US20120219961A1 - Methods of running assays using intrachain fluorophore-quencher FRET-aptamers - Google Patents

Methods of running assays using intrachain fluorophore-quencher FRET-aptamers Download PDF

Info

Publication number
US20120219961A1
US20120219961A1 US12/931,309 US93130911A US2012219961A1 US 20120219961 A1 US20120219961 A1 US 20120219961A1 US 93130911 A US93130911 A US 93130911A US 2012219961 A1 US2012219961 A1 US 2012219961A1
Authority
US
United States
Prior art keywords
fret
aptamers
target molecule
aptamer
fluorescence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/931,309
Inventor
John G. Bruno
Joseph Chanpong
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
PRONUCLEOTEIN BIOTECHNOLOGIES LLC
Original Assignee
PRONUCLEOTEIN BIOTECHNOLOGIES LLC
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 PRONUCLEOTEIN BIOTECHNOLOGIES LLC filed Critical PRONUCLEOTEIN BIOTECHNOLOGIES LLC
Priority to US12/931,309 priority Critical patent/US20120219961A1/en
Publication of US20120219961A1 publication Critical patent/US20120219961A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • 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
    • G01N33/5308Immunoassay; Biospecific binding assay; Materials therefor for analytes not provided for elsewhere, e.g. nucleic acids, uric acid, worms, mites
    • 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
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • G01N33/533Production of labelled immunochemicals with fluorescent label
    • 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
    • G01N33/536Immunoassay; Biospecific binding assay; Materials therefor with immune complex formed in liquid phase
    • G01N33/542Immunoassay; Biospecific binding assay; Materials therefor with immune complex formed in liquid phase with steric inhibition or signal modification, e.g. fluorescent quenching
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/16Aptamers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/10Applications; Uses in screening processes

