US20120208189A1 - Methods for isolation, identification, and quantification of mirnas - Google Patents

Methods for isolation, identification, and quantification of mirnas Download PDF

Info

Publication number
US20120208189A1
US20120208189A1 US13/350,277 US201213350277A US2012208189A1 US 20120208189 A1 US20120208189 A1 US 20120208189A1 US 201213350277 A US201213350277 A US 201213350277A US 2012208189 A1 US2012208189 A1 US 2012208189A1
Authority
US
United States
Prior art keywords
mirna
beads
sample
probe
nucleic acid
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
US13/350,277
Inventor
Tom Xu
Christopher Trinh
Carole Bornarth
Brian Evans
Mousumi Rath
Kathy Tran
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.)
Life Technologies Corp
Original Assignee
Life Technologies Corp
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 Life Technologies Corp filed Critical Life Technologies Corp
Priority to US13/350,277 priority Critical patent/US20120208189A1/en
Assigned to Life Technologies Corporation reassignment Life Technologies Corporation ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BORNARTH, CAROLE, EVANS, BRIAN, RATH, MOUSUMI, TRAN, KATHY, TRINH, CHRISTOPHER, XU, TOM
Publication of US20120208189A1 publication Critical patent/US20120208189A1/en
Priority to US15/682,103 priority patent/US11046727B2/en
Priority to US17/304,883 priority patent/US20210323994A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/02Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with ribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1003Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor
    • C12N15/1006Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor by means of a solid support carrier, e.g. particles, polymers
    • 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
    • C12Q1/6809Methods for determination or identification of nucleic acids involving differential detection
    • 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
    • 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
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • 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
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/178Oligonucleotides characterized by their use miRNA, siRNA or ncRNA

Definitions

  • This application relates to method and compositions and kits for isolation, identification, and quantification of miRNAs and other RNAs, including but not limited to, siRNAs, mRNAs, and snRNAs.
  • MicroRNAs regulate virtually every aspect of biology, including developmental timing, differentiation, proliferation, antiviral defense and metabolism.
  • MicroRNAs are ⁇ 22-nucleotide-long RNAs that are generated by sequential processing from longer transcripts that contain a stem-loop.
  • One strand is loaded into the miRNA-induced silencing complex (miRISC), which contains the proteins argonaute (Ago) and Tnrc6 (trinucleotide repeat—containing 6; GW182).
  • miRISC miRNA-induced silencing complex
  • the mature miRNA guides the miRISC to partially complementary sequences, termed miRNA recognition elements (MREs), in target mRNAs to repress mRNA translation, promote transcript decay or both.
  • MREs miRNA recognition elements
  • miRNAs have been reported from all living organisms, including humans, bacteria, viruses, plants, worms, and others. About 3% of human genes encode for miRNAs and about 30% of genes are believed to be regulated by miRNAs. It has also been widely reported that an abnormal level of miRNA expression or presence is associated with diseases, such as cancer, heart attack, diabetes, etc.
  • miRNAs Quantitative and qualitative isolation of miRNAs from various biological samples has been hampered for several reasons, including, but not limited to, labor-intensive and time consuming protocols; the nature of small size of miRNA leads to easy loss of the targets during extraction; miRNAs, like other RNAs, are not stable and can be degraded easily during processing and storage; the miRNA detection rate from real patient samples is usually low or undetectable, i.e., not quantitative; and further the extracted miRNA targets normally are poorly correlated between related study objects (e.g. placenta vs. blood).
  • related study objects e.g. placenta vs. blood.
  • miRNA or RNA
  • isolation reagents or kits that are commercially available (from QIAGEN—PAXGENE blood RNA kit, AMBION—LEUKOLOCK total RNA kit, and others), suffer from one or more of the following drawbacks: low sensitivity and detectability; low yield; not reproducible; large sample volume requirement; miRNA extraction does not scale well; time-consuming workflows, 4-5 hours workflow in general; low throughput and not automatable.
  • FIG. 1 Schematic for making anti-miRNA probe beads using chemical synthesis.
  • FIG. 2 Schematic for enzymatic synthesis of anti-miRNA probe beads.
  • FIG. 3A Non-anti-probe beads hybridization with Cy3-laneled Let-7a.
  • FIG. 3B Anti-Let-7a-probe beads hybridization with Cy3-labeled Let-7a.
  • FIG. 3C FACS after de-hybridization of Cy3-labeled Let-7a bond from anti-Let-7a-probe beads.
  • FIG. 4 Let-7 family miRNA profiling by C t values.
  • FIG. 5 miRNA detection after addition (spiking) to plasma.
  • FIG. 6A Quantitative real-time PCR Ct profiles of 96 miRNAs from blood.
  • FIG. 6B Quantitative real-time PCR Ct profiles of 95 miRNAs from blood.
  • FIG. 7 Saliva miRNA profiling using anti-miRNA probe beads.
  • FIG. 8 A384C6 Beads showing Sensitivity and Dynamic Range of anti-miRNA probe beads.
