ligation products in an RNA DAPPL
  • FIG. 25C depicts Sanger sequencing confirming the expected sequences of the ligated products after performing an RNA DAPPL (rDAPPL) assay to detect interactions between MSI1 and Qkl with their known RNA sequences, such as those depicted in FIG. 25A and FIG 25B.
  • rDAPPL RNA DAPPL
  • FIG. 26A depicts a schematic of the methods described herein to determine antibody- protein interactions (aDAPPL).
  • Ag Antigen (target analyte),
  • Ab Antibody.
  • FIG. 26B depicts a schematic of the preparation and conjugation of an antibody with a polynucleotide for use in the methods described herein to determine antibody-protein interactions (aDAPPL).
  • FIG. 26C depicts a polyacrylamide gel stained with Coommaisse (top)
  • FIG. 26D depicts a schematic of a design for an exemplary aDAPPL assay and corresponding chip (left) and an agarose gel of PCR ligation products in an aDAPPL assay
  • FIG. 26E depicts labeling of antibodies with maleimide proximity oligos (POs) and labeling of address oligos with biotin and streptavidin.
  • FIG. 26F depicts an exemplary process for generating cohesive sticky ends of address oligos and proximity oligos for aDAPPL.
  • FIG. 26G depicts an exemplary process and design for probing macrocycle and protein binding partners of FKBPl A (GST tagged).
  • FIG. 26H depicts a stained agarose gel of primary PCR products.
  • FIG. 261 depicts a stained agarose gel of the primary PCR products after a cleaning step (left) and a stained agarose gel of secondary PCR products using AO Fl and PO R2-2 primers as shown in FIG. 26G.
  • FIG. 27A depicts a schematic of the methods described herein to determine protein- protein interactions (pDAPPL).
  • FIG. 27B depicts a schematic of the preparation and conjugation of a protein with a polynucleotide for use in the methods described herein to determine protein-protein interactions (pDAPPL).
  • proteins are expressed with a glutathione S-transferase (GST) tag. Conjugation of the proteins to a polynucleotide is predominantly through a thiol group on the GST tag.
  • GST glutathione S-transferase
  • FIG. 27C depicts a polyacrylamide gel stained with Coommaisse (top)
  • FIG. 28 depicts a schematic of the methods described herein to perform a multiplexed chromatin-immunoprecipitation coupled deep-sequencing (ChlP-seq) method to enable simultaneous detection of transcription factor (TF) binding sites in chromatin.
  • ChlP-seq multiplexed chromatin-immunoprecipitation coupled deep-sequencing
  • a splint polynucleotide is then added to facilitate ligation between captured genomic DNA fragments to the address polynucleotides.
  • the ligated products are then amplified and sequenced. Bioinformatics analyses are performed to deconvolute the sequencing data and identify TF binding sites.
  • Bioinformatics analyses are performed to deconvolute the sequencing data and identify TF binding sites.
  • a given TF as represented by a particular address polynucleotide is connected to the genomic sequences that it binds to, and hence, resulted in mapping of its chromatin binding sites.
  • FIG. 29 depicts a schematic of the methods described herein for quantification of proteins of interest and/or posttranslational modifications of proteins of interest from single cells. Direct detection of phosphoproteins is depicted on the left. Briefly, a group of single cells (e.g., -100-1,000) or a group of lysates each extracted from a single cell thereof is placed into separate spots on an array or into a well of a 384- or 1536-well titer dish with a unique address polynucleotide (AO).
  • AO polynucleotide
  • a library of proximity probes comprising a plurality of binding moieties (e.g., Abs) specific to proteins of interest or posttranslational modifications of proteins of interest is then added to each well, the proximity polynucleotides are then ligated to address polynucleotides on the array or in each well, and the ligated products are amplified and sequenced. Identity of each single cell is represented by the address polynucleotides and identity of the proteins of interest or posttranslational modifications of proteins of interest is represented by the proximity polynucleotide sequences of the proximity probes. Detection of the phospho- tyrosine phosphorylome in a sandwich format is depicted on the right.
  • binding moieties e.g., Abs
  • an antibody that detects a desired posttranslational modification e.g., phospho-tyrosine
  • a desired posttranslational modification e.g., phospho-tyrosine
  • a group of lysate extracted from single cells is then place into separate spots on the array or a particular well in the titer dish.
  • a library of proximity probes comprising a plurality of binding moieties (e.g., Abs) specific to proteins of interest is then added to the array or to each well; the proximity polynucleotides are then ligated to address polynucleotides, and the ligated products are amplified and sequenced.
  • Identity of each single cell is represented by the address polynucleotides and identity of the tyrosine-phosphorylated proteins is represented by the proximity polynucleotide sequences of the proximity probes.
  • FIG. 30 depicts a schematic of the methods described herein for detection and quantification of specific posttranslational modifications (PTMs) on histone proteins from single cells.
  • PTMs posttranslational modifications
  • a generic anti-histone antibody is coated on bottom of each well in a titer dish.
  • a group of lysates extracted from single cells is then placed into separate wells in the titer dish.
  • a library of proximity probes comprising a plurality of binding moieties (e.g., Abs) specific to a desired histone PTM is then added to each well, the proximity polynucleotides are then ligated to address polynucleotides, and the ligated products are amplified and sequenced.
  • binding moieties e.g., Abs
  • FIG. 31A depicts a schematic of the methods described herein to screen and identify DNA or RNA aptamers that specifically bind transcription factors and protein kinases with mono-specificity and high affinity. Briefly, a library of DNA or RNA aptamers is incubated on an array of containing transcription factors and/or protein kinases of interest. Bound aptamers are then recovered and amplified. Asymmetric amplification of the amplified products is then performed to regenerate the DNA aptamers. In the case of using an RNA aptamer library, in vitro transcription is then performed to regenerate the RNA aptamers. This process is repeated for 4 cycles.
  • bound aptamers are ligated to address polynucleotides, amplified, and sequenced to identify transcription factors and/or protein kinases recognized by the aptamers. This process is repeated for a 6 th and 7 th cycle. Sequencing data from cycles 5, 6, and 7 can be compared to identify high affinity aptamers specific to transcription factors and/or protein kinases.
  • FIG. 31B depicts graphs comparing aptamer-protein interactions from the indicated cycles using the method depicted in FIG. 31A.
  • FIG. 31C depicts an alignment of aptamer sequences identified using the method depicted in FIG. 31 A and aptamer consensus sequences for binding to IKZF1. Identification of significant consensus aptamer sequences are is a good indicator of aptamer sequence
  • FIG. 31D depicts exemplary structural consensus sequences from aptamers identified using the method depicted in FIG. 31 A.
  • FIG. 31E depicts agarose gels of DNA template (left), transcribed RNA probes (middle), and PCR amplified ligated rDAPPL products from the 3 cycle using the method depicted in FIG. 31 A.
  • FIG. 32A depicts a schematic of the methods described herein to screen and identify synthetic heavy chain variable region (V H ) and light chain variable region (V L ) single domains that can specifically recognize human proteins with high affinity (e.g., recDAPPL).
  • V H heavy chain variable region
  • V L light chain variable region
  • a synthetic pool of single chain V H or V L CDNA sequences are in vitro transcribed and tethered with a puromycin-labeled DNA polynucleotide to the 3 '-ends of the transcribed RNA species.
  • In vitro translation reaction is then performed and the RNA templates are tethered to the translated protein products ⁇ e.g., a library of V H or V L single domains).
  • corresponding cDNAs are reverse-transcribed with a primer complementary to the puromycin-labeled DNA moiety, followed by an RNase treatment to remove the RNA moieties. If necessary, translated single domains can be purified with the HisX6 tag (6-His) using a nickel column to ensure full-length products. After being incubated on an array of target proteins, tethered cDNAs are amplified and this process is repeated for 6 cycles. During the 7 th cycle, tethered cDNAs are ligated to address polynucleotides, amplified, and sequenced to identify proteins recognized single chain V H or V L domains. If necessary, tethered cDNAs can be ligated to the address polynucleotides in cycles 4, 5, or 6.
  • FIG. 32B depicts an exemplary design schematic for the methods depicted in FIG. 32A to screen affinity reagents, e.g., heavy chain variable region (V H ) and light chain variable region (V L ) single domains, against the human proteome.
  • affinity reagents e.g., heavy chain variable region (V H ) and light chain variable region (V L ) single domains, against the human proteome.
  • FIG. 32C depicts an exemplary schematic of the methods described herein to screen and identify synthetic V H single domains that can specifically recognize human RAS with high affinity.
  • FIG. 32D depicts agarose gels stained for DNA and visualization on the same agarose gels demonstrating in vitro transcription of RNA and DNA-puromycin linker ligation.
  • FIG. 32E depicts a polyacrylamide gel stained with Coommaisse (right) showing in vitro translation of anti-RAS V H and a Western blot with an anti-FLAG antibody (left).
  • FIG. 33 depicts a schematic of the methods described herein to apply previously identified aptamers that specifically bind to a transcription factor to perform a comprehensive chromatin-IP assay against a plurality of human TFs simultaneously with a mixture of their corresponding aptamers (ChlP-omix).
  • a library of identified aptamers that contain adaptor sequences on their ends, and that specifically bind to a target protein are biotinylated and mixed with chromatin preparations in which genomic DNA from cells has been sheered and modified to contain "Y" adapters on their ends.
  • the ends of the aptamers are then ligated to the "Y" adapter ends of the sheered genomic DNA. Beads coated in streptavidin are added to the mixture. After washing, bound ligation products are amplified and sequenced.
  • the sequence of an aptamer can be used to identify the TF and the genomic DNA sequences ligated to this aptamer can be used to identify chromatin sequences to which that TF binds.
  • FIG. 34 depicts a schematic of the methods described herein to perform small molecule inhibitor screens against ion channels using an array of virion particles containing
  • Virion-displayed ion channels e.g., 64 recombinant virions
  • the dyes e.g., ANG-2 for sodium channel imaging
  • a collection of compounds e.g., Sigma LOP AC and Microsource Spectrum
  • the plates will be loaded onto a BD Pathway Imager to establish a baseline, followed by adding stimulus buffer.
  • the fluorescence signals are continuously detected for 180 sec. Signals obtained from WT virions will sever as baseline of fluorescent signal detection. Compared to the WT virions, a compound that causes a signal reduction >3 standard deviations in activity will be scored as a hit.
  • FIG. 35A depicts an exemplary design of address oligos, a 40-mer DNA aptamer library, and sequences reads designated as indicating a positive interaction for a dDAPPL method as described herein.
  • FIG. 35B depicts a graph of the distribution of aptamer sequences identified using a 40- mer DNA aptamer library in a dDAPPL method as described herein.
  • FIG. 36A depicts an agarose gel showing unexpected DAPPL ligation products at higher than expected molecular weights when an aptamer-protein DAPPL screen was performed according to the accompanying schematic (left).
  • FIG 36B depicts an agarose gel showing a single clean band corresponding to a DAPPL ligation product when a modified aptamer-protein DAPPL screen was performed according to the accompanying schematic (left). The redesigned DAPPL produced a clean, single-banded ligation product of 1 19 bp.
  • FIG. 37A depicts exemplary macrocycles utilized to screen and identify macrocycle- protein interactions.
  • FIG. 37B depicts a schematic of the methods described herein to screen and identify macrocycle-protein interactions.
  • FIG. 37C depicts an agarose gel of eluted DAPPL products that were amplified using PCR for 40 cycles. 10 colonies were chosen for Sanger sequencing; 7 were confirmed correctly ligated.
  • FIG. 37D depicts Sanger sequencing confirming the expected sequences of the ligated products after performing a macrocycle-protein DAPPL assay to detect interactions between Src macrocycle 1 and IDE macrocycle 6 with Srcl or IDE6.
  • FIG. 38A depicts an exemplary DAPPL-PPI method schematic. FK506, GST and printing buffer were mixed with different address oligos and then printed on an array.
  • FIG. 38B depicts labeling of FKBP1 A (GST tagged) and GST with different 5' maleimide proximity oligos (POs).
  • FIG. 38C depicts an exemplary process for generating cohesive sticky ends of address oligos and proximity oligos.
  • FIG. 38D depicts an exemplary process for probing macrocycle and protein binding partners of FKBPl A (GST tagged).
  • FKBPl A binds to FK506 and rapamycin.
  • Proximity oligo #1 is close to address oligo #1 and #2 and ready for ligation.
  • GST with proximity oligo #2 is washed away.
  • FIG. 38E depicts an exemplary process and design for probing macrocycle and protein binding partners of FKBPl A (GST tagged).
  • FIG. 38F depicts a stained agarose gel of primary PCR products.
  • FIG. 38G depicts a stained agarose gel of the primary PCR products after a cleaning step (left) and a stained agarose gel of secondary PCR products using AO Fl and PO R2-2 primers as shown in FIG. 38E.
  • FIG. 39 A depicts a schematic of the methods described herein to perform RNA aptamer screening screens against human kinases to identify phospho-specific RNA aptamers potential or application as therapies.
  • RNA aptamers can be induced to express in cells and phospho-specific RNA aptamers can serve as a unique set of molecular tools for dissecting protein kinase functions in cells.
  • FIG. 39B depicts a stained agarose gel showing DAPPL product detection using the RNA aptamer screening methodology as shown in FIG. 39A.
  • FIG. 40 depicts a schematic of address polynucleotides comprising an affinity tag (GST) and target analyte associated with a solid support (glutathione bead).
  • FIG. 41 depicts a schematic of an array, wherein wells of the array are coated with an affinity ligand (GST protein binding residue).
  • FIG. 41 further depicts address polynucleotides comprising an affinity tag (GST) and target analyte associated with the affinity ligand.
  • “about” can mean a range of up to 20%, up to 10%, up to 5%, or up to 1% of a given value. Alternatively, particularly with respect to biological systems or processes, the term can mean within an order of magnitude, within 5 -fold, and more preferably within 2-fold, of a value. Where particular values are described in the application and claims, unless otherwise stated the term “about” meaning within an acceptable error range for the particular value should be assumed.
  • the term “about” has the meaning as commonly understood by one of ordinary skill in the art. In some embodiments, the term “about” refers to ⁇ 10%. In some embodiments, the term “about” refers to ⁇ 5%.
  • attach refers to covalent interactions (e.g., by chemically coupling), or non-covalent interactions (e.g., ionic interactions, hydrophobic interactions, hydrogen bonds, hybridization, etc.).
  • non-covalent interactions e.g., ionic interactions, hydrophobic interactions, hydrogen bonds, hybridization, etc.
  • “specific”, “specifically”, or specificity” refer to the preferential recognition, contact, and formation of a stable complex between a first molecule and a second molecule compared to that of the first molecule with any one of a plurality of other molecules (e.g., substantially less to no recognition, contact, or formation of a stable complex between the first molecule and any one of the plurality of other molecules).
  • two molecules may be specifically attached, specifically bound, specifically coupled, or specifically linked.
  • specific hybridization between a first polynucleotide and a second polynucleotide can refer to the binding, duplexing, or hybridizing of the first polynucleotide preferentially to a particular nucleotide sequence of the second polynucleotide under stringent conditions.
  • a sufficient number complementary base pairs in a polynucleotide sequence may be required to specifically hybridize with a target nucleic acid sequence.
  • a high degree of complementarity may be needed for specificity and sensitivity involving hybridization, although it need not be 100%.
  • barcoded to refers to a relationship between molecules where a first molecule contains a barcode that can be used to identify a second molecule.
  • Proximity or "in proximity to” refers to a distance between two locations or molecules relative to each other that allows a reaction to take place.
  • the distance can be a length that permits the address polynucleotide of a first discrete location of a discrete region to be coupled, such as through ligation, to a proximity probe when the proximity probe is bound to a target analyte at a second discrete location of the discrete region.
  • the distance can be a length that permits the address polynucleotide to be in contact with a proximity polynucleotide when joined by a splint polynucleotide when the proximity probe is bound to a target analyte.
  • the present invention relates to methods, kits, and compositions for Digital Affinity Profiling via Proximity Ligation (DAPPL) that can be used to screen multiple binding moieties against multiple target analytes using proximity coupling and deep sequencing techniques in an extremely high-throughput manner.
  • the present invention relates to a proximity-probe based detection assay, (e.g., a proximity ligation assay (PL A)), for detecting binding of a binding moiety to an analyte in a sample, such as a target analyte on a solid support.
  • a proximity-probe based detection assay e.g., a proximity ligation assay (PL A)
  • PL A proximity ligation assay
  • Proximity ligation assays rely on proximal binding of proximity probes to an analyte to generate a signal from a ligation reaction involving or mediated by (e.g. between and/or templated by) nucleic acid domains of the proximity assays.
  • Proximity-probe based detection assays permit sensitive, rapid, and convenient detection and/or quantification of one or more analytes in a sample by converting the presence of such an analyte into a readily detectable or quantifiable nucleic acid-based signal, and can be performed in homogeneous or heterogeneous formats.
  • Proximity probes of the art are generally used in pairs, and individually consist of an analyte-binding domain with specificity to the target analyte, and a functional domain, e.g. a nucleic acid domain coupled thereto.
  • the methods, kits, and compositions described herein rely on the principle of proximity probing, wherein a binding moiety' s interaction with an analyte is detected through the coupling of multiple (e.g., two or three or four or more) polynucleotide probes, which when brought into proximity by interaction of a binding moiety to an analyte, allow a signal to be generated.
  • multiple e.g., two or three or four or more
  • At least one of the proximity probes comprises a nucleic acid domain (e.g., a polynucleotide) linked to an analyte-binding domain (e.g., a binding moiety).
  • Generation of a signal can involve an interaction between a nucleic acid moiety of the proximity probe and a nucleic acid domain (e.g., a polynucleotide) comprised on another probe comprising a polynucleotide, such as an address polynucleotide.
  • Generation of a signal can depend on or indicate an interaction between the probes exists.
  • binding of binding moiety of a proximity probe to a target analyte brings a nucleic acid domain of the proximity probe into proximity to a nucleic acid domain (e.g., a polynucleotide) comprised on another probe, such as an address polynucleotide
  • generation of a signal can depend on, indicate, or be used to determine, for example, an interaction, affinity, specificity or a combination thereof, between the binding moiety and the target analyte.
  • an affinity, or strength of an interaction between a binding moiety and a target analyte can be determined.
  • lack of signal generation can indicate or be used to determine an interaction between an analyte-binding domain and a target analyte does not exist.
  • polynucleotides which interact with one or more proximity probes in a proximity-dependent manner, can be used in the methods described herein, for example, to determine binding partners, affinities of one or more binding moieties to one or more target analytes, and/or specificities of one or more binding moieties for one or more target analytes.
  • the multiplex assays described herein utilize a unique label (or barcode) to be attached to each analyte and probe such that positive hits can be deconvoluted at the end using high- throughput methodologies.
  • a unique label or barcode
  • mAbs can be simultaneously screened for their binding partners in a multiplex format (e.g., on a human proteome (HuProt) array, harboring over 17,000 full-length human proteins).
  • mAb-binding moiety pairs e.g., mAb-polypeptide pairs
  • Binding specificity of a given binding moiety e.g., a mAb
  • binding specificity of a given binding moiety e.g., a mAb
  • the methods and compositions can reveal the wide or narrow spectrum of target analytes to which one or more binding moieties interact. Because the methods and compositions described herein provide unique digital sequencing reads barcoded to each binding moiety and target analyte, the total number of resulting sequencing reads can serve as a unique determinant for the affinity of each binding moiety-target analyte pair.
  • DAPPL methods and compositions can be used to perform functional genome wide associations of S Ps, map transcription factors to DNase I hypersensitive sites (DHSs), determine protein and RNA (coding or non-coding) interactions, determine antigen and antibody interactions, determine protein to protein interactions, determine peptide to protein interactions, screen for aptamer binding partners, determine protein-DNA interactions (e.g., transcription factor-DNA interactions), determine small molecule to protein interactions, perform serum profiling, and much more.
  • DHSs map transcription factors to DNase I hypersensitive sites
  • determine protein and RNA (coding or non-coding) interactions determine antigen and antibody interactions
  • determine protein to protein interactions determine peptide to protein interactions
  • screen for aptamer binding partners determine protein-DNA interactions (e.g., transcription factor-DNA interactions)
  • determine small molecule to protein interactions perform serum profiling, and much more.
  • an antibody can be an autoantibody.
  • an antigen can be an autoantigen.
  • An analyte, or target analyte can be, but is not limited to, a polypeptide, a protein, a protein fragment, a tagged protein, a fusion protein, an antibody, an antibody fragment, a small molecule, a virus particle (e.g., a virus particle comprising a transmembrane protein), or a cell.
  • a target analyte does not base pair with an address polynucleotide in proximity thereto.
  • a target analyte comprises at least two amide bonds.
  • a target analyte does not comprise a phosphodiester linkage.
  • a target analyte is not DNA or RNA.
  • a target analyte comprises a polypeptide, protein, or fragment thereof.
  • Polypeptide and “protein” are used interchangeably and refer to a polymer of two or more amino acids joined by a covalent bond (e.g., an amide bond).
  • Polypeptides as described herein can include full length proteins (e.g., fully processed proteins) as well as shorter amino acid sequences (e.g., fragments of naturally-occurring proteins or synthetic polypeptide fragments).
  • Polypeptides can include naturally occurring amino acids (e.g., one of the twenty amino acids commonly found in peptides synthesized in nature, and known by the one letter abbreviations A, R, N, C, D, Q, E, G, H, I, L, K, M, F, P, S, T, W, Y and V) and non-naturally occurring amino acids (e.g., amino acids which is not one of the twenty amino acids commonly found in peptides synthesized in nature, including synthetic amino acids, amino acid analogs, and amino acid mimetics).
  • a target analyte can comprise an isolated polypeptide, a purified polypeptide, or a polypeptide within a virus particle.
  • a target analyte can comprise a polypeptide is within a virus particle membrane.
  • a virus particle refers to a fully or partially assembled capsid of a virus surrounded by a lipid envelope.
  • a viral particle may or may not contain nucleic acids.
  • a target analyte can comprise an antibody or fragment thereof.
  • a target analyte can comprise a transcription factor.
  • a target analyte can comprise a receptor.
  • a target analyte can comprise a transmembrane receptor.
  • Target analytes include isolated, purified, and/or recombinant polypeptides.
  • Target analytes include target analytes present in a mixture of analytes (e.g., a lysate).
  • target analytes include target analytes present in a lysate from a plurality of cells or from a lysate of a single cell.
  • a target analyte comprises a small molecule.
  • a target analyte can comprise a drug.
  • a target analyte can comprise a compound.
  • a target analyte can comprise an organic compound.
  • a target analyte comprises a small molecule with a molecular weight of 900 Daltons or less.
  • a target analyte comprises a small molecule with a molecular weight of about 100, 200, 300, 400, 500, 600, or about 700 Daltons or more.
  • Small molecules may be obtained, for example, from a library of naturally occurring or synthetic molecules, including a library of compounds produced through combinatorial means, i.e.
  • Combinatorial libraries as well as methods for their production and screening, are known in the art and described in: US 5,741,713; 5,734,018; 5,731,423; 5,721,099; 5,708, 153; 5,698,673; 5,688,997; 5,688,696; 5,684,71 1 ; 5,641,862; 5,639,603; 5,593,853; 5,574,656; 5,571,698; 5,565,324;
  • a target analyte can comprise a member of a specific binding pair (e.g., a ligand).
  • a target analyte can be monovalent (monoepitopic) or polyvalent (polyepitopic).
  • a target analyte can be antigenic or haptenic.
  • a target analyte can be a single molecule or a plurality of molecules that share at least one common epitope or determinant site.
  • a target analyte can be a part of a cell (e.g., a bacteria cell, a plant cell, or an animal cell).
  • a target cell can be either in a natural environment (e.g., tissue), a cultured cell, or a microorganism (e.g., a bacterium, fungus, protozoan, or virus), or a lysed cell.
  • a target analyte can be further modified (e.g. chemically), to provide one or more additional binding sites such as, but not limited to, a dye (e.g., a fluorescent dye), a polypeptide modifying moiety such as a phosphate group, a carbohydrate group, and the like, or a polynucleotide modifying moiety such as a methyl group.
  • a target analyte comprises at least one potential binding site for a binding moiety.
  • a target analyte comprises one binding site. In some instances, a target analyte comprises at least two binding sites. For example, a target analyte can comprise 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more binding sites.
  • a target analyte is a molecule found in a sample from a host.
  • a sample from a host includes a body fluid (e.g., urine, blood, plasma, serum, saliva, semen, stool, sputum, cerebral spinal fluid, tears, mucus, and the like).
  • a sample can be examined directly or may be pretreated to render the target analyte more readily detectible.
  • Samples include a quantity of a substance from a living thing or formerly living things.
  • a sample can be natural, recombinant, synthetic, or not naturally occurring.
  • a target analyte can be expressed from a cell naturally or recombinantly, in a cell lysate or cell culture medium, an in vitro translated sample, or an immunoprecipitation from a sample ⁇ e.g., a cell lysate).
  • a target analyte is expressed in a cell-free system or in vitro.
  • a target analyte can be in a cell extract containing a nucleotide template and raw materials for translation of the target analyte.
  • a target analyte can be in a cell extract containing a DNA template, and reagents for transcription and translation.
  • Exemplary sources of cell extracts that can be used include wheat germ, Escherichia coli, rabbit
  • Exemplary cell-free methods that can be used to express target polypeptides ⁇ e.g., to produce target polypeptides on an array) include Protein in situ arrays (PISA), Multiple spotting technique (MIST), Self-assembled mRNA translation, Nucleic acid programmable protein array (NAPPA), nanowell NAPPA, DNA array to protein array (DAP A), membrane-free DAP A, nanowell copying and ⁇ -microintaglio printing, and pMAC - protein microarray copying ⁇ See Kilb et al, Eng. Life Sci. 2014, 14, 352-364).
  • PISA Protein in situ arrays
  • MIST Multiple spotting technique
  • NAPPA Nucleic acid programmable protein array
  • DAP A DNA array to protein array
  • membrane-free DAP A nanowell copying and ⁇ -microintaglio printing
  • pMAC - protein microarray copying ⁇ See Kilb et al, Eng. Life Sci. 2014, 14, 352-364).
  • a target analyte is synthesized in situ ⁇ e.g., on a solid substrate of an array) from a DNA template.
  • a plurality of target analytes is synthesized in situ from a plurality of corresponding DNA templates in parallel or in a single reaction.
  • Exemplary methods for in situ target polypeptide expression include those described in Stevens, Structure 8(9): R177-R185 (2000); Katzen et al, Trends Biotechnol. 23(3); 150-6. (2005); He et al, Curr. Opin. Biotechnol 19(l );4-9. (2008); Ramachandran et al, Science 305(5680):86-90. (2004); He et al, Nucleic Acids Res. 29(15);E73-3 (2001); Angenendt et al., Mol. Cell
  • target analyte polypeptide synthesis is carried out on a solid surface (e.g., an array surface) coated with a protein-capturing reagent or antibody.
  • the target polypeptides comprise a tag ⁇ e.g., polyhistidine or GST) that is bound by the capture reagent or antibody, thus coupling the target polypeptides to the solid surface ⁇ e.g., a nucleic acid programmable protein array (NAPPA)).
  • the DNA template is immobilized onto the same protein-capture surface.
  • the DNA template can be biotinylated and bound to avidin pre-coated onto the protein capture surface.
  • the DNA template is not coupled to the solid support.
  • the DNA template is added as a free molecule in the reaction synthesis mixture ⁇ e.g., a protein in situ array (PISA)).
  • PISA protein in situ array
  • in situ puromycin-capture methods can be used to express target polypeptides.
  • the template DNA can be transcribed to mRNA, and a single- stranded DNA oligonucleotide modified with biotin and puromycin on each end can be hybridized to the 3 '-end of the mRNA.
  • the mRNAs can be coupled to the surface e.g., by the binding of biotin to streptavidin that is pre-coated on the surface. Cell extract can then be added to initiate in situ translation.
  • the ribosome When the ribosome reaches the hybridized oligonucleotide, it stalls and incorporates the puromycin molecule to the nascent polypeptide chain, thereby attaching the newly synthesized protein to the surface via the DNA oligonucleotide.
  • Purified target polypeptides may be obtained after the mRNA is removed ⁇ e.g., digested with RNase).
  • DNA array to protein array (DAP A) methods can be used to repeatedly produce protein arrays by printing them from a single DNA template array, on demand.
  • An array of immobilized DNA templates on a substrate is assembled face-to-face with a second substrate pre-coated with a protein-capturing reagent, and a membrane soaked with a cell extract is placed between the two substrates for transcription and translation.
  • the synthesized target polypeptides are then immobilized onto a substrate to form the array.
  • a target analyte can comprise a plurality of target analytes.
  • a target analyte can comprise a plurality of target analytes representing a substantial portion or an entire organism's proteome, such as a bacterial, viral, fungal, plant, or animal proteome.
  • a target analyte can comprise a plurality of target analytes representing a substantial portion or an entire proteome of an insect or mammal, such as a mouse, rat, rabbit, cat, dog, monkey, goat, or human.
  • a target analyte can comprise a plurality of target analytes representing at least 0.5%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, or 100% of an organism's proteome.
  • a target analyte can comprise a plurality of target analytes comprising at least 2 different target analytes.
  • a target analyte can comprise a plurality of target analytes comprising at least 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 1 1,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 21,000, 22,000, 23,000, 24,000, or 25,000 different target analytes.
  • target analytes can comprise a tag.
  • the tag is an affinity tag.
  • affinity tags include, but are not limited to, Glutathione- S- transferase (GST), Maltose binding protein (MBP), Green Fluorescent Protein (GFP), AviTag (a peptide allowing biotinylation by the enzyme BirA and so the protein can be isolated by streptavidin), Calmodulin-tag (a peptide bound by the protein calmodulin), polyglutamate tag (a peptide binding efficiently to anion-exchange resin such as Mono-Q), FLAG-tag (a peptide recognized by an antibody), HA-tag (a peptide recognized by an antibody), His tag (generally 5- 0 histidines which are bound by a nickel or cobalt chelate), Myc-tag (a short peptide
  • GST Glutathione- S- transferase
  • MBP Maltose binding protein
  • GFP Green Fluorescent Protein
  • AviTag a peptide allowing biot
  • a proteinaceous target analyte can comprise a fusion tag.
  • a proteinaceous target analyte can comprise a GST-tag, His-tag, FLAG-tag, T7 tag, S tag, PKA tag, HA tag, c-Myc tag, Trx tag, Hsv tag, CBD tag, Dsb tag, pelB/ompT, KSI, MBP tag, VSV-G tag, 3-Gal tag, GFP tag, or a combination thereof, or other similar tags.
  • the protein tag binder is a group which binds an endogenous protein tag (e.g., an epitope on the protein).
  • the protein tag binder can typically be an antibody or antibody fragment which is sufficient to form a non-covalent association complex with the protein tag or epitope.
  • the polypeptide target analytes comprise PTMs including, but not limited to, glycosylation, phosphorylation, acetylation, methylation, myristoylation, prenylation, or proteolytic processing.
  • a polypeptide target analyte is homologous to a native polypeptide.
  • a target analyte comprises a contiguous span of at least 6 amino acids, for example, least 8, 9, 10, 12, 15, 20, 25, 30, 40, 50, or 100 amino acids of a reference sequence.
  • a target analyte comprises a contiguous stretch of amino acids comprising a site of a mutation or functional mutation, including a deletion, addition, swap, or truncation of the amino acids in a polypeptide sequence.
  • Polypeptides may be isolated from human or mammalian tissue samples or expressed from human or mammalian genes.
  • Polypeptides may be made using routine expression methods known in the art.
  • a polynucleotide encoding a desired polypeptide may be inserted into an expression vector suitable for any convenient host. Both eukaryotic and prokaryotic host systems can be used in forming recombinant polypeptides.
  • a polypeptide may be isolated from lysed cells or from the culture medium and purified to the extent needed for its intended use. ⁇ See, e.g., WO2012103260 and WO2011159959).
  • Purification may be by any technique known in the art, for example, differential extraction, salt fractionation, chromatography, centrifugation, and the like ⁇ See, e.g., Abbondanzo et al, (1993) Methods in Enzymology, Academic Press, New York. pp. 803-23).
  • shorter protein fragments may be produced by chemical synthesis.
  • proteins of the presently disclosed subject matter are extracted from cells or tissues of humans or non-human animals.
  • Methods for purifying proteins are known in the art, and include the use of detergents or chaotropic agents to disrupt particles followed by differential extraction and separation of the polypeptides by ion exchange chromatography, affinity chromatography, sedimentation according to density, and gel electrophoresis, for example.