Definitions

  • Applicants' invention relates to the field of aptamer- and nucleic acid-based diagnostics. More particularly, it relates to methods for the production and use of single chain (single-stranded) fluorescence resonance energy transfer (“FRET”) DNA or RNA aptamers containing fluorophores (“F”) and quenchers (“Q”) at various loci within their structures.
  • FRET single chain fluorescence resonance energy transfer
  • F fluorophores
  • Q quenchers
  • FRET-aptamers are a new class of compounds, consisting in part of single-stranded oligonucleotides, desirable for their use in rapid (within minutes), one-step, homogeneous assays involving no wash steps (simple bind and detect quantitative assays).
  • FRET-aptamer methods for various target analytes that consist of placing the F and Q moieties either on the 5′ and 3′ ends respectively to act like a “molecular (aptamer) beacon” or placing only F in the heart of the aptamer structure to be “quenched” by another proximal F or the DNA or RNA itself.
  • F-dNTPs fluorophore-labeled deoxynucleotides
  • Q-dNTP highly efficient spectrally matched quencher
  • the advantage of this F and Q “doping” method is two-fold: 1) the method allows nature to take its course and select the most sensitive FRET-aptamer target interactions in solution, and 2) the positions of F and Q within the aptamer structure can be determined via exonuclease digestion of the FRET-aptamer followed by mass spectral analysis of the resulting fragments, thereby eliminating the need to “engineer” the F and Q moieties into a prospective aptamer binding pocket or loop. Sequence and mass spectral data can be used to further optimize the FRET-aptamer assay performance after natural selection as well.
  • beacons that snap open upon binding to an analyte or upon hybridizing to a complementary sequence, but beacons are always end-labeled with F and Q at the 3′ and 5′ ends.
  • FRET-aptamers may be labeled anywhere in their structure that places the F and Q within the Förster distance of approximately 60-85 Angstroms to achieve quenching prior to or after target analyte binding to the aptamer “binding pocket” (typically a “loop” in the secondary structure).
  • “Signaling aptamers” do not include a Q in their structures, but rather appear to rely upon the “self-quenching” of two adjacent fluorophores or the mild quenching ability of the nucleic acid itself. Both of these methods of quenching are relatively poor, because eventually F-emitted photons escape into the environment and are detectable, thereby contributing to background light and limiting the sensitivity of the FRET assay.
  • True quenchers such as dabcyl (“D”), the “Black Hole Quenchers” (“BHQs”), and the QSY family of dyes (QSY-5, QSY-7, or QSY-9) are broad spectrum absorbing molecules that appear dark or even black in color, because they absorb many wavelengths of light and do not re-emit photons.
  • the inclusion of a Q in the intrachain FRET-aptamer structure or the competitive aptamer FRET format reduces background fluorescence intensity significantly, thereby increasing signal-to-noise ratios and improving assay sensitivity.
  • the method of selecting single intrachain FRET-aptamers based on differential molecular weight and fluorescence intensity of the target analyte-aptamer bound subset fractions is a novel FRET-aptamer development method.
  • the F and Q molecules used can include any number of appropriate fluorophores and quenchers as long as they are spectrally matched so the emission spectrum of F overlaps significantly (almost completely) with the absorption spectrum of Q.
  • the present invention describes a single chain (single-stranded intrachain) FRET assay approach in which F and Q are incorporated into an aptamer population via their nucleotide triphosphate derivatives (for example, ALEXA FLUORTM-NTPs, CASCADE BLUE®-NTPs, CHROMATIDE®-NTPs, fluorescein-NTPs, rhodamine-NTPs, RHODAMINE GREENTM-NTPs, tetramethylrhodamine-dNTPs, OREGON GREEN®-NTPs, and TEXAS RED®-NTPs may be used to provide the fluorophores, while dabcyl-NTPs, Black Hole Quencher or BHQTM-NTPs, and QSYTM dye-NTPs may be used for the quenchers) by PCR after several rounds of selection and amplification without the F- and Q-modified bases. This process is generally referred to as “do
  • the single chain or intrachain FRET-aptamers in the population that still bind the intended target are purified by size-exclusion chromatography columns, spin columns, gel electrophoresis or other means. Once bound and separated based on weight or other physical properties, the brightest fluorescing FRET-aptamer-target complexes are selected because they are clearly the optimal FRET candidates.
  • the FRET-aptamers are separated from the targets by heating or chemical means (urea, formamide, etc.) and purified again by size-exclusion chromatography or other means.
  • the resulting oligonucleotide fragments are subjected to mass spectral analysis which can reveal the nucleotide sequences as well as the positions of F and Q within the FRET-aptamers.
  • FRET-aptamer sequence Once the FRET-aptamer sequence is known with the positions of F and Q, it can be further manipulated during solid-phase DNA or RNA synthesis in an attempt to make the FRET assay more sensitive and specific.
  • the single-chain FRET-aptamers developed by the present invention including quantifiable fluorescence assays for small molecules including pesticides, natural and synthetic amino acids and their derivatives (e.g., histidine, histamine, homocysteine, DOPA, melatonin, nitrotyrosine, etc.), short chain proteolysis products such as cadaverine, putrescine, the polyamines spermine and spermidine, nitrogen bases of DNA or RNA, nucleosides, nucleotides, and their cyclical isoforms (e.g., cAMP and cGMP), cellular metabolites (e.g., urea, uric acid), pharmaceuticals (therapeutic drugs), drugs of abuse (e.g., narcotics, hallucinogens, gamma-hydroxybutyrate, etc.), cellular mediators (e.g., cytokines, chemokines, immune modulators, neural modulators,
  • single-chain FRET-aptamers include use in quantifiable fluorescence assays for macromolecules including proteins such as biotoxins including botulinum toxins, Shiga toxins (See FIG. 2 ), staphylococcal enterotoxins, other bacterial toxins, prions such as bovine spongiform encephalopathies (“BSEs”) and transmissible spongiform encephalopathies (“TSEs”), glycoproteins, lipids, glycolipids, triglycerides, nucleic acids, polysaccharides, lipopolysaccharides, etc.
  • proteins such as biotoxins including botulinum toxins, Shiga toxins (See FIG. 2 ), staphylococcal enterotoxins, other bacterial toxins, prions such as bovine spongiform encephalopathies (“BSEs”) and transmissible spongiform encephalopathies (“TSEs”), glycoproteins, lipids, glycolipids, triglycerides, nucleic
  • the single-chain FRET-aptamers may also be used in quantifiable fluorescence assays for subcellular and whole cell targets including subcellular organelles such as ribosomes, Golgi apparatus, vesicles, microfilaments, microtubules, etc., viruses, virions, rickettsiae, bacteria, protozoa, plankton, parasites such as Cryptosporidium species, Giardia species, mammalian cells such as various classes of leukocytes, neurons, stem cells, cancer cells, etc.
  • subcellular organelles such as ribosomes, Golgi apparatus, vesicles, microfilaments, microtubules, etc.
  • parasites such as Cryptosporidium species, Giardia species, mammalian cells such as various classes of leukocytes, neurons, stem cells, cancer cells, etc.
  • unlabeled aptamer sequence information and secondary stem-loop structures may aid in the determination and engineered optimization of F or Q placement within the aptamer structure to maximize FRET assay sensitivity and specificity.
  • “within” and “interior” are defined as directing F or Q placement, or labeling, interior to, or between, the ends of the aptamer; the ends of the aptamer being the 3′ and 5′ ends.
  • FIG. 1 is a schematic illustration of the single chain (intrachain) FRET-aptamer selection method.
  • FIG. 2 is a bar graph showing toxin concentration mapped with fluorescence intensity and illustrating a “lights off” FRET with shiga-like toxin 1 and round 5 aptamers.
  • FIG. 3 is a schematic illustration that illustrates a comparison of possible nucleic acid FRET assay formats.
  • FIG. 4 illustrates sample aptamer sequences.
  • FIGS. 4A-4B are schematic illustrations of the secondary structures of selected aptamer sequences shown in FIG. 4 .
  • FIG. 5 is a line graph correlating absorbance with BoNT A concentration.
  • FIGS. 6A-6B are line graphs mapping fluorescence intensity against time.
  • FIG. 1 illustrates a single chain (intrachain) FRET-aptamer selection method.
  • This method consists of several steps. First the random DNA library of oligonucleotides (randomized region of 20 or more bases flanked by known primer regions) is “doped” with F-dNTPs Q-dNTPs by the PCR ( 10 ). The F and Q doped library is then exposed to a protein or other target molecule ( 12 ). Some members of the doped library will bind to the target protein ( 14 ).
  • the target molecule is a larger water-soluble molecule such as a protein, glycoprotein, or other water soluble macromolecule
  • exposure of the nascent F-labeled and Q-labeled DNA or RNA random library to the free target analyte is done in solution.
  • the target is a soluble protein or other larger water-soluble molecule
  • the optimal FRET-aptamer-target complexes are separated by size-exclusion chromatography.