  • FIG. 9 miRNA Detectability and reproducibility.
  • FIG. 10A Let7a—miRNA profiling from human bladder LCM samples; LCM area and Ct correlation.
  • FIG. 10B Let7c—miRNA profiling from human bladder LCM samples; LCM area and Ct correlation.
  • FIG. 10C miR-16—miRNA profiling from human bladder LCM samples; LCM area and Ct correlation.
  • FIG. 11 Breast cancer FFPE sample miRNA profiling.
  • FIG. 12 Difference in expression of miRNA levels between breast cancer lines: MBA-MB-21 versus MCF-7 (delta Ct is shown).
  • FIG. 13 miRNA profiling from raw cow milk.
  • FIG. 14 miRNA profiling from urine from three donors.
  • FIG. 15 Megaplex A miRNA profiling from 25 microL serum.
  • FIG. 16 Megaplex B miRNA profiling from 25 microL serum.
  • FIG. 17 miRNA expression before and at 15-week of pregnancy.
  • the invention relates to a method for isolating, identifying, or quantifying an miRNA from a sample of interest, comprising: contacting the sample of interest with an anti-miRNA probe covalently attached to a bead; incubating the sample of interest—bead mixture under suitable conditions to form hybridized complexes between the miRNA in the sample of interest and the anti-miRNA probes on the beads; washing the beads under suitable conditions to remove the unbound sample material; and isolating, identifying, or quantifying the miRNA bound to the anti-miRNA probes.
  • the miRNA is isolated from the anti-miRNA probe beads before identifying or quantifying the miRNA.
  • the identifying or quantifying the miRNA is comprises using reverse transcription followed by conventional or real-time or quantitative polymerase chain reaction (q-PCR) or other methods of amplification.
  • q-PCR quantitative polymerase chain reaction
  • the identifying or quantifying a miRNA involves non-qPCR techniques.
  • the beads are magnetic beads. In another embodiment of the invention, the beads are non-magnetic beads. In one embodiment of the invention, the beads are carboxylic acid functionalized beads. In another embodiment of the invention, the beads are DNA beads. In another embodiment of the invention, the beads are P1 DNA beads.
  • the anti-miRNA probe is attached to the beads using chemical synthesis.
  • the anti-miRNA probe is attached to the beads using enzymatic synthesis.
  • the enzymatic synthesis comprises: taking an oligonucleotide that has a sequence complementary to an miRNA of interest (anti-miRNA probe); adding a terminal didexoy nucleotide to the 3′ end using a terminal transferase; adding a 5′-phosphate group to the oligonucleotide using a kinase; and ligating the oligonucleotide to DNA beads using a single-strand DNA ligase.
  • the kinase is a T4 polynucleotide kinase.
  • a single species of ant-miRNA probe is attached to an individual bead. In another embodiment of the invention more than one species of anti-miRNA probes are attached to an individual bead.
  • sample of interest is a biological sample.
  • the biological sample is selected from the group consisting of blood, serum, plasma, urine, saliva, cerebrospinal fluid, wound exudates, biopsies, autopsies, tissues, formalin-fixed, paraffin-embedded (FFPE) samples, or organs.
  • the biological sample is a biological fluid.
  • the biological fluid is blood.
  • the biological fluid is plasma.
  • the biological fluid is serum.
  • the biological fluid is saliva.
  • the sample could be a fossil or a fossilized rock or sediment.
  • a method of diagnosing a disease or disease progression comprising: identifying a set of miRNA markers that are differentially regulated during a causation or progression of a disease; isolating and measuring the levels of such miRNA markers from a suitable biological sample from a patient in need thereof, at different time points if necessary; and diagnosing the disease or its progression over time in the patient.
  • a kit for isolating, identifying, or quantifying an miRNA from a sample of interest comprising: beads comprising anti-miRNA probe molecules covalently attached thereto; a wash buffer; and an elution buffer.
  • an instruction leaflet may be included describing the product and protocol to carry out the inventive method.
  • an “amplicon” typically refers to a molecule made by copying or transcribing another molecule, e.g., as occurs in transcription, cloning, and/or in a polymerase chain reaction (“PCR”) (e.g., strand displacement PCR amplification (SDA), duplex PCR amplification, etc.) or other nucleic acid amplification technique.
  • PCR polymerase chain reaction
  • an amplicon is a copy of a selected nucleic acid or a portion thereof (e.g., a template or target nucleic acid) or is complementary thereto.
  • amplifying or “amplification” in the context of nucleic acids typically refers to the production of multiple copies of a polynucleotide, or a portion of the polynucleotide, typically starting from a small amount of the polynucleotide (e.g., a single polynucleotide molecule), where the amplification products or amplicons are generally detectable. Any of several methods may be used to amplify the target nucleic acid from the sample.
  • amplifying which typically refers to an “exponential” increase in target nucleic acid is being used herein to describe both linear and exponential increases in the numbers of a select target sequence of nucleic acid.