  • Reference cDNA may be used to express polypeptides.
  • a nucleic acid encoding a polypeptide to be expressed can be operably linked to a promoter in an expression vector using conventional cloning technology.
  • a polypeptide in an expression vector may comprise the full coding sequence for the polypeptide or a portion thereof.
  • a target analyte is a membrane bound protein.
  • the membrane bound protein is CD4, a classical type I membrane protein with a single transmembrane (TM) domain. (Carr et al, (1989) J. Biol. Chem. 264:21286-95).
  • the membrane bound protein is GPR77, a multi- spanning, G-protein coupled receptor (GPCR) membrane protein. (Cain & Monk, (2002) J. Biol. Chem. 277:7165- 69).
  • Additional exemplary membrane bound proteins include, but are not limited to, GPCRs ⁇ e.g. adrenergic receptors, angiotensin receptors, cholecystokinin receptors, muscarinic acetylcholine receptors, neurotensin receptors, galanin receptors, dopamine receptors, opioid receptors, erotonin receptors, somatostatin receptors, etc.), ion channels ⁇ e.g., nicotinic acetylcholine receptors, sodium channels, potassium channels, etc.), receptor tyrosine kinases, receptor serine/threonine kinases, receptor guanylate cyclases, growth factor and hormone receptors ⁇ e.g., epidermal growth factor (EGF) receptor), and others.
  • GPCRs ⁇ e.g. adrenergic receptors, angiotensin receptors, cholecystokinin receptors, muscarinic acet
  • Mutant or modified variants of membrane-bound proteins may also be used.
  • some single or multiple point mutations of GPCRs retain function and are involved in disease ⁇ See, e.g., Stadel et al, (1997) Trends in Pharmacological Review 18:430-37).
  • a target analyte can comprise a plurality of target analytes that are specific to a common pathway.
  • Target analytes belong to a common pathway when they share one or more attributes in common in a gene ontology, a collection that assigns defined characteristics to a set of genes and their products.
  • the ontology administered by the Gene Ontology ("GO") Consortium is particularly useful in this regard.
  • Target analytes belonging to common pathways can be identified by searching gene ontology, such as GO, for genes sharing one or more attributes.
  • the common attribute could be, for example, a common structural feature, a common location, a common biological process or a common molecular function.
  • target analytes in a common pathway are the expression product of genes involved in the same biological process or molecular function as annotated by gene ontology, ⁇ e.g., genes involved in the response to DNA damage or gene products of transcription factors, such as of a particular tissue, cell type or organ, such as the brain).
  • genes involved in the same biological process or molecular function as annotated by gene ontology, ⁇ e.g., genes involved in the response to DNA damage or gene products of transcription factors, such as of a particular tissue, cell type or organ, such as the brain.
  • target analytes in a common pathway are the expression product of genes involved in the different biological process or molecular function as annotated by gene ontology.
  • target analytes in a common pathway are small molecules that affect the expression product of genes involved in the same biological process or molecular function as annotated by gene ontology.
  • target analytes in a common pathway are the gene products of genes whose transcript levels or proteins levels change upon treatment or exposure to the same stimulus and are thus co-regulated ⁇ e.g., target analytes that are induced or repressed upon treatment to UV radiation).
  • target analytes in a common pathway are small molecules that affect the gene products of genes whose transcript levels or proteins levels change upon treatment or exposure to the same small molecule and are thus co-regulated.
  • target analytes in a common pathway are target analytes that contain similar sequence features. These features may be a DNA sequence motif, collection of DNA sequence motifs, or enrichment of higher order sequence features that are distinguishable from a background model of random genomic sequences.
  • a DNA sequence motif can either be defined by a consensus sequence or a probability matrix where the identity of each base at each position of a motif is defined as a probability.
  • the members of the pathway share a common structural or functional attribute.
  • the target analytes could share a common sequence motif, such as a zinc finger or a transmembrane region.
  • target analytes in a common pathway can be the gene products of genes whose sequences, transcripts or proteins are connected via metabolic transformations and/or physical protein-protein, protein-DNA and protein-compound interactions. Enzymes catalyze these reactions, and often require dietary minerals, vitamins and other cofactors in order to function properly. Because of the many chemicals that may be involved, pathways can be quite elaborate.
  • target analytes in a common pathway are gene products of genes which are all bound by the same transcription factor protein, complex of transcription factor proteins, other nucleic acid binding proteins, or other molecules. These interactions may occur in a living cell (in vivo) or in a solution of purified molecules (in vitro).
  • the target analytes in a common pathway can belong to the same signal transduction pathway.
  • signal transduction refers to any process by which a cell converts one kind of signal or stimulus into another, most often involving ordered sequences of biochemical reactions inside the cell that are carried out by enzymes, activated by second messengers resulting in what is thought of as a signal transduction pathway.
  • signal transduction involves the binding of extracellular signaling molecules (or ligands) to cell- surface receptors that face outwards from the plasma membrane and trigger events inside the cell.
  • intracellular signaling cascades can be triggered through cell- sub stratum interactions, as in the case of integrins which bind ligands found within the extracellular matrix.
  • Steroids represent another example of extracellular signaling molecules that may cross the plasma membrane due to their lipophilic or hydrophobic nature. Many steroids, but not all, have receptors within the cytoplasm and usually act by stimulating the binding of their receptors to the promoter region of steroid responsive genes. Within multicellular organisms there are a diverse number of small molecules and polypeptides that serve to coordinate a cell's individual biological activity within the context of the organism as a whole. Examples include hormones (e.g. melatonin), growth factors (e.g. epidermal growth factor), extra-cellular matrix components (e.g. fibronectin), cytokines (e.g. interferon-gamma), chemokines (e.g. RANTES), neurotransmitters (e.g. acetylcholine), and neurotrophins (e.g. nerve growth factor).
  • hormones e.g. melatonin
  • growth factors e.g. epidermal growth factor
  • extra-cellular matrix components e.g.
  • Environmental stimuli may also be molecular in nature or more physical, such as, light striking cells in the retina of the eye, odorants binding to odorant receptors in the nasal epithelium, bitter and sweet tastes stimulating taste receptors in the taste buds, UV light altering DNA in a cell, and hypoxia activating a series of events in cells.
  • Certain microbial molecules e.g. viral nucleotides, bacterial lipopolysaccharides, or protein target analytes are able to elicit an immune system response against invading pathogens, mediated via signal
  • Gene activation leads to further cellular effects, since the protein products of many of the responding genes include enzymes and transcription factors themselves. Transcription factors produced as a result of a signal transduction cascade can in turn activate yet more genes. Therefore an initial stimulus can trigger the expression of an entire cohort of genes, and this in turn can lead to the activation of any number of complex physiological events. These events include, for example, the increased uptake of glucose from the blood stream stimulated by insulin and the migration of neutrophils to sites of infection stimulated by bacterial products.
  • target analytes in a common pathway are part of an oncology pathway.
  • Target analytes in an oncology pathway are those gene products of genes involve in the development of hyperplasia, neoplasia and/or cancer.
  • oncology pathways include, but are not limited to, hypoxia, DNA damage, apoptosis, cell cycle, and p53 pathway.
  • target analytes in a common pathway are part of a membrane pathway.
  • membrane pathways include, but are not limited to, transport protein, G-coupled receptor, ion channel, cell-adhesion protein, and receptor pathways.
  • target analytes in a common pathway are part of a nuclear receptor pathway.
  • target analytes in a nuclear receptor pathway include, but are not limited to, gene products regulated by the glucocorticoid receptor protein, estrogen receptor proteins, peroxisome proliferator-activated receptor proteins, androgen receptor proteins, and transporter proteins, including ABC and SLC transporters.
  • target analytes in a common pathway are part of a neuronal pathway.
  • target analytes in a neuronal pathway include, but not limited to, gene products of genes expressed in neurons such as neurotransmitters and cell adhesion proteins.
  • target analytes in a common pathway are part of a vascular pathway.
  • target analytes in a vascular pathway include, but not limited to, target analytes involved in angiogenesis, lipid metabolism, and inflammation.
  • target analytes in a common pathway are part of a signaling pathway.
  • target analytes in a signaling pathway include, but are not limited to, gene products involved in cell-to-cell signaling, hormones, hormone receptors, cAMP response, and cytokines.
  • target analytes in a common pathway are part of an enzymatic pathway.
  • target analytes in a enzymatic pathway include, but are not limited to, gene products of genes involved in glycolysis, anaerobic respiration, Krebs cycle / Citric acid cycle, Oxidative phosphorylation, fatty acid oxidation ( ⁇ -oxidation), gluconeogenesis, HMG-CoA reductase pathway, pentose phosphate pathway, porphyrin synthesis (or heme synthesis) pathway, urea cycle, photosynthesis (plants, algae, cyanobacteria) and chemosynthesis (some bacteria).
  • the present invention also provides a library of target analytes comprising a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100%) of the target analytes are part of a common pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of the target analytes are part of an oncology pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of the target analytes are part of a hypoxia pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of the target analytes are part of a DNA-damage pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of the target analytes are part of an apoptosis pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of the target analytes are part of a cell cycle pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of the target analytes are part of a p53 pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of the target analytes are differently selected from the group consisting of hypoxia pathway, DNA-damage pathway, apoptosis pathway, cell cycle pathway, and p53 pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100%) of the target analytes are part of a membrane bound pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of the target analytes are part of a nuclear receptor pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%), 99% or 100%) of the target analytes are part of a glucocorticoid receptor pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of the target analytes are part of a peroxisome proliferator-activated receptor pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of the target analytes are part of an estrogen receptor pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%), 99% or 100%) of the target analytes are part of an androgen receptor pathway
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of the target analytes are part of a cytochrome P450 receptor pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of the target analytes are part of a transporter receptor pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100%) of the target analytes are differently selected from the group consisting of glucocorticoid receptor pathway, peroxisome proliferator-activated receptor pathway, estrogen receptor pathway, androgen receptor pathway, cytochrome P450 pathway, and transporter pathways.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of the target analytes are part of a vascular pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%), 80%), 90%), 99%) or 100%) of the target analytes are part of a neuronal pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of the target analytes are part of a transcription factor pathway.
  • a library of target analytes comprises a plurality of target analytes in which at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of the target analytes are part of a signaling pathway.
  • the present invention also provides a library of target analytes in which the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of all the target analytes that are part of a common pathway in the genome.
  • the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of all the target analytes that are part of an oncology pathway in the genome.
  • the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%), 99% or 100%) of all the target analytes that are part of a hypoxia pathway in the genome.
  • the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of all the target analytes that are part of a DNA-damage pathway in the genome. In some embodiments, the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of all the target analytes that are part of an apoptosis pathway in the genome. In some embodiments, the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of all the target analytes that are part of a cell cycle pathway in the genome.
  • the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of all the target analytes that are part of a p53 pathway in the genome. In some embodiments, the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of all the target analytes that are part of a membrane bound pathway in the genome. In some embodiments, the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of all the target analytes that are part of a nuclear receptor pathway in the genome. In some
  • the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%), 99% or 100%) of all the target analytes that are part of a glucocorticoid receptor pathway in the genome. In some embodiments, the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of all the target analytes that are part of a peroxisome proliferator-activated receptor pathway in the genome. In some embodiments, the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of all the target analytes that are part of an estrogen receptor pathway in the genome.
  • the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%), 99%) or 100%) of all the target analytes that are part of an androgen receptor pathway in the genome. In some embodiments, the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of all the target analytes that are part of a cytochrome P450 receptor pathway in the genome. In some embodiments, the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of all the target analytes that are part of a transporter receptor pathway in the genome.
  • the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of all the target analytes that are part of a neuronal pathway in the genome. In some embodiments, the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% or 100% of all the target analytes that are part of a signaling pathway in the genome, n some
  • the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%), 99% or 100%) of all the target analytes that are part of a vascular pathway in the genome. In some embodiments, the library represents at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%), 80%), 90%), 99% or 100%> of all the target analytes that are part of a transcription factor pathway in the genome.
  • a target analyte can be coupled to a solid support (e.g., an array or bead).
  • a target analyte is non-covalently coupled to a solid support.
  • a non- covalent interaction can be an ionic interaction or a van der Waals interaction.
  • a target analyte is covalently coupled to a solid support.
  • a target analyte is reversibly coupled to a solid support.
  • a target analyte is irreversibly coupled to a solid support.
  • a surface of a solid support can be coated with a functional group and a target analyte can be attached to the solid support through the functional group.
  • a solid support can be coated with a first functional group and a target analyte comprising a second functional group can be attached to the solid support by reacting the first functional group with the second functional group.
  • a surface of a solid support can be coated with streptavidin and a biotinylated target analyte can be attached thereto.
  • Exemplary couplings of a target analyte include streptavidin- or avidin- to biotin interactions; hydrophobic interactions; magnetic interactions; polar interactions, (e.g., associations between two polar surfaces); formation of a covalent bond (e.g., an amide bond, disulfide bond, thioether bond, or via crosslinking agents; and via an acid-labile linker.
  • the surface of a solid support can be coated with an affinity ligand.
  • an affinity ligand can include, but is not limited to an antigen, an antibody, an antibody fragment, glutathione, calmodulin, biotin, streptavidin, streptactin, amylose, an amon-exchange resin such as Mono-Q, FlAsH and ReAsH biarsenical compounds, pilin-C protein, SpyCatcher protein or a metal chelate.
  • the metal chelate can include but is not limited to nickel, cobalt, zinc, mercury, cupper or iron chelate.
  • the solid support can be coated entirely. In some embodiments, the solid support can be coated partially.
  • target analytes can comprise an affinity tag and solid support can comprise an affinity ligand, thus coupling the target analytes to the solid support by reacting the affinity tag with the affinity ligand.
  • a target analyte is coupled to a solid surface through a linker.
  • a first functional group of a linker attached to a solid surface can be coupled to a target analyte, thereby coupling the target analyte to the solid surface.
  • a first functional group of a linker can be coupled to a target analyte and a second functional group of the linker can be coupled to a solid support, thereby coupling the target analyte to the solid surface, target analyte can be coupled to a solid surface through a linker.
  • a linker comprising a first and a second functional group can be attached to the solid support via the second functional group after the first functional group is coupled to the target analyte. In some instances, a linker comprising a first and a second functional group can be attached to the solid support via the second functional group before the first functional group is coupled to the target analyte.
  • a target analyte is coupled to a solid surface via an antibody.
  • an antibody linker can be attached to a solid surface and a target analyte to which the antibody specifically bind can be linked to the solid support by binding to the antibody linker.
  • the coupling is photocleavable.
  • target analytes can comprise a tag that is directly coupled to a solid surface.
  • a proteinaceous target analyte can comprise a fusion tag that is directly conjugated to the solid surface.
  • a fusion tag that is directly conjugated to the solid surface.
  • proteinaceous target analyte can comprise a GST-tag, His-tag, FLAG-tag, or other similar tags and the tag can be directly coupled to the solid surface instead of the proteinaceous target analyte itself.
  • target analyte e.g., a polypeptide, a protein, an antibody, an antibody fragment, a small molecule, a virus particle, or a cell
  • suitable coupling chemistries can be employed to couple the target analyte to a solid surface.
  • a covalent amide bond can be formed between a target analyte and a solid support.
  • a covalent amide bond can be formed by reacting a carboxyl-functionalized target analyte with an amino-functionalized solid support.
  • a covalent amide bond can be formed by reacting an amide-functionalized target analyte with a carboxyl-functionalized solid support.
  • Thiol groups can be used to couple or immobilize target analytes to a solid surface.
  • target analytes having or functionalized with thiol groups with may be immobilized on surfaces presenting, e.g., maleimide, aryl- or carbon-carbon double-bond-containing groups through formation of stable carbon-sulfur bonds, or through interactions with aziridine- functionalized surfaces.
  • Disulfide exchange reactions with thiol-functionalized surfaces may also be used.
  • Target analytes having or functionalized with thiol groups may be immobilized on gold surfaces through semi-covalent interactions between gold and sulphur groups.
  • Carboxylic acid-functionalized surfaces may also be used to immobilize target analytes functionalized with carbodiimide and diazoalkane groups. Solid surfaces presenting hydroxyl groups may be used to immobilize isocyanate- and epoxide-functionalized target analytes.
  • Functionalized target analytes may also be immobilized through cycloaddition reactions between functional groups having a conjugated diene and groups having a substituted alkene through Diels- Alder chemistry, or using "click" chemistry, through reactions between nitrile and azine groups.
  • the target analytes -surface orientation of functional groups may be reversed.
  • An alternative means of covalent attachment not utilizing a derivatized binding agent utilizes array surfaces having photoreactive groups such as benzophenone, diazo, diazirine, phthalamido and arylazide groups.
  • Non-covalent immobilization may involve electrostatic interactions between target analytes and surfaces modified to contain positively- or negatively-charged groups, such as amine or carboxy groups, respectively.
  • Target analytes may be non-covalently immobilized in a defined orientation, for example, using fluorophilic, biotin-streptavidin, histidine-Ni, histidine- Co, and complementary single-stranded DNA interactions between tagged target analytes and binding partner-coated surfaces, in either orientation.
  • Appropriate agents for coupling of target analytes to a solid surface include a variety of agents that are capable of reacting with a functional group present on a surface of the target analyte and with a functional group present on the solid surface.
  • Reagents capable of such reactivity include homo- and hetero-bifunctional reagents, many of which are known in the art.
  • Exemplary bifunctional cross-linking agents include is N-succinimidyl(4-iodoacetyl)
  • SATA N-succinimidyl-3-(2-pyridyldithio) propionate
  • SPDP succinimidyl 4-(N-maleimidomethyl) cyclohexane-l-carboxylate
  • HYNIC 6-hydrazinonicotimide
  • Any suitable nucleophile reactive group can be used including - R 1 - H 2 (hydrazide), NH 2 (carbonylhydrazide), (thiocarbonylhydrazide), -(S0 2 ) Ri H 2
  • the nucleophilic moiety can include any suitable nucleophile, e.g., hydrazide, hydroxylamine, semicarbazide, or carbonylhydrazide.
  • a target analyte may be deposited onto a substrate or support by any suitable technique.
  • a target analyte may be deposited as a monolayer (e.g., a self-assembled monolayer), a continuous layer or as a discontinuous (e.g., patterned) layer.
  • a target analyte may be deposited or coupled to a support or substrate by modification of the substrate or support by chemical reaction (See, e.g., U. S. Pat. No.
  • target analytes may be directly spotted onto a surface (e.g., a planar glass surface).
  • glycerol (30-40%) may be employed, and/or spotting can be carried out in a humidity-controlled environment.
  • An address polynucleotide is a polynucleotide containing a sequence barcoded to a target analyte or discrete region containing a target analyte.
  • Polynucleotide “nucleic acid sequence”, and “nucleic acid” are used interchangeably and refer to deoxyribonucleotides or ribonucleotides and polymers thereof in either single-stranded or double-stranded form. Nucleic acid sequences can contain known nucleotide analogs or modified backbone residues or linkages.
  • Nucleic acid sequences implicitly encompass conservatively modified variants thereof (e.g., degenerate codon substitutions/mutations) and complementary sequences.
  • Polynucleotides include, among others, single-stranded DNA, double-stranded DNA, DNA that is a mixture of single- and double-stranded regions, single-stranded RNA, double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, and hybrid molecules comprising DNA and RNA.
  • an address polynucleotide does not substantially interact with a target analyte.
  • an address polynucleotide interacts with, or can be coupled to, a proximity polynucleotide when the address polynucleotide is in proximity to the proximity polynucleotide.
  • an address polynucleotide can be coupled to a proximity polynucleotide when the binding moiety of a proximity probe binds to a target analyte in proximity to the address polynucleotide.
  • An address polynucleotide can be coupled directly or indirectly to a solid support.
  • An address polynucleotide can be coupled covalently or non-covalently to a solid support.
  • An address polynucleotide can be coupled to a solid support.
  • An address polynucleotide can be coupled to a solid surface at a particular address (e.g., a discrete region), of the solid support.
  • An address polynucleotide can be coupled to a solid surface at a discrete location within a particular address of the solid support.
  • An address polynucleotide can be coupled to a solid support at a first location within a discrete region of the solid support comprising a target analyte.
  • An address polynucleotide can be coupled to a solid support at a first location within a discrete region of the solid support and a target analyte can be coupled to the solid support at a second location within the discrete region of the solid support.
  • An address polynucleotide coupled to a solid support within a first discrete region of the solid support can be different than an address polynucleotide within a second discrete region of the solid support.
  • An address polynucleotide coupled to a solid support at a first location within a first discrete region of the solid support can be different than an address polynucleotide coupled to the solid support at a first location within a second discrete region of the solid support.
  • An address polynucleotide can comprise a plurality of address polynucleotides, each barcoded to a particular target analyte.
  • a first address polynucleotide can be barcoded to a first target analyte and a second address polynucleotide can be barcoded to a second target analyte.
  • An address polynucleotide of the invention can be used to identify a target analyte to which it is barcoded.
  • a barcode of an address polynucleotide can correspond to a target analyte.
  • a barcode of a first address polynucleotide can correspond to a first target analyte and a barcode of a second address polynucleotide can correspond to a second target analyte.
  • a sequence of an address polynucleotide can be used to identify a target polynucleotide.
  • An address polynucleotide can comprise a plurality of segments.
  • polynucleotide can comprise an address barcode sequence.
  • An address polynucleotide can comprise an address linker sequence.
  • An address polynucleotide can comprise an address primer binding sequence.
  • An address polynucleotide can comprise an address spacer sequence.
  • An address polynucleotide can comprise an address polynucleotide linker sequence, an address barcode, an address primer binding sequence, an address spacer, or any combination thereof, arranged in a particular order.
  • an address polynucleotide can be arranged in the order of the address polynucleotide linker sequence, the address barcode, the address primer binding sequence, and the address spacer propagating toward the solid support.
  • An address polynucleotide can comprise an address barcode sequence, an address linker sequence, an address primer binding sequence an address spacer sequence, or any combination thereof.
  • An address polynucleotide can further comprise an affinity tag.
  • polynucleotide can be arranged in an order such that an address linker sequence is located at one end of the address polynucleotide.
  • An address polynucleotide can be arranged in an order such that it contains an address barcode upstream of the address linker sequence.
  • An address polynucleotide can comprise an address spacer sequence between the address barcode and the address linker sequence.
  • An address polynucleotide can be arranged in an order such that it contains an address primer binding sequence upstream of the address barcode.
  • An address polynucleotide can comprise an address spacer sequence between the address barcode and the address primer binding sequence.
  • An address polynucleotide can comprise an address spacer sequence between the address barcode and the address primer binding sequence.
  • An address polynucleotide can be arranged in an order such that an address spacer sequence is located upstream or downstream of the proximity primer binding sequence.
  • An address polynucleotide can be arranged in an order such that an address spacer sequence is located upstream of the proximity barcode sequence.
  • An address polynucleotide can be arranged in an order such that an address spacer sequence is located at one end of the address polynucleotide, for example, an end of the address polynucleotide that does not contain the address linker sequence.
  • an address polynucleotide can be arranged in an order of the address linker sequence, the address barcode, the address primer binding sequence, and the address spacer sequence.
  • an address polynucleotide can be arranged in an order of the address linker sequence, the address barcode, the address primer binding sequence, and the address spacer sequence propagating toward the solid surface.
  • an address polynucleotide can be arranged in the order of the address linker sequence, the address barcode, the address primer binding sequence, and the address spacer sequence from the 5' end to the 3' end.
  • an address polynucleotide can comprise a 5' end address linker sequence, a unique address barcode sequence, a reverse address primer binding sequence, and a 3 ' address spacer sequence attached to a solid support directly or indirectly through a linker (e.g., via a primary amine group attached to the 3 'end) in that order.
  • an address polynucleotide attached to a solid support can be arranged, propagating toward the solid support, in the order of the address linker sequence, the address barcode sequence, the address proximity primer binding sequence, and the address spacer sequence.
  • An address polynucleotide can comprise a plurality of address polynucleotides.
  • the plurality of address polynucleotides can be comprised by a plurality of discrete regions on a solid support.
  • an address polynucleotide can comprise a plurality of at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 address polynucleotides.
  • a plurality of address polynucleotide can comprise a plurality of at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 address polynucleotides comprised by a plurality of at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 discrete regions of a solid support.
  • An address polynucleotide can comprise an address linker sequence.
  • An address linker sequence is a sequence or end of an address polynucleotide that can be coupled to a proximity polynucleotide, (e.g., a proximity address linker sequence).
  • a proximity polynucleotide e.g., a proximity address linker sequence
  • an address linker sequence can be indirectly hybridized to a proximity polynucleotide through use of a splint polynucleotide.
  • an address linker sequence can be hybridized to a proximity polynucleotide directly.
  • an end of an address linker sequence can be ligated to an end of a proximity polynucleotide.
  • 3' end of an address polynucleotide comprising an address linker sequence can be ligated to a 5' end of a proximity polynucleotide (e.g., a proximity linker sequence).
  • An address linker sequence can be located at a terminus or an end of an address polynucleotide.
  • an address linker sequence can be a 3' terminus or end of an address polynucleotide.
  • An address linker sequence can be interposed between an end of an address polynucleotide and an address primer binding sequence of an address polynucleotide.
  • An address linker sequence can be located downstream of an address primer binding sequence.
  • an address linker sequence can be located 3' to an address primer binding sequence.
  • An address linker sequence can be located downstream of an address barcode sequence of an address polynucleotide. For example, an address linker sequence can be located 3' to an address barcode sequence. An address linker sequence can be located downstream of an address spacer sequence of an address polynucleotide. For example, an address linker sequence can be located 3' to an address spacer sequence.
  • An address linker sequence can be at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, or more consecutive nucleotides.
  • An address linker sequence can be a sequence of known length.
  • An address linker sequence of each address proximity polynucleotide of a plurality of address polynucleotides can be a unique or a same linker sequence.
  • any one address linker sequence of a plurality of address linker sequences can be a unique linker sequence.
  • each address linker sequence of a plurality of address linker sequences is the same sequence.
  • each address linker sequence of a plurality of address linker sequences can comprise a same sequence that hybridizes to a splint
  • each address linker sequence of a plurality of address linker sequences can comprise a same sequence that hybridizes to a same sequence of a splint polynucleotide.
  • each address linker sequence of a plurality of address linker sequences can comprise a same sequence that hybridizes to a same sequence of a splint polynucleotide, wherein the splint polynucleotide can hybridize to a proximity linker sequence (thus, coupling the address polynucleotide and the proximity polynucleotide).
  • each address polynucleotide can comprise the same address linker sequence.
  • an address linker sequence can be a universal address linker sequence.
  • An address linker sequence can comprise a randomly assembled sequence of nucleotides.
  • An address linker sequence can be a sequence of known length.
  • An address linker sequence can be a known sequence.
  • An address linker sequence can be a predefined sequence.
  • An address linker sequence can be an unknown sequence of known length.
  • An address linker sequence can be an unknown sequence of known length.
  • an address linker sequence is a universal sequence such that coupling of each address linker sequence of a plurality of address polynucleotides to a proximity polynucleotide can be carried out with a universal splint polynucleotide.
  • a universal splint polynucleotide that hybridizes to each of the address linker sequences can be utilized to couple all address polynucleotides to a proximity polynucleotide in a single reaction, simultaneously, and/or without bias for the coupling reaction.
  • An address polynucleotide can comprise an address barcode sequence or compliment thereof.
  • a barcode or barcode sequence relates to a natural or synthetic nucleic acid sequence comprised by a polynucleotide allowing for unambiguous identification of the polynucleotide and other sequences comprised by the polynucleotide having said barcode sequence.
  • an address barcode comprised by an address polynucleotide can allow for
  • the number of different barcode sequences theoretically possible can be directly dependent on the length of the barcode sequence; e.g., if a DNA barcode with randomly assembled adenine, thymidine, guanosine and cytidine nucleotides can be used, the theoretical maximal number of barcode sequences possible can be 1,048,576 for a length of ten nucleotides, and can be 1,073,741,824 for a length of fifteen nucleotides.
  • An address barcode sequence can be used to identify a specific target analyte.
  • An address barcode sequence can be barcoded to a target analyte in proximity to the address polynucleotide containing the address barcode.