  • the FRET-aptamer-target complex population of molecules is the heaviest subset in solution and will emerge from a size-exclusion column first, followed by unbound FRET-aptamers and unbound proteins or other targets.
  • analyte-bound aptamers there will be heterogeneity in the numbers of F- and Q-NTPs that are incorporated as well as nucleotide sequence differences, which will again effect the mass, electrical charge, and weak interaction capabilities (e.g., hydrophobicity and hydrophilicity) of each analyte-aptamer complex.
  • These differences in physical properties of the aptamer-analyte complexes can then be used to separate out or partition the bound from unbound analyte-aptamer complexes.
  • the target is a small molecule (generally defined as a molecule with molecular weight of ⁇ 1,000 Daltons)
  • exposure of the nascent F-labeled and Q-labeled DNA or RNA random library to the target is done by immobilizing the target.
  • the small molecule can be immobilized on a column, membrane, plastic or glass bead, magnetic bead, or other matrix. If no functional group is available on the small molecule for immobilization, the target can be immobilized by the Mannich reaction (formaldehyde-based condensation reaction) on a device similar to a PHARMALINKTM column. Elution of bound DNA from the small molecule affinity column, membrane, beads or other matrix by use of 0.2-3.0M sodium acetate at a pH ranging between 3 and 7, although the optimal pH is approximately 5.2.
  • aptamer-protein aggregates or selected aptamers-protein aggregates
  • SephadexTM or other gel materials in the column ( 16 ). Since they vary in weight due to variations in aptamers sequences and degree of labeling, they can be separated into fractions with different fluorescence intensities. Purification methods such as capillary electrophoresis or preparative gel electrophoresis are possible as well. Small volume fractions ( ⁇ 1 mL) can be collected from the column and analyzed for absorbance at 260nm and 280nm which are characteristic wavelengths for DNA, RNA and proteins.
  • the heaviest materials come through a size-exclusion column first. Therefore, the DNA-protein complexes or RNA-protein complexes will come out of the column before either the DNA or protein alone when using an appropriate grade of column matrix material (e.g., various grades of Sephadex, Superdex, Sepharose, or Sephacryl, etc.).
  • an appropriate grade of column matrix material e.g., various grades of Sephadex, Superdex, Sepharose, or Sephacryl, etc.
  • Means of separating FRET-aptamer-target complexes from solution by alternate techniques include, without limitation, various molecular weight cut off (“MWCO”) spin columns, dialysis, gel electrophoresis, thin layer chromatography (“TLC”), and differential centrifugation or ultracentrifugation using density gradient materials.
  • MWCO molecular weight cut off
  • TLC thin layer chromatography
  • a preferred FRET-aptamer is selected.
  • a preferred FRET-aptamer is one that exhibits a measurable change in fluorescence between the fluorescence of said FRET-aptamer when it is not bound to said target molecule compared to the fluorescence of said FRET-aptamer when it is bound to said target molecule.
  • the optimal (most sensitive or highest signal to noise ratio) FRET-aptamers among the bound class of FRET-aptamer-target complexes are identified by assessment of fluorescence intensity for various fractions of the FRET-aptamer-target class.
  • the separated DNA-protein complexes will exhibit the highest absorbance at established wavelengths, such as 260nm and 280nm similar to that graphed in FIG. 1 ( 18 ).
  • the fractions showing the highest absorbance at the given wavelengths, such as 260nm and 280nm, are then further analyzed for fluorescence and those fractions exhibiting the greatest fluorescence are selected for separation and sequencing ( 20 ).
  • FRET-aptamers may be further separated using techniques such as ion pair reverse-phase HPLC, ion-exchange chromatography (“IEC”, either low pressure or HPLC versions of IEC), TLC, capillary electrophoresis, or similar techniques.
  • IEC ion pair reverse-phase HPLC
  • IEC ion-exchange chromatography
  • TLC capillary electrophoresis
  • the final FRET-aptamers are able to act as one-step “lights on” or “lights off' binding and detection components in assays.
  • Intrachain FRET-aptamers that are to be used in assays with long shelf-lives may be lyophilized (freeze dried) and reconstituted as needed.
  • FIG. 2 is a bar graph showing toxin concentration mapped with fluorescence intensity and illustrating a “lights off' FRET response with shiga-like toxin 1 and round 5 aptamers. If the fluorescence intensity of the DNA aptamers is correlated to the concentration of the surface protein and the fluorescence intensity decreases as a function of increasing analyte concentration, then it is referred to as a “lights off' assay. If the fluorescence intensity increases as a function of increasing analyte concentration, then it is referred to as a “lights on” assay. Intrachain FRET-aptamer assay data are shown for detection of E. coli shiga-like toxin I protein resulting in a “lights off' FRET reaction as a function of toxin concentration. Fluorescence readings were obtained within five minutes of toxin addition.
  • FIG. 3 illustrates a comparison of possible nucleic acid FRET assay formats.
  • Upper left is a molecular beacon ( 30 ) which may or may not be an aptamer, but is typically a short oligonucleotide used to hybridize to other DNA or RNA molecules and exhibit FRET upon hybridizing.
  • Molecular beacons are only labeled with F and Q at the ends of the DNA molecule.
  • Lower left is a signaling aptamer ( 32 ), which does not contain a quencher molecule, but relies upon fluorophore self-quenching or weak intrinsic quenching capacity of the DNA or RNA to achieve limited FRET.
  • Upper right is an intrachain FRET-aptamer ( 34 ) containing F and Q molecules built into the interior structure of the aptamer. Intrachain FRET-aptamers are naturally selected and characterized by the processes described herein.
  • Lower right shows a competitive aptamer FRET ( 36 ) motif in which the aptamer contains either F or Q and the target molecule ( 38 ) is labeled with the complementary F or Q. Introduction of unlabeled target molecules ( 40 ) then shifts the equilibrium so that some labeled target molecules ( 38 ) are liberated from the labeled aptamer ( 36 ) and modulate the fluorescence level of the solution up or down thereby achieving FRET.
  • a target analyte ( 40 ) is either unlabeled or labeled with a quencher (Q). F and Q can be switched or swapped from placement in the aptamer ( 36 ) to placement in the target analyte ( 40 ) and vice versa.
  • FIG. 4 illustrates sample aptamer sequences for botulinum toxin A (BoNT A) in which all sequences are arranged 5′ to 3′ from left to right.
  • the actual degenerate (randomized) aptamer regions are bolded. Clear consensus regions are bolded and italicized. Flanking sequences match with the primers used in the PCR scheme or the complementary strand primer sequences except in highlighted cases. Most sequences end in a 3′ A (added by Taq, underlined).
  • BoNT A botulinum A
  • FIGS. 4A-4D illustrate secondary structures of selected aptamer sequences listed in FIG. 4 .
  • FIG. 5 is a line graph correlating absorbance with BoNT A concentration. It illustrates that aptamer-peroxidase colorimetric plate binding assay results using polyclonal BoNT A aptamers and BoNT A holotoxin. Two different trials or runs are shown. Absorbance was quantified at 405 nm using standard ABTS substrate and hydrogen peroxide activator reagents. The curves illustrate binding and sensitive detection of BoNT A by the aptamers at a level of at least 12.5 ng/mL.
  • FIGS. 6A-6B are line graphs mapping the fluorescence intensity of the DNA aptamers such as those shown in FIGS. 4A-4D against time in minutes.
  • DNA aptamers such as those shown in FIGS. 4A-4D , bind and inhibit the enzymatic activity of BoNT A.
  • the inhibition of BoNT A's enzymatic activity is further proof of tight aptamer binding to the toxin.
  • FIG. 6A shows assay results using the BoNT A holotoxin
  • FIG. 6B shows results using the isolated 50 kD enzymatic subunit of BoNT A.
  • the positive control line shows greater fluorescence intensity over time for the uninhibited BoNT SNAPTIDETM assay and the “Test with Aptamer” line shows consistent suppression of the fluorescence intensity of the SNAPTIDETM assay further proving aptamer binding and aptamer-mediated inhibition of BoNT A enzymatic activity.
  • FIG. 4 gives an example of BoNT A aptamer sequences that are claimed as unlabeled sequences, resulting in secondary stem-loop structures from energy minimization software using 25° C. as the nominal binding temperature.
  • the stem-loop structures shown in FIGS. 4A-4D may be especially useful in determining if the F and Q locations are indeed logical (i.e., fall in or near a binding loop structure).
  • Aptamers were incorporated into plasmids.
  • the plasmids were purified and sequenced by capillary electrophoresis following PCR.
  • BoNT A functionality of the aptamer sequences (ability to bind and inhibit BoNT A) shown in FIGS. 4 and 4 A- 4 D were confirmed by colorimetric plate assay binding data ( FIG. 5 ) and SNAPTIDETM FRET assay data showing inhibition of BoNT A enzymatic activity by the “polyclonal” family of BoNT A aptamers ( FIG. 6 ).