  • amplification reaction mixture refers to an aqueous solution comprising the various reagents used to amplify a target nucleic acid. These include enzymes, aqueous buffers, salts, amplification primers, target nucleic acid, and nucleoside triphosphates. Depending upon the context, the mixture can be either a complete or incomplete amplification reaction mixture.
  • the method used to amplify the target nucleic acid may be any available to one of skill in the art. Any in vitro means for multiplying the copies of a target sequence of nucleic acid may be utilized. These include linear, logarithmic, and/or any other amplification method. Exemplary methods include polymerase chain reaction (PCR; see, e.g., U.S.
  • RNA polymerases see, e.g., WO 2006/081222
  • strand displacement see, e.g., U.S. Pat. No. RE39007E
  • partial destruction of primer molecules see, e.g., WO2006087574
  • LCR ligase chain reaction
  • RNA transcription-based systems e.g., TAS, 3SR
  • rolling circle amplification RCA
  • RCA rolling circle amplification
  • SDA strand displacement amplification
  • Many systems are suitable for use in amplifying target nucleic acids and are contemplated herein as would be understood by one of skill in the art.
  • any of several methods may be used to detect target nucleic acids using various primers and/or probes.
  • Many different reagents, systems, and/or detectable labels may be used in the methods described herein. These include, for example, TaqMan® systems, detectable label-quencher systems (e.g., FRET, salicylate/DTPA ligand systems (see, e.g., Oser et al. Angew. Chem. Int. Engl. 29(10):1167 (1990), displacement hybridization, homologous probes, assays described in EP 070685), molecular beacons (e.g., NASBA), Scorpion, locked nucleic acid (LNA) bases (Singh, et al.
  • detectable label-quencher systems e.g., FRET, salicylate/DTPA ligand systems
  • displacement hybridization e.g., homologous probes, assays described in EP 070685
  • molecular beacons e.g.
  • fluorescent dyes are used to label probes and/or amplified products.
  • the probes bind to single-stranded and/or double-stranded amplified products, and/or the dyes intercalate into the double-stranded amplified products, and consequently, the resulting fluorescence increases as the amount of amplified product increases.
  • the T m is ascertained by observing a fluorescence decrease as the double-stranded amplified product dissociates and the intercalating dye is released therefrom.
  • the amount of fluorescence may be quantitated using standard equipment such as a spectra-fluorometer, for example.
  • the use of other methods and/or reagents is also contemplated herein.
  • TaqMan® One exemplary method for amplifying and detecting target nucleic acids is commercially available as TaqMan® (see, e.g., U.S. Pat. Nos. 4,889,818; 5,079,352; 5,210,015; 5,436,134; 5,487,972; 5,658,751; 5,210,015; 5,487,972; 5,538,848; 5,618,711; 5,677,152; 5,723,591; 5,773,258; 5,789,224; 5,801,155; 5,804,375; 5,876,930; 5,994,056; 6,030,787; 6,084,102; 6,127,155; 6,171,785; 6,214,979; 6,258,569; 6,814,934; 6,821,727; 7,141,377; and/or 7,445,900).
  • TaqMan® assays are typically carried out by performing nucleic acid amplification on a target polynucleotide using a nucleic acid polymerase having 5′-3′ nuclease activity, a primer capable of hybridizing to said target polynucleotide, and an oligonucleotide probe capable of hybridizing to said target polynucleotide 3′ relative to said primer.
  • the oligonucleotide probe typically includes a detectable label (e.g., a fluorescent reporter molecule) and a quencher molecule capable of quenching the fluorescence of said reporter molecule.
  • the detectable label and quencher molecule are part of a single probe.
  • the polymerase digests the probe to separate the detectable label from the quencher molecule.
  • the detectable label e.g., fluorescence
  • detection of the label corresponds to the occurrence of nucleic acid amplification (e.g., the higher the signal the greater the amount of amplification).
  • Variations of TaqMan® assays e.g., LNATM spiked TaqMan® assay are known in the art and would be suitable for use in the methods described herein.
  • the probe typically includes two complementary oligonucleotides of different lengths where one includes a detectable label and the other includes a quencher molecule. When not bound to a target nucleic acid, the quencher suppresses the signal from the detectable label. The probe becomes detectable upon displacement hybridization with a target nucleic acid. Multiple probes may be used, each containing different detectable labels, such that multiple target nucleic acids may be queried in a single reaction.
  • molecular beacons are single-stranded hairpin shaped oligonucleotide probes. In the presence of the target sequence, the probe unfolds, binds and emits a signal (e.g., fluoresces).
  • a molecular beacon typically includes at least four components: 1) the “loop”, an 18-30 nucleotide region which is complementary to the target sequence; 2) two 5-7 nucleotide “stems” found on either end of the loop and being complementary to one another; 3) at the 5′ end, a detectable label; and 4) at the 3′ end, a quencher dye that prevents the detectable label from emitting a single when the probe is in the closed loop shape (e.g., not bound to a target nucleic acid).
  • the “stem” portion of the beacon separates out resulting in the probe hybridizing to the target.
  • Other types of molecular beacons are also known and may be suitable for use in the methods described herein.