  • An address barcode sequence can be barcoded to a discrete region of a solid support, such as a discrete region comprising a target analyte.
  • an address barcode sequence can be barcoded to a discrete region comprising a target analyte in proximity to the address polynucleotide, wherein the address polynucleotide is in the same discrete region as the target analyte.
  • An address barcode can be a unique barcode sequence.
  • any one address barcode of a plurality of address barcodes can be a unique barcode sequence.
  • An address barcode can be used to identify the target analyte to which it is barcoded from a plurality of target analytes ⁇ e.g., a plurality of different target analytes or same target analytes from different samples or sources).
  • An address barcode can be used to identify the region, location, or position of a target analyte on a solid support from a plurality of discrete regions, locations, or positions on the solid support.
  • An address barcode can be used to identify a target analyte on a solid support to which the address polynucleotide is in proximity from a plurality of target analytes on the solid support to which the address polynucleotide is not in proximity.
  • An address barcode can be used identify a target analyte that interacts with a binding moiety from a plurality of target analytes.
  • an address barcode can be used identify a target analyte from a plurality of target analytes and a binding moiety that interacted with the identified target analyte.
  • An address barcode can be barcoded to a target analyte exclusively.
  • An address barcode can be barcoded to a discrete region on a solid support exclusively.
  • An address barcode sequence can comprise a sequence of at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 40, 45, or 50 or more consecutive nucleotides.
  • An address polynucleotide can comprise two or more address barcode sequences or compliments thereof.
  • An address barcode sequence can comprise a randomly assembled sequence of nucleotides.
  • An address barcode sequence can be a degenerate sequence.
  • An address barcode sequence can be a known sequence.
  • An address barcode sequence can be a predefined sequence.
  • an address barcode sequence is a known, unique sequence that is barcoded a target analyte in a discrete region of a solid support such that a signal containing the address barcode sequence (e.g., a sequence read) or compliment thereof can be used to identify a target analyte of a plurality of target analytes that interacted with a binding moiety of a plurality of binding moieties.
  • a signal containing the address barcode sequence e.g., a sequence read
  • compliment thereof can be used to identify a target analyte of a plurality of target analytes that interacted with a binding moiety of a plurality of binding moieties.
  • An address primer binding sequence can be used as a primer binding site for a reaction, such as amplification or sequencing.
  • a primer binding sequence relates to a nucleic acid sequence that specifically hybridizes to a predefined amplification primer under conditions typicality used in PCR or other nucleic acid amplifying methods.
  • An address primer binding sequence can be a first primer binding sequence of a primer pair used for a reaction (e.g., amplification or sequencing).
  • an address primer binding sequence can be a forward or reverse primer binding site.
  • an address primer binding site can be a forward primer binding site and a proximity primer binding sequence can be a reverse primer binding sequence.
  • an address primer binding sequence is a universal primer binding sequence.
  • An address primer binding sequence and a proximity primer binding sequence can comprise melting temperatures that differ by no more than 6, 5, 4, 3, 2, or 1 degree Celsius.
  • the nucleotide sequence of a proximity primer binding sequence and an address primer binding sequence of an address polynucleotide can differ such that a polynucleotide that hybridizes to one does not hybridize to the other.
  • An address primer binding sequence can be located upstream of an address barcode.
  • an address primer binding sequence can be 3' to an address barcode or compliment thereof.
  • An address primer binding sequence can be located upstream of an address linker sequence.
  • an address primer binding sequence can be 3' to an address linker sequence.
  • An address primer binding sequence can be located upstream of a proximity linker sequence when the address polynucleotide is coupled to a proximity polynucleotide.
  • an address primer binding sequence can be 3 ' to a proximity linker sequence when the address polynucleotide is coupled to a proximity polynucleotide.
  • An address primer binding sequence can be located upstream of a proximity barcode sequence when the address polynucleotide is coupled to a proximity polynucleotide.
  • an address primer binding sequence can be 3' to a proximity barcode sequence when the address polynucleotide is coupled to a proximity polynucleotide.
  • a spacer ⁇ e.g., a spacer sequence
  • a spacer can also include natural or synthetic nucleic acid sequences, peptides, or other chemical entities, interposed between two amino acid sequences that do not naturally link the two polypeptide domains in nature.
  • An address polynucleotide can comprise an address spacer.
  • An address spacer sequence is a sequence used to increase the length of the address polynucleotide or to separate one or more of an address barcode, an address linker, an address primer binding site, and a solid support to which the address polynucleotide is attached, from each other.
  • an address spacer sequence can be interposed between an address primer binding sequence of an address polynucleotide and a solid support to which an end or other portion of the address polynucleotide is attached.
  • a spacer can be interposed between a primer binding sequence and a binding moiety of an address polynucleotide.
  • an address polynucleotide does not comprise a spacer sequence.
  • an address polynucleotide can be coupled to a solid support at an end of the address polynucleotide comprising an address primer binding site.
  • an address spacer is attached to a solid support.
  • an address spacer is located upstream of an address primer binding sequence.
  • an address spacer can be located 3' to an address primer binding sequence.
  • an address spacer is located downstream of an address primer binding sequence.
  • an address spacer can be located 5' to an address primer binding sequence.
  • an address spacer is located upstream of an address barcode.
  • an address spacer can be located 3' to an address barcode.
  • an address spacer is located downstream of an address barcode.
  • an address spacer sequence can be located 5' to an address barcode.
  • an address spacer is located upstream of an address linker sequence.
  • an address spacer sequence can be located 3 ' to an address linker sequence.
  • an address spacer is interposed between an address primer binding sequence and a solid support to which the address polynucleotide is attached.
  • an address spacer sequence can be located 3 ' to an address primer binding sequence and a 3 ' end of the address polynucleotide attached to a solid support.
  • an address spacer is interposed between an address primer binding sequence and an address barcode.
  • an address spacer sequence can be located 5' to an address primer binding sequence and 3 ' to an address barcode.
  • an address spacer is interposed between an address linker sequence and an address barcode.
  • an address spacer sequence can be located 5' to a proximity barcode and 3 ' to a proximity linker sequence.
  • An address spacer sequence can be at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 200, 250, 300, 400, 500 or more consecutive nucleotides.
  • An address spacer sequence can comprise a randomly assembled sequence of nucleotides.
  • An address spacer sequence can be a sequence of known length.
  • An address spacer sequence can be a known sequence.
  • An address spacer sequence can be a predefined sequence.
  • An address spacer sequence can be an unknown sequence of known length.
  • An address spacer sequence can be a known sequence of known length.
  • An address polynucleotide can be coupled to a solid support.
  • an address polynucleotide can be immobilized on a solid substrate.
  • An address polynucleotide can be coupled to the solid support through covalent or non-covalent interactions.
  • address polynucleotide can be coupled to the solid support non-covalently through hydrophobic bonding, hydrogen bonding, Van der Waals interactions, ionic bonding, etc.
  • an address polynucleotide is coupled reversibly.
  • an address polynucleotide is coupled irreversibly.
  • An address polynucleotide can be coupled to a solid support at any internal position along its length or at either the 5' or 3 ' position.
  • a solid support-coupled address polynucleotide is then able to undergo interactions at positions distant from the solid support.
  • the coupling allows removal of undesired molecules on the solid support (e.g., molecules that non-specifically interact with the solid support or components on the solid support) by washing.
  • An address polynucleotide can be coupled a solid support through a functional group (e.g., a reactive group).
  • An address polynucleotide can comprise any suitable functional group for coupling to a solid support. Any suitable methods and reagent for modifying the ends of polynucleotides and/or for incorporating bases modified with functional groups into polynucleotides can be used. (See, e.g., Atherton et al, (1989) Tet Lett 30(15): 1927-30;
  • an address polynucleotide can be phosphorylated at the 5'-terminus ⁇ e.g., with phosphokinase) and covalently attached to an amino-activated substrate through a phosphoramidate or phosphodiester linkage.
  • an address polynucleotides modified is modified at its 3'- or 5 '-termini with a primary amino group and coupled to a carboxy-activated substrate.
  • the functional group may be selected to covalently or non- covalently couple the address polynucleotide to the surface. Coupling can be at an internal position or at either the 5' or 3' position of an address polynucleotide.
  • a surface of a solid support can be coated with a functional group and an address polynucleotide can be attached to the solid support through the functional group.
  • a solid support can be coated with a first functional group and an address polynucleotide comprising a second functional group can be attached to the solid support by binding or reacting the first and second functional groups.
  • a surface of a solid support can be coated with streptavidin and a biotinylated address polynucleotide can be attached thereto.
  • address polynucleotide can comprise a tag.
  • the tag is an affinity tag. Examples of such affinity tags include, but are not limited to,
  • Glutathione-S-transferase GST
  • Maltose binding protein MBP
  • Green Fluorescent Protein GFP
  • AviTag a peptide allowing biotinylation by the enzyme BirA and so the protein can be isolated by streptavidin
  • Calmodulin-tag a peptide bound by the protein calmodulin
  • polyglutamate tag a peptide binding efficiently to anion-exchange resin such as Mono-Q
  • FL AG-tag a peptide recognized by an antibody
  • HA-tag a peptide recognized by an antibody
  • His tag generally 5-10 histidines which are bound by a nickel or cobalt chelate
  • Myc-tag a short peptide recognized by an antibody
  • S-tag, SBP-tag a peptide which binds to streptavidin
  • Softag 1, Strep-tag a peptide which binds to streptavidin or the modified streptavidin called streptactin
  • TC tag a te
  • a surface of a solid support can be coated with an affinity ligand and an address polynucleotide comprising an affinity tag can be attached thereto.
  • address polynucleotides are synthesized directly on a solid substrate (e.g., a hydroxy-activated solid surface), such as by using phosphoramidite synthesis reagents, photoprotected phosphoramidites, or photolithographic methods (See, e.g., U.S. Pat. No. 5,744,305).
  • address polynucleotides can be covalently attached to a substrate via its 3 '-terminus via a phosphodiester linkage.
  • polynucleotide can be deposited on a solid surface (e.g., an array or bead) by any suitable technique.
  • an address polynucleotide or functional group may be deposited as a self-assembled monolayer, modification of the solid substrate by chemical reaction (See, e.g., U.S. Pat. No. 6,444,254), reactive plasma etching, corona discharge treatment, a plasma deposition process, spin coating, dip coating, spray painting, deposition, printing, stamping, etc.
  • An address polynucleotide or functional group may be deposited as a continuous layer or as a discontinuous (e.g., patterned) layer.
  • An address polynucleotide or functional group may be deposited via diffusion, adsorption/ab sorption, or covalent cross-linking.
  • address polynucleotides or functional groups are spotted onto a glass surface.
  • a solid support is modified to achieve better binding capacity.
  • a glass surface may be coated with a thin nitrocellulose membrane or poly-L-lysine such that an address
  • polynucleotide can be passively adsorbed onto the modified surface via non-specific
  • a surface of the solid substrate can be coated with streptavidin and a biotinylated address polynucleotide can be coupled thereto.
  • the surface of a solid support can be coated with an affinity ligand.
  • an affinity ligand can include, but is not limited to an antigen, an antibody, an antibody fragment, glutathione, calmodulin, biotin, streptavidin, streptactin, amylose, an anion-exchange resin such as Mono-Q, FiAsH and ReAsH biarsenicai compounds, pilin-C protein, SpyCatcher protein or a metal chelate and an address polynucleotide can be coupled thereto.
  • the metal chelate can include but is not limited to nickel, cobalt, zinc, mercury, cupper or iron chelate.
  • the solid support can be coated entirely. In some embodiments, the solid support can be coated partially.
  • Examples of solid surface materials and corresponding functional groups include gold, silver, copper, cadmium, zinc, palladium, platinum, mercury, lead, iron, chromium, manganese, tungsten, and any alloys thereof.
  • Exemplary functional groups of solid surfaces include sulfur- containing functional groups such as thiols, sulfides, disulfides (e.g., -SR or -SSR where R is H, alkyl, or aryl), and the like; doped or undoped silicon with silanes and chlorosilanes (e.g., - SiR 2 Cl where R is H, alkyl, or aryl); metal oxides (e.g., silica, alumina, quartz, glass, and the like) with carboxylic acids; platinum and palladium with nitrites and isonitriles; copper with hydroxamic acids; benzophenones; acid chlorides; anhydrides; epoxides; sulfonyl groups;
  • Address polynucleotides can optionally be coupled to a solid support through one or more bifunctional linkers (e.g., the linkers comprising one functional group capable of forming a linkage with a solid substrate and another functional group capable of forming a linkage with another linker molecule or the address polynucleotides.
  • linkers may be long or short, flexible or rigid, charged or uncharged, and/or hydrophobic or hydrophilic.
  • an address polunucleotide can be coupled to a solid support by a linker.
  • a linker can be a linker described herein.
  • a linker can be a protein or a fragment thereof, for example BSA or the like.
  • a proximity probe comprises a binding moiety and a proximity polynucleotide.
  • a proximity barcode of the proximity probe's proximity polynucleotide can be used to identify the one or more binding moieties that the proximity probe comprises.
  • the proximity polynucleotide is attached covalently or non-covalently to the binding moiety.
  • the proximity polynucleotide is an extension of the binding moiety, for example, when the binding moiety is a polynucleotide.
  • a proximity probe comprises a single binding moiety.
  • proximity probes are multivalent proximity probes.
  • Multivalent proximity probes comprise at least two analyte-binding domains conjugated to one or more nucleic acid(s).
  • multivalent proximity probes may comprise at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 200, 500, or 1,000 analyte-binding domains conjugated to at least one, or more than one, nucleic acid (e.g., a proximity polynucleotide).
  • a proximity probe comprises a single proximity polynucleotide. In some embodiments, a proximity probe comprises 2 or more proximity polynucleotides. For example, a proximity probe can comprise at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 200, 500, or 1,000 proximity polynucleotides conjugated to at least one, or more than one, binding moiety. In some embodiments, a proximity probe comprises 2 or more proximity
  • polynucleotides containing a same proximity barcode sequence polynucleotides containing a same proximity barcode sequence.
  • An analyte-binding moiety also referred to as a binding moiety (or domain) is the region, molecule, domain, portion, fragment, or moiety of a proximity probe that binds to a target analyte.
  • a binding moiety confers the ability to bind or specifically bind to a given target analyte.
  • a binding moiety does not substantially interact with an address polynucleotide.
  • an analyte-binding moiety does not prevent coupling of the proximity polynucleotide to an address polynucleotide in proximity thereto.
  • a binding moiety does not substantially interact with an address polynucleotide.
  • a binding moiety is a molecule that can contain a nucleic acid, or to which a nucleic acid can be attached, without substantially abolishing the binding of the analyte-binding moiety to its target analyte.
  • An analyte-binding moiety can be a nucleic acid molecule or can be proteinaceous. Binding moieties include, but are not limited to, RNAs, DNAs, RNA-DNA hybrids, small molecules (e.g., drugs), aptamers, polypeptides, proteins, antibodies, viruses, virus particles, cells, fragments thereof, and combinations thereof. (See, e.g., Fredriksson et al., (2002) Nat Biotech 20:473-77; Gullberg et al, (2004) PNAS, 101 :8420-24).
  • a binding moiety can be a single-stranded RNA, a double- stranded RNA, a single-stranded DNA, a double- stranded DNA, a DNA or RNA comprising one or more double stranded regions and one or more single stranded regions, an RNA-DNA hybrid, a small molecule, an aptamer, a
  • polypeptide a protein, an antibody, an antibody fragment, a mixture of antibodies, a virus particle, a cell, or any combination thereof.
  • a binding moiety is a polypeptide, a protein, or any fragment thereof.
  • a binding moiety can be a purified polypeptide, an isolated polypeptide, a fusion tagged polypeptide, a polypeptide attached to or spanning the membrane of a cell or a virus or virion, a cytoplasmic protein, an intracellular protein, an extracellular protein, a kinase, a phosphatase, an aromatase, a helicase, a protease, an oxidoreductase, a reductase, a transferase, a hydrolase, a lyase, an isomerase, a glycosylase, a extracellular matrix protein, a ligase, an ion transporter, a channel, a pore, an apoptotic protein, a cell adhesion protein, a pathogenic protein, an aberrantly expressed protein, an transcription factor,
  • a binding moiety is a heterologous polypeptide.
  • a binding moiety is a protein overexpressed in a cell using molecular techniques, such as transfection.
  • a binding moiety is recombinant polypeptide.
  • a binding moiety can comprise samples produced in bacterial (e.g., E. coli), yeast, mammalian, or insect cells (e.g., proteins overexpressed by the organisms).
  • a binding moiety is a polypeptide containing a mutation, insertion, deletion, or polymorphism.
  • a binding moiety is an antigen, such as a polypeptide used to immunize an organism or to generate an immune response in an organism, such as for antibody production.
  • a binding moiety is an antibody or fragment thereof.
  • An antibody can specifically bind to a particular spatial and polar organization of another molecule.
  • An antibody can be monoclonal, polyclonal, or a recombinant antibody, and can be prepared by techniques that are well known in the art such as immunization of a host and collection of sera (polyclonal) or by preparing continuous hybrid cell lines and collecting the secreted protein (monoclonal), or by cloning and expressing nucleotide sequences, or mutagenized versions thereof, coding at least for the amino acid sequences required for specific binding of natural antibodies.
  • a naturally occurring antibody can be a protein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds.
  • Each heavy chain can be comprised of a heavy chain variable region (V H ) and a heavy chain constant region.
  • the heavy chain constant region can be comprised of three domains, C HI , Cm and C H3 -
  • Each light chain can be comprised of a light chain variable region (V L ) and a light chain constant region.
  • the light chain constant region can be comprised of one domain, C L .
  • the V H and V L regions can be further subdivided into regions of hypervariability, termed complementary determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementary determining regions
  • Each V H and V L can be composed of three CDRs and four FRs arranged from amino-terminus to carboxy-terminus in the following order: FRi, CDRi, FR 2 , CDR 2 , FR 3 , CDR 3 , and FR4.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Cl q) of the classical complement system.
  • the antibodies can be of any isotype (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., lgGi, lgG 2 , lgG 3 , lgG 4 , IgAi and lgA 2 ), subclass or modified version thereof.
  • Antibodies may include a complete immunoglobulins or fragments thereof.
  • An antibody fragment can refer to one or more fragments of an antibody that retain the ability to specifically bind to a target analyte, such as an antigen.
  • aggregates, polymers, and conjugates of immunoglobulins or their fragments can be used where appropriate so long as binding affinity for a particular molecule is maintained.
  • antibody fragments include a Fab fragment, a monovalent fragment consisting of the V L , V H , C L and C HI domains; a F(ab) 2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; an Fd fragment consisting of the V H and C HI domains; an Fv fragment consisting of the V L and V H domains of a single arm of an antibody; a single domain antibody (dAb) fragment (Ward et al, (1989) Nature 341 :544-46), which consists of a V H domain; and an isolated CDR and a single chain Fragment (scFv) in which the V L and V H regions pair to form monovalent molecules (known as single chain Fv (scFv); See, e.g., Bird et al, (1988) Science 242:423-26; and Huston et al, (1988) PNAS 85:5879-83).
  • scFv single
  • antibody fragments include Fab, F(ab) 2 , scFv, Fv, dAb, and the like.
  • V L and V H are coded for by separate genes, they can be joined, using recombinant methods, by an artificial peptide linker that enables them to be made as a single protein chain.
  • single chain antibodies include one or more antigen binding moieties.
  • Antibodies can be human, humanized, chimeric, isolated, dog, cat, donkey, sheep, any plant, animal, or mammal.
  • a binding moiety is a polymeric form of ribonucleotides and/or deoxyribonucleotides (adenine, guanine, thymine, or cytosine), such as DNA or RNA ⁇ e.g., mRNA).
  • DNA includes double-stranded DNA found in linear DNA molecules ⁇ e.g., restriction fragments), viruses, plasmids, and chromosomes.
  • a polynucleotide binding moiety is single-stranded, double stranded, small interfering RNA (siRNA), messenger RNA (mRNA), transfer RNA (tRNA), a chromosome, a gene, a noncoding genomic sequence, genomic DNA ⁇ e.g., fragmented genomic DNA), a purified polynucleotide, an isolated polynucleotide, a hybridized polynucleotide, a transcription factor binding site, mitochondrial DNA, ribosomal RNA, a eukaryotic polynucleotide, a prokaryotic polynucleotide, a synthesized polynucleotide, a ligated polynucleotide, a recombinant polynucleotide, a polynucleotide containing a nucleic acid analogue, a methylated polynucleotide, a demethylated polynucleo
  • a binding moiety is a polynucleotide comprising double stranded region and an end that is not double stranded ⁇ e.g., a 5' or 3' overhang region.
  • a binding moiety is a polynucleotide comprising double stranded region that is hybridized and a double stranded end comprising two non-hybridized single strands ⁇ e.g., two single stranded overhangs at an end such as a "Y- adapter" depicted in FIG. 28).
  • a binding moiety is a polynucleotide comprising a double stranded region that is hybridized and two double stranded ends each comprising two non-hybridized single strands ⁇ e.g., both ends comprise two single stranded overhangs, such as a polynucleotide comprising two "Y-adapters" as depicted in FIG. 28).
  • a binding moiety is a recombinant polynucleotide.
  • a binding moiety is a heterologous polynucleotide.
  • a binding moiety can comprise polynucleotides produced in bacterial ⁇ e.g., E.
  • a binding moiety is a polynucleotide containing a mutation, insertion, deletion, or polymorphism.
  • a binding moiety is an aptamer.
  • An aptamer is an isolated nucleic acid molecule that binds with high specificity and affinity to a target analyte, such as a protein.
  • An aptamer is a three dimensional structure held in certain conformation(s) that provides chemical contacts to specifically bind its given target.
  • aptamers are nucleic acid based molecules, there is a fundamental difference between aptamers and other nucleic acid molecules such as genes and mRNA. In the latter, the nucleic acid structure encodes information through its linear base sequence and thus this sequence is of importance to the function of information storage. In complete contrast, aptamer function, which is based upon the specific binding of a target molecule, is not entirely dependent on a conserved linear base sequence (a non-coding sequence), but rather a particular secondary/tertiary/quaternary structure. Any coding potential that an aptamer may possess is entirely fortuitous and plays no role whatsoever in the binding of an aptamer to its cognate target.
  • Aptamers must also be differentiated from the naturally occurring nucleic acid sequences that bind to certain proteins. These latter sequences are naturally occurring sequences embedded within the genome of the organism that bind to a specialized sub-group of proteins that are involved in the transcription, translation, and transportation of naturally occurring nucleic acids (e.g., nucleic acid-binding proteins).
  • Aptamers on the other hand are short, isolated, non-naturally occurring nucleic acid molecules. While aptamers can be identified that bind nucleic acid-binding proteins, in most cases such aptamers have little or no sequence identity to the sequences recognized by the nucleic acid- binding proteins in nature. More importantly, aptamers can bind virtually any protein (not just nucleic acid-binding proteins) as well as almost any target of interest including small molecules, carbohydrates, peptides, etc. For most targets, even proteins, a naturally occurring nucleic acid sequence to which it binds does not exist.
  • aptamers are capable of specifically binding to selected targets and modulating the targets activity or binding interactions, e.g., through binding, aptamers may block their target's ability to function.
  • the functional property of specific binding to a target is an inherent property an aptamer.
  • a typical aptamer is 6-35 kDa in size (20-100 nucleotides), binds its target with micromolar to sub-nanomolar affinity, and may discriminate against closely related targets (e.g., aptamers may selectively bind related proteins from the same gene family). Aptamers are capable of using commonly seen intermolecular interactions such as hydrogen bonding, electrostatic
  • An aptamer can comprise a molecular stem and loop structure formed from the hybridization of complementary polynucleotides that are covalently linked (e.g., a hairpin loop structure).
  • the stem comprises the hybridized polynucleotides and the loop is the region that covalently links the two complementary polynucleotides.
  • a binding moiety is a small molecule.
  • a small molecule can be a macrocyclic molecule, an inhibitor, a drug, or chemical compound.
  • a small molecule contains no more than five hydrogen bond donors.
  • a small molecule contains no more than ten hydrogen bond acceptors.
  • a small molecule has a molecular weight of 500 Daltons or less.
  • a small molecule has a molecular weight of from about 180 to about 900 Daltons.
  • a small molecule contains an octanol-water partition coefficient lop P of no more than five.
  • a small molecule has a partition coefficient log P of from -0.4 to 5.6. In some embodiments, a small molecule has a molar refractivity of from 40 to 130. In some embodiments, a small molecule contains from about 20 to about 70 atoms. In some embodiments, a small molecule has a polar surface area of 140 Angstroms 2 or less.
  • a binding moiety is a cell.
  • a binding moiety can be an intact cell, a cell treated with a compound (e.g., a drug), a fixed cell, a lysed cell, or any combination thereof.
  • a binding moiety is a single cell.
  • a binding moiety is a plurality of cells.
  • a binding moiety is a plurality of binding moieties, such as a mixture or library of binding moieties.
  • a binding moiety is a plurality of different binding moieties.
  • a binding moiety can comprise a plurality of at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 binding moieties.
  • a binding moiety can comprise a plurality of at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000,
  • a binding moiety is a plurality of different binding moieties that represents an entire, or portion of, a proteome of an organism.
  • a binding moiety can comprise a plurality of proteins representing at least about 0.5%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 10%, 80%), 90%), or 100% of an organism's proteome.
  • a binding moiety can comprise a plurality of antibodies that bind to at least about 0.5%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, or 100% of the proteins of an organism's proteome.
  • the proteome can be a bacterial, viral, fungal proteome.
  • the proteome can be of an insect or mammal, such as a mouse, rat, rabbit, cat, dog, monkey, goat, or human. In some embodiments, the proteome is human.
  • the proteome can be of an animal or a non-human animal, such as a bovine, avian, canine, equine, feline, ovine, porcine, or primate.
  • the proteome can be of a mammal, such as a mouse, rat, rabbit, cat, dog monkey, or goat.
  • a proximity polynucleotide is a region, molecule, domain, portion, fragment, or moiety of a proximity probe that can be coupled to an address polynucleotide when in proximity to the address polynucleotide.
  • a proximity polynucleotide can be coupled directly or indirectly to a binding moiety.
  • a proximity polynucleotide can be coupled covalently or non-covalently to a binding moiety. In preferred embodiments, a proximity polynucleotide does not substantially interact with a target analyte.
  • a proximity polynucleotide interacts with, or can be coupled to, an address polynucleotide when the proximity polynucleotide is in proximity to the address polynucleotide.
  • a proximity polynucleotide interacts with, or can be coupled to, an address polynucleotide when the binding moiety of a proximity probe binds to a target analyte in proximity to the address polynucleotide.
  • a proximity polynucleotide can comprise a plurality of proximity polynucleotides.
  • the plurality of proximity polynucleotides can be comprised by a plurality of proximity probes.
  • a proximity polynucleotide can comprise a plurality of at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 proximity polynucleotides.
  • a plurality of at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 proximity polynucleotides can be comprised by a plurality of at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 proximity probes.
  • a proximity polynucleotide can comprise a proximity barcode sequence, a proximity linker sequence, a proximity primer binding sequence, a proximity spacer sequence, or any combination thereof.
  • a proximity polynucleotide can be arranged in an order such that a proximity linker sequence is located at one end of the proximity polynucleotide.
  • a proximity polynucleotide can be arranged in an order such that it contains a proximity barcode upstream of the proximity linker sequence.
  • a proximity polynucleotide can comprise a proximity linker sequence between the proximity barcode and the proximity linker sequence.
  • a proximity polynucleotide can be arranged in an order such that it contains a proximity primer binding sequence upstream of the proximity barcode.
  • a proximity polynucleotide can comprise a proximity linker sequence between the proximity barcode and the proximity primer binding sequence.
  • a proximity polynucleotide can comprise a proximity linker sequence between the proximity barcode and the proximity primer binding sequence.
  • a proximity polynucleotide can be arranged in an order such that a proximity spacer sequence is located upstream or
  • a proximity polynucleotide can be arranged in an order such that a proximity spacer sequence is located upstream of the proximity barcode sequence.
  • a proximity polynucleotide can be arranged in an order such that a proximity spacer sequence is located at one end of the proximity polynucleotide, for example, an end of the proximity polynucleotide that does not contain the proximity linker sequence.
  • a proximity polynucleotide can be arranged in an order of the proximity linker sequence, the proximity barcode, the proximity primer binding sequence, and the proximity spacer sequence.
  • a proximity polynucleotide can be arranged in an order of the proximity linker sequence, the proximity barcode, the proximity primer binding sequence, and the proximity spacer sequence propagating toward the binding moiety.
  • a proximity polynucleotide can be arranged in an order such that a proximity spacer sequence is located upstream of the proximity barcode sequence.
  • a proximity polynucleotide can be arranged in an order such
  • a proximity polynucleotide can be arranged in the order of the proximity linker sequence, the proximity barcode, the proximity primer binding sequence, and the proximity spacer sequence from the 5' end to the 3' end.
  • a proximity polynucleotide can comprise a 5' end proximity linker sequence, a unique proximity barcode sequence, a reverse proximity primer binding sequence, and a 3' proximity spacer sequence attached to a binding moiety (e.g., via a primary amine group attached to the 3 'end) in that order.
  • a proximity polynucleotide attached to a binding moiety can be arranged, propagating toward the binding moiety, in the order of the proximity linker, the proximity barcode, the proximity primer binding site, and the proximity spacer.
  • a proximity polynucleotide can comprise a proximity linker sequence.
  • a proximity linker sequence is a sequence or end of a proximity polynucleotide that can be coupled to an address polynucleotide, (e.g., an address linker sequence).
  • a proximity linker sequence can be indirectly hybridized to an address polynucleotide through use of a splint polynucleotide.
  • a proximity linker sequence can be hybridized to an address polynucleotide directly.
  • an end of a proximity linker sequence can be ligated to an end of an address polynucleotide.
  • 3' end of a proximity polynucleotide comprising a proximity linker sequence can be ligated to a 5' end of an address polynucleotide (e.g., an address linker sequence).
  • a proximity linker sequence can be located at a terminus or an end of a proximity polynucleotide.
  • a proximity linker sequence can be a 3' terminus or end of a proximity polynucleotide.
  • a proximity linker sequence can be interposed between an end of a proximity polynucleotide and a proximity primer binding sequence of a proximity polynucleotide.
  • a proximity linker sequence can be located downstream of a proximity primer binding sequence.
  • a proximity linker sequence can be located 3' to a proximity primer binding sequence.
  • a proximity linker sequence can be located downstream of a proximity barcode sequence of a proximity polynucleotide.
  • a proximity linker sequence can be located 3' to a proximity barcode sequence.
  • a proximity linker sequence can be located downstream of a proximity spacer sequence of a proximity polynucleotide.
  • a proximity linker sequence can be located 3' to a proximity spacer sequence.
  • a proximity linker sequence can be at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, or more consecutive nucleotides.
  • a proximity linker sequence can be a sequence of known length.
  • a proximity linker sequence of each proximity polynucleotide of a plurality of proximity probes can be a unique or a same linker sequence.
  • any one proximity linker sequence of a plurality of proximity linker sequences can be a unique linker sequence.