Abstract

The present invention describes methods for the production and use of single chain (single-stranded) fluorescence resonance energy transfer (“FRET”) DNA or RNA aptamers containing fluorophores (F) and quenchers (Q) at various loci within their structures, such that when its specific matching analyte is bound and the FRET-aptamers are excited by specific wavelengths of light, the fluorescence intensity of the system is modulated (increased or decreased) in proportion to the amount of analyte added. F and Q are covalently linked to nucleotide triphosphates (NTPs), which are incorporated by various nucleic acid polymerases such as Taq polymerase during the polymerase chain reaction (PCR) and then selected by affinity chromatographic, size-exclusion or molecular sieving, and fluorescence techniques. Further separation of related FRET-aptamers can be achieved by ion-pair reverse phase high performance liquid chromatography (HPLC) or other types of chromatography. Finally, FRET-aptamer structures and the specific locations of F and Q within FRET-aptamer structures are determined by digestion with exonucleases and mass spectral nucleotide sequencing analysis.

Description

  • This application is based upon and claims priority from U.S. Utility application Ser. No. 11/433,009, which is incorporated herein by reference.
  • BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • Applicants' invention relates to the field of aptamer- and nucleic acid-based diagnostics. More particularly, it relates to methods for the production and use of single chain (single-stranded) fluorescence resonance energy transfer (“FRET”) DNA or RNA aptamers containing fluorophores (“F”) and quenchers (“Q”) at various loci within their structures.
  • 2. Background Information
  • FRET-aptamers are a new class of compounds, consisting in part of single-stranded oligonucleotides, desirable for their use in rapid (within minutes), one-step, homogeneous assays involving no wash steps (simple bind and detect quantitative assays). Several individuals and groups have published and patented FRET-aptamer methods for various target analytes that consist of placing the F and Q moieties either on the 5′ and 3′ ends respectively to act like a “molecular (aptamer) beacon” or placing only F in the heart of the aptamer structure to be “quenched” by another proximal F or the DNA or RNA itself. These preceding FRET-aptamer methods are all highly engineered and based on some prior knowledge of particular aptamer sequences and secondary structures, thereby enabling clues as to where F might be placed in order to optimize FRET results.
  • Until now, no individual or group has described a method for natural selection of single chain (intrachain) FRET-aptamers that contain both fluorophore-labeled deoxynucleotides (“F-dNTPs”) and highly efficient spectrally matched quencher (“Q-dNTP”) moieties in the heart of an aptamer binding loop or pocket by polymerase chain reaction (“PCR”). The advantage of this F and Q “doping” method is two-fold: 1) the method allows nature to take its course and select the most sensitive FRET-aptamer target interactions in solution, and 2) the positions of F and Q within the aptamer structure can be determined via exonuclease digestion of the FRET-aptamer followed by mass spectral analysis of the resulting fragments, thereby eliminating the need to “engineer” the F and Q moieties into a prospective aptamer binding pocket or loop. Sequence and mass spectral data can be used to further optimize the FRET-aptamer assay performance after natural selection as well.
  • Others have described nucleic acid-based “molecular beacons” that snap open upon binding to an analyte or upon hybridizing to a complementary sequence, but beacons are always end-labeled with F and Q at the 3′ and 5′ ends. FRET-aptamers may be labeled anywhere in their structure that places the F and Q within the Förster distance of approximately 60-85 Angstroms to achieve quenching prior to or after target analyte binding to the aptamer “binding pocket” (typically a “loop” in the secondary structure).
  • “Signaling aptamers” do not include a Q in their structures, but rather appear to rely upon the “self-quenching” of two adjacent fluorophores or the mild quenching ability of the nucleic acid itself. Both of these methods of quenching are relatively poor, because eventually F-emitted photons escape into the environment and are detectable, thereby contributing to background light and limiting the sensitivity of the FRET assay. True quenchers such as dabcyl (“D”), the “Black Hole Quenchers” (“BHQs”), and the QSY family of dyes (QSY-5, QSY-7, or QSY-9) are broad spectrum absorbing molecules that appear dark or even black in color, because they absorb many wavelengths of light and do not re-emit photons. The inclusion of a Q in the intrachain FRET-aptamer structure or the competitive aptamer FRET format, reduces background fluorescence intensity significantly, thereby increasing signal-to-noise ratios and improving assay sensitivity.
  • In addition to the novelty of the quencher introduction into the assay formats and advantages conferred in terms of sensitivity by cutting background fluorescence, the method of selecting single intrachain FRET-aptamers based on differential molecular weight and fluorescence intensity of the target analyte-aptamer bound subset fractions is a novel FRET-aptamer development method. The F and Q molecules used can include any number of appropriate fluorophores and quenchers as long as they are spectrally matched so the emission spectrum of F overlaps significantly (almost completely) with the absorption spectrum of Q.
  • SUMMARY OF THE INVENTION
  • The present invention describes a single chain (single-stranded intrachain) FRET assay approach in which F and Q are incorporated into an aptamer population via their nucleotide triphosphate derivatives (for example, ALEXA FLUOR™-NTPs, CASCADE BLUE®-NTPs, CHROMATIDE®-NTPs, fluorescein-NTPs, rhodamine-NTPs, RHODAMINE GREEN™-NTPs, tetramethylrhodamine-dNTPs, OREGON GREEN®-NTPs, and TEXAS RED®-NTPs may be used to provide the fluorophores, while dabcyl-NTPs, Black Hole Quencher or BHQ™-NTPs, and QSY™ dye-NTPs may be used for the quenchers) by PCR after several rounds of selection and amplification without the F- and Q-modified bases. This process is generally referred to as “doping” with F-NTPs and Q-NTPs.
  • Thereafter, the single chain or intrachain FRET-aptamers in the population that still bind the intended target (after the doping process) are purified by size-exclusion chromatography columns, spin columns, gel electrophoresis or other means. Once bound and separated based on weight or other physical properties, the brightest fluorescing FRET-aptamer-target complexes are selected because they are clearly the optimal FRET candidates. The FRET-aptamers are separated from the targets by heating or chemical means (urea, formamide, etc.) and purified again by size-exclusion chromatography or other means.
  • These intrachain FRET-aptamers cannot be cloned for sequencing due to the need for determining the locations of F and Q in their structures. Cloning would lead to replication of the FRET-aptamer insert in the plasmid and either dilution of the desired FRET-aptamer or alteration of its F and Q locations within the aptamer. Therefore, the candidate FRET-aptamers are separated based on physical properties such as charge or weak interactions by various types of high performance liquid chromatography (“HPLC”), digested at each end with specific exonucleases (snake venom phosphodiesterase on the 3′ end and calf spleen phosphodiesterase on the 5′ end). The resulting oligonucleotide fragments, each one base shorter than the predecessor, are subjected to mass spectral analysis which can reveal the nucleotide sequences as well as the positions of F and Q within the FRET-aptamers. Once the FRET-aptamer sequence is known with the positions of F and Q, it can be further manipulated during solid-phase DNA or RNA synthesis in an attempt to make the FRET assay more sensitive and specific.