  • Molecular beacons may be used in a variety of assay systems.
  • One such system is nucleic acid sequence-based amplification (NASBA®), a single step isothermal process for amplifying RNA to double stranded DNA without temperature cycling.
  • a NASBA reaction typically requires avian myeloblastosis virus reverse transcriptase (AMV-RT or AMV), T7 RNA polymerase, RNase H, and two oligonucleotide primers.
  • AMV-RT or AMV avian myeloblastosis virus reverse transcriptase
  • T7 RNA polymerase T7 RNA polymerase
  • RNase H oligonucleotide primers
  • the amplified target nucleic acid may be detected using a molecular beacon.
  • Other uses for molecular beacons are known in the art and would be suitable for use in the methods described herein.
  • Scorpion primers are bi-functional molecules in which a primer is covalently linked to the probe, along with a detectable label (e.g., a fluorophore) and a quencher. In the presence of a target nucleic acid, the detectable label and the quencher separate which leads to an increase in signal emitted from the detectable label.
  • a primer used in the amplification reaction includes a probe element at the 5′ end along with a “PCR blocker” element (e.g., HEG monomer) at the start of the hairpin loop.
  • the probe typically includes a self-complementary stem sequence with a detectable label at one end and a quencher at the other.
  • PCR amplification cycle
  • the Scorpion system may be used to examine and identify point mutations using multiple probes that may be differently tagged to distinguish between the probes.
  • the newly synthesized target region will be attached to the same strand as the probe.
  • the probe and the target hybridize.
  • the hairpin sequence then hybridizes to a part of the newly produced PCR product. This results in the separation of the detectable label from the quencher and causes emission of the signal.
  • Other uses for molecular beacons are known in the art and would be suitable for use in the methods described herein.
  • One or more detectable labels and/or quenching agents are typically attached to an oligonucleotide primer and/or probe.
  • the detectable label may emit a signal when free or when bound to the target nucleic acid.
  • the detectable label may also emit a signal when in proximity to another detectable label.
  • Detectable labels may also be used with quencher molecules such that the signal is only detectable when not in sufficiently close proximity to the quencher molecule.
  • the assay system may cause the detectable label to be liberated from the quenching molecule. Any of several detectable labels may be used to label the primers and probes used in the methods described herein.
  • the detectable label may be attached to a probe, which may be incorporated into a primer, or may otherwise bind to amplified target nucleic acid (e.g., a detectable nucleic acid binding agent such as an intercalating or non-intercalating dye).
  • a detectable nucleic acid binding agent such as an intercalating or non-intercalating dye.
  • Exemplary detectable labels include, for instance, a fluorescent dye or fluorophore (e.g., a chemical group that can be excited by light to emit fluorescence or phosphorescence), “acceptor dyes” capable of quenching a fluorescent signal from a fluorescent donor dye, and the like.
  • a fluorescent dye or fluorophore e.g., a chemical group that can be excited by light to emit fluorescence or phosphorescence
  • acceptor dyes capable of quenching a fluorescent signal from a fluorescent donor dye
  • Suitable detectable labels may include, for example, fluorosceins (e.g., 5-carboxy-2,7-dichlorofluorescein; 5-Carboxyfluorescein (5-FAM); 5-HAT (Hydroxy Tryptamine); 5-Hydroxy Tryptamine (HAT); 6-JOE; 6-carboxyfluorescein (6-FAM); FITC); Alexa fluors (e.g., 350, 405, 430, 488, 500, 514, 532, 546, 555, 568, 594, 610, 633, 635, 647, 660, 680, 700, 750); BODIPY fluorophores (e.g., 492/515, 493/503, 500/510, 505/515, 530/550, 542/563, 558/568, 564/570, 576/589, 581/591, 630/650-X, 650/665-X, 665/676, FL, FL ATP,
  • EGFP blue fluorescent protein
  • BFP blue fluorescent protein
  • EBFP EBFP2, Azurite, mKalama1
  • cyan fluorescent protein e.g., ECFP, Cerulean, CyPet
  • yellow fluorescent protein e.g., YFP, Citrine, Venus, YPet
  • FRET donor/acceptor pairs e.g., fluorescein/tetramethylrhodamine, IAEDANS/fluorescein, EDANS/dabcyl, fluorescein/fluorescein, BODIPY FL/BODIPY FL, Fluorescein/QSY7 and QSY9
  • LysoTracker and LysoSensor e.g., LysoTracker Blue DND-22, LysoTracker Blue-White DPX, LysoTracker Yellow HCK-123, LysoTracker Green DND-26, LysoTracker Red DND-99, LysoSensor Blue
  • Nucleic acid binding agents may also be used to detect nucleic acids amplified using the methods described herein. Many suitable detectable nucleic acid binding agents are available to one of skill in the art and may be used alone or in combination with other agents and/or components of an assay system. Exemplary DNA binding agents may include, for example, acridines (e.g., acridine orange, acriflavine), actinomycin D (Jain, et al. J. Mol. Biol. 68:21 (1972)), anthramycin, BOBOTM-1, BOBOTM-3, BO-PROTM-1, cbromomycin, DAPI (Kapuseinski, et al. Nuc. Acids Res.