  • each proximity linker sequence of a plurality of proximity linker sequences is the same sequence.
  • each proximity linker sequence of a plurality of proximity linker sequences can comprise a same sequence that hybridizes to a splint
  • each proximity linker sequence of a plurality of proximity linker sequences can comprise a same sequence that hybridizes to a same sequence of a splint polynucleotide.
  • each address linker sequence of a plurality of address linker sequences can comprise a same sequence that hybridizes to a same sequence of a splint polynucleotide, wherein the splint polynucleotide can hybridize to a proximity linker sequence (thus, coupling the address polynucleotide and the proximity polynucleotide).
  • each proximity polynucleotide can comprise the same proximity linker sequence.
  • a proximity linker sequence can be a universal sequence.
  • a proximity linker sequence can comprise a randomly assembled sequence of nucleotides.
  • a proximity linker sequence can be a sequence of known length.
  • a proximity linker sequence can be a known sequence.
  • a proximity linker sequence can be a predefined sequence.
  • a proximity linker sequence can be an unknown sequence of known length.
  • a proximity linker sequence can be an known sequence of known length.
  • a proximity linker sequence is a universal sequence such that coupling of each proximity linker sequence of a plurality of proximity probes to an address polynucleotide can be carried out with a universal splint polynucleotide.
  • a universal splint polynucleotide that hybridizes to each of the proximity linker sequences can be utilized to couple all proximity probes to an address polynucleotide in a single reaction, simultaneously, and/or without bias for the coupling reaction.
  • a proximity polynucleotide can comprise a proximity barcode sequence or compliment thereof.
  • a proximity barcode can allow for identification of a proximity probe comprising the proximity barcode.
  • a proximity barcode can allow for identification of a binding moiety to which the proximity barcode is attached.
  • a proximity barcode can be used to identify a binding moiety from a plurality of binding moieties that binds to a target analyte.
  • a proximity barcode can be barcoded to a proximity probe exclusively.
  • a proximity barcode can be barcoded to a binding moiety exclusively.
  • a proximity barcode sequence can be barcoded to a specific binding moiety.
  • a proximity barcode can be a unique barcode sequence.
  • any one proximity barcode of a plurality of proximity barcodes can be a unique barcode sequence.
  • the number of different barcode sequences theoretically possible can be directly dependent on the length of the barcode sequence. For example, if a DNA barcode with randomly assembled adenine, thymidine, guanosine and cytidine nucleotides can be used, the theoretical maximal number of barcode sequences possible can be 1,048,576 for a length of ten nucleotides, and can be 1,073,741,824 for a length of fifteen nucleotides.
  • a proximity barcode sequence can comprise a sequence of at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 40, 45, or 50 or more consecutive nucleotides.
  • a proximity polynucleotide can comprise two or more proximity barcode sequences or compliments thereof.
  • a proximity barcode sequence can comprise a randomly assembled sequence of nucleotides.
  • a proximity barcode sequence can be a degenerate sequence.
  • a proximity barcode sequence can be a known sequence.
  • a proximity barcode sequence can be a predefined sequence.
  • a proximity barcode sequence is a known, unique sequence that is barcoded to a binding moiety to which it is coupled such that a signal containing the proximity barcode (e.g., a sequence read) or compliment thereof can be used to identify a binding moiety of a plurality of binding moieties that interacted with a target analyte of a plurality of target analytes.
  • a signal containing the proximity barcode e.g., a sequence read
  • compliment thereof can be used to identify a binding moiety of a plurality of binding moieties that interacted with a target analyte of a plurality of target analytes.
  • a proximity primer binding sequence can be used as a primer binding site for a reaction, such as amplification or sequencing.
  • a proximity primer binding sequence can be a first primer binding sequence for a pair of primers used for a reaction, such as amplification or sequencing.
  • a proximity primer binding sequence can be a forward primer binding site.
  • a proximity primer binding site can be a reverse primer binding site.
  • a proximity primer binding site can be a forward primer binding site and an address primer binding sequence can be a reverse primer binding sequence.
  • a proximity primer binding sequence is a universal primer binding sequence.
  • a proximity primer binding sequence and an address primer binding sequence can comprise melting temperatures that differ by no more than 6, 5, 4, 3, 2, or 1 degree Celsius.
  • the nucleotide sequence of a proximity primer binding sequence and an address primer binding sequence of an address polynucleotide can differ such that a polynucleotide that hybridizes to the proximity primer binding sequence does not hybridize to the address primer binding sequence.
  • the nucleotide sequence of a proximity primer binding sequence and an address primer binding sequence of an address polynucleotide can differ such that a polynucleotide that hybridizes to the address primer binding sequence does not hybridize to the proximity primer binding sequence.
  • a proximity primer binding sequence can be located upstream of an address barcode.
  • a proximity primer binding sequence can be 5' to a proximity barcode.
  • a proximity primer binding sequence can be located upstream of a proximity linker sequence.
  • a proximity primer binding sequence can be 5' to a proximity linker sequence.
  • a proximity primer binding sequence can be located upstream of an address linker sequence when the proximity polynucleotide is coupled to an address polynucleotide.
  • a proximity primer binding sequence can be 5' to an address linker sequence when the proximity polynucleotide is coupled to an address polynucleotide.
  • a proximity primer binding sequence can be located upstream of an address barcode sequence when the proximity polynucleotide is coupled to an address polynucleotide.
  • a proximity primer binding sequence can be 5' to an address barcode sequence when the proximity polynucleotide is coupled to an address polynucleotide.
  • a proximity polynucleotide can comprise a proximity spacer sequence.
  • a proximity spacer sequence is a sequence used to increase the length of the proximity polynucleotide or to separate one or more of a proximity barcode, proximity linker, a proximity primer binding site, and a binding moiety from each other.
  • a proximity polynucleotide does not comprise a proximity spacer sequence.
  • a proximity polynucleotide can be coupled to a binding moiety at an end of the proximity polynucleotide comprising a proximity primer binding site.
  • a proximity spacer sequence is attached to a binding moiety of a proximity probe.
  • a proximity spacer is located upstream of a proximity primer binding sequence.
  • a proximity spacer sequence can be located 5' to a proximity primer binding sequence.
  • a proximity spacer is located downstream of a proximity primer binding sequence.
  • a proximity spacer sequence can be located 3 ' to a proximity primer binding sequence.
  • a proximity spacer is located upstream of a proximity barcode.
  • a proximity spacer sequence can be located 5' to a proximity barcode.
  • a proximity spacer is located downstream of a proximity barcode.
  • a proximity spacer sequence can be located 3 ' to a proximity barcode.
  • a proximity spacer is located upstream of a proximity linker sequence.
  • a proximity spacer sequence can be located 5' to a proximity linker sequence.
  • a proximity spacer is interposed between a proximity primer binding sequence and a binding moiety of a proximity probe.
  • a proximity spacer sequence can be located 5' to a proximity primer binding sequence and 5' end of the proximity polynucleotide containing the proximity linker sequence can be attached to a binding moiety of a proximity polynucleotide.
  • a proximity spacer is interposed between a proximity primer binding sequence and a proximity barcode.
  • a proximity spacer sequence can be located 3 ' to a proximity primer binding sequence and 5' to a proximity barcode.
  • a proximity spacer is interposed between a proximity linker sequence and a proximity barcode.
  • a proximity spacer sequence can be located 3' to a proximity barcode and 5' to a proximity linker sequence.
  • a proximity spacer sequence can be at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 200, 250, 300, 400, 500 or more consecutive nucleotides.
  • a proximity spacer sequence can comprise a randomly assembled sequence of nucleotides.
  • a proximity spacer sequence can be a sequence of known length.
  • a proximity spacer sequence can be a known sequence.
  • a proximity spacer sequence can be a predefined sequence.
  • a proximity spacer sequence can be an unknown sequence of known length.
  • a proximity spacer sequence can be a known sequence of known length.
  • a binding moiety can be coupled directly or indirectly (e.g., via a linker) to a proximity polynucleotide.
  • a binding moiety can be coupled covalently (e.g. via chemical cross-linking) or non-covalently (e.g., via streptavidin-biotin) to a proximity polynucleotide.
  • proximity probes The design and preparation of proximity probes is widely described in the art, for example various different binding moieties which may be used, the design of proximity polynucleotides for proximity ligation assays, and the coupling of such o proximity polynucleotides to the binding moieties to form the proximity probes.
  • the details and principles described in the art may be applied to the design of the proximity probes for use in the methods of the invention (See, e.g., WO2007107743, and U. S. Pat. Nos. 7,306,904 and 6,878,515).
  • a direct coupling reaction between a proximity polynucleotide and a binding moiety may be utilized, for example, where each possesses a functional group (e.g., a substituent or chemical handle) capable of reacting with a functional group on the other.
  • Functional groups may be present on the proximity polynucleotide or binding moiety, or introduced onto these components (e.g. via oxidation reactions, reduction reactions, cleavage reactions and the like).
  • Functional groups of an antibody or a polypeptide that can be used for coupling to a proximity polynucleotide include, but are not limited to carbohydrates, thiol groups (HS-) of amino acids, amine groups (H 2 N-) of amino acids, and carboxy groups of amino acids.
  • thiol groups can be reacted with a thiol-reactive group to form a thioether or disulfide.
  • free thiol groups of proteins may be introduced into proteins by thiolation or splitting of disulfides in native cysteine residues.
  • an amino group e.g., of an amino-terminus or an omega amino group of a lysine residue
  • an electrophilic group e.g., an activated carboxy group
  • a carboxy group e.g., a carboxy-terminus or a carboxy group of a diacidic alpha amino acid
  • Other exemplary functional groups include, e.g., SPDP, carbodiimide, glutaraldehyde, and the like.
  • a proximity polynucleotide is covalently coupled to a binding moiety using a commercial kit ("All-in-One Antibody-Oligonucleotide Conjugation Kit"; Solulink, Inc.).
  • a 3 '-amino- proximity polynucleotide can be derivatized with Sulfo-S-4FB.
  • a binding moiety can be modified with an S-HyNic group.
  • the HyNic-modified binding moiety can be reacted with the 4FB -modified proximity
  • the polynucleotide to yield a bis-arylhydrazone mediated proximity probe.
  • Excess 4FB-modified proximity polynucleotide can be further removed via a magnetic affinity matrix.
  • the overall binding moiety recovery can be at least about 95%, 96%, 97%, 98%, 99%, or 100% free of HyNic-modified binding moiety and 4FB-modified proximity polynucleotide.
  • the bis- arylhydrazone bond can be stable to both heat (e.g., 94° C) and pH (e.g., 3-10).
  • linking groups may be chosen to provide for covalent attachment or non-covalent attachment of the binding domain and proximity polynucleotide through the linking group.
  • suitable linkers are known t in the art.
  • the linker is at least about 50 or 100 Daltons 100 Daltons. In some embodiments, the linker is at most about 300; 500; 1,000; 10,000, or 100,000 Daltons.
  • a linker can comprise a functional group at either end with a reactive functionality capable of bonding to the proximity polynucleotide.
  • a linker can comprise a functional group at either end with a reactive functionality capable of bonding to the binding moiety.
  • Functional groups may be present on the proximity polynucleotide, binding moiety, and/or linker, or introduced onto these components (e.g. via oxidation reactions, reduction reactions, cleavage reactions and the like).
  • linkers include polymers, aliphatic hydrocarbon chains, unsaturated hydrocarbon chains, polypeptides, polynucleotides, cyclic linkers, acyclic linkers,
  • linkers include nucleophilic functional groups (e.g., amines, amino groups hydroxy groups, sulfhydryl groups, amino groups, alcohols, thiols, and hydrazides), electrophilic functional groups (e.g., aldehydes, esters, vinyl ketones, epoxides, isocyanates, and maleimides), and functional groups capable of cycloaddition reactions, forming disulfide bonds, or binding to metals.
  • nucleophilic functional groups e.g., amines, amino groups hydroxy groups, sulfhydryl groups, amino groups, alcohols, thiols, and hydrazides
  • electrophilic functional groups e.g., aldehydes, esters, vinyl ketones, epoxides, isocyanates, and maleimides
  • functional groups capable of cycloaddition reactions forming disulfide bonds, or binding to metals.
  • linkers can be primary amines, secondary amines, hydroxamic acids, N-hydroxysuccinimidyl esters, N-hydroxysuccinimidyl carbonates, oxycarbonylimidazoles, nitrophenylesters, trifluoroethyl esters, glycidyl ethers, vinylsulfones, maleimides, azidobenzoyl hydrazide, N-[4-(p-azidosalicylamino)butyl]-3'-[2'- pyridyldithio]propionamid), bis-sulfosuccinimidyl suberate, dimethyladipimidate,
  • the proximity probes may be produced using in vitro protocols that yield nucleic acid-protein conjugates (e.g. molecules having nucleic acids covalently bonded to a protein), such as producing the binding domain in vitro from vectors which encode the proximity probe.
  • nucleic acid-protein conjugates e.g. molecules having nucleic acids covalently bonded to a protein
  • the methods provided herein comprise forming complexes.
  • a complex refers to an association between at least two moieties (e.g. chemical or biochemical) that have an affinity for one another.
  • the methods provided herein comprise forming a complex between a target analyte and a binding moiety.
  • the methods comprise forming a complex between a target analyte and a single binding moiety.
  • the methods comprise forming a complex between a target analyte and a complex of two or more binding moieties.
  • the methods comprise forming a complex between a target analyte and a complex of two or more binding moieties.
  • the methods comprise forming a complex between two or more target analytes and a complex of two or more binding moieties. In some embodiments, the methods comprise forming a complex between a first complex comprising a target analyte and another moiety (e.g., a polypeptide, polynucleotide, or small molecule) and a binding moiety. In some embodiments, the methods comprise forming a complex between a first complex comprising a target analyte and another moiety (e.g., a polypeptide, polynucleotide, or small molecule) and a second complex comprising two or more binding moieties. For example, complexes can be formed between a target analyte coupled to a solid support, and a proximity probe comprising a binding moiety and a proximity
  • Complexes include a proximity probe bound to a target analyte.
  • Complexes include a binding moiety of a proximity probe bound to a target analyte and a proximity polynucleotide of the proximity probe coupled to an address polynucleotide.
  • Complexes include a binding moiety (e.g., a binding moiety of a proximity probe) bound to a target analyte.
  • complexes can include antibody-polypeptide complexes, polypeptide- polypeptide complexes, polypeptide-DNA complexes, polypeptide-RNA complexes,
  • polypeptide-aptamer complexes virus particle-antibody complexes, virus particle-polypeptide complexes, virus particle-DNA complexes, virus particle-RNA complexes, virus particle- aptamer complexes, cell-antibody complexes, cell-polypeptide complexes, cell-DNA complexes, cell-RNA complexes, cell-aptamer complexes, small molecule-polypeptide complexes, small molecule-DNA complexes, small molecule-aptamer complexes, small molecule-cell complexes, small molecule- virus particle complexes, and combinations thereof.
  • Complexes that may be excluded in some instances include complexes consisting of an address polynucleotide bound directly to a target analyte. Complexes that may be excluded in some instances include complexes consisting of an address polynucleotide bound directly to a binding moiety. Complexes that may be excluded in some instances include complexes consisting of a proximity polynucleotide bound directly to a target analyte. [00245] In some instances, a complex comprises a polypeptide interacting with a single-stranded DNA. In some instances, a complex comprises a tagged protein interacting with a single- stranded DNA.
  • a complex comprises an antibody interacting with a single- stranded DNA. In some instances, a complex comprises a virus particle interacting with a single- stranded DNA. In some instances, a complex comprises a cell interacting with a single-stranded DNA. In some instances, a complex comprises a small molecule interacting with a single- stranded DNA. In some instances, a complex comprises polypeptide interacting with a double- stranded DNA. In some instances, a complex comprises a tagged protein interacting with a double-stranded DNA. In some instances, a complex comprises an antibody interacting with a double-stranded DNA. In some instances, a complex comprises a virus particle interacting with a double-stranded DNA.
  • a complex comprises a cell interacting with a double-stranded DNA. In some instances, a complex comprises a small molecule interacting with a double-stranded DNA. In some instances, a complex comprises a polypeptide interacting with a single- stranded RNA. In some instances, a complex comprises a tagged protein interacting with a single-stranded RNA. In some instances, a complex comprises an antibody interacting with a single-stranded RNA. In some instances, a complex comprises a virus particle interacting with a single-stranded RNA. In some instances, a complex comprises a cell interacting with a single-stranded RNA.
  • a complex comprises a small molecule interacting with a single- stranded RNA.
  • a complex comprises a polypeptide interacting with a double-stranded RNA.
  • a complex comprises a tagged protein interacting with a double-stranded RNA.
  • a complex comprises an antibody interacting with a double-stranded RNA.
  • a complex comprises a virus particle interacting with a double-stranded RNA.
  • a complex comprises a cell interacting with a double-stranded RNA.
  • a complex comprises a small molecule interacting with a double-stranded RNA.
  • a complex comprises a polypeptide interacting with a RNA-DNA hybrid. In some instances, a complex comprises a tagged protein interacting with a RNA-DNA hybrid. In some instances, a complex comprises an antibody interacting with a RNA-DNA hybrid. In some instances, a complex comprises a virus particle interacting with a RNA-DNA hybrid. In some instances, a complex comprises a cell interacting with a RNA-DNA hybrid. In some instances, a complex comprises a small molecule interacting with a RNA-DNA hybrid. In some instances, a complex comprises a small molecule interacting with a double-stranded RNA.
  • a complex comprises a polypeptide interacting with a methylated polynucleotide.
  • a complex comprises a tagged protein interacting with a methylated polynucleotide.
  • a complex comprises an antibody interacting with a methylated polynucleotide.
  • a complex comprises a virus particle interacting with a methylated polynucleotide.
  • a complex comprises a cell interacting with a methylated polynucleotide.
  • a complex comprises a small molecule interacting with a methylated polynucleotide.
  • a complex comprises a polypeptide interacting with an unmethylated polynucleotide. In some instances, a complex comprises a tagged protein interacting with an unmethylated polynucleotide. In some instances, a complex comprises an antibody interacting with an unmethylated polynucleotide. In some instances, a complex comprises a virus particle interacting with an unmethylated polynucleotide. In some instances, a complex comprises a cell interacting with an unmethylated polynucleotide. In some instances, a complex comprises a small molecule interacting with an unmethylated polynucleotide.
  • a complex comprises a polypeptide interacting with a
  • a complex comprises a tagged protein interacting with a polynucleotide-coupled small molecule. In some instances, a complex comprises an antibody interacting with a polynucleotide-coupled small molecule. In some instances, a complex comprises a virus particle interacting with a polynucleotide-coupled small molecule. In some instances, a complex comprises a cell interacting with a polynucleotide- coupled small molecule.
  • a complex comprises a polypeptide interacting with an aptamer.
  • a complex comprises a tagged protein interacting with an aptamer.
  • a complex comprises an antibody interacting with an aptamer.
  • a complex comprises a virus particle interacting with an aptamer.
  • a complex comprises a cell interacting with an aptamer.
  • a complex comprises a small molecule interacting with an aptamer.
  • a complex comprises a polypeptide interacting with another polypeptide.
  • a complex comprises a tagged protein interacting with a polypeptide.
  • a complex comprises an antibody interacting with a polypeptide.
  • a complex comprises a virus particle interacting with a polypeptide.
  • a complex comprises a cell interacting with a polypeptide.
  • a complex comprises a small molecule interacting with a polypeptide.
  • a complex comprises a tagged protein interacting with an antibody.
  • a complex comprises an antibody interacting with another antibody.
  • a complex comprises a virus particle interacting with an antibody.
  • a complex comprises a cell interacting with an antibody. In some instances, a complex comprises a small molecule interacting with an antibody. In some instances, a complex comprises a tagged protein interacting with a virus particle. In some instances, a complex comprises a virus particle interacting with another virus particle. In some instances, a complex comprises a cell interacting with a virus particle. In some instances, a complex comprises a small molecule interacting with a virus particle. In some instances, a complex comprises a tagged protein interacting with a cell. In some instances, a complex comprises a cell interacting with another cell. In some instances, a complex comprises a small molecule interacting with a cell.
  • a complex comprises one or more polypeptides bound to one or more other polypeptides, one or more polynucleotides (e.g. DNAs, RNAs aptamers), one or more tagged proteins, one or more antibodies, one or more virus particles, one or more cells, one or more small molecules, or any combination thereof.
  • a complex comprises one or more tagged proteins bound to one or more polynucleotides (e.g. DNAs, RNAs aptamers), one or more other tagged proteins, one or more antibodies, one or more virus particles, one or more cells, one or more small molecules, or any combination thereof.
  • a complex comprises one or more antibodies bound to one or more polynucleotides (e.g. DNAs, RNAs aptamers), one or more other antibodies, one or more virus particles, one or more cells, one or more small molecules, or any combination thereof.
  • a complex comprises one or more virus particles bound to one or more polynucleotides (e.g. DNAs, RNAs aptamers), one or more other virus particles, one or more cells, one or more small molecules, or any combination thereof.
  • a complex comprises one or more cells bound to one or more polynucleotides (e.g. DNAs, RNAs aptamers), one or more other cells, one or more small molecules, or any combination thereof.
  • a complex comprises one or more small molecules bound to one or more polynucleotides (e.g. DNAs, RNAs aptamers), one or more other small molecules, or any combination thereof.
  • the methods disclosed herein can also include coupling a proximity polynucleotide in proximity to an address polynucleotide.
  • the proximity linker sequence and the address linker sequence are generally of a length sufficient to allow coupling to each other.
  • the proximity linker sequence and the address linker sequence can be of a length to permit hybridization of the two polynucleotides.
  • the proximity linker sequence can be of a length to permit splint polynucleotide-mediated interactions with the address linker sequence (e.g., when a binding moiety to which the proximity linker sequence is coupled is bound to a target analyte (e.g., in proximity to the address polynucleotide).
  • Proximity linker sequences and address linker sequences can be from about 8 up to about 1,000 nucleotides in length, about 8 to about 500 nucleotides in length, about 8 to about 250 nucleotides in length, about 8 to about 160 nucleotides in length, about 12 to about 150 nucleotides in length, about 14 to about 130 nucleotides in length, about 16 to about 110 nucleotides in length, about 8 to about 90 nucleotides in length, about 12 to about 80 nucleotides in length, about 14 to about 75 nucleotides in length, about 16 to about 70 nucleotides in length, about 16 to about 60 nucleotides in length, and so on.
  • the proximity linker sequences and address linker sequences may range in length from about 10 to about 80 nucleotides in length, from about 12 to about 75 nucleotides in length, from about 14 to about 70 nucleotides in length, from about 34 to about 60 nucleotides in length, and any length between the stated ranges. In some embodiments, the proximity linker sequences and address linker sequences are not more than about 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 44, 46, 50, 55, 60, 65, or 70 nucleotides in length.
  • a splint polynucleotide in proximity ligation assays is known in the art.
  • the splint polynucleotide may accordingly be viewed a "connector" polynucleotide which acts to connect or “hold together” the proximity linker sequence and the address linker sequence, such that they may interact, or may be coupled together.
  • a splint polynucleotide is generally of a length sufficient to allow coupling of the proximity linker sequence and the address linker sequence to each other.
  • the sequence of the splint polynucleotide may be chosen or selected with respect to the proximity linker sequence and the address linker sequence.
  • the sequence of the proximity linker sequence and the address linker sequence may be chosen or selected with respect to a splint polynucleotide. Thus, these sequences are not critical as long as the proximity linker sequence and the address linker sequence may hybridize to the splint polynucleotide However, the proximity linker sequences and the address linker sequences should be chosen to avoid the occurrence of hybridization events other than between the proximity linker sequence and the address linker sequence with that of the splint polynucleotide. Once the proximity linker sequence and the address linker sequence are selected or identified, the splint polynucleotide sequence may be synthesized using any convenient method.
  • the splint polynucleotide can be a short single-stranded molecule complementary to an end of the address polynucleotide linker and an end of the proximity linker. Hence, the splint polynucleotide will bring the termini of the address polynucleotide linker and the proximity linker into position for a ligase to join the two ends. The splint can then be removed by using exonucleases e.g.
  • the splint polynucleotide can be at least 2 nucleotides, e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more nucleotides) in length.
  • the splint polynucleotide hybridizes with the proximity linker sequences and the address linker sequences.
  • a splint polynucleotide can hybridize (anneal) simultaneously or in the same reaction with each of a plurality of proximity linker sequences.
  • a splint polynucleotide can hybridize simultaneously or in the same reaction with each of a plurality of address linker sequences.
  • a splint polynucleotide can hybridize simultaneously or in the same reaction with each of a plurality of proximity linker sequences and each of a plurality of address linker sequences.
  • the hybridization of the proximity linker sequences of each proximity probe and the address polynucleotide to each other increases the avidity of the proximity probe-target analyte complex upon binding to the target analyte. This avidity effect contributes to the sensitivity of the assays by supporting the formation of signal-generating proximity probe-target analyte complexes.
  • a proximity linker sequence and an address linker sequence can be coupled when in proximity to each other.
  • a proximity linker sequence and an address linker sequence can be coupled using any method that permits amplification and/or detection of the address barcode and the proximity barcode such that the two barcodes are known to arise from a sample molecule or complex of molecules.
  • a proximity linker sequence and an address linker sequence are coupled by ligating the two polynucleotides to each other.
  • Ligation involves creating a phosphodiester bond between the 3' hydroxyl of one nucleotide and the 5' phosphate of another.
  • a suitable ligase and any reagents that are necessary and/or desirable are contacted to the solid support or spot on a solid support and maintained under conditions sufficient for ligation of the proximity linker sequence and address linker sequence to occur.
  • Ligases catalyze the formation of a phosphodiester bond between juxtaposed 3 '-hydroxyl and 5 '-phosphate termini of two immediately adjacent nucleic acids when they are annealed or hybridized to a third nucleic acid sequence to which they are complementary (e.g. a splint polynucleotide).
  • ligases e.g., temperature sensitive and thermostable ligases
  • representative ligases of interest include, but are not limited to bacteriophage T4 DNA ligase, Taq ligase, Tth ligase, Ampligase®, Pfu ligase, Thermus thermophilics ligase, Thermus acquaticus ligase, Pyrococcus ligase, bacteriophage T7 ligase, and E. coli ligase.
  • Thermostable ligase may be obtained from thermophilic or hyperthermophilic organisms (e.g., prokaryotic, eukaryotic, or archael organisms). Certain RNA ligases may also be employed in the methods of the invention.
  • Ligation reaction conditions are well known to those of skill in the art. Ligation can be carried out at 4-45°C in the presence of a ligase enzyme (e.g., a DNA ligase). For example, during ligation, the reaction mixture may be maintained at a temperature ranging from about 4°C to about 50°C, or 20°C to about 37°C; and for a period of time ranging from about 5 seconds to about 16 hours, such as from about 1 minute to about 1 hour.
  • a ligase enzyme e.g., a DNA ligase
  • the reaction mixture may be maintained at a temperature ranging from about 35°C to about 45°C, such as from about 37°C to about 42°C (e.g., at or about 38°C, 39°C, 40°C or 4FC), for a period of time ranging from about 5 seconds to about 16 hours, such as from about 1 minute to about 1 hour, including from about 2 minutes to about 8 hours.
  • a ligation reaction mixture can include, for example, 50 mM Tris pH7.5, 10 mM MgCl 2 , 10 mM DTT, 1 mM ATP, 25 mg/ml BSA, 0.25 units/ml RNase inhibitor, and T4 DNA ligase at 0.125 units/ml.
  • a ligation reaction mixture can include, for example, 2.125 mM magnesium ion, 0.2 units/ml RNase inhibitor; and 0.125 units/ml DNA ligase.
  • the ligation conditions may depend on the ligase enzyme used in the methods of the invention.
  • the above-described ligation conditions are merely a representative example and the parameters may be varied according to well-known protocols.
  • a ligase namely Ampligase®
  • the alteration of one parameter e.g. temperature
  • Such manipulation of the methods is routine in the art.
  • ligating comprises bringing an end of a proximity polynucleotide adjacent to an end of an address polynucleotide. In some instances, bringing an end of a proximity polynucleotide adjacent to an end of an address polynucleotide comprises hybridizing a splint polynucleotide to the proximity linker sequence and the address polynucleotide linker sequence. In some instances, ligating comprises hybridizing a splint polynucleotide to the proximity linker sequence and the address polynucleotide linker sequence.
  • Ligation of a proximity polynucleotide to an address polynucleotide wherein the proximity polynucleotide and address polynucleotide are hybridized to a splint polynucleotide can be achieved by contacting a ligating activity thereto (e.g. provided by a suitable nucleic acid ligase) and maintaining the mixture under conditions sufficient for ligation of the proximity linker sequence and address linker sequence to occur.
  • a proximity linker sequence and an address linker sequence can be coupled to each other by ligating an end of the proximity linker sequence to an end of the address linker sequence.
  • a proximity linker sequence and an address linker sequence can be coupled to each other by ligating a 5' end of the address polynucleotide to a 3 ' end of the proximity polynucleotide.
  • the methods provide for ligating a proximity polynucleotide to an address polynucleotide, wherein the address polynucleotide is coupled to a solid support and in proximity to a target analyte or in proximity to a proximity probe bound to the target analyte.
  • a ligated product of the resulting ligation reaction between the proximity polynucleotide and the address polynucleotide, or an amplified product thereof, can then be detected and/or amplified.
  • coupling comprises hybridizing an address linker sequence to a proximity linker sequence.
  • Such a coupled product can be subjected to extension of one or both ends of the hybridized linker sequences.
  • Such a coupled product containing one or both extended ends of the hybridized linker sequences can then be amplified as described herein (e.g., such that the amplified products contain the proximity barcode and the address barcode.
  • the new paired barcoded polynucleotide composition generated using the methods of the invention can serve multiple functions.
  • the paired barcoded polynucleotide allows for quantitative and/or qualitative detection of target analytes, binding moieties, and affinities and specificities between target analytes and binding, on multiplex and multiplex-on- multiplex formats.
  • the paired barcoded polynucleotide can serve to pair or join a binding event between a single target analyte and a single binding moiety from a plurality of target analytes and a plurality of binding moieties.
  • the nucleic acid domains of the proximity probes when in proximity may template the ligation of one or more added
  • oligonucleotides to each other (which may be the nucleic acid domain of one or more proximity probes), including an intramolecular ligation to circularize an added linear oligonucleotide, for example based on the so-called padlock probe principle, wherein analogously to a padlock probe, the ends of the added linear oligonucleotide are brought into juxtaposition for ligation by hybridizing to a template, here a nucleic acid domain of the proximity probe (in the case of a padlock probe the target nucleic acid for the probe).
  • nucleic acid domains may be joined to form a new nucleic acid sequence generally by means of a ligation reaction, which may be templated by a splint polynucleotide added to the reaction, the splint polynucleotide containing regions of complementarity for the ends of the respective polynucleotide domains of the barcoded proximity probe and the barcoded address polynucleotide.
  • a ligation reaction which may be templated by a splint polynucleotide added to the reaction, the splint polynucleotide containing regions of complementarity for the ends of the respective polynucleotide domains of the barcoded proximity probe and the barcoded address polynucleotide.