  • There are a number of uses of the single-chain FRET-aptamers developed by the present invention, including quantifiable fluorescence assays for small molecules including pesticides, natural and synthetic amino acids and their derivatives (e.g., histidine, histamine, homocysteine, DOPA, melatonin, nitrotyrosine, etc.), short chain proteolysis products such as cadaverine, putrescine, the polyamines spermine and spermidine, nitrogen bases of DNA or RNA, nucleosides, nucleotides, and their cyclical isoforms (e.g., cAMP and cGMP), cellular metabolites (e.g., urea, uric acid), pharmaceuticals (therapeutic drugs), drugs of abuse (e.g., narcotics, hallucinogens, gamma-hydroxybutyrate, etc.), cellular mediators (e.g., cytokines, chemokines, immune modulators, neural modulators, inflammatory modulators such as prostaglandins, etc.), or their metabolites, explosives (e.g., trinitrotoluene) and their breakdown products or byproducts, peptides and their derivatives. Other uses of the single-chain FRET-aptamers include use in quantifiable fluorescence assays for macromolecules including proteins such as biotoxins including botulinum toxins, Shiga toxins (See FIG. 2), staphylococcal enterotoxins, other bacterial toxins, prions such as bovine spongiform encephalopathies (“BSEs”) and transmissible spongiform encephalopathies (“TSEs”), glycoproteins, lipids, glycolipids, triglycerides, nucleic acids, polysaccharides, lipopolysaccharides, etc. The single-chain FRET-aptamers may also be used in quantifiable fluorescence assays for subcellular and whole cell targets including subcellular organelles such as ribosomes, Golgi apparatus, vesicles, microfilaments, microtubules, etc., viruses, virions, rickettsiae, bacteria, protozoa, plankton, parasites such as Cryptosporidium species, Giardia species, mammalian cells such as various classes of leukocytes, neurons, stem cells, cancer cells, etc.
  • The use of unlabeled aptamer sequence information and secondary stem-loop structures may aid in the determination and engineered optimization of F or Q placement within the aptamer structure to maximize FRET assay sensitivity and specificity. As described and claimed herein, “within” and “interior” are defined as directing F or Q placement, or labeling, interior to, or between, the ends of the aptamer; the ends of the aptamer being the 3′ and 5′ ends. Although, an anticipated step in the present method of natural selection of F- and/or Q-labeled aptamers to form solution phase interactions with their target analytes, additional sequence and secondary structure information can be used to confirm and enhance F and Q placement to optimize assay performance.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1. is a schematic illustration of the single chain (intrachain) FRET-aptamer selection method.
  • FIG. 2. is a bar graph showing toxin concentration mapped with fluorescence intensity and illustrating a “lights off” FRET with shiga-like toxin 1 and round 5 aptamers.
  • FIG. 3. is a schematic illustration that illustrates a comparison of possible nucleic acid FRET assay formats.
  • FIG. 4. illustrates sample aptamer sequences.
  • FIGS. 4A-4B. are schematic illustrations of the secondary structures of selected aptamer sequences shown in FIG. 4.
  • FIG. 5. is a line graph correlating absorbance with BoNT A concentration.
  • FIGS. 6A-6B. are line graphs mapping fluorescence intensity against time.
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT
  • Referring to the figures, FIG. 1. illustrates a single chain (intrachain) FRET-aptamer selection method. This method consists of several steps. First the random DNA library of oligonucleotides (randomized region of 20 or more bases flanked by known primer regions) is “doped” with F-dNTPs Q-dNTPs by the PCR (10). The F and Q doped library is then exposed to a protein or other target molecule (12). Some members of the doped library will bind to the target protein (14).
  • If the target molecule is a larger water-soluble molecule such as a protein, glycoprotein, or other water soluble macromolecule, then exposure of the nascent F-labeled and Q-labeled DNA or RNA random library to the free target analyte is done in solution. If the target is a soluble protein or other larger water-soluble molecule, then the optimal FRET-aptamer-target complexes are separated by size-exclusion chromatography. The FRET-aptamer-target complex population of molecules is the heaviest subset in solution and will emerge from a size-exclusion column first, followed by unbound FRET-aptamers and unbound proteins or other targets. Among the subset of analyte-bound aptamers there will be heterogeneity in the numbers of F- and Q-NTPs that are incorporated as well as nucleotide sequence differences, which will again effect the mass, electrical charge, and weak interaction capabilities (e.g., hydrophobicity and hydrophilicity) of each analyte-aptamer complex. These differences in physical properties of the aptamer-analyte complexes can then be used to separate out or partition the bound from unbound analyte-aptamer complexes.
  • If the target is a small molecule (generally defined as a molecule with molecular weight of ≦1,000 Daltons), then exposure of the nascent F-labeled and Q-labeled DNA or RNA random library to the target is done by immobilizing the target. The small molecule can be immobilized on a column, membrane, plastic or glass bead, magnetic bead, or other matrix. If no functional group is available on the small molecule for immobilization, the target can be immobilized by the Mannich reaction (formaldehyde-based condensation reaction) on a device similar to a PHARMALINK™ column. Elution of bound DNA from the small molecule affinity column, membrane, beads or other matrix by use of 0.2-3.0M sodium acetate at a pH ranging between 3 and 7, although the optimal pH is approximately 5.2.
  • These complexes can be separated from the non-binding doped DNA molecules by running the aptamer-protein aggregates (or selected aptamers-protein aggregates) through a size-exclusion column, by means of size-exclusion chromatography using Sephadex™ or other gel materials in the column (16). Since they vary in weight due to variations in aptamers sequences and degree of labeling, they can be separated into fractions with different fluorescence intensities. Purification methods such as capillary electrophoresis or preparative gel electrophoresis are possible as well. Small volume fractions (≦1 mL) can be collected from the column and analyzed for absorbance at 260nm and 280nm which are characteristic wavelengths for DNA, RNA and proteins. The heaviest materials come through a size-exclusion column first. Therefore, the DNA-protein complexes or RNA-protein complexes will come out of the column before either the DNA or protein alone when using an appropriate grade of column matrix material (e.g., various grades of Sephadex, Superdex, Sepharose, or Sephacryl, etc.).
  • Means of separating FRET-aptamer-target complexes from solution by alternate techniques (other than size-exclusion chromatography) include, without limitation, various molecular weight cut off (“MWCO”) spin columns, dialysis, gel electrophoresis, thin layer chromatography (“TLC”), and differential centrifugation or ultracentrifugation using density gradient materials.
  • A preferred FRET-aptamer is selected. A preferred FRET-aptamer is one that exhibits a measurable change in fluorescence between the fluorescence of said FRET-aptamer when it is not bound to said target molecule compared to the fluorescence of said FRET-aptamer when it is bound to said target molecule.
  • The optimal (most sensitive or highest signal to noise ratio) FRET-aptamers among the bound class of FRET-aptamer-target complexes are identified by assessment of fluorescence intensity for various fractions of the FRET-aptamer-target class. The separated DNA-protein complexes will exhibit the highest absorbance at established wavelengths, such as 260nm and 280nm similar to that graphed in FIG. 1 (18). The fractions showing the highest absorbance at the given wavelengths, such as 260nm and 280nm, are then further analyzed for fluorescence and those fractions exhibiting the greatest fluorescence are selected for separation and sequencing (20).
  • These-similar FRET-aptamers may be further separated using techniques such as ion pair reverse-phase HPLC, ion-exchange chromatography (“IEC”, either low pressure or HPLC versions of IEC), TLC, capillary electrophoresis, or similar techniques.