  • acridines e.g., acridine orange, acriflavine
  • actinomycin D Jain, et al. J. Mol. Biol. 68:21 (1972)
  • anthramycin BOBOTM-1, BOBOTM
  • SYBR® Green II SYTOX blue, SYTOX green, SYTO® 43, SYTO® 44, SYTO® 45, SYTOX® Blue, TO-PRO®-1, SYTO® 11, SYTO® 13, SYTO® 15, SYTO® 16, SYTO® 20, SYTO® 23, thiazole orange (Aldrich Chemical Co., Milwaukee, Wis.), TOTOTM-3, YO-PRO®-1, and YOYO®-3 (Molecular Probes, Inc., Eugene, Oreg.), among others.
  • SYBR® Green I see, e.g., U.S. Pat. Nos.
  • the primary interaction between the antiparallel polynucleotides is typically base specific, e.g., A/T and G/C, by Watson/Crick and/or Hoogsteen-type interactions. It is not a requirement that two polynucleotides have 100% complementarity over their full length to achieve hybridization.
  • a hybridization complex can form from intermolecular interactions, or alternatively, can form from intramolecular interactions.
  • Hybridization occurs due to a variety of well-characterized forces, including hydrogen bonding, solvent exclusion, and base stacking.
  • An extensive guide to nucleic hybridization may be found in, Laboratory Techniques in Biochemistry and Molecular Biology-Hybridization with Nucleic Acid Probes, part I, chapter 2, “Overview of principles of hybridization and the strategy of nucleic acid probe assays,” Elsevier (1993).
  • a “mixture” refers to a combination of two or more different components.
  • a “reaction mixture” refers a mixture that comprises molecules that can participate in and/or facilitate a given reaction or assay.
  • an amplification reaction mixture generally includes a solution containing reagents necessary to carry out an amplification reaction, and typically contains primers, a biocatalyst (e.g., a nucleic acid polymerase, a ligase, etc.), dNTPs, and a divalent metal cation in a suitable buffer.
  • a reaction mixture is referred to as complete if it contains all reagents necessary to carry out the reaction, and incomplete if it contains only a subset of the necessary reagents.
  • reaction components are routinely stored as separate solutions, each containing a subset of the total components, for reasons of convenience, storage stability, or to allow for application-dependent adjustment of the component concentrations, and that reaction components are combined prior to the reaction to create a complete reaction mixture.
  • reaction components are packaged separately for commercialization and that useful commercial kits may contain any subset of the reaction or assay components, which includes the biomolecules of the invention.
  • a “moiety” or “group” refers to one of the portions into which something, such as a molecule, is or can be divided (e.g., a functional group, substituent group, or the like).
  • an oligonucleotide described herein includes at least one donor moiety and/or at least one acceptor moiety in certain embodiments.
  • mutation refers to a nucleic acid that has been altered in its nucleic acid sequence or an encoded protein product of a nucleic acid that has been altered in its amino acid sequence relative to an unaltered or native form of the nucleic acid or encoded protein product. Such alterations include, for example, point mutations or substitutions, deletions and insertions.
  • nucleic acid refers to a polymer that can be corresponded to a ribose nucleic acid (RNA) or deoxyribose nucleic acid (DNA) polymer, or an analog thereof. This includes polymers of nucleotides such as RNA and DNA, as well as modified forms thereof, peptide nucleic acids (PNAs), locked nucleic acids (LNATM), and the like.
  • a nucleic acid can be a polymer that includes multiple monomer types, e.g., both RNA and DNA subunits.
  • a nucleic acid can be or can include, e.g., a chromosome or chromosomal segment, a vector (e.g., an expression vector), an expression cassette, a naked DNA or RNA polymer, the product of a polymerase chain reaction (PCR), an oligonucleotide, a probe, a primer, etc.
  • a nucleic acid may be, e.g., single-stranded, double-stranded, triple-stranded (and the like) and is not limited to any particular length. Unless otherwise indicated, a particular nucleic acid sequence optionally comprises or encodes complementary sequences, in addition to any sequence explicitly indicated.
  • Nucleic acids are not limited to molecules having naturally occurring polynucleotide sequences or structures, naturally occurring backbones, and/or naturally occurring internucleotide linkages.
  • nucleic acids containing one or more carbocyclic sugars are also included within this definition (Jenkins et al. (1995) Chem. Soc. Rev. pp 169-176, which is incorporated by reference).
  • a nucleic acid will generally contain phosphodiester bonds, in some cases nucleic acid analogs are included that have alternate backbones. These may include, without limitation, phosphoramide (Beaucage et al. (1993) Tetrahedron 49(10): 1925 and the references therein; Letsinger (1970) J. Org.