  • the splint polynucleotide to template ligation of the nucleic acid domains of two proximity probes is carried on a third proximity probe.
  • multivalent proximity probes comprise at least two, but as many as 100, analyte-binding domains conjugated to at least one, and preferably more than one, nucleic acid(s).
  • Coupling can comprise hybridizing an address linker sequence to a proximity linker sequence via enzymatic and non-enzymatic (e.g., chemical) methods.
  • ligation reactions that are non-enzymatic include the non-enzymatic ligation techniques described in U.S. Pat. Nos. 5,780,613 and 5,476,930.
  • a ligase for example a DNA ligase or RNA ligase is used for coupling.
  • Ligation techniques comprise blunt-end ligation and sticky-end ligation.
  • Ligation reactions may include DNA ligases such as DNA ligase I, DNA ligase III, DNA ligase IV, and T4 DNA ligase.
  • Ligation reactions may include RNA ligases such as T4 RNA ligase I and T4 RNA ligase II.
  • Multiple ligases are known in the art, and include, without limitation NAD + -dependent ligases including tRNA ligase, Taq DNA ligase, Thermus filiformis DNA ligase, Escherichia coli DNA ligase, Tth DNA ligase, Thermus scotoductus DNA ligase (I and II), thermostable ligase, Ampligase thermostable DNA ligase, VanC-type ligase, 9° N DNA Ligase, Tsp DNA ligase, and novel ligases discovered by bioprospecting; ATP-dependent ligases including T4 RNA ligase, T4 DNA ligase, T3 DNA ligase, T7 DNA ligase, Pfu DNA ligase, DNA ligase 1, DNA ligase III,
  • ligation can be between polynucleotides having hybridizable sequences, such as complementary overhangs. In some instances, ligation can be between polynucleotides having.
  • a 5' phosphate is utilized in a ligation reaction. The 5' phosphate can be provided by the target polynucleotide, the adaptor oligonucleotide, or both. 5' phosphates can be added to or removed from polynucleotides to be joined, as needed. Methods for the addition or removal of 5' phosphates are known in the art, and include without limitation enzymatic and chemical processes. Enzymes useful in the addition and/or removal of 5' phosphates include kinases, phosphatases, and polymerases. In some embodiments, 3 ' phosphates are removed prior to ligation.
  • the coupling makes use of CLICK chemistry.
  • CLICK chemistry Suitable methods to link various molecules using CLICK chemistry are known in the art (for CLICK chemistry linkage of oligonucleotides, see, e.g. El-Sagheer et al. (PNAS, 108:28, 1 1338-1 1343, 201 1). Click chemistry may be performed in the presence of Cul .
  • the coupling makes use of topoisomerase, e.g., a Vaccinia virus topoisomerase I.
  • the coupling makes use of restriction enzyme known in the art that produces blunt ends.
  • a 3' overhang can be added to the blunt ends.
  • a 3 ' overhang can be added using terminal transferase in the presence of dNTPs.
  • a 3 ' overhang can be added using a polymerase in the presence of dNTPs.
  • the polymerase can be a polymerase lacking proofreading activity.
  • the polymerase can be a Taq polymerase.
  • topoisomerase I bonded to can ligate the polynucleotides.
  • coupling can comprise incubation with Vaccinia virus topoisomerase I using any method as provided herein, processing with a blunt end cutting restriction enzyme, incubation with an enzyme (e.g., Taq polymerase) that adds a residue to each blunt end, and ligation via the topoisomerase I.
  • an enzyme e.g., Taq polymerase
  • End repair can include the generation of blunt ends, non-blunt ends (i.e. sticky or cohesive ends), or single base overhangs, such as the addition of a single dA nucleotide to the 3 '-end of the double-stranded polynucleotide product by a polymerase lacking 3 '-exonuclease activity.
  • end repair is performed to produce blunt ends wherein the ends contain 5' phosphates and 3 ' hydroxyls. End repair can be performed using any number of enzymes and/or methods known in the art.
  • An overhang can comprise about, more than, less than, or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides.
  • a sticky end refers to an end of a double stranded nucleic acid wherein the 5' or the 3 ' end has an extension of one or more nucleotides and which do not form a base pair. This is in contrast to a blunt end wherein the terminal 5 ' polynucleotide forms a basepair with the 3 ' terminal polynucleotide.
  • Blunt ends can be generated by the use of a single strand specific DNA exonuclease such as for example exonuclease 1, exonuclease 7 or a combination thereof to degrade overhanging single stranded ends.
  • blunt ends can be generated by the use of a single stranded specific DNA endonuclease for example but not limited to mung bean endonuclease or SI endonuclease.
  • blunt ends can be generated by the use of a polymerase that comprises single stranded exonuclease activity such as for example T4 DNA polymerase, any other polymerase comprising single stranded exonuclease activity or a combination thereof to degrade the overhanging single stranded ends.
  • the polymerase comprising single stranded exonuclease activity can be incubated in a reaction mixture that does or does not comprise one or more dNTPs.
  • a combination of single stranded nucleic acid specific exonucleases and one or more polymerases can be used to generate blunt ends.
  • products of an extension reaction can be made blunt ended by filling in the overhanging single stranded ends of the double stranded polynucleotides.
  • the polynucleotides can be incubated with a polymerase such as T4 DNA polymerase or Klenow polymerase or a combination thereof in the presence of one or more dNTPs to fill in single stranded portions of the double stranded polynucleotides.
  • the polynucleotides can be made blunt by a combination of a single stranded overhang degradation reaction using exonucleases and/or polymerases, and a fill-in reaction using one or more polymerases in the presence of one or more dNTPs.
  • a polymerase without terminal transferase activity or with proofreading activity can be used for coupling the address polynucleotide to the proximity polynucleotide.
  • DNA polymerization with these DNA polymerase enzymes can result in double stranded DNA with blunt ends, without overhang or recessive end at the 3' end.
  • Enzymes within this class are for example Klenow polymerase and several polymerases which have polymerase activity below 95 °C such as pfu polymerase.
  • the methods provided herein can comprise an amplification step.
  • a determining step comprises amplification.
  • Amplification can be used in the methods described herein to increase the number of copies of a nucleic acid sequence, such as through the use of enzymes.
  • detection can comprise amplifying a sequence of a polynucleotide comprising an address barcode.
  • detection can comprise amplifying a sequence of a polynucleotide comprising a proximity barcode.
  • detection can comprise amplifying a sequence of a polynucleotide comprising an address barcode and a proximity barcode.
  • detection can comprise amplifying a coupled ⁇ e.g., ligated) polynucleotide containing the proximity barcode and the address barcode and/or complements thereof.
  • detection can comprise amplifying a sequence of a polynucleotide comprising an address barcode.
  • detection can comprise amplifying a sequence of a polynucleotide comprising a proximity barcode.
  • detection can comprise amplifying a sequence of a polynucleotide comprising an address barcode and a proximity barcode.
  • detection can comprise amplifying a sequence of a polynucleotide that is a ligated product, such as a ligated product containing the proximity barcode and the address barcode and their complementary sequence.
  • the methods described herein can be used to amplify coupled polynucleotides (e.g., an address polynucleotide coupled to a proximity polynucleotide).
  • the methods described herein can employ amplification, such as to increase in the number of copies of a sequence and/or compliment thereof, of a target polynucleotide, such as a ligated product containing a proximity barcode and an address barcode and their complementary sequence.
  • Amplification may be performed using any method known in the art.
  • a variety of amplification processes are known.
  • One of the most commonly used is the polymerase chain reaction (PCR).
  • the PCR process of Mullis is described in U. S. Pat. Nos. 4,683, 195 and 4,683,202. Any type of PCR may be used.
  • the PCR amplification process involves an enzymatic chain reaction for preparing exponential quantities of a specific nucleic acid sequence. It requires a small amount of a sequence to initiate the chain reaction and
  • polynucleotide primers that will hybridize to the sequence.
  • primers are annealed to denatured nucleic acid followed by extension with an inducing agent (enzyme) and nucleotides. This results in newly synthesized extension products. Since these newly synthesized sequences become templates for the primers, repeated cycles of denaturing, primer annealing, and extension results in exponential accumulation of the specific sequence being amplified.
  • the extension product of the chain reaction will be a discrete nucleic acid duplex with a termini corresponding to the ends of the specific primers employed.
  • Amplification methods also include methods performed at a single temperature (isothermal).
  • amplifying nucleic acid examples include, for example, reverse transcription-PCR, real-time PCR, quantitative real-time PCR, digital PCR (dPCR), digital emulsion PCR (dePCR), clonal PCR, amplified fragment length polymorphism PCR (AFLP PCR), allele specific PCR, assembly PCR, asymmetric PCR (in which a great excess of primers for a chosen strand can be used), colony PCR, helicase-dependent
  • HDA Hot Start PCR
  • IPCR inverse PCR
  • in situ PCR long PCR (extension of DNA greater than about 5 kilobases)
  • multiplex PCR multiplex PCR
  • nested PCR uses more than one pair of primers
  • single-cell PCR touchdown PCR
  • LAMP loop-mediated isothermal PCR
  • RPA recombinase polymerase amplification
  • NASBA nucleic acid sequence based amplification
  • LCR ligase chain reaction
  • SDA strand displacement amplification
  • QPRA QP replicase amplification
  • TACL Target Amplification by Capture and Ligation
  • RACE RACE amplification
  • Amplification may be performed by amplifying a sequence of a polynucleotide, such as a ligated product, as a single amplification product (e.g., a single amplified amplicon).
  • a primer may be selected such that one amplified product can include all target sequences, such as an address barcode sequence and a proximity barcode sequence, contained in one ligated product.
  • Amplification may be performed by amplifying a sequence of a polynucleotide, such as a ligated product, that has a length of about 5,000 nucleotides or less.
  • the length of the ligated product may be a length of 4,500; 4,000; 3,500; 3,000; 3,000; 2,500; 2,000; 1,500; 1,000; 800; 600; 400; 200; or 100 nucleotides or less.
  • Amplification may be performed by amplifying a sequence of a polynucleotide, such as a ligated product, that has a length of about 10 or more nucleotides.
  • the length of the ligated product may be a length of 4,500; 4,000; 3,500; 3,000; 3,000; 2,500; 2,000; 1,500; 1,000; 800; 600; 400; 200; or 100 nucleotides or more.
  • the length of the ligated product may be a length of from about 10-5,000; 10-4,500; 10-4,000; 10-3,500; 10-3,000; 10-3,000; 10-2,500; 10-2,000; 10-1,500; 10-1,000; 10- 800; 10-600; 10-400; 10-200; 15-5,000; 15-4,500; 15-4,000; 15-3,500; 15-3,000; 15-3,000; 15- 2,500; 15-2,000; 15-1,500; 15-1,000; 15-800; 15-600; 15-400; 15-200; 18-4,000; 18-3,500; 18- 3,000; 18-3,000; 18-2,500; 18-2,000; 18-1,500; 18-1,000; 18-800; 18-600; 18-400; 18-200; 21- 4,000; 21-2,000; or 21-1,000 nucleot
  • RNA in a sample can be converted to cDNA by using reverse transcription.
  • the methods may further comprise, producing a DNA (cDNA) complementary to the target nucleic acid by reverse- transcribing the target nucleic acid.
  • Reverse-transcribing may be performed before or after forming a ligation product. It is known in the art that the reverse-transcribing produces a DNA complementary strand using an RNA strand using a reverse transcriptase.
  • the method may further include ligating an adaptor sequence to at least one of the 3 '-terminus and the 5 '-terminus of an RNA before reverse-transcribing the RNA, such that the resulting RNA ligation product or cDNA thereof contains a sequence complimentary to the RNA and a sequence complimentary to the adaptor sequence, which complimentary sequence also can be an adaptor sequence.
  • the adaptor sequence may be specifically ligated to at least one of the 3 '-terminal and the 5 '-terminal of the nucleic acid.
  • the method may include reverse transcribing RNA, and subsequently attaching, such as ligating, one or more adaptor and primer (or primer binding) sequences to at least one of the 3 '-terminal and the 5 '-terminal of cDNA of nucleic acid after the reverse- transcribing.
  • the adaptor sequence may be specifically ligated to at least one of the 3 '-terminal and the 5 '-terminal of the target nucleic acid.
  • the ligating the adaptor sequence can be as described above.
  • Amplification, reverse transcription, sequencing, and combinations thereof can be performed with one or more primers, or one or more primer sets.
  • a primer is polynucleotide, the sequence of at least a portion of which is complementary to a segment of a template
  • a primer is capable of acting as a point of initiation of nucleic acid synthesis when placed under conditions in which synthesis of a primer extension product which is complementary to a nucleic acid strand is induced, e.g., in the presence of nucleotides and an agent for polymerization such as DNA polymerase, reverse transcriptase and the like, and at a suitable temperature and pH.
  • the primer is preferably single stranded for maximum efficiency, but may alternatively be in double stranded form. If double stranded, the primer is first treated to separate it from its complementary strand before being used to prepare extension products.
  • a primer must be sufficiently long to prime the synthesis of extension products in the presence of the agents for polymerization. The exact lengths of the primers will depend on many factors, including temperature and the source of primer.
  • the primers used herein are selected to be substantially complementary to the different strands of each specific sequence to be amplified, reverse transcribed, or sequenced, and preferably non-randomly hybridize with its respective template strand. Therefore, the primer sequence may or may not reflect the exact sequence of the template.
  • Primers can be prepared using any suitable method, such as, for example, the phosphotriester or phosphodiester methods described in Narang et al, (1979) Meth Enzymol, 68:90; Brown et al, (1979) Meth Enzymol, 68: 10; and U.S. Pat. Nos. 4,356,270; 4,458,066; 4,416,988; and 4,293,652.
  • Exemplary reverse transcription primers include poly-A primers, random primers, sequence specific primers, and gene specific primers.
  • a primer for use in the methods described herein can be substantially complementary to an address polynucleotide primer binding sequence.
  • An amplification primer for use in the methods described herein can be substantially complementary to a proximity polynucleotide primer binding sequence.
  • a primer pair can comprise a first primer substantially complementary to an address polynucleotide primer binding sequence and a second primer substantially complementary to a proximity polynucleotide primer binding sequence.
  • Amplification of a polynucleotide comprising a proximity polynucleotide coupled to an address polynucleotide can be used to amplify the polynucleotide comprising a proximity polynucleotide coupled to an address polynucleotide such that amplified products produced contain the proximity barcode or compliment thereof.
  • Amplification of a polynucleotide comprising a proximity polynucleotide coupled to an address polynucleotide can be used to amplify the polynucleotide comprising a proximity polynucleotide coupled to an address polynucleotide such that amplified products produced contain the address barcode or compliment thereof.
  • Amplification of a polynucleotide comprising a proximity polynucleotide coupled to an address polynucleotide can be used to amplify the polynucleotide comprising a proximity polynucleotide coupled to an address polynucleotide such that amplified products produced contain both the proximity barcode or compliment thereof and the address barcode or compliment thereof.
  • a primer used in amplification can have any suitable sequence for amplification.
  • an amplification primer does not have a sequence complementary to a proximity barcode, such as an address barcode contained in a ligated product.
  • an amplification primer does not have a sequence complementary to an address barcode, such as an address barcode contained in a ligated product.
  • an amplification primer binds to a polynucleotide comprising a proximity polynucleotide coupled to an address polynucleotide at a region upstream of the address barcode.
  • an amplification primer binds to a polynucleotide comprising a proximity polynucleotide coupled to an address polynucleotide at a region upstream of the address barcode.
  • Amplification can be performed using a primer set comprising a first primer and a second primer.
  • amplification can be performed using a primer set comprising a forward primer and a reverse primer.
  • a forward primer can be complementary to a region of a ligated product that is upstream of a proximity barcode.
  • a reverse primer can be complementary to a region of a ligated product that is upstream of an address barcode.
  • an amplification primer set comprises a first primer (e.g., a forward primer) and a second primer (e.g., a reverse primer) that bind to a polynucleotide comprising a proximity polynucleotide coupled to an address polynucleotide, wherein the first primer binds to a region upstream of the address barcode, and wherein the second primer binds to a region upstream of the proximity barcode.
  • a primer can be a universal primer.
  • a universal primer contains a unique amplification or sequencing priming region that is, for example, about 5, 7, 10, 13, 15, 17, 20, 22, or 25 nucleotides in length, and is present on each polynucleotide of a plurality of polynucleotides to be amplified.
  • a universal primer can be used to amplify multiple polynucleotides simultaneously, in a single reaction, and/or with similar amplification efficiencies.
  • a primer can comprise a universal adaptor.
  • a primer can comprise a universal sequencing primer binding region such that amplified products contain the universal sequencing primer region.
  • the methods described herein can comprise detecting a product (or amplified product thereof) comprising an address polynucleotide coupled to a proximity polynucleotide.
  • the detecting can comprise sequencing.
  • the detecting can comprise sequencing the proximity barcode and the address barcode of a product comprising an address polynucleotide coupled to a proximity polynucleotide.
  • methods for sequencing products (or amplified products thereof) comprising an address polynucleotide coupled to a proximity polynucleotide using one or more primers or primer pairs located upstream of the proximity barcode and address barcode.
  • a sequence read can comprise an address barcode sequence and a proximity barcode sequence on the same sequence read. Any sequencing technique described herein or known to one skilled in the art can be used in the methods herein.
  • Sequencing methods include deep sequencing methods.
  • the detecting can comprise deep sequencing (i.e., ultra-deep sequencing or next generation sequencing (NGS)) which is directed to an enhanced sequencing method enabling the rapid parallel sequencing of multiple nucleic acid sequences.
  • deep sequencing methods include sequencing nucleic acids to a depth that allows each base to be read hundreds of times, typically at least about 500, 1,000, or 1,500 times.
  • nucleic acids e.g. DNA fragments
  • a reaction platform e.g., flow cell, microarray, and the like.
  • polynucleotides are amplified in situ and used as templates for synthetic sequencing (e.g., sequencing by synthesis) using a detectable label (e.g. fluorescent reversible terminator deoxyribonucleotide).
  • a detectable label e.g. fluorescent reversible terminator deoxyribonucleotide
  • Representative reversible terminator deoxyribonucleotides may include 3'-0-azidomethyl-2'-deoxynucleoside triphosphates of adenine, cytosine, guanine and thymine, each labeled with a different recognizable and removable fluorophore, optionally attached via a linker.
  • the identity of the inserted based may be determined by excitation (e.g., laser-induced excitation) of the fluorophores and imaging of the resulting immobilized growing duplex nucleic acid.
  • excitation e.g., laser-induced excitation
  • the fluorophore, and optionally linker may be removed by methods known in the art, thereby regenerating a 3 ' hydroxyl group ready for the next cycle of nucleotide addition.
  • Exemplary suitable deep sequencing methods include single molecule real time
  • SMRTTM sequencing Pacific Biosciences
  • Ion Torrent sequencing MiSeq sequencing
  • HiSeq sequencing massively parallel signature sequencing
  • MPSS sequencing by synthesis
  • SBS SBS pyro sequencing (454 Life Sciences)
  • SOLiDTM sequencing by ligation Applied Biosystems
  • a sequencing technique used in the methods of the provided invention generates at least 100 reads per run, at least 200 reads per run, at least 300 reads per run, at least 400 reads per run, at least 500 reads per run, at least 600 reads per run, at least 700 reads per run, at least 800 reads per run, at least 900 reads per run, at least 1,000 reads per run, at least 5,000 reads per run, at least 10,000 reads per run, at least 50,000 reads per run, at least 100,000 reads per run, at least 500,000 reads per run, at least 1,000,000 reads per run, at least 2,000,000 reads per run, at least 3,000,000 reads per run, at least 4,000,000 reads per run at least 5,000,000 reads per runs at least 6,000,000 reads per run at least 7,000,000 reads per run at least 8,000,000 reads per runs at least 9,000,000 reads per run, or at least 10,000,000 reads per run.
  • the methods, kits, and compositions described herein can be used for numerous applications, including identification of binding partners, determination of affinities of binding moieties to target analytes, determination of specificities of binding moieties to target analytes, quantification of target analytes in a sample, quantification of binding events, identification of biomarkers of a disease or condition, drug discovery, molecular biology, immunology and toxicology.
  • Arrays can be used for large scale binding assays in numerous diagnostic and screening applications. These methods of use include, but are not limited to, high-content, high- throughput assays for screening for binding moieties that interact with target analytes.
  • Additional methods of use include medical diagnostic, proteomic, and biosensor assays.
  • the multiplexed measurement of quantitative variation in levels of large numbers of target analytes allows the recognition of patterns defined by several to many different target analytes.
  • the multiplexed identification of large numbers of interactions between target analytes and binding moieties allows for the recognition of binding and interaction patterns defined by several to many different interactions between target analytes and binding moieties.
  • the assays used with the arrays of the presently disclosed subject matter may be direct, noncompetitive assays or indirect, competitive assays.
  • the affinity for a target analyte to a binding moiety can be determined directly.
  • the target analyte can be directly exposed to a binding moiety.
  • the binding moiety may be labeled or unlabeled.
  • a label refers to a molecule that, when attached to another molecule provides or enhances a means of detecting the other molecule.
  • a signal emitted from a label can allow detection of the molecule or complex to which it is attached, and/or the label itself.
  • a label can be a molecular species that elicits a physical or chemical response that can be observed or detected by the naked eye or by means of instrumentation such as, without limitation, scintillation counters, colorimeters, UV spectrophotometers and the like.
  • Labels include but are not limited to, radioactive isotopes, fluorophores, chemiluminescent dyes, chromophores, enzymes, enzymes substrates, enzyme cofactors, enzyme inhibitors, dyes, metal ions, nanoparticles, metal sols, ligands (such as biotin, avidin, streptavidin or haptens) and the like.
  • a fluorescence or fluorescent label or tag emits detectable light at a particular wavelength when excited at a different wavelength.
  • a radiolabel or radioactive tag emits radioactive particles detectable with an instrument such as, without limitation, a scintillation counter.
  • Other signal generation detection methods include: chemiluminescence detection, electrochemiluminescence detection, Raman energy detection, colorimetric detection, hybridization protection assays, and mass spectrometry.
  • the methods of detection could include fluorescence, luminescence, radioactivity, and the like. If the binding moiety is unlabeled, the detection of binding would be based on a change in some physical property of the target analyte. Such physical properties could include, for example, a refractive index or electrical impedance. The detection of binding of unlabeled binding moiety could include, for example, mass spectroscopy. In competitive methods, binding-site occupancy may be determined indirectly. In this method, the target analytes can be exposed to a solution containing a cognate labeled binding moiety and an unlabeled moiety.
  • the labeled cognate binding moiety and the unlabeled moiety compete for the binding sites on the target analyte.
  • the affinity of the unlabeled moiety for the target analyte relative to the labeled cognate binding moiety is determined by the decrease in the amount of binding of the labeled binding moiety.
  • the detection of binding can also be carried out using sandwich assays, in which after the initial binding, the array is incubated with a second solution containing molecules such as labeled antibodies that have an affinity for the binding moiety bound to the target analyte, and the amount of binding is determined based on the amount of binding of the labeled antibodies to the binding moiety.
  • the detection of binding can be carried out using a displacement assay in which after the initial binding of a labeled moiety, the array is incubated with a second solution containing unlabeled binding moiety.
  • the binding capability and the amount of binding of the binding moiety are determined based on the decrease in number of the pre-bound labeled moieties in the target analytes.
  • the arrays of the presently disclosed subject matter may also be used in a method for screening for binding moieties, wherein a potential binding moiety candidate is screened directly for its ability to bind or otherwise interact with a plurality of target analytes on the array.
  • a plurality of potential binding moieties may be screened in parallel for their ability to bind or otherwise interact with one or more types of target analytes on the array.
  • the screening process may involve assaying for the interaction, such as binding, of at least one binding moiety of a sample with one or more target analytes on the array, both in the presence and absence of the potential binding moiety candidate. This allows for a potential binding moiety to be tested for its ability to act as an inhibitor of the interaction or interactions originally being assayed.
  • the arrays of the presently disclosed subject matter may also be used in a method for screening a plurality of target analytes for their ability to bind a particular binding moiety of a sample containing a plurality of binding moieties.
  • the sample can be contacted to an array comprising target analytes and the presence or amount of the particular binding moiety retained at each microspot can be detected, either directly or indirectly, or by sequencing.
  • the method further comprises characterizing the particular binding moiety retained on at least one microspot.
  • Also disclosed herein are methods for determining a quantity, amount, or concentration of a target analyte in a sample wherein the determining comprises determining a number of sequence reads having a proximity barcode sequence corresponding to the binding moiety and an address barcode sequence corresponding to the target analyte, wherein the number of the sequence reads is proportional to the quantity, amount, or concentration of the target analyte in the sample.
  • the determining can comprise determining a number of sequence reads having a proximity barcode sequence corresponding to the binding moiety and an address barcode sequence corresponding to the target analyte and comparing the number of reads to a standard curve, such as a standard curve generated using a same method using a particular binding moiety known to interact with a particular target analyte, wherein the particular target analyte is present at one or more known concentrations.
  • a standard curve such as a standard curve generated using a same method using a particular binding moiety known to interact with a particular target analyte, wherein the particular target analyte is present at one or more known concentrations.
  • Also disclosed herein is a method of determining a relative binding affinity of a binding moiety for a target analyte, wherein the determining comprises determining a number of sequence reads having a proximity barcode sequence corresponding to the binding moiety and an address barcode sequence corresponding to the target analyte, wherein the number of sequence reads is proportional to the relative binding affinity.
  • Also provided herein is a method of determining a relative binding affinity of a binding moiety for a target analyte, the method comprising amplifying coupled proximity polynucleotide and address polynucleotide products; measuring an amount of sequence reads having the proximity barcode sequence and the address barcode sequence from the amplified product; and determining a relative binding affinity of the binding moiety for the target analyte by using the measured amount.
  • the relative binding affinity of a binding moiety for a target analyte may be measured by measuring or counting the coupled product and/or amplified products thereof by using any suitable method known in the art.
  • the determining may be performed by standardizing an amount of sequence reads having the proximity barcode sequence and the address barcode sequence with respect to a predetermined value, for example, a threshold value, or comparing the amount of sequence reads having the proximity barcode sequence and the address barcode sequence with a standard value.
  • the determining may be performed by standardizing an amount of sequence reads having the proximity barcode sequence and the address barcode sequence with respect to a control, for example, an amount of sequence reads generated from a control reaction.
  • the determining of a relative binding affinity of the binding moiety' s binding to the target analyte may be used to determine whether an association between the target nucleic acid and various physiological conditions or diseases exists.
  • a method herein can include further determining a relative binding specificity of the binding moiety for the target analyte, wherein the determining can include determining the number of sequence reads having the same address barcode but different proximity barcodes. The number or quantity of sequence reads having a different target analyte barcode can be inversely proportional to the binding specificity.
  • the relative binding specificity of the binding moiety for the target analyte may be measured according to coupled product and/or amplified products thereof using any method known in the art.
  • the determining may be performed by standardizing the amount of sequence reads having a same address barcode but different proximity barcodes and/or a same address barcode and a same proximity barcode, with respect to a predetermined value, for example, a threshold value, or comparing the amount of sequence reads a same address barcode but different proximity barcodes and/or a same address barcode and a same proximity barcode with a standard value.
  • a predetermined value for example, a threshold value
  • the subject methods may also be used to screen for agents that modulate the interaction between a binding moiety of a proximity probe with a target analyte to which it binds.
  • modulating includes both decreasing (e.g., inhibiting) and enhancing the interaction between the two molecules.
  • the screening method may be an in vitro or in vivo format, where both formats are readily developed by those of skill in the art.
  • Candidate agents encompass numerous chemical classes including, but not limited to, peptides, polynucleotides, and organic molecules (e.g., small organic compounds having a molecular weight of more than 50 and less than about 2,500 Daltons).
  • Candidate agents can comprise functional groups for structural interaction with target analytes, such as hydrogen bonding, and can include at least one or at least two of an amine, carbonyl, hydroxyl or carboxyl group.
  • the candidate agents can comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more functional groups.
  • Candidate agents can be biomolecules including peptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogs or combinations thereof.
  • Candidate agents can be obtained from a wide variety of sources including libraries of synthetic or natural compounds. For example, numerous means are available for random and directed synthesis of a wide variety of organic compounds and biomolecules, including expression of randomized polynucleotides and polypeptides.
  • libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced. Additionally, natural or synthetically produced libraries and compounds are readily modified through conventional chemical, physical and biochemical means, and may be used to produce combinatorial libraries.
  • pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, esterification, acidification, etc. to produce structural analogs.
  • Agents identified find uses in a variety of methods, including methods of modulating the activity of a target analyte, and conditions related to the presence, activity, and/or interactions of a target analyte
  • a selected binding moiety is identified as monospecific.
  • at most about 0.01% of the screened binding moieties can be monospecific.
  • binding of a binding moiety to a target analyte can be validated or determined by various established methods known in the art and include ELISA, FACS, Western Blot, ImmunoBlot, MSD, BIAcore and SET; and these values can be compared to the corresponding binding affinities determined using the methods described herein.
  • a binding moiety can be deemed to be a binding partner for a target analyte if the binding moiety is demonstrated to be able to bind to a specific target analyte at least 2 -fold, 3-fold, 4-fold, 5-fold, 6- fold, 7-fold, 8- fold, 9-fold, 10-fold, 20-fold, 50-fold, 100-fold, 500-fold, or 1,000-fold over background or a negative control reaction.
  • a binding moiety can be deemed to be a binding partner for a target analyte if the number of sequence reads containing an address barcode sequence corresponding to that target analyte and a proximity barcode corresponding to the binding moiety is at least about 2-fold, 3-fold, 4-fold, 5-fold, 6- fold, 7-fold, 8-fold, 9-fold, 10-fold, 20- fold, 50-fold, 100-fold, 500-fold, or 1,000-fold higher than the number of sequence reads containing an address barcode sequence that does not correspond to that target analyte and a proximity barcode corresponding to the binding moiety.
  • a binding moiety can be deemed monospecific for a target analyte if the binding moiety is demonstrated to be able to bind to a specific target analyte at least 2 -fold, 3-fold, 4-fold, 5- fold, 6- fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 50-fold, 100-fold, 500-fold, or 1,000-fold more than the binding moiety binds to any other target analyte of a plurality of target analytes.
  • a binding moiety can be deemed monospecific for a target analyte if the binding moiety is demonstrated to be able to bind to a specific target analyte at least 2 -fold, 3-fold, 4- fold, 5-fold, 6- fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 50-fold, 100-fold, 500-fold, or 1,000-fold more than the binding moiety binds to any other target analyte of a plurality of at least about 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,00 target analytes.