  • The final FRET-aptamers are able to act as one-step “lights on” or “lights off' binding and detection components in assays.
  • Intrachain FRET-aptamers that are to be used in assays with long shelf-lives may be lyophilized (freeze dried) and reconstituted as needed.
  • FIG. 2. is a bar graph showing toxin concentration mapped with fluorescence intensity and illustrating a “lights off' FRET response with shiga-like toxin 1 and round 5 aptamers. If the fluorescence intensity of the DNA aptamers is correlated to the concentration of the surface protein and the fluorescence intensity decreases as a function of increasing analyte concentration, then it is referred to as a “lights off' assay. If the fluorescence intensity increases as a function of increasing analyte concentration, then it is referred to as a “lights on” assay. Intrachain FRET-aptamer assay data are shown for detection of E. coli shiga-like toxin I protein resulting in a “lights off' FRET reaction as a function of toxin concentration. Fluorescence readings were obtained within five minutes of toxin addition.
  • FIG. 3. illustrates a comparison of possible nucleic acid FRET assay formats. Upper left is a molecular beacon (30) which may or may not be an aptamer, but is typically a short oligonucleotide used to hybridize to other DNA or RNA molecules and exhibit FRET upon hybridizing. Molecular beacons are only labeled with F and Q at the ends of the DNA molecule. Lower left is a signaling aptamer (32), which does not contain a quencher molecule, but relies upon fluorophore self-quenching or weak intrinsic quenching capacity of the DNA or RNA to achieve limited FRET. Upper right is an intrachain FRET-aptamer (34) containing F and Q molecules built into the interior structure of the aptamer. Intrachain FRET-aptamers are naturally selected and characterized by the processes described herein. Lower right shows a competitive aptamer FRET (36) motif in which the aptamer contains either F or Q and the target molecule (38) is labeled with the complementary F or Q. Introduction of unlabeled target molecules (40) then shifts the equilibrium so that some labeled target molecules (38) are liberated from the labeled aptamer (36) and modulate the fluorescence level of the solution up or down thereby achieving FRET. A target analyte (40) is either unlabeled or labeled with a quencher (Q). F and Q can be switched or swapped from placement in the aptamer (36) to placement in the target analyte (40) and vice versa.
  • FIG. 4 illustrates sample aptamer sequences for botulinum toxin A (BoNT A) in which all sequences are arranged 5′ to 3′ from left to right. The actual degenerate (randomized) aptamer regions are bolded. Clear consensus regions are bolded and italicized. Flanking sequences match with the primers used in the PCR scheme or the complementary strand primer sequences except in highlighted cases. Most sequences end in a 3′ A (added by Taq, underlined). Aptamer sequences that bind and inhibit the action of botulinum A (BoNT A) 150 kD holotoxin and the 50 kD enzymatic light chain or subunit of BoNT A, and which may be useful in single chain FRET-aptamer or competitive aptamer-FRET assays for detection and quantification of BoNT A.
  • FIGS. 4A-4D. illustrate secondary structures of selected aptamer sequences listed in FIG. 4. Various botulinum A (BoNT A) DNA aptamer secondary (two dimensional) stem-loop structures that bind the holotoxin (FIG. 4A, which exemplifies a sequence that occurred in four different clones), and bind and inhibit (See FIGS. 6A and 6B) the small (50 kD) enzymatic subunit (FIGS. 4B-4D, showing the secondary structures for three different sequences that produced similar secondary structures).
  • FIG. 5. is a line graph correlating absorbance with BoNT A concentration. It illustrates that aptamer-peroxidase colorimetric plate binding assay results using polyclonal BoNT A aptamers and BoNT A holotoxin. Two different trials or runs are shown. Absorbance was quantified at 405 nm using standard ABTS substrate and hydrogen peroxide activator reagents. The curves illustrate binding and sensitive detection of BoNT A by the aptamers at a level of at least 12.5 ng/mL.
  • FIGS. 6A-6B. are line graphs mapping the fluorescence intensity of the DNA aptamers such as those shown in FIGS. 4A-4D against time in minutes. DNA aptamers, such as those shown in FIGS. 4A-4D, bind and inhibit the enzymatic activity of BoNT A. Here the inhibition of BoNT A's enzymatic activity is further proof of tight aptamer binding to the toxin. FIG. 6A shows assay results using the BoNT A holotoxin and FIG. 6B shows results using the isolated 50 kD enzymatic subunit of BoNT A. The positive control line shows greater fluorescence intensity over time for the uninhibited BoNT SNAPTIDE™ assay and the “Test with Aptamer” line shows consistent suppression of the fluorescence intensity of the SNAPTIDE™ assay further proving aptamer binding and aptamer-mediated inhibition of BoNT A enzymatic activity.
  • EXAMPLE 1
  • Single (Intrachain) Chain FRET-Aptamer Assay for a Protein (E. coli Shiga-Like Toxin I).
  • Following five rounds of systematic evolution of ligands by exponential enrichment (“SELEX”) an aptamer family was subjected to PCR in the presence of 3 μM CHROMATIDE™-dUTP and 40μM Dabcyl-dUTP using a standard PCR mix formulation and Taq enzyme at 1 Unit per 50 μL reaction. This led to incorporation of the FRET (F and Q) pair which demonstrated the lowest background fluorescence of all F:Q ratios tested (nearly 1,200 fluorescence units for the baseline reading without the toxin target). Fluorescence readings in FIG. 2 were taken with a handheld fluorometer. Error bars in FIG. 2 represent the standard deviation of three trials and the bar heights represent the means of the 3 measurements. At the level of 40,000 picograms per milliliter (pg/mL) or 40 nanograms (ng) of Shiga-like toxin I, a definitive “lights off’ FRET effect is noted. Since the mean fluorescence at 40 ng of added toxin is far greater than two standard deviations below any of the other treatment groups, it must be considered statistically significant.
  • EXAMPLE 2
  • Use of Unlabeled Aptamer Nucleotide Sequences and Secondary (Stem-Loop) Structures that Can Confirm, Enhance, and Optimize FRET-Aptamer Assays.
  • The present method enables the natural selection of FRET-aptamers. However, the method can be confirmed and enhanced by knowledge of the unlabeled aptamer sequences and structures that were selected from several rounds of SELEX before the aptamer population was “doped” with F-dNTPs and/or Q-dNTPs. FIG. 4 gives an example of BoNT A aptamer sequences that are claimed as unlabeled sequences, resulting in secondary stem-loop structures from energy minimization software using 25° C. as the nominal binding temperature. The stem-loop structures shown in FIGS. 4A-4D may be especially useful in determining if the F and Q locations are indeed logical (i.e., fall in or near a binding loop structure). In addition, if F and/or Q loci are found to be distal, information such as the secondary structures in FIGS. 4A-4D could be instrumental in slightly relocating the F and Q moieties to enhance or optimize the FRET assay results in terms of assay sensitivity and specificity.
  • Aptamers were incorporated into plasmids. The plasmids were purified and sequenced by capillary electrophoresis following PCR.
  • The BoNT A functionality of the aptamer sequences (ability to bind and inhibit BoNT A) shown in FIGS. 4 and 4A-4D were confirmed by colorimetric plate assay binding data (FIG. 5) and SNAPTIDE™ FRET assay data showing inhibition of BoNT A enzymatic activity by the “polyclonal” family of BoNT A aptamers (FIG. 6).
  • Although the invention has been described with reference to ‘specific embodiments, this description is not meant to be construed in a limited sense. Various modifications of the disclosed embodiments, as well as alternative embodiments of the inventions will become apparent to persons skilled in the art upon the reference to the description of the invention. It is, therefore, contemplated that the appended claims will cover such modifications that fall within the scope of the invention.