  • nucleic acid analogs In addition to naturally occurring heterocyclic bases that are typically found in nucleic acids (e.g., adenine, guanine, thymine, cytosine, and uracil), nucleic acid analogs also include those having non-naturally occurring heterocyclic or other modified bases. For instance, certain bases used in nucleotides that act as melting temperature (T m ) modifiers are optionally included.
  • T m melting temperature
  • 7-deazapurines e.g., 7-deazaguanine, 7-deazaadenine, and the like
  • pyrazolo[3,4-d]pyrimidines propynyl-dN (e.g., propynyl-dU, propynyl-dC, and the like)
  • hypoxanthine inosine; xanthine; 8-aza derivatives of 2-aminopurine, 2,6-diaminopurine, 2-amino-6-chloropurine, hypoxanthine, inosine and xanthine
  • 6-azacytosine 5-fluorocytosine; 5-chlorocytosine; 5-iodocytos
  • Nucleoside typically refers to a nucleic acid component that comprises a base or basic group (e.g., comprising at least one homocyclic ring, at least one heterocyclic ring, at least one aryl group, and/or the like) covalently linked to a sugar moiety (e.g., a ribose sugar, etc.), a derivative of a sugar moiety, or a functional equivalent of a sugar moiety (e.g., an analog, such as carbocyclic ring).
  • a base is typically linked to a 1′-position of that sugar moiety.
  • a base can be naturally occurring (e.g., a purine base, such as adenine (A) or guanine (G), a pyrimidine base, such as thymine (T), cytosine (C), or uracil (U)), or non-naturally occurring (e.g., a 7-deazapurine base, a pyrazolo[3,4-d]pyrimidine base, a propynyl-dN base, etc.).
  • exemplary nucleosides include ribonucleosides, deoxyribonucleosides, dideoxyribonucleosides, carbocyclic nucleosides, and the like.
  • nucleotide refers to an ester of a nucleoside, e.g., a phosphate ester of a nucleoside.
  • a nucleotide can include 1, 2, 3, or more phosphate groups covalently linked to a 5′ position of a sugar moiety of the nucleoside.
  • Nucleoside triphosphates and 2′-deoxynucleoside triphosphates or their chemically modified versions may be used as substrates for multiple-nucleotide extension by APP where, for example, one nucleotide is incorporated the extending strand can be further extended.
  • 2′,3′-dideoxynucleoside triphosphates may be used as terminators for further extension may be used for single-nucleotide extension.
  • 2′,3′-dideoxynucleoside triphosphates may be labeled with radioactivity or fluorescence dye for differentiation from the 3′ terminal dideoxynucleotide of oligonucleotide P*.
  • Mixtures of nucleoside triphosphates or 2′-deoxynucleoside triphosphates and 2′,3′-dideoxynucleoside triphosphates may also be used.
  • a “nucleotide incorporating biocatalyst” refers to a catalyst that catalyzes the incorporation of nucleotides into a nucleic acid. Nucleotide incorporating biocatalysts are typically enzymes. An “enzyme” is a protein- and/or nucleic acid-based catalyst that acts to reduce the activation energy of a chemical reaction involving other compounds or “substrates.” A “nucleotide incorporating enzyme” refers to an enzyme that catalyzes the incorporation of nucleotides into a nucleic acid, e.g., during nucleic acid amplification or the like.
  • Exemplary nucleotide incorporating enzymes include, e.g., polymerases, terminal transferases, reverse transcriptases (e.g., RQY polymerase described above), telomerases, polynucleotide phosphorylases, and the like.
  • a “thermostable enzyme” refers to an enzyme that is stable to heat, is heat resistant, and retains sufficient catalytic activity when subjected to elevated temperatures for selected periods of time. For example, a thermostable polymerase retains sufficient activity to effect subsequent primer extension reactions when subjected to elevated temperatures for the time necessary to effect denaturation of double-stranded nucleic acids.
  • thermostable polymerase refers to an enzyme that is suitable for use in a temperature cycling reaction, such as a polymerase chain reaction (“PCR”).
  • PCR polymerase chain reaction
  • enzymatic activity refers to the catalysis of the combination of the nucleotides in the proper manner to form primer extension products that are complementary to a template nucleic acid.
  • Exemplary thermostable polymerases are described herein, and others may available to the skilled artisan may also be suitable.
  • TdT terminal deoxynucleotidyl transferase catalyses the addition of nucleotides to the 3′ terminus of a DNA molecule. Unlike most DNA polymerases it does not require a template.
  • the preferred substrate of this enzyme is a 3′-overhang, but it can also add nucleotides to blunt or recessed 3′ ends.
  • Cobalt is a necessary cofactor; however the enzyme catalyzes reaction upon Mg and Mn administration in vitro.
  • CircLigaseTM (Epicentre Biotechnologies), ssDNA Ligase is a thermostable ATP-dependent ligase that catalyzes intramolecular ligation (i.e. circularization) of ssDNA templates having a 5′-phosphate and a 3′-hydroxyl group.
  • CircLigase ssDNA Ligase ligates ends of ssDNA in the absence of a complementary sequence. The enzyme is therefore useful for making circular ssDNA molecules from linear ssDNA.