  • a binding moiety can be deemed monospecific for a target analyte if the number of sequence reads containing an address barcode sequence corresponding to that target analyte and a proximity barcode corresponding to the binding moiety is at least about 2-fold, 3-fold, 4-fold, 5-fold, 6- fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 50-fold, 100-fold, 500-fold, or 1,000-fold higher the number of sequence reads containing an address barcode sequence corresponding to any other target analyte of a plurality of target analytes and a proximity barcode corresponding to the binding moiety.
  • a binding moiety can be deemed monospecific for a target analyte if the number of sequence reads containing an address barcode sequence corresponding to that target analyte and a proximity barcode corresponding to the binding moiety is at least about 2- fold, 3-fold, 4-fold, 5-fold, 6- fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 50-fold, 100-fold, 500-fold, or 1,000-fold higher the number of sequence reads containing an address barcode sequence corresponding to any other target analyte of a plurality of at least about 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,00 target analytes and a proximity barcode corresponding
  • the methods and apparatus disclosed herein can be used to screen for various diseases or conditions, including an alteration in the state of the body or of some of the organs, interrupting or disturbing the performance of the functions and/or causing symptoms such as discomfort, dysfunction, distress, or even death to the person afflicted or those in contact with a person.
  • a disease or condition can also include a distemper, ailing, ailment, malady, disorder, sickness, illness, complain, interdisposition and/or affectation.
  • samples containing binding moieties from a diseased animal can be simultaneously screened for the binding moieties' ability to interact with target analytes on an array. These interactions can be compared to those of samples from individuals that are not in a disease state, not presenting symptoms of persons in the disease state, or presenting symptoms of the disease state.
  • the levels of target analytes in samples from a diseased animal can be simultaneously determined. These levels can be compared to those of samples from individuals that are not in a disease state, not presenting symptoms of persons in the disease state, or presenting symptoms of the disease state.
  • the methods, kits, and compositions described herein can be used in medical diagnostics, drug discovery, molecular biology, immunology and toxicology.
  • Arrays can be used for large scale binding assays in numerous diagnostic and screening applications.
  • the multiplexed measurement of quantitative variation in levels of large numbers of target analytes allows the recognition of patterns defined by several to many different target analytes.
  • the multiplexed identification of large numbers of interactions between target analytes and binding moieties allows for the recognition of binding and interaction patterns defined by several to many different interactions between target analytes and binding moieties. Many physiological parameters and disease-specific patterns can be simultaneously assessed.
  • One embodiment involves the separation, identification and characterization of proteins present in a biological sample. For example, by comparison of disease and control samples, it is possible to identify disease specific target analytes. These target analytes can be used as targets for drug development or as molecular markers of disease.
  • the level of a target analyte For many diagnostic and investigative purposes, it can be useful to determine the binding specificity and strength of the binding moiety. This application can be important for the discovery and diagnosis of clinically useful markers that correlate with a particular diagnosis or prognosis. For example, by monitoring a range of antibody or T-cell receptor specificities in parallel, one may determine the levels and kinetics of antibodies during the course of
  • novel markers and interactions between markers associated with a disease of interest can be developed by comparing normal and diseased samples, or by comparing clinical samples at different stages of a disease.
  • Detection a level of one or more target analyte or detection of interactions between binding moieties and target analytes can lead to a medical diagnosis.
  • the identity of a pathogenic microorganism can be established unambiguously by binding a sample of the unknown pathogen to an array containing many types of antibodies specific for known pathogenic antigens.
  • the sample can be a sample from a subject with a condition or disease.
  • a sample can be a diseased tissue or cell, such as a breast cancer, ovarian cancer, lung cancer, colon cancer, hyperplastic polyp, adenoma, colorectal cancer, high grade dysplasia, low grade dysplasia, prostatic hyperplasia, prostate cancer, melanoma, pancreatic cancer, brain cancer (such as a glioblastoma), hematological malignancy, hepatocellular carcinoma, cervical cancer, endometrial cancer, head and neck cancer, esophageal cancer, gastrointestinal stromal tumor (GIST), renal cell carcinoma (RCC) or gastric cancer tissue or cell.
  • GIST gastrointestinal stromal tumor
  • RRCC renal cell carcinoma
  • the sample can be from a subject with a disease or condition such as a cancer, inflammatory disease, immune disease, autoimmune disease, cardiovascular disease, neurological disease, infectious disease, metabolic disease, or a perinatal condition.
  • a disease or condition such as a cancer, inflammatory disease, immune disease, autoimmune disease, cardiovascular disease, neurological disease, infectious disease, metabolic disease, or a perinatal condition.
  • the disease or condition can be a tumor, neoplasm, or cancer.
  • the cancer can be, but is not limited to, breast cancer, ovarian cancer, lung cancer, colon cancer, hyperplastic polyp, adenoma, colorectal cancer, high grade dysplasia, low grade dysplasia, prostatic hyperplasia, prostate cancer, melanoma, pancreatic cancer, brain cancer (such as a glioblastoma), hematological malignancy, hepatocellular carcinoma, cervical cancer, endometrial cancer, head and neck cancer, esophageal cancer, gastrointestinal stromal tumor (GIST), renal cell carcinoma (RCC) or gastric cancer.
  • the colorectal cancer can be CRC Dukes B or Dukes C-D.
  • the hematological malignancy can be B-Cell Chronic Lymphocytic Leukemia, B-Cell Lymphoma-DLBCL, B-Cell Lymphoma-DLBCL-germinal center-like, B- Cell Lymphoma-DLBCL-activated B-cell-like, or Burkitt's lymphoma.
  • the disease or condition can also be a premalignant condition, such as Barrett's Esophagus.
  • the disease or condition can also be an inflammatory disease, immune disease, or autoimmune disease.
  • the disease may be inflammatory bowel disease (IBD), Crohn' s disease (CD), ulcerative colitis (UC), pelvic inflammation, vasculitis, psoriasis, diabetes, autoimmune hepatitis, Multiple Sclerosis, Myasthenia Gravis, Type I diabetes, Rheumatoid Arthritis, Psoriasis, Systemic Lupus Erythematosis (SLE), Hashimoto's Thyroiditis, Grave's disease, Ankylosing Spondylitis Sjogrens Disease, CREST syndrome, Scleroderma, Rheumatic Disease, organ rejection, Primary Sclerosing Cholangitis, or sepsis.
  • IBD inflammatory bowel disease
  • CD Crohn' s disease
  • UC ulcerative colitis
  • pelvic inflammation vasculitis
  • vasculitis vasculitis
  • psoriasis psoriasis
  • diabetes autoimmune hepatitis
  • Multiple Sclerosis Multiple Sclerosis
  • the disease or condition can also be a cardiovascular disease, such as atherosclerosis, congestive heart failure, vulnerable plaque, stroke, or ischemia.
  • the cardiovascular disease or condition can be high blood pressure, stenosis, vessel occlusion or a thrombotic event.
  • the disease or condition can also be a neurological disease, such as Multiple Sclerosis (MS), Parkinson' s Disease (PD), Alzheimer' s Disease (AD), schizophrenia, bipolar disorder, depression, autism, Prion Disease, Pick's disease, dementia, Huntington disease (HD), Down's syndrome, cerebrovascular disease, Rasmussen's encephalitis, viral meningitis, neuropsychiatric systemic lupus erythematosus (NPSLE), amyotrophic lateral sclerosis, Creutzfeldt- Jacob disease, Gerstmann-Straussler-Scheinker disease, transmissible spongiform encephalopathy, ischemic reperfusion damage (e.g. stroke), brain trauma, microbial infection, or chronic fatigue syndrome.
  • MS Multiple Sclerosis
  • PD Parkinson' s Disease
  • AD Alzheimer' s Disease
  • AD Alzheimer' s Disease
  • schizophrenia bipolar disorder
  • depression depression
  • autism autism
  • Prion Disease Pick's disease
  • dementia Huntington disease
  • HD Huntington disease
  • the condition may also be fibromyalgia, chronic neuropathic pain, or peripheral neuropathic pain.
  • the disease or condition may also be an infectious disease, such as a bacterial, viral or yeast infection.
  • the disease or condition may be Whipple' s Disease, Prion Disease, cirrhosis, methicillin-resistant staphylococcus aureus, HIV, hepatitis, syphilis, meningitis, malaria, tuberculosis, or influenza.
  • the disease or condition can also be a perinatal or pregnancy related condition (e.g. preeclampsia or preterm birth), or a metabolic disease or condition, such as a metabolic disease or condition associated with iron metabolism.
  • a substrate can be composed of any material which will permit coupling of an address polynucleotide and/or a target analyte, which will not melt or otherwise substantially degrade under the conditions used to hybridize and/or denature nucleic acids.
  • a substrate can be composed of any material which will permit coupling of an address
  • a substrate can be composed of any material which permit washing or physical or chemical manipulation without dislodging an address polynucleotide or target moiety from the solid support.
  • Substrates can be fabricated by the transfer of target analyte and or address
  • the techniques for fabrication of a substrate of the invention include, but are not limited to, photolithography, ink jet and contact printing, liquid dispensing and piezoelectrics.
  • the patterns and dimensions of arrays are to be determined by each specific application. The sizes of each target analyte spots may be easily controlled by the users.
  • a method of making a solid substrate can comprise contacting or coupling an address polynucleotide to a first discrete location of a discrete region on a solid support, and contacting or coupling a target analyte to a second discrete location of the discrete region on the solid support, wherein the target analyte is in proximity to the address polynucleotide.
  • the coupling can include any of the coupling methods described herein or otherwise known in the art.
  • a solid support is coated with an affinity ligand as described herein and contacting or coupling a target analyte thereto in proximity to an address polynucleotide.
  • a method of making an array can comprise contacting or coupling a first address polynucleotide to a first discrete location of a first discrete region on a solid support, and contacting or coupling a first target analyte to a second discrete location of the first discrete region on the solid support, wherein the first target analyte is in proximity to the first address polynucleotide; and contacting or coupling a second address polynucleotide to a first discrete location of a second discrete region on the solid support, and contacting or coupling a second target analyte to a second discrete location of the second discrete region on the solid support, wherein the second target analyte is in proximity to the second address polynucleotide.
  • a substrate may take a variety of configurations ranging from simple to complex, depending on the intended use of the array.
  • a substrate can have an overall slide or plate configuration, such as a rectangular or disc configuration.
  • a standard microplate configuration can be used.
  • the surface may be smooth or substantially planar, or have irregularities, such as depressions or elevations.
  • the substrates of the presently disclosed subject matter can include at least one surface on which a pattern of recombinant virion microspots can be coupled or deposited.
  • a substrate may have a rectangular cross-sectional shape, having a length of from about 10-200 mm, 40-150 mm, or 75- 125 mm; a width of from about 10-200 mm, 20-120 mm, or 25-80 mm, and a thickness of from about 0.01-5.0 mm, 0.1-2 mm, or 0.2 to 1 mm.
  • a support may be organic or inorganic; may be metal (e.g., copper or silver) or non- metal; may be a polymer or nonpolymer; may be conducting, semiconducting or nonconducting (insulating); may be reflecting or nonreflecting; may be porous or nonporous; etc.
  • a solid support as described above can be formed of any suitable material, including metals, metal oxides, semiconductors, polymers (particularly organic polymers in any suitable form including woven, nonwoven, molded, extruded, cast, etc.), silicon, silicon oxide, and composites thereof.
  • Suitable materials for use as substrates include, but are not limited to, polycarbonate, gold, silicon, silicon oxide, silicon oxynitride, indium, tantalum oxide, niobium oxide, titanium, titanium oxide, platinum, iridium, indium tin oxide, diamond or diamond-like film, acrylic, styrene-methyl methacrylate copolymers, ethylene/acrylic acid, acrylonitrile-butadiene-styrene (ABS), ABS/polycarbonate, ABS/polysulfone, ABS/polyvinyl chloride, ethylene propylene, ethylene vinyl acetate (EVA), nitrocellulose, nylons (including nylon 6, nylon 6/6, nylon 6/6-6, nylon 6/9, nylon 6/10, nylon 6/12, nylon 1 1 and nylon 12), polyacrylonitrile (P
  • polybutylene terephthalate PBT
  • PE poly(ethylene)
  • PP poly(propylene)
  • PP poly(propylene)
  • PB poly(butadiene)
  • PS polystyrene
  • PC polycarbonate
  • PECL poly(epsilon-caprolactone)
  • PMMA poly(methyl methacrylate) and its homologs, poly(methyl acrylate) and its homologs
  • PLA poly(lactic acid)
  • PLA poly(glycolic acid
  • polyorthoesters poly(anhydrides), nylon, polyimides, polydimethylsiloxane (PDMS), polybutadiene (PB), polyvinylene terephthalate
  • solid supports examples include polypropylene, polystyrene, polyethylene, dextran, nylon, amylases, glass, natural and modified celluloses (e.g.,
  • the solid support can be silica or glass because of its great chemical resistance against solvents, its mechanical stability, its low intrinsic fluorescence properties, and its flexibility of being readily
  • the substrate is glass, particularly glass coated with nitrocellulose, more particularly a nitrocellulose-coated slide (e.g., FAST slides).
  • a substrate may be modified with one or more different layers of compounds or coatings that serve to modify the properties of the surface in a desirable manner.
  • a substrate may further comprise a coating material on the whole or a portion of the surface of the substrate.
  • a coating material enhances the affinity of the target analyte, and address polynucleotide, or another moiety (e.g., a functional group) for the substrate.
  • the coating material can be nitrocellulose, silane, thiol, disulfide, or a polymer.
  • the substrate may comprise a gold-coated surface and/or the thiol comprises hydrophobic and hydrophilic moieties.
  • the substrate comprises glass and the silane may present terminal moieties including, for example, hydroxyl, carboxyl, phosphate, glycidoxy, sulfonate, isocyanato, thiol, or amino groups.
  • the coating material may be a derivatized monolayer or multilayer having covalently bonded linker moieties.
  • the monolayer coating may have thiol (e.g., a thioalkyl selected from the group consisting of a thioalkyl acid (e.g., 16- mercaptohexadecanoic acid), thioalkyl alcohol, thioalkyl amine, and halogen containing thioalkyl compound), disulfide or silane groups that produce a chemical or physicochemical bonding to the substrate.
  • thiol e.g., a thioalkyl selected from the group consisting of a thioalkyl acid (e.g., 16- mercaptohexadecanoic acid), thioalkyl alcohol, thioalkyl amine, and halogen containing thioalkyl compound
  • disulfide or silane groups that produce a chemical or physicochemical bonding to the substrate.
  • the attachment of the monolayer to the substrate may also be achieved by non-covalent interactions or by
  • the coating may comprise at least one functional group.
  • functional groups on the monolayer coating include, but are not limited to, carboxyl, isocyanate, halogen, amine or hydroxyl groups.
  • these reactive functional groups on the coating may be activated by standard chemical techniques to corresponding activated functional groups on the monolayer coating (e.g., conversion of carboxyl groups to anhydrides or acid halides, etc.).
  • Exemplary activated functional groups of the coating on the substrate for covalent coupling to terminal amino groups include anhydrides, N-hydroxysuccinimide esters or other common activated esters or acid halides
  • Exemplary activated functional groups of the coating on the substrate include anhydride derivatives for coupling with a terminal hydroxyl group; hydrazine derivatives for coupling onto oxidized sugar residues of the linker compound; or maleimide derivatives for covalent attachment to thiol groups of the linker compound.
  • at least one terminal carboxyl group on the coating can be activated to an anhydride group and then reacted, for example, with a linker compound.
  • the functional groups on the coating may be reacted with a linker having activated functional groups (e.g., N-hydroxysuccinimide esters, acid halides, anhydrides, and isocyanates) for covalent coupling to reactive amino groups on the coating.
  • a linker having activated functional groups e.g., N-hydroxysuccinimide esters, acid halides, anhydrides, and isocyanates
  • a substrate can contain a linker (e.g., to indirectly couple a moiety to the substrate).
  • a linker has one terminal functional group, a spacer region and a target analyte adhering region.
  • the terminal functional groups for reacting with functional groups on an activated coating include halogen, amino, hydroxyl, or thiol groups.
  • a terminal functional group is selected from the group consisting of a carboxylic acid, halogen, amine, thiol, alkene, acrylate, anhydride, ester, acid halide, isocyanate, hydrazine, maleimide and hydroxyl group.
  • the spacer region may include, but is not limited to, poly ethers, polypeptides, polyamides, polyamines, polyesters, polysaccharides, polyols, multiple charged species or any other combinations thereof.
  • Exemplary spacer regions include polymers of ethylene glycols, peptides, glycerol, ethanolamine, serine, inositol, etc.
  • the spacer region may be hydrophilic in nature.
  • the spacer region may be hydrophobic in nature.
  • the spacer has n oxy ethylene groups, where n is between 2 and 25.
  • a region of a linker that adheres to an address polynucleotide, target analyte, or other moiety is hydrophobic or amphiphilic with straight or branched chain alkyl, alkynyl, alkenyl, aryl, arylalkyl, heteroalkyl, heteroalkynyl, heteroalkenyl, heteroaryl, or heteroarylalkyl.
  • a region of a linker that adheres to an address polynucleotide, target analyte, or other moiety comprises a C 10 -C25 straight or branched chain alkyl or heteroalkyl hydrophobic tail.
  • a linker comprises a terminal functional group on one end, a spacer, a target analyte adhering region, and a hydrophilic group on another end.
  • the hydrophilic group at one end of the linker may be a single group or a straight or branched chain of multiple hydrophilic groups (e.g., a single hydroxyl group or a chain of multiple ethylene glycol units).
  • the a support can be planar. In some instances, the support can be spherical. In some instances, the support can be a bead. In some instances, a support can be magnetic. In some instances, a magnetic solid support can comprises magnetite, maghemitite, FePt, SrFe, iron, cobalt, nickel, chromium dioxide, ferrites, or mixtures thereof. In some instances, a support can be nonmagnetic. In some embodiments, the nonmagnetic solid support can comprise a polymer, metal, glass, alloy, mineral, or mixture thereof. In some instances a nonmagnetic material can be a coating around a magnetic solid suppoort.
  • a magnetic material may be distributed in the continuous phase of a magnetic material.
  • the solid support comprises magnetic and nonmagnetic materials.
  • a solid support can comprise a combination of a magnetic material and a nonmagnetic material.
  • the magnetic material is at least about 5, 10, 20, 30, 40, 50, 60, 70, or about 80 % by weight of the total composition of the solid support.
  • the bead size can be quite large, on the order of 100-900 microns or in some cases even up to a diameter of 3 mm. In other embodiments, the bead size can be on the order of 1-150 microns.
  • the average particle diameters of beads of the invention can be in the range of about 2 um to several millimeters, e.g., diameters in ranges having lower limits of 2 um, 4 ⁇ , 6 rn, 8 um, 10 ⁇ , 20 um, 30 ⁇ , 40 um, 50 um, 60 ⁇ , 70 um, 80 ⁇ , 90 ⁇ , 100 ⁇ , 150 ⁇ , 200 ⁇ , 300 ⁇ , or 500 um, and upper limits of 20 ⁇ , 30 um, 40 um, 50 um, 60 um, 70 ⁇ , 80 ⁇ , 90 ⁇ , 100 ⁇ , 150 um, 200 ⁇ , 300 um, 500 ⁇ , 750 ⁇ , 1 mm, 2 mm, or 3 mm.
  • a support or substrate can be an array.
  • a solid support comprises an array.
  • An array of the invention can comprise an ordered spatial arrangement of two or more discrete regions. Address, spot, microspot, and discrete region are terms used interchangeably and refer to a particular position, such as on an array.
  • An array can comprise target analytes located at known or unknown discrete regions.
  • An array can comprise address polynucleotides located at known or unknown discrete regions.
  • Each of two or more discrete regions can comprise an address polynucleotide.
  • Each of two or more discrete regions can comprise a target analyte.
  • Each of two or more discrete regions can comprise an address polynucleotide and a target analyte.
  • the two or more discrete regions of an array can comprise two or more first discrete locations and two or more second discrete locations.
  • Each first discrete location can comprise a coupled address polynucleotide.
  • Each second discrete location can comprise a target analyte.
  • An address polynucleotide in a discrete region can be in proximity to the target analyte within the same discrete region.
  • An address polynucleotide in a discrete region can be barcoded to the target analyte within the same discrete region.
  • An address polynucleotide can be used to identify the target polynucleotide in the same region.
  • an array can comprise a first discrete region comprising a first address polynucleotide and a first target analyte, and a second discrete region comprising a second address polynucleotide and a second target analyte.
  • an array can comprise a first discrete region comprising a first address polynucleotide at a first discrete location within the first discrete region and a first target analyte at a second discrete location within the first discrete region, and a second discrete region comprising a second address polynucleotide at a first discrete location within the second discrete region and a second target analyte at a second discrete location within the second discrete region.
  • Row and column arrangements of arrays can be selected due to the relative simplicity in making such arrangements.
  • the spatial arrangement can, however, be essentially any form selected by the user, and optionally, in a pattern.
  • Microspots of an array may be any convenient shape, including circular, ellipsoid, oval, annular, or some other analogously curved shape, where the shape may, in certain embodiments, be a result of the particular method employed to produce the array.
  • the microspots may be arranged in any convenient pattern across or over the surface of the array, such as in rows and columns so as to form a grid, in a circular pattern, and the like, where generally the pattern of spots will be present in the form of a grid across the surface of the substrate.
  • An array can comprise an ordered spatial arrangement of two or more target analytes, two or more address polynucleotides, or a combination thereof, on a solid surface.
  • an array can comprise at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 target analytes.
  • An array can comprise at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 antibodies specific for a target analyte.
  • the target analytes can be linked to the array by the antibodies.
  • an array can comprise at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 target analytes linked to the array by at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 antibodies specific for the target analytes.
  • an array can comprise at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 address polynucleotides.
  • an array can comprise at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 target analytes and at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 address polynucleotides.
  • An array can comprise an ordered spatial arrangement of two or more same or different target analytes, two or more same or different address polynucleotides, or a combination thereof, on a solid surface.
  • an array can comprise at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 same or different target analytes.
  • an array can comprise at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 same or different address
  • an array can comprise at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 same or different target analytes and at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 25,000, or 30,000 same or different address polynucleotides.
  • An array can be a high-density array.
  • a high-density array can comprise tens, hundreds, thousands, tens-of-thousands or hundreds-of-thousands of target analytes and/or address polynucleotides.
  • the density of microspots of an array may be at least about 1/cm 2 or at least about 10/cm 2 , up to about 1,000/cm 2 or up to about 500/cm 2 .
  • the density of all the microspots on the surface of the substrate may be up to about 400/cm 2 , up to about 300/cm 2 , up to about 200/cm 2 , up to about 100/cm 2 , up to about 90/cm 2 , up to about 80/cm 2 , up to about 70/cm 2 , up to about 60/cm 2 , or up to about 50/cm 2 .
  • an array can comprise at least 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, or 1,000 distinct antibodies per a surface area of less than about 1 cm 2 .
  • an array can comprise 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 350 or 400 discrete regions in an area of about 16 mm 2 , or 2,500 discrete regions/cm 2 .
  • target analytes, address polynucleotides, linkers, or another moiety in each discrete region are present in a defined amount (e.g., between about 0.1 femtomoles and 100 nanomoles).
  • an array can comprise at least about 2 target analytes and/or address polynucleotides per cm 2 .
  • an array can comprise at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 1 1,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 21,000, 22,000, 23,000, 24,000, 25,000, or more target analytes and/or address polynucleotides.
  • an array can be a high-density protein array comprising at least about 10 target analytes and/or address polynucleotides per cm 2 .
  • an array can comprise at least about 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 1 1,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 21,000, 22,000, 23,000, 24,000, 25,000, or more target analytes and/or address polynucleotides per cm 2 .
  • kits that find use in practicing the subject methods, as mentioned above.
  • a kit can include one or more of the compositions described herein.
  • a kit includes at least one proximity probe.
  • a kit can include at least one proximity polynucleotide.
  • a kit can include at least one address polynucleotide.
  • a kit can include at least one target analyte.
  • a kit can include at least one binding moiety.
  • a kit can include at least one splint polynucleotide.
  • a kit can include at least one proximity probe.
  • a kit can include at least one proximity polynucleotide.
  • a kit can include at least one address polynucleotide.
  • a kit can include at least one target analyte.
  • a kit can include at least one binding moiety.
  • a kit can include at least one splint polynucleotide.
  • a kit can include at least one proximity
  • a kit can include a reagent for coupling at least one proximity polynucleotide and at least one binding moiety.
  • a kit can include a solid support.
  • the solid support is already functionalized with at least one address polynucleotide and/or at least one target analyte. In some embodiments, the solid support is not functionalized with at least one address
  • kits can include a reagent for coupling at least one address polynucleotide to the solid support.
  • a kit can include a reagent for coupling at least one target analyte to the solid support.
  • a kit can include one or more reagents for performing amplification, including suitable primers, enzymes, nucleobases, and other reagents such as PCR amplification reagents (e.g., nucleotides, buffers, cations, etc.), and the like. Additional reagents that are required or desired in the protocol to be practiced with the kit components may be present.
  • suitable primers e.g., primers, enzymes, nucleobases, and other reagents
  • PCR amplification reagents e.g., nucleotides, buffers, cations, etc.
  • Additional reagents that are required or desired in the protocol to be practiced with the kit components may be present.
  • Such additional reagents include, but are not limited to, one or more of the following an enzyme or combination of enzymes such as a polymerase, reverse transcriptase, nickase, restriction endonuclease, uracil- DNA glycosylase enzyme, enzyme that methylates or demethylates DNA, endonuclease, ligase, etc.
  • an enzyme or combination of enzymes such as a polymerase, reverse transcriptase, nickase, restriction endonuclease, uracil- DNA glycosylase enzyme, enzyme that methylates or demethylates DNA, endonuclease, ligase, etc.
  • kits include two or more distinct sets of proximity probes, proximity polynucleotides, binding moieties, address polynucleotides, and/or target analytes.
  • kit components may be present in separate containers, or one or more of the components may be present in the same container, where the containers may be storage containers and/or containers that are employed during the assay for which the kit is designed.
  • the subject kits may further include instructions for practicing the subject methods. These instructions may be present in the subject kits in a variety of forms, such as printed information on a suitable medium or substrate (e.g., a piece or pieces of paper on which the information is printed), in the packaging of the kit, in a package insert, etc. Yet another means would be a computer readable medium (e.g., diskette, CD, etc.), on which the information has been recorded. Yet another means that may be present is a website address which may be used via the internet to access the information at a removed site.
  • a suitable medium or substrate e.g., a piece or pieces of paper on which the information is printed
  • a computer readable medium e.g., diskette, CD, etc.
  • a website address which may be used via the internet to access the information at a removed site.
  • ranges include the range endpoints. Additionally, every sub range and value within the rage is present as if explicitly written out.
  • the term “about” or “approximately” can mean within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, e.g., the limitations of the measurement system. For example, “about” can mean within 1 or more than 1 standard deviation, per the practice in the art. Alternatively, “about” can mean a range of up to 20%, up to 10%, up to 5%), or up to 1% of a given value.
  • the term can mean within an order of magnitude, within 5- fold, or within 2-fold, of a value.
  • a proximity polynucleotide is covalently crosslinked to a binding moiety, (e.g., a mAb) using a commercial kit (Solulink, Inc.).
  • a 3 '-amino-oligo proximity polynucleotide
  • Sulfo-S-4FB a 3 '-amino-oligo
  • mAb proteins are modified with S-HyNic groups.
  • the HyNic-modified mAb is reacted with the 4FB-modified proximity polynucleotide to yield a bis-arylhydrazone mediated conjugate. Excess 4FB-modified proximity polynucleotide can be further removed via magnetic affinity matrix.
  • the overall yield of the antibody-proximity polynucleotide conjugate is 30-50% based on the mAb recovery and is >95% free from HyNic- modified mAb and 4FB-modified polynucleotides.
  • the bis-arylhydrazone bond is stable to heat (e.g., 94°C) and pH (e.g., 3-10).
  • Example 2 Annotating an Address of a Target analyte
  • address polynucleotides are designed and individually synthesized with primary amine groups attached to their 5 '-ends and arrayed in 384-well titer dishes. An aliquot of the arrayed address polynucleotide is then added to the protein source plates also arrayed in the 384-well format.
  • polynucleotide comprising a unique barcode to form a barcoded array.
  • the splint polynucleotide brings together the proximity polynucleotide conjugated to the mAb and the address polynucleotide attached to the glass surface, leaving a nick between the two
  • DNA ligase is then added to covalently ligate the address polynucleotide and the proximity polynucleotide.
  • the ligation products are then PCR-amplified using universal primer pairs, followed by deep-sequencing to determine the sequences of the ligated products.
  • a mAb When a mAb recognizes a particular protein on a barcoded HuProt array, its barcode and the protein address barcode appear in the same sequence. By counting the number of the reads for the same sequences, the relative strength of the mAb can be determined. By counting the number of different address polynucleotide barcodes coexisting with a given mAb' s proximity polynucleotide barcode, binding specificity of the mAb can be determined.
  • polynucleotide (tethered with streptavidin).
  • streptavidin The resulting mixtures are transferred to a 384-well ELISA plate to allow immobilization of the total cell lysate proteins and address polynucleotides to the bottom of the well.
  • bovine serum albumin (BSA) is added to each well to block further absorption of proteins.
  • a group of antibodies (e.g., 100) that each specifically recognizes a particular protein is tethered with a unique probe polynucleotide either via Cys or Lys residues of the antibodies.
  • Probe polynucleotide-tethered antibodies are mixed and added to the ELISA plate.
  • the ELISA plate After incubation at RT, the ELISA plate is washed extensively. Next, the splint polynucleotide is added to each well to carry out the proximity ligation. After the ligation reaction, ligated products are PCR amplified, pooled, and subjected to deep-sequencing. Identity of each single cell is represented by the address polynucleotides; identity of proteins is revealed by the proximity polynucleotide. Similar to other DAPPL data, the number of each probe polynucleotide counts serves as the proxy of the protein concentration in each single cell detected by the corresponding antibodies.
  • the ligation products are amplified with PCR reactions, pooled, and subjected to deep-sequencing. Identity of each antibody and the corresponding single cell is represented by the address polynucleotides; DNA sequences chromatin immunoprecipitated by each antibody are revealed by sequencing from the opposite end of the PCR products. Similar to other DAPPL data, the number or counts of each immunoprecipitated sequence reveals the binding sites by each transcription factor that the corresponding antibodies recognize.
  • a multiplexed chromatin-precipitation coupled deep-sequencing method is for simultaneous detection of transcription factor binding sites in chromatin.
  • the principle of this idea is illustrated in FIG. 28.
  • First, multiple monoclonal antibodies against hundreds to thousands of human transcription factors are spotted onto a nitrocellulose-coated slide (e.g., anti- TF mAb array) with address polynucleotides.
  • Second, cells of interest are crosslinked, sheared, and ligated to a Y-shaped DNA adapter.
  • the ligated chromatin preps are incubated on the anti-TF mAb array to allow capture of the chromatin-DNA complexes by the mAbs spotted on the array.
  • the array is washed thoroughly, and the splint polynucleotide is added to facilitate ligation between captured genomic DNA fragments to the address polynucleotides at each mAb spot.
  • the splint polynucleotide is added to facilitate ligation between captured genomic DNA fragments to the address polynucleotides at each mAb spot.
  • polynucleotide tethered with streptavidin.
  • the resulting mixtures are transferred to a 384-well ELISA plate to allow immobilization of the total cell lysate proteins and address polynucleotides to the bottom of the well (FIG. 29; left panel).