Claims (9)

1. A method of assaying a target molecule in a solution using single-stranded fluorescence resonance energy transfer (“FRET”)-aptamers, comprising:
selecting FRET-aptamers that bind with said target molecule;
adding said selected FRET-aptamers to said solution,
wherein said FRET-aptamers have at least one fluorophore (“F”) and at least one quencher (“Q”) incorporated into said FRET-aptamers in the interior of said FRET-aptamers;
wherein said F and said Q are spectrally matched such that an absorption spectrum of said Q overlaps significantly with an emission spectrum of said F;
wherein prior to binding said target molecule said location of said F is within a Förster distance of said Q such that said Q interacts with said F such that minimal fluorescence is detectable;
wherein said FRET-aptamers emit increased detectable fluorescence after binding said target molecule due to an increase in said Förster distance between said Q and said F, and said detectable fluorescence changes proportionately in response to the amount of said target molecule in said solution;
measuring said fluorescence light level; and
determining the presence or absence of said target molecules in said solution.
2. The method of claim 1, further comprising calculating the amount of said target molecules in said solution
3. The method of claim 2, wherein said assay is used for detecting and quantifying a target molecule, wherein said target molecule is less than 1,000 Daltons.
4. The method of claim 3, wherein said target molecule is selected from the group consisting of:
pesticides, natural and synthetic amino acids, histidine, histamine, homocysteine, DOPA, melatonin, nitrotyrosine, short chain proteolysis products, cadaverine, putrescine, polyamines, spermine, spermidine, nitrogen bases of DNA or RNA, nucleosides, nucleotides, nucleotide cyclical isoforms, cAMP, cGMP, cellular metabolites, urea, uric acid, pharmaceuticals, therapeutic drugs, narcotics, hallucinogens, gamma-hydroxybutyrate, cellular mediators, cytokines, chemokines, immune modulators, neural modulators, inflammatory modulators, prostaglandins, prostaglandin metabolites, explosives, trinitrotoluene, peptides, macromolecules, proteins, bacterial surface proteins, glycoproteins, lipids, glycolipids, nucleic acids, polysaccharides, lipopolysaccharides, whole cells, and subcellular organelles or cellular fractions.
5. The method of claim 2, wherein said assay is used for detecting and quantifying a target molecule, wherein said target molecule is equal to or greater than 1,000 Daltons and water- soluble.
6. A method of assaying a target molecule in a solution using single-stranded fluorescence resonance energy transfer (“FRET”)-aptamers, comprising:
selecting FRET-aptamers that bind with said target molecule;
adding said selected FRET-aptamers to said solution,
wherein said FRET-aptamers have at least one fluorophore (“F”) and at least one quencher (“Q”) incorporated into said FRET-aptamers in the interior of said FRET-aptamers;
wherein said F and said Q are spectrally matched such that an absorption spectrum of said Q overlaps significantly with an emission spectrum of said F;
wherein prior to binding said target molecule said location of said F is beyond a Förster distance of said Q such that said Q does not interact with said F such that said F emits detectable fluorescence
wherein said FRET-aptamers emit decreased detectable fluorescence after binding said target molecule due to a decrease in the Förster distance between said F and said Q, and said detectable fluorescence changes proportionately in response to the amount of said target molecule in said solution;
measuring said fluorescence light level; and
determining the presence or absence of said target molecules in said solution.
7. The method of claim 6, wherein said assay is used for detecting a target molecule, wherein said target molecule is less than 1,000 Daltons.
8. The method of claim 7, wherein said target molecule is selected from the group consisting of: pesticides, natural and synthetic amino acids, histidine, histamine, homocysteine, DOPA, melatonin, nitrotyrosine, short chain proteolysis products, cadaverine, putrescine, polyamines, spermine, spermidine, nitrogen bases of DNA or RNA, nucleosides, nucleotides, nucleotide cyclical isoforms, cAMP, cGMP, cellular metabolites, urea, uric acid, pharmaceuticals, therapeutic drugs, narcotics, hallucinogens, gamma-hydroxybutyrate, cellular mediators, cytokines, chemokines, immune modulators, neural modulators, inflammatory modulators, prostaglandins, prostaglandin metabolites, explosives, trinitrotoluene, peptides, macromolecules, proteins, bacterial surface proteins, glycoproteins, lipids, glycolipids, nucleic acids, polysaccharides, lipopolysaccharides, whole cells, and subcellular organelles or cellular fractions.
9. The method of claim 6, wherein said assay is used for detecting a target molecule, wherein said target molecule is equal to or greater than 1,000 Daltons and water-soluble.
US12/931,309 2006-05-12 2011-01-28 Methods of running assays using intrachain fluorophore-quencher FRET-aptamers Abandoned US20120219961A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/931,309 US20120219961A1 (en) 2006-05-12 2011-01-28 Methods of running assays using intrachain fluorophore-quencher FRET-aptamers

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US11/433,009 US7906280B2 (en) 2005-05-13 2006-05-12 Methods of producing intrachain fluorophore-quencher FRET-aptamers and assays
US12/931,309 US20120219961A1 (en) 2006-05-12 2011-01-28 Methods of running assays using intrachain fluorophore-quencher FRET-aptamers

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/433,009 Continuation US7906280B2 (en) 2005-05-13 2006-05-12 Methods of producing intrachain fluorophore-quencher FRET-aptamers and assays

Publications (1)

Publication Number Publication Date
US20120219961A1 true US20120219961A1 (en) 2012-08-30

Family

ID=46719228

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/433,009 Expired - Fee Related US7906280B2 (en) 2005-05-13 2006-05-12 Methods of producing intrachain fluorophore-quencher FRET-aptamers and assays
US12/931,309 Abandoned US20120219961A1 (en) 2006-05-12 2011-01-28 Methods of running assays using intrachain fluorophore-quencher FRET-aptamers
US12/931,305 Abandoned US20120270221A1 (en) 2006-05-12 2011-01-28 Methods of sequencing fluorophore-quencher FRET-aptamers

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/433,009 Expired - Fee Related US7906280B2 (en) 2005-05-13 2006-05-12 Methods of producing intrachain fluorophore-quencher FRET-aptamers and assays

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/931,305 Abandoned US20120270221A1 (en) 2006-05-12 2011-01-28 Methods of sequencing fluorophore-quencher FRET-aptamers

Country Status (1)

Country Link
US (3) US7906280B2 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103760139A (en) * 2013-05-14 2014-04-30 江南大学 Method for identifying commercially available orange juice beverage based on three-dimensional fluorescence spectrum and robust mahalanobis distance
WO2014093698A1 (en) * 2012-12-12 2014-06-19 The Methodist Hospital Research Institute Multi-aptamer-based, cell-specific, one-step tumor cell detection assays
WO2014124046A1 (en) * 2013-02-08 2014-08-14 Bio-Rad Laboratories, Inc. Affinity-based partition assay for detection of target molecules
CN104568867A (en) * 2014-11-30 2015-04-29 陈燕婷 Homocysteine (HCy) kit based on aptamer fluorescent probe, and detection method of HCy kit
CN104561010A (en) * 2014-11-30 2015-04-29 陈燕婷 Homocysteine aptamer HCy5 and preparation method thereof
CN104597246A (en) * 2014-11-30 2015-05-06 陈燕婷 Homocysteine kit based on aptamer fluorescence probe HCy2 and detection method thereof
CN104677868A (en) * 2014-11-30 2015-06-03 陈燕婷 Homocysteine (HCy) kit based on aptamer fluorescence probe and detection method for concentration of homocysteine (HCy) by using homocysteine (HCy) kit
CN105300942A (en) * 2015-10-29 2016-02-03 锦州奥鸿药业有限责任公司 Method for detecting adenosine monophosphate content in deproteinised calf serum injection
US9788776B1 (en) 2014-09-22 2017-10-17 Verily Life Sciences Llc Protein M-based in vivo diagnostic system and detection method
US9927442B1 (en) 2014-10-31 2018-03-27 Verily Life Sciences Llc Biosensor for in vitro detection system and method of use
CN110174386A (en) * 2019-06-11 2019-08-27 青岛科技大学 A kind of Subjective and Objective sensing pair and its application detecting spermine