  • Circular ssDNA molecules can be used as substrates for rolling-circle replication or rolling-circle transcription.
  • Linear ssDNA of >30 bases is circularized by CircLigase enzyme. Under standard reaction conditions, virtually no linear concatamers or circular concatamers are produced.
  • CircLigase enzyme also has activity in ligating a single-stranded nucleic acid having a 3′-hydroxyl ribonucleotide and a 5′-phosphorylated ribonucleotide or deoxyribonucleotide.
  • oligonucleotide refers to a nucleic acid that includes at least two nucleic acid monomer units (e.g., nucleotides), typically more than three monomer units, and more typically greater than ten monomer units. The exact size of an oligonucleotide generally depends on various factors, including the ultimate function or use of the oligonucleotide.
  • the nucleoside monomers are linked by phosphodiester bonds or analogs thereof, including phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phosphoranilidate, phosphoramidate, and the like, including associated counterions, e.g., H + , NH 4 + , Na + , and the like, if such counterions are present.
  • the 3′ end linkage of P* is a phosphodiester bond.
  • Oligonucleotides may be prepared by any suitable method, including, but not limited to, isolation of an existing or natural sequence, DNA replication or amplification, reverse transcription, cloning and restriction digestion of appropriate sequences, or direct chemical synthesis by a method such as the phosphotriester method of Narang et al. ((1979) Meth. Enzymol. 68:90-99); the phosphodiester method of Brown et al. ((1979) Meth. Enzymol. 68:109-151); the diethylphosphoramidite method of Beaucage et al. ((1981) Tetrahedron Lett. 22:1859-1862); the triester method of Matteucci et al. ((1981) J. Am. Chem. Soc. 103:3185-3191); automated synthesis methods; or the solid support method described in U.S. Pat. No. 4,458,066, and/or other methods known to those skilled in the art.
  • a “primer nucleic acid” or “primer” is a nucleic acid that can hybridize to a target or template nucleic acid and permit chain extension or elongation using, e.g., a nucleotide incorporating biocatalyst, such as a polymerase under appropriate reaction conditions.
  • a primer nucleic acid is typically a natural or synthetic oligonucleotide (e.g., a single-stranded oligodeoxyribonucleotide). Although other primer nucleic acid lengths are optionally utilized, they typically comprise hybridizing regions that range from about 8 to about 100 nucleotides in length. Shorter primer nucleic acids generally require lower temperatures to form sufficiently stable hybrid complexes with template nucleic acids.
  • a primer nucleic acid that is at least partially complementary to a subsequence of a template nucleic acid is typically sufficient to hybridize with the template for extension to occur.
  • a primer nucleic acid may be labeled, if desired, by incorporating a detectable label as described herein.
  • probe nucleic acid refers to a labeled or unlabeled oligonucleotide capable of selectively hybridizing to a target or template nucleic acid under suitable conditions.
  • a probe is sufficiently complementary to a specific target sequence contained in a nucleic acid sample to form a stable hybridization duplex with the target sequence under selected hybridization conditions.
  • a hybridization assay carried out using a probe under sufficiently stringent hybridization conditions permits the selective detection of a specific target sequence.
  • hybridizing region refers to that region of a nucleic acid that is exactly or substantially complementary to, and therefore capable of hybridizing to, the target sequence.
  • the hybridizing region is typically from about 8 to about 100 nucleotides in length.
  • the hybridizing region generally refers to the entire oligonucleotide, the probe may include additional nucleotide sequences that function, for example, as linker binding sites to provide a site for attaching the probe sequence to a solid support.
  • a probe of the invention may be generally included in a nucleic acid that comprises one or more labels (e.g., donor moieties, acceptor moieties, and/or quencher moieties), such as a 5′-nuclease probe, a hybridization probe, a fluorescent resonance energy transfer (FRET) probe, a hairpin probe, or a molecular beacon, which can also be utilized to detect hybridization between the probe and target nucleic acids in a sample.
  • the hybridizing region of the probe is completely complementary to the target sequence. However, in general, complete complementarity is not necessary (e.g., nucleic acids can be partially complementary to one another); stable hybridization complexes may contain mismatched bases or unmatched bases.
  • Modification of the stringent conditions may be necessary to permit a stable hybridization complex with one or more base pair mismatches or unmatched bases.
  • Sambrook et al., Molecular Cloning: A Laboratory Manual, 3rd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (2001) provides guidance for suitable modification.
  • Stability of the target/probe hybridization complex depends on a number of variables including length of the oligonucleotide, base composition and sequence of the oligonucleotide, temperature, and ionic conditions.
  • probe nucleic acids can also be used as primer nucleic acids.
  • a target nucleic acid is any nucleic acid for which an assay system is designed to identify or detect as present (or not), and/or quantify in a test sample.
  • Such nucleic acids may include, for example, those of infectious agents (e.g., virus, bacteria, parasite, and the like), a disease process such as cancer, diabetes, or the like, or to measure an immune response.