  • BSA is added to each well to block further absorption of proteins.
  • a group of antibodies e.g., 100 that each specifically recognizes a particular protein is tethered with a unique proximity
  • the proximity polynucleotide- tethered antibodies are mixed and added to the ELISA plate. After incubation at RT, the ELISA plate is washed extensively. Next, the splint polynucleotide is added to each well to carry out the proximity ligation. After the ligation reaction, ligated products are PCR amplified, pooled, and subjected to deep-sequencing. Identity of each single cell is represented by the address polynucleotides; identity of proteins is revealed by the proximity polynucleotides. Similar to other DAPPL data, the number of each probe polynucleotide counts serves as a measure of the protein concentration in each single cell detected by the corresponding antibodies.
  • each FACS- sorted single cell is collected in a 96-well, plated, lysed, mixed with a particular address polynucleotide, and transferred to another 96-well plate, in which each well is already coated with anti-pTyr antibodies (e.g., 4G10 and pTyrlOO) and an address polynucleotide. After overnight incubation, a mixture of proximity polynucleotide-tethered antibodies that each recognizes a particular protein is added to each well and allowed to incubate at RT for 2 hr.
  • anti-pTyr antibodies e.g., 4G10 and pTyrlOO
  • AO Address polynucleotides
  • the ligation products are amplified with PCR reactions, pooled, and subjected to deep-sequencing. Identity of each antibody and the corresponding single cell are represented by the address polynucleotides; DNA sequences immunoprecipitated by each antibody are revealed by sequencing from the opposite end of the PCR products. Similar to other DAPPL data, the number or counts of each immunoprecipitated sequence reveal the binding sites by each transcription factor that the corresponding antibodies recognize.
  • a DAPPL-based approach to enable quantitative detection of histone PTM abundance in a single cell is performed.
  • a generic anti-histone antibody is first used to coat each well with a particular address polynucleotide in a 96-well dish.
  • Each FACS- sorted single cell is collected in another 96-well plate, lysed, and transferred to histone antibody- coated plates.
  • a mixture of anti-histone mark mAbs e.g., anti-H3K27Ac and -H3K4me3
  • each tethered with a particular proximity polynucleotide is added to each well.
  • AO Address polynucleotides
  • a DAPPL approach is applied to select DNA/RNA aptamers that can recognize human transcription factors and protein kinases with mono-specificity and high affinity.
  • 20- and 40-mer random DNA polynucleotides with fixed flanking sequences on both sides are used. They are heated at 98°C for 10 min and slowly cooled down to allow formation of secondary or tertiary structures (FIG. 31A). The mixture of DNA aptamers is incubated on the human TF array at 4°C overnight. After several washes, the bound DNA aptamers are recovered from the slide and PCR-amplified.
  • asymmetric PCR is performed using an aliquot of the PCR products to regenerate DNA aptamers, which go through the same procedure 4 times.
  • the bound DNA aptamers are ligated to the address polynucleotides spotted on the TF array and an aliquot of the recovered ligation products is deep-sequenced.
  • the same procedure is continued for two more cycles, and at the ends of cycles 6 and 7, the ligated products are deep-sequenced.
  • mono-specific DNA aptamers of high affinity can be selected.
  • the same approach is applied to identify aptamers on HuProt arrays.
  • a summary of the sequencing data can be seen in the table below. 1 olsil rends (S Ken (Is w it h # ⁇ unique # ⁇ unique # of
  • RNA aptamers are screened against human proteins kinases (>500 proteins, including some splice variants). The procedure is almost the same as above, except a step of in vitro transcription after PCR amplification is added to convert double-stranded DNA templates to RNA aptamers. Similarly, recovered RNA aptamers at the end of cycles 5, 6, and 7 are deep- sequenced to reveal their identity.
  • DNA aptamers to perform Western analysis (WB), immunoprecipitation (IP), chromatin precipitation (ChIP), and/or immunohistochemistry analysis (IHC), is performed by selecting a small random set ⁇ e.g. , 20-40) of identified aptamers and synthesis of them with a biotin moiety attached to their 5'- or 3 '-ends. Using HRP-conjugated streptavidin, these DNA aptamers are tested in at least 6 cell lines. A single band in WB analysis with a particular aptamer is optimal. Cell lines transfected with FLAG-tagged target constructs are used to perform aptamer-assisted IP assay. The success of IP is detected with anti-FLAG WB. Similarly, the success of aptamer-assisted ChIP is determined by comparison between the
  • RNA aptamers can effectively inhibit autophosphorylation activity of their targets.
  • autophosphorylation assays with ⁇ - 2 ⁇ - ⁇ on a kinase array in the presence or absence of the mixture of identified RNA aptamers is performed.
  • purified kinase proteins are spotted on an epoxy surface to form a kinase array.
  • the immobilized kinases are preincubated with a mixture of identified RNA aptamers at different concentrations for 1 hr. at RT.
  • Kinase reaction buffer containing ⁇ - 2 ⁇ - ⁇ are added to the kinase array and the autophosphorylation reactions are carried out for 20 min at 30°C.
  • the autophosphorylation signals are detected by exposure of the kinase array to a piece of X-ray film.
  • a kinase array without the RNA aptamer treatment is carried out in parallel, signals of which are used as positive controls.
  • Those RNA aptamers that can significantly reduce autophosphorylation signals are selected for further in vivo validation.
  • Kinases that are well- studied with known downstream substrates are selected for the in vivo validation. Their corresponding RNA aptamers are overexpressed in cell lines by transfecting constructs carrying the cDNAs of the RNA aptamers.
  • the kinase's autophosphorylation level and the phosphorylation level of the kinase's downstream targets are expected to be reduced as detected with a phospho-specific antibody, when an RNA aptamer effectively inhibits its target kinase in cells.
  • a DAPPL approach is also applied for detection of small molecule and protein interactions.
  • a protein array with Src and IDE as positive controls is generated, and 10 other random proteins as negative controls. Each protein is spotted with a unique address
  • Two DNA-templated macrocycles are first tested individually on the array at a wide range of concentration ⁇ e.g., pM to attoM). Interactions with their expected targets ⁇ e.g., Src and IDE) are confirmed by Sanger sequencing the PCR products of the ligated products. The two small molecules are tested again if successful in the context of a compound mixture.
  • V H synthetic heavy chain variable
  • V L light chain variable
  • polynucleotide sequence encoding Hisx6 and FLAG epitopes is added to the 3 '-end of the polynucleotide.
  • a protein pool of either V H or V L single domains is generated that are tethered with its coding RNA sequence via a puromycin moiety. Reverse transcription is applied to create the cDNA strands. This mixture is incubated on the human protein microarray. After stringent washes, the captured single domains are recovered from the microarray and the tethered cDNA is PCR-amplified. The PCR products serve as the templates for the next round of screen. This screen will be performed for 6 cycles.
  • the cDNAs tethered to the single domains are ligated to the address polynucleotide spotted together with each protein on the array, followed by PCR amplification.
  • the PCR products are deep-sequenced to determine which protein on the array is recognized by which single domain.
  • aptamers are used to perform a comprehensive ChIP assay against all human TFs at once with a mixture of their corresponding aptamers.
  • a set of oligos that each encode a particular aptamer sequence flanked by two fiexed sequence tags is generated first.
  • a biotin moiety is added to either the 5'- or 3 '-end of the aptamers.
  • Equal amounts of all the synthesized aptamers are mixed together to various final concentrations (e.g., 10 nM to 1 ⁇ ). Meanwhile, standard chromatin preparation are carried out in selected cell lines (e.g., ENCODE cell lines).
  • the shared genomic DNAs are end-repaired and ligated to a Y-shaped DNA adapter.
  • the aptamer mixture is added to the chromatin preps and allowed to incubate overnight.
  • the mixture is diluted at least 10-fold and a splint polynucleotide is added to faciltate ligation between the aptamer ends (e.g., 5'- or 3 '-fixed sequences) and the single- stranded sequences of the Y-shaped adapters.
  • streptavidin-coated beads are added to the mixture to capture the aptamer- TF-DNA complexes via the biotin moiety on the aptamers.
  • the beads are washed under stringent conditions and the bound DNA ligation products are recovered by boiling the beads.
  • the ligation products are PCR-amplifed and subjected to Hi-seq analysis to identify chromatin locations to which each TF bind.
  • the ChlP-omix assays are performed in both labeling orientations. Given the complexity of the chromatin distribution of the TFs as a whole, a single run of Hi-seq (e.g., 300 M reads) may not be sufficient to fully cover all the possibilities. If this is the case, more Hi-seq runs are used to generate up to 3 billion reads.
  • Hi-seq e.g. 300 M reads
  • the ChIP peaks identified with the ChlP-omix approach are exepected to show a significant overlap with those identified with the traditional antibody-based ChlP-seq approach.
  • the identified DNA/RNA aptamers are used for proteome-wide detection of protein abundance inside a cell or tissue (FIG. 8).
  • the identity of captured aptamers can be revealed by hybridiuzing recovered aptamer, end-labeled with a fluorphore (e.g., Cy5), to a DNA polynucleotide array that encodes the complementary sequences of the entire aptamer set used in this assay.
  • the number of reads per aptamer serve as a determination of a relative abundance of the targeted proteins.
  • a foreign protein e.g., GFP
  • V5-tag is spiked into the lysate at a known concentration (e.g., 1 nM).
  • An aptamer recognizing V5 epitope is included in the aptamer mixture to serve as a
  • the previously identified aptamers are used to determine protein-protein interactions by testing all possible combinations in a cell line and/or tissue.
  • the an exemplary IP-omix method utilizes a mixture of DNA/RNA aptamers that each recognizes a unique human protein to examine all possible combinations of protein-protein interactions inside a cell or tissue.
  • total protein lysates obtained using standard protocols from cultured cell lines or primary tissues are lightly biotinylated.
  • Second, a mxiture of hundreds to thousands of aptamers are mixed and added to the lysates. When two proteins form a complex inside the cell, their corresponding aptamers are brought to proximity.
  • a splint polynucleotide is added to the mixture and allowed to aneal to the 5'- and 3 '-ends of the fixed sequences on the aptamers.
  • the two aptamers bound to the same protein complex form an aptamer dimer.
  • biotinylated protein complexes are purified with streptavidin beads and washed under stringent conditions. The ligated aptamer dimers are then recovered from the beads and PCR- amplified.
  • PCR products are deep-sequenced to identify all possible aptamer pairs.
  • standard co-IP experiments are performed using a random set of candidate protein pairs (e.g., 20-40 pairs).
  • candidate protein pairs e.g. 20-40 pairs.
  • cells are co-transfected with X and Y expression constructs each tagged with a different epitope (e.g., V5 and FLAG).
  • epitope e.g., V5 and FLAG
  • Peptide ligands for 128 orphan GPCRs are identified by performing high-throughput screening against a 10-mer random peptide library in a microarray format (e.g., a human GPCR VirD array).
  • a microarray format e.g., a human GPCR VirD array.
  • Class A is the largest with 79 orphans, such as those for somatostatin, relaxin, prokineticin, and peptide ligands. Therefore, these 79 orphan GPCRsre included.
  • GPCR ORFs are selects from a human ORFeome collection to generate recombinant viruses. Individually purified virions are spotted on a glass slide to form an orphan GPCR VirD array.
  • a peptide library comprised of random 10-mer peptide sequences is constructed using the mRNA-display method.
  • a 10-mer peptide pool contains >lxl0 13 peptide species, much more complex than phage- or bacteria- display libraries.
  • a pool of DNA oligo templates is synthesized, each encoding a 30-mer random nucleotide sequence flanked by an upstream T7:Kozak sequence to facilitate in vitro
  • step 4 in cycle 6 the cDNA-mRNA-peptide conjugate are treated with RNase to remove the RNA moiety.
  • the captured cDNA-peptide conjugates are ligated to the address polynucleotides.
  • the ligation products After recovery of the ligation products from the VirD array, they are PCR-amplified and a fraction of the products are deep-sequenced (FIG. 8; step 7). A fraction of the remaining product serves as the template for cycle 7 screen using the same procedure as cycle 6.
  • Bioinformatics analysis sithen performed to identify statistically enriched peptide-GPCR interactions. The selection is performed against 128 orphan GPCRs in parallel.
  • GPCR ligands identified above are confirmed with a different system.
  • a heterologous cell-based Ca 2+ imaging assay is employed for further characterization of these identified peptide ligands.
  • At least 5 positive orphan GPCRs ⁇ e.g., Mrgs) coming out of the VirD array assays are validated employing Ca 2+ imaging assays.
  • Heterologous cells, which do not express endogenously a GPCR-of-interest are used. The parental cells without GPCR expression are included in the experiments as negative controls.
  • agonist versus antagonist ligands are identified, respectively.
  • validated ligands are counter-screened in heterologous cells expressing unrelated GPCR, and in parental cells to ensure target specificity.
  • the DAPPL and VirD approaches are employed to select DNA/RNA aptamers that can recognize human transmembrane proteins with mono-specificity and high affinity.
  • 20- and 40-mer random DNA polynucleotides are generated with fixed flanking sequences on both sides. They are heated at 98°C for 10 min and slowly cooled down to allow formation of secondary or tertiary structures (FIGs. 7A and 7B).
  • a VirD array comprised of human GPCRs (e.g., -300 non-odorant GPCRs), 300 ion channels, and 100 members of immunoglobulin superfamily is constructed.
  • the mixture of DNA aptamers is incubated on the human transmembrane VirD array (-700 recombinant virions) at 4°C overnight. After several washes, the bound DNA aptamers are recovered from the slide and PCR-amplified. Next, asymmetric PCR is performed using an aliquot of the PCR products to regenerate DNA aptamers, which goes through the same procedure 4 times. At the DNA aptamer incubation step on the VirD array in cycle 5, the bound DNA aptamers are ligated to the address polynucleotides spotted on the VirD array and an aliquot of the recovered ligation products is deep-sequenced.
  • DAPPL-assisted virion (VirD) technology is employed to identify aptamers that can recognize conformational epitopes in the ecto-domains of 58 receptor tyrosine kinases (RTKs).
  • RTKs 58 receptor tyrosine kinases
  • a virion-displayed RTK VirD array is generated.
  • a mixture of DNA aptamers is incubated on the human RTK VirD array at 4°C overnight. After several washes, the bound DNA aptamers are recovered from the slide and PCR-amplified.
  • asymmetric PCR is performed using an aliquot of the PCR products to regenerate DNA aptamers, which goes through the same procedure 4 times.
  • the bound DNA aptamers are ligated to the address polynucleotides spotted on the VirD array and an aliquot of the recovered ligation products is deep-sequenced. The same procedure continues for two more cycles, and at the ends of cycles 6 and 7, the ligated products are deep-sequenced. By comparing the deep-sequencing data obtained at cycles 5, 6, and 7, mono-specific DNA aptamers of high affinity are selected. [00374] To determine whether a positive aptamer activates or inhibits its target RTK, a cell- based system is employed.
  • a given aptamer can block the corresponding RTK signaling
  • pretreatment of cells with this aptamer abolishes/reduces autophosphorylation signals of the RTK as judged by Western Blot (WB) analysis with antibodies that specifically recognize the autophosphorylated form of the RTK.
  • WB Western Blot
  • an aptamer can activate the RTK signaling, incubation with this aptamer in the absence of the canonical ligand is sufficient to induce
  • a highly multiplexed platform for inhibitor screens against human ion channels is also employed. Because the VirD technology offers a cell-free system, multiple ion channels can be simultaneously screened against a compound library, allowing for both specific target screen and simultaneous counter- screens against all other ion channels. As the viral envelope is almost identical to plasma membrane, ion channels displayed on virions are functional. 10 sodium and 55 (40 voltage-gated and 15 inwardly rectifying) potassium channels are used. Opening of these channels can be readily detected by a high-content imaging system using fluorescent dyes as a reporter. Such a screen scheme has established using several high-content, automated imaging systems, such as BD Pathway Imager.
  • a robotic microarrayer (NanoPrint, Arraylt) is used to spot a total of 65 virion-displayed ion channels in duplicate at the bottom of wells in a 96-well plate (FIG. 34); WT virions are included as negative controls.
  • WT virions are included as negative controls.
  • the dyes e.g., ANG-2 for sodium channel imaging
  • 3,280 compounds Sigma LOP AC and Microsource Spectrum
  • a compound that causes a signal reduction >3 standard deviations in activity is scored as a hit.
  • Z-scores are calculated for each interaction. To avoid potential side effects, those hits that only specifically inhibit a single channel in the assays are selected for further validation.
  • PPC population patch clamp
  • FIG. 19A and FIG. 19B A DAPPL approach was applied to establish a comprehensive screen for protein-DNA interactions. As illustrated in FIG. 19A and FIG. 19B, -1,600 human TF proteins were each mixed with a unique address polynucleotide and spotted to form a TF protein microarray.
  • a pool of DNA polynucleotides was synthesized that each contains eight random nucleotides with a CpG in the middle and two fixed sequences on both sides.
  • bacterial enzyme Sssl is used to methylate the CpG, followed by treatment with a T4 DNA polymerase to generate a 5'- overhang.
  • This pool of 65,536 species was then incubated on the TF array, washed, and the captured DNA fragments were ligated to the address polynucleotides.
  • a mCpG containing 8-mer sequence is now connected to an address oligo representing the TF protein that has captured this 8-mer in the binding assay. Therefore, it is an "all-to-all" screening approach. In other words, 4 8 (i.e., 65,536) 8-mer species can be screened simultaneously against 1,620 TFs in a single experiment, representing 1 x 10 9 combinations. The ligated products were then recovered from the slide and PCR-amplified. This pool of DNA was then re-screened using the same protocol for a total of six cycles, and the products of cycles 4-6 were deep-sequenced. Bioinformatics analyses were performed to determine whether there was any enrichment of consensus motifs for each TF protein.
  • Address oligos were designed such that there were at least 3 nt differences between all other address oligos and were designed to avoid self-annealing. A total of 22, 168 address oligos were designed and screen against 1,632 TF proteins. For some of these experiments, address oligos that differed by at least 4 nts were utilized. The initial complexity of the DNA aptamer library was about lxl 0 19 . A computer algorithm was designed to extract sequences with an address oligo (AO) and a 40-mer DNA aptamer sequence with fixed regions as shown in FIG. 35A
  • Example 21 Global mapping of interactions between DNasel hypersensitive sites (DHS) and nuclear proteins
  • Enhancers are highly enriched in DHSs. DHS-nuclear protein interactions are comprehensively profiled using a DAPPL approach. To capture dynamic changes of the 4-D nucleome, different DHS pools are obtained during a time course of matrix-compliance-induced morphological changes of mouse embryonic stem (ES) cells. Each DHS pool is separated in an ultra-centrifuge in order to recover DHS species around 150 bps. The recovered DHSs are end- fixed and ligated to a Y-shaped adapter DNA. Each DHS pool is incubated on a human protein microarray containing -4,200 nuclear proteins each spotted with a unique address
  • a splint polynucleotide is added to the array that anneals to the constant region at one end of the address oligo and to the single-stranded sequence of the adapter.
  • the ligated DNA is recovered, PCR-amplified, and deep-sequenced using Hi-Seq. Bioinformatics analysis of the sequences is used to determine which DHS sequence is recognized by which nuclear protein(s).
  • the resulting DHS-protein interaction networks obtained at different time points in the process of matrix-compliance- induced morphological changes is compiled together, and global DHS-protein interaction network with a temporal resolution is generated. Selected predictions (e.g., important TF protein candidates) made from these networks are examined using traditional methodologies.
  • nucleosome pools are obtained during a time course of matrix-compliance-induced morphological changes of mouse ES cells. Each nucleosome pool is separated to recover nucleosome species. After coupling a proximity polynucleotide is coupled to the nucleosomes, each nucleosome pool is incubated on a human protein microarray containing -4,200 nuclear proteins each spotted with a unique address polynucleotide. After washing, a splint polynucleotide is added to the array that anneals to the constant region at one end of the address oligo and to the single-stranded sequence of the adapter.
  • nucleosome-protein interaction networks obtained at different time points in the process of matrix-compliance- induced morphological changes is compiled together, and global nucleosome-protein interaction network with a temporal resolution is generated. Selected predictions made from these networks are examined using traditional methodologies.
  • RNA sequences on a protein microarray comprised of -1,600 TF and -1,000 annotated RNA-binding proteins.
  • a DNA polynucleotide pool was synthesized that each contains a 12-mer random sequence with a T7 sequence and a fixed sequence to its 5'- and 3 '-ends.
  • in vitro transcription was performed to generate the RNA molecules with a complexity of -16 million. This mixture of RNAs was incubated on the protein microarray and, after stringent washes, the captured RNA molecules were ligated to the free 5 '-end of the address
  • RNA-binding proteins complementary DNA strand of the ligated DNA-RNA fragments, followed by PCR- amplification with a primer pair that adds back the T7 primer sequences to the ds-DNA templates.
  • the recovered DNA templates were subjected to the same screening process for 5 more cycles and the ligated products from cycles 4 to 6 were deep-sequenced. Similar bioinformatics analyses were performed to determine whether statistically significant consensus RNA motifs were obtained for the RNA-binding proteins.
  • the recovered ligation products between the RNA probe and the address polynucleotides from two RNA-binding proteins, MSI1 and QK1PCR amplification could be readily PCR amplified as demonstrated by probing separately for their known RNA sequences. Sanger sequencing confirmed the expected ligation sequences (FIG. 25C).
  • Example 24 PTM-omix approach to globally quantify posttranslationally modified (PTM) human proteome in cells and tissues
  • Identified DNA/RNA aptamers can be used to enable proteome-wide detection of posttranslationally modified human proteome inside a cell or tissue.
  • Total protein lysates are obtained using standard protocols from cultured cell lines or primary tissues and are lightly biotinylated. A mixture of hundreds to thousands of aptamers that each specifically recognized a PTM-modified proteins are mixed and added to the lysates. After incubation at RT, the aptamer- protein complexes will be purified using streptavidin beads, followed by stringent washes. After recovery of the aptamers captured by proteins, they are PCR-amplified and deep-sequenced.
  • the number of reads per aptamer serves as the proxy for the relative abundance of the PTM- modified proteins.
  • a foreign protein e.g., e.g., GFP
  • a V5-tag is spiked into the lysate at a known concentration (e.g., 1 nM).
  • An aptamer recognizing V5 epitope is included in the aptamer mixture to serve as a normalization control.
  • Example 25 Aptamer-based perturbation in cells and tissues
  • RNA aptamers that each encodes specific inhibition activity against a particular enzyme (e.g., protein kinases, phosphatases, (de)acetyltransferases, deubiquitinases, etc.) are cloned into an inducible mammalian expression constructed and transfected to human cell lines. Upon induction, the encoded RNA aptamers are expressed and targeted to their corresponding enzyme target, and result in inhibition of the enzyme activity.
  • Each DNA/RNA aptamer with specific inhibition activity against a particular enzyme is packaged into viruses and transfected into human cell lines or tissues to inhibit one or many enzymes of interest.
  • Example 26 Aptamer-based scaffolds to dictate protein-protein and enzyme-substrate interactions in cells and tissues
  • aptamers either DNA or RNA
  • a polynucleotide link as a single molecule to create a dimeric or multimeric aptamer scaffold, a tailor-made molecular scaffold that can be used to dictate formation of protein homo- or heterodimers.
  • the aptamer scaffold brings the two desired proteins into proximity and facilitates homo- or heterodimeric protein complex formation or promotes enzyme- substrate interactions (FIG. 11).
  • an aptamer scaffold can act as a molecular chaperon to deliver its cargo, namely the protein that interacts with the other aptamer of the dimeric aptamer scaffold, to the desired subcellular compartment, such as the mitochondria, Golgi, lipid rafts, to name a few (FIG. 11).
  • this aptamer scaffold can be used to deliver chromatin modification enzymes, such as histone acetylases, deacetylases, methylases, demethylases, and DNA methylases, to particular chromatin regions and modify the local histones or DNAs (FIG. 11).
  • chromatin modification enzymes such as histone acetylases, deacetylases, methylases, demethylases, and DNA methylases
  • a desired protein complex is dictated to form inside a cell (FIG. 11).
  • a group of metabolic enzymes when brought together in the order of the metabolism cascade via this type of multimeric aptamer, it may greatly enhance the production of the desired metabolites.
  • a series of protein kinases in the same signaling cascade such as MAP kinases, are brought together to form a multi-member kinase sink that may greatly amplify the phosphorylation signals in cells.
  • the multimeric aptamer serves as a signaling dock to facilitate signal transduction.
  • immunoglobulins such as IgG/IgM/IgA/IgE, are isolated from serum samples collected from a cohort of patients and healthy controls (e.g., >30 subject in each category), using Protein A/G or L conjugated beads. After a stringent wash step to remove nonspecific proteins, the captured immunoglobulins are eluted at low pH (e.g., glycine-HCl, pH 2). Each immunoglobulin mixture of a particular subject is mixed with a unique address
  • a mixture of DNA/RNA aptamers with fixed sequences flanking the variable regions will be pre-incubated with a mixture of commercial human IgG/IgM/IgA/IgE at RT for 1 hr, followed by adding Protein A/G or L conjugated beads to deplete those aptamers that can directly recognize these immunoglobulin.
  • the depleted aptamer pool are added to the array and incubated in the presence of a mixture of human IgG/IgM/IgA/IgE for 1-3 hr at RT, in order to further eliminate aptamers that can recognize human immunoglobulin (FIG. 10).
  • This step allows formation of a sandwich-like complex, comprised of autoantibodies immobilized on the surface, captured antigens in the middle, and aptamers on top.
  • stringent washes e.g., 300 mM NaCl with 0.1% Tween 100 at pH 7
  • the captured aptamers are recovered and PCR-amplified.
  • the recovered aptamers are regenerated and probed to the same autoantibody array.
  • the same procedure can be repeated for at least 4-6 times.
  • DAPPL reactions are carried out to ligate address polynucleotides to the captured aptamer sequences and the products are deep-sequenced.
  • the address polynucleotide represents the identity of the patient or healthy subject tested; whereas the aptamer sequence(s) ligated to the address polynucleotide indicates the autoantigen(s) that it recognizes. Because in each sequence read an aptamer sequence to ligated to an address polynucleotide, representing a subject identity (e.g., an IBD patient or healthy subject), its sensitivity and specificity are readily known.
  • a subject identity e.g., an IBD patient or healthy subject
  • each identified aptamer is individually synthesized, pooled, and probed to the HuProt® array. After ligation between the captured aptamers and the address polynucleotides takes place on the HuProt® array, the ligation products are recovered, PCR-amplified, and deep-sequenced. Alternatively, the identity of captured aptamers can be revealed by hybridiuzing recovered aptamer, end- labeled with a fluorphore (e.g., Cy5), to a DNA polynucleotide array that encodes the complementary sequences of the entire aptamer set used in this assay.
  • a fluorphore e.g., Cy5
  • this aptamer In the case that a given aptamer fails to recognize any protein on the HuProt® array, presumably due to a lack of proper protein posttranslational modifications, this aptamer is resynthesized with an affinity tag (e.g., biotin) used to pull down protein(s) directly from total proteins extracted from cell lines or tissues.
  • an affinity tag e.g., biotin
  • the identity of the captured proteins can be revealed by mass spectrometry (e.g., MS/MS).
  • mass spectrometry e.g., MS/MS.
  • total proteins extracted from tumors of cancer patients are used to incubate with the autoantibody arrays instead of total protein lysates.
  • the mixture of total proteins extracted from the microbiome of an IBD patient cohort is incubated on the autoantibody arrays, comprised of purified IgG/IgM/IgA/IgE from IBD patients and healthy controls.
  • the identified disease-specific aptamer(s) is synthesized with an affinity tag (e.g., biotin) and used to pull down the candidate antigens from the total proteins extracted from the microbiome of IBD patients.
  • the identity of the antigens can be revealed by mass spectrometry (e.g., MS/MS).
  • MS/MS mass spectrometry
  • immunoglobulin in IBD patients may tightly bind to human proteins (e.g., autoantigens) and therefore, some aptamers may unavoidably recognize these human autoantigens. Because these autoantigens are also valuable in IBD diagnosis or prognosis, all the identified aptamers are probed to the HuProt® arrays to determine the autoantigens using the same approach as described above.
  • human proteins e.g., autoantigens
  • Example 28 Alternative aptamer-protein screening methodology
  • FIG. 26A A DAPPL approach is applied to identify high affinity antibodies (FIG. 26A) or protein (FIG. 27A) binding partners that interact with protein targets of interest.
  • ELIS A plate was prepared by coating the surface of a 384 well-PCR plate with a 0.01% poly-L-Lysine solution and incubating for 20 min. The plate was then rinsed with water and allowed to dry. 10 ⁇ of a biotinylated address oligo mixture was mixed with streptavidin (1.7 ⁇ ) and capture antibody (500ng) and incubated for lhr (FIG. 26E, bottom). The Klenow polymerase was added with a primer (FIG. 26F, left panel, right to left arrow; and FIG.
  • the "chew-back" reaction was mediated by T4 DNA polymerase to generate cohesive ends to the AG overhang.
  • 20 ⁇ of IL-10 antigen (100 pg) was added and incubated for 2 hrs and washed with lx PBST for 5 min twice.
  • 20 ⁇ of Oligo-labeled detection Antibody was incubated for 2 hrs and washed with lx PBST twice, then rinsed with water for 5 min. Ligation was carried out at RT for 1 hr and washed with lx PBST for 10 min twice, followed by a water wash. The plate was then twice heated for 20 min. Ligated products were harvested and transferred to a PCR tube (around 20 ⁇ ).
  • a 1 PCR reaction was performed (30 cycles); PCR products were separated by gel electrophoresis, and then purified.
  • a 2 nd PCR reaction was then performed (35 cycles) using inner nested primers. PCR products were separated by gel electrophoresis, purified, ligated, and transformed into bacteria. DNA from the transformed bacteria was then sequenced and analyzed.
  • FKBP1 A is a protein known to bind FK506 and rapamycin.
  • GST-tagged FKBPIA and GST (negative control) were labeled with different barcoded oligos.
  • FK506 and rapamycin were printed on an array with different address oligos along with a negative control (printing buffer).
  • GST-tagged FKBPIA and GST were probed on the array for peptidyl-prolyl isomerase (PPI) activity.
  • DAPPL methods were carried out after binding PCR amplification of the DAPPL product was then performed. The PCR products were then cloned into E.coli and verified by sequencing
  • the array was blocked with 5% BSA in PBS for 1 hour.
  • the address oligos on the array and the barcode oligos on FKBPIA and GST were then filled in by Klenow enzyme to generate double stranded DNA.
  • the "chew-back" reaction was mediated by T4 DNA polymerase to generate cohesive ends to the AG overhang.
  • a mixture of FKBPIA and GST was incubated on the array for 1 hour.
  • the array was then washed with lx TBST for 10 mins 3 times then dried. Ligation was carried out at RT for lhr and washed twice with lx TBST+lOmM EDTA for 10 min followed by a water wash.
  • nitrocellulose membrane on the array was harvested and transferred to a 1.5 ml tube. 30 ⁇ of ddH 2 0 was added and boiled for 10 min. The tube was spun and the supernatant was transferred to a new tube (PCR template). A 1 PCR reaction was performed (30 cycles); PCR products were separated by gel electrophoresis, and then purified. A 2 nd PCR reaction was then performed (35 cycles) using inner nested primers. PCR products were separated by gel electrophoresis, purified, ligated, and transformed into bacteria. DNA from the transformed bacteria was then, mini-prepped, sequenced and analyzed.
  • Example 31 Phospho-specific aptamer screens.
  • RNA aptamer screening was performed against human kinases to identify phospho- specific RNA aptamers potential for application as therapies.
  • RNA aptamers can be induced to express in cells and phospho-specific RNA aptamers can serve as a unique set of molecular tools for dissecting protein kinase functions in cells. Briefly, a library of DNA or RNA aptamers is incubated on an array of containing protein kinases of interest that have been
  • RNA aptamers autophosphorylated, treated with kinases, or treated with phosphatases. Bound aptamers are then recovered and amplified. Asymmetric amplification of the amplified products is then performed to regenerate the DNA aptamers. In the case of using an RNA aptamer library, in vitro transcription is then performed to regenerate the RNA aptamers. This process is repeated for 4 cycles. During the 5 th cycle, bound aptamers are ligated to address polynucleotides, amplified, and sequenced to identify phospho-specific RNA aptamers. This process is repeated for a 6 th and 7 th cycle. Sequencing data from cycles 5, 6, and 7 can be compared to identify high affinity phospho-specific RNA aptamers.
  • GST tagged target analytes and GST tagged address polynucleotides can be coupled to a glutathione bead.
  • a GST tagged target analyte and a corresponding barcoded GST tagged address polynucleotide can be coupled to a glutathione bead.
  • a plurality of glutathione beads comprising GST tagged target analyte and a corresponding barcoded GST tagged address polynucleotide can be combined and a proximity probe can thereafter be a screened.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Genetics & Genomics (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Hematology (AREA)
  • Biophysics (AREA)
  • General Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pathology (AREA)
  • Medicinal Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