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100896987B1 (en) * 2007-03-14 2009-05-14 한국과학기술연구원 Detection method for target protein using aptamer and the detection kit
EP2197350A2 (en) * 2007-09-11 2010-06-23 Baxter International Inc. Infusion therapy sensor system
CN102323400B (en) * 2011-06-08 2013-11-27 中国人民解放军第三军医大学第一附属医院 Method for detecting and identifying variety of snake venom by utilizing adaptor technology
CN109804087A (en) * 2016-08-19 2019-05-24 昆塔波尔公司 Using quencher based on optical nano-pore sequencing
CN107449899A (en) * 2017-08-14 2017-12-08 天津科技大学 A kind of malachite green immunoaffinity gel detection column and preparation method thereof
WO2020010133A1 (en) * 2018-07-03 2020-01-09 Rutgers, The State University Of New Jersey A luminescent layered composition and a method for using the composition
WO2020046809A1 (en) * 2018-08-27 2020-03-05 The Regents Of The University Of California Reporter nucleic acids for type v crispr-mediated detection
EP3650559B1 (en) * 2018-11-08 2022-06-29 Siemens Healthcare GmbH Direct rna nanopore sequencing with help of a stem-loop polynucleotide
EP4288546A1 (en) 2021-02-04 2023-12-13 Fresenius Medical Care Holdings, Inc. Lipopolysaccharide (lps) aptamers and associated methods

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5795782A (en) * 1995-03-17 1998-08-18 President & Fellows Of Harvard College Characterization of individual polymer molecules based on monomer-interface interactions
US5866336A (en) * 1996-07-16 1999-02-02 Oncor, Inc. Nucleic acid amplification oligonucleotides with molecular energy transfer labels and methods based thereon
AU5492499A (en) * 1998-08-21 2000-03-14 University Of Virginia Patent Foundation Signal generating oligonucleotide-based biosensor
US6573047B1 (en) * 1999-04-13 2003-06-03 Dna Sciences, Inc. Detection of nucleotide sequence variation through fluorescence resonance energy transfer label generation
US6680377B1 (en) * 1999-05-14 2004-01-20 Brandeis University Nucleic acid-based detection
AU5723700A (en) * 1999-05-25 2000-12-12 Praelux Incorporated Method for sequency and characterizing polymeric biomolecules using aptamers anda method for producing aptamers

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Hamaguchi et al. (Analytical Biochemistry, 2001, vol. 294, 126-131) *
Mann et al. (JBC, 246(19):6106-6114) *
McKeen et al. (Org. Biomol. Chem., 2003, vol. 1, 2267-2275) *
Osborne et al. (Curr Opinion in Chemical Biology, 1997, vol.1, p. 5-9) *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014093698A1 (en) * 2012-12-12 2014-06-19 The Methodist Hospital Research Institute Multi-aptamer-based, cell-specific, one-step tumor cell detection assays
US10018633B2 (en) 2012-12-12 2018-07-10 The Methodist Hospital Research Institute Multi-aptamer-based, cell-specific, one-step tumor cell detection assays
WO2014124046A1 (en) * 2013-02-08 2014-08-14 Bio-Rad Laboratories, Inc. Affinity-based partition assay for detection of target molecules
US9850515B2 (en) 2013-02-08 2017-12-26 Bio-Rad Laboratories, Inc. Affinity-based partition assay for detection of target molecules
CN105102696A (en) * 2013-02-08 2015-11-25 生物辐射实验室股份有限公司 Affinity-based partition assay for detection of target molecules
CN103760139A (en) * 2013-05-14 2014-04-30 江南大学 Method for identifying commercially available orange juice beverage based on three-dimensional fluorescence spectrum and robust mahalanobis distance
US9788776B1 (en) 2014-09-22 2017-10-17 Verily Life Sciences Llc Protein M-based in vivo diagnostic system and detection method
US9927442B1 (en) 2014-10-31 2018-03-27 Verily Life Sciences Llc Biosensor for in vitro detection system and method of use
CN104568867A (en) * 2014-11-30 2015-04-29 陈燕婷 Homocysteine (HCy) kit based on aptamer fluorescent probe, and detection method of HCy kit
CN104677868A (en) * 2014-11-30 2015-06-03 陈燕婷 Homocysteine (HCy) kit based on aptamer fluorescence probe and detection method for concentration of homocysteine (HCy) by using homocysteine (HCy) kit
CN104597246A (en) * 2014-11-30 2015-05-06 陈燕婷 Homocysteine kit based on aptamer fluorescence probe HCy2 and detection method thereof
CN104561010A (en) * 2014-11-30 2015-04-29 陈燕婷 Homocysteine aptamer HCy5 and preparation method thereof
CN105300942A (en) * 2015-10-29 2016-02-03 锦州奥鸿药业有限责任公司 Method for detecting adenosine monophosphate content in deproteinised calf serum injection
CN110174386A (en) * 2019-06-11 2019-08-27 青岛科技大学 A kind of Subjective and Objective sensing pair and its application detecting spermine

Also Published As

Publication number Publication date
US20120270221A1 (en) 2012-10-25
US7906280B2 (en) 2011-03-15
US20060257914A1 (en) 2006-11-16

Similar Documents

Publication Publication Date Title
US20120219961A1 (en) Methods of running assays using intrachain fluorophore-quencher FRET-aptamers
US20060257915A1 (en) Methods of producing competitive aptamer fret reagents and assays
Bruno et al. A novel screening method for competitive FRET-aptamers applied to E. coli assay development
US11214841B2 (en) Rapid low-cost detection of valley fever using isothermal amplification and sensing methods
US20090186342A1 (en) Methods of producing competitive aptamer fret reagents and assays
Ma et al. Femtogram ultrasensitive aptasensor for the detection of OchratoxinA
US20160033495A1 (en) Detection of Non-Nucleic Acid Analytes Using Strand Displacement Exchange Reactions
JP2008259496A (en) Method and kit for detecting target protein using dna aptamer
Ma et al. A simple and rapid sensing strategy based on structure-switching signaling aptamers for the sensitive detection of chloramphenicol
JP2007525661A (en) Biosensors for detection of large molecules and other analytes
Cui et al. Low-background and visual detection of antibiotic based on target-activated colorimetric split peroxidase DNAzyme coupled with dual nicking enzyme signal amplification
Zhao et al. Aptamer binding assays and molecular interaction studies using fluorescence anisotropy-a review
Nie et al. A self-assemble aptamer fragment/target complex based high-throughput colorimetric aptasensor using enzyme linked aptamer assay
Ye et al. Orientation selection of broad-spectrum aptamers against lipopolysaccharides based on capture-SELEX by using magnetic nanoparticles
KR20190135468A (en) 2-piece medium probe
Tombelli et al. Aptamers biosensors for pharmaceutical compounds
EP3180463A1 (en) Detecting residual host cell proteins in recombinant protein preparations
US20220243196A1 (en) Screening method of aptamer and immunoassay using the aptamer
US20100291553A1 (en) Nucleic acid strand useful in detecting substance and method thereof
US20110306553A1 (en) Methods and uses involving heme binding protein 1
US20140349873A1 (en) Methods of Producing Competitive Aptamer FRET Reagents and Assays
Ge et al. Cocaine detection in blood serum using aptamer biosensor on gold nanoparticles and progressive dilution
US20140011200A1 (en) Methods of producing competitive aptamer fret reagents and assays
He et al. Selected ssDNA aptamers–graphene oxide‐based fluorescent biosensor to detect sulfameter in milk
US20120094277A1 (en) Methods of producing competitive aptamer FRET reagents and assays

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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