  • infectious agents e.g., virus, bacteria, parasite, and the like
  • a disease process such as cancer, diabetes, or the like
  • test samples include various types of samples, such as biological samples.
  • Exemplary biological samples include, for instance, a bodily fluid (e.g., blood, saliva, or spinal fluid), a tissue sample, a food (e.g., meat) or beverage (e.g., milk) product, or the like.
  • Expressed nucleic acids may include, for example, genes for which expression (or lack thereof) is associated with medical conditions such as infectious disease (e.g., bacterial, viral, fungal, protozoal infections) or cancer.
  • infectious disease e.g., bacterial, viral, fungal, protozoal infections
  • the methods described herein may also be used to detect contaminants (e.g., bacteria, virus, fungus, and/or protozoan) in pharmaceutical, food, or beverage products.
  • the methods may be useful to, for example, detect minimal residual disease (e.g., rare remaining cancer cells during remission, especially mutations in the p53 gene or other tumor suppressor genes previously identified within the tumors), and/or measure mutation load (e.g., the frequency of specific somatic mutations present in normal tissues, such as blood or urine).
  • minimal residual disease e.g., rare remaining cancer cells during remission, especially mutations in the p53 gene or other tumor suppressor genes previously identified within the tumors
  • measure mutation load e.g., the frequency of specific somatic mutations present in normal tissues, such as blood or urine.
  • Kits for performing the methods described herein are also provided.
  • the kit typically includes at least a pair of oligonucleotides (e.g., at least one of the pair being a P* oligonucleotide) for amplifying at least one target nucleic acid from a sample, one or more polyphosphorolyzing agents described herein, a biocatalyst (e.g., DNA polymerase) and/or corresponding one or more probes labeled with a detectable label.
  • the kit may also include samples containing pre-defined target nucleic acids to be used in control reactions.
  • the kit may also optionally include stock solutions, buffers, enzymes, detectable labels or reagents required for detection, tubes, membranes, and the like that may be used to complete the amplification reaction. In some embodiments, multiple primer sets are included. Other embodiments of particular systems and kits are also contemplated.
  • Magnetic polymer particles are of general utility in various medical and biochemical fields, for example as transport vehicles for the delivery of pharmaceutical products, for diagnostic purposes, for separation and for synthetic purposes. Such particles rely upon their magnetic properties in order to perform these functions: in diagnostic assay applications, for example, application of a magnetic field to a sample containing an analyte bound to magnetic polymer particles allows the isolation of the analyte without the use of centrifugation or filtration; and in therapeutic applications, for example, application of a magnetic field to the patient may serve to target drug-carrying magnetic polymer particles to a desired body site.
  • maghemite- or magnetite-containing polymer particles from pre-formed magnetic iron oxides, e.g. magnetite.
  • the magnetic polymer particles used in the process of the invention may be any magnetic polymer particle e.g. as described in U.S. Pat. No. 4,654,267.
  • the particles are preferably porous to allow the presence of the superparamagnetic crystals in the pores thereof.
  • the surface of the magnetic particles is normally functionalized to allow coupling of a ligand to the polymer particle, e.g. it may be functionalized to carry any known surface structure such as carboxyl groups, tosyl groups, amino groups, epoxy groups, maleamido groups, thiol groups etc.
  • the surface may be amine functionalized.
  • an amine functionalized surface can itself be further functionalized to carry other functional groups, e.g. COOH groups.
  • the polymer particle is preferably made from combinations of vinylic polymers (e.g. styrene), acrylates and/or methacrylates.
  • the polymeric material may optionally be cross-linked, for example by incorporation of cross-linking agents, for example as comonomers, e.g. divinylbenzene (DVB) or ethyleneglycol dimethacrylate.
  • cross-linking agents for example as comonomers, e.g. divinylbenzene (DVB) or ethyleneglycol dimethacrylate.
  • the polymer is a cross-linked styrenic polymer (e.g.
  • a styrene-divinylbenzene polymer surface functionalized by the use of a nitro-group containing comonomer, e.g. nitro-styrene, and subsequent reduction) or a cross-linked (meth)acrylic polymer surface functionalized by the use of an epoxy-group containing co-monomer (e.g. glycidylmethacrylate) and subsequent amination (e.g. by reaction with ethylene diamine).
  • a nitro-group containing comonomer e.g. nitro-styrene, and subsequent reduction
  • a cross-linked (meth)acrylic polymer surface functionalized by the use of an epoxy-group containing co-monomer (e.g. glycidylmethacrylate) and subsequent amination e.g. by reaction with ethylene diamine.
  • the superparamagnetic crystals in the polymer particles used in the process of the invention may be of any material capable of being deposited in superparamagnetic crystalline form in the porous polymer particles.
  • Magnetic iron oxides e.g. magnetite or maghemite are preferred; however the crystals may be of mixed metal oxides or other magnetic material if desired.
  • the total quantity of crystalline magnetic material present is generally more than 1%, preferably more than 3%, desirably more than or equal to 5% (by weight, e.g. up to 40% wt. The percentage is calculated