Des procédés de séquençage et de couplage de proximité pour cribler, identifier, valider et/ou caractériser les interactions entre des analytes et des fragments de liaison, sont divulgués. L'invention concerne également des méthodes de séquençage et de couplage de proximité pour déterminer ou quantifier les niveaux d'analytes cibles. Les méthodes divulguées peuvent être multiplexées en deux dimensions, et peuvent être utilisées pour déterminer l'affinité et la spécificité de chaque fragment d'une pluralité de fragments de liaison pour chaque analyte d'une pluralité d'analytes cibles. Des substrats, des jeux ordonnés et des réactifs destinés à être utilisés dans les méthodes, ainsi que des méthodes pour leur préparation sont également divulgués.
PCT/US2017/014151 2016-01-20 2017-01-19 Méthodes et compositions pour identifier, quantifier, et caractériser des analytes cibles et des fragments de liaison WO2017127556A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662281111P 2016-01-20 2016-01-20
US62/281,111 2016-01-20

Publications (1)

Publication Number Publication Date
WO2017127556A1 true WO2017127556A1 (fr) 2017-07-27

Family

ID=59362702

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/014151 WO2017127556A1 (fr) 2016-01-20 2017-01-19 Méthodes et compositions pour identifier, quantifier, et caractériser des analytes cibles et des fragments de liaison

Country Status (1)

Country Link
WO (1) WO2017127556A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110412277A (zh) * 2019-06-26 2019-11-05 四川大学华西医院 Snap91自身抗体检测试剂在制备肺癌筛查试剂盒中的用途
WO2021021348A1 (fr) * 2019-07-31 2021-02-04 The Regents Of The University Of California Détection in situ de molécules de signalisation par combinaison de la modification chimique de protéines endogènes et du dosage par ligature de proximité

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050053964A1 (en) * 2000-11-15 2005-03-10 Minerva Biotechnologies Corporation Oligonucleotide identifiers

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050053964A1 (en) * 2000-11-15 2005-03-10 Minerva Biotechnologies Corporation Oligonucleotide identifiers

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110412277A (zh) * 2019-06-26 2019-11-05 四川大学华西医院 Snap91自身抗体检测试剂在制备肺癌筛查试剂盒中的用途
CN110412277B (zh) * 2019-06-26 2022-09-09 四川大学华西医院 Snap91自身抗体检测试剂在制备肺癌筛查试剂盒中的用途
WO2021021348A1 (fr) * 2019-07-31 2021-02-04 The Regents Of The University Of California Détection in situ de molécules de signalisation par combinaison de la modification chimique de protéines endogènes et du dosage par ligature de proximité

Similar Documents

Publication Publication Date Title
US20170212101A1 (en) Methods and compositions to identify, quantify, and characterize target analytes and binding moieties
US20240254475A1 (en) Proteomic analysis with nucleic acid identifiers
US20240294974A1 (en) Methods, compositions, and kits for determining the location of an analyte in a biological sample
CN110475864B (zh) 用于识别或量化在生物样品中的靶标的方法和组合物
JP5415264B2 (ja) 検出可能な核酸タグ
US10288608B2 (en) Polynucleotide conjugates and methods for analyte detection
EP2369015B1 (fr) Analyse pour la détection localisée d'analytes
US8916504B2 (en) Methods of RNA display
CN111500680A (zh) 检测样品中生物靶标的空间分布的方法和系统
US20110143955A1 (en) Protein Capture, Detection and Quantitation
WO2009151149A1 (fr) Procédé pour détecter ou quantifier de l’adn
JP2017536846A (ja) 方法
WO2017031378A1 (fr) Systèmes et procédés permettant une analyse d'activité protéomique à l'aide de sondes codées par l'adn
WO2017127556A1 (fr) Méthodes et compositions pour identifier, quantifier, et caractériser des analytes cibles et des fragments de liaison
WO2022040269A1 (fr) Systèmes de capteurs et sondes basés sur l'hybridation
JP2020507326A (ja) 組換え事象および再編成事象の検出に関する方法および組成物
WO2021157589A1 (fr) Procédé de détection d'une séquence de bases d'acide nucléique cible en parallèle à un niveau monocellulaire
CA3161615A1 (fr) Procede et kit pour l'amplification et l'analyse du genome entier de molecules cibles dans un echantillon biologique
JP2024525171A (ja) ポリペプチドライブラリーを生成および分析する方法、システムおよび組成物
WO2023086824A1 (fr) Procédés d'identification de molécules de liaison à l'antigène
RU2497128C2 (ru) Способ одновременного определения двух аналитов биолюминесцентным молекулярным микроанализом

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17741934

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17741934

Country of ref document: EP

Kind code of ref document: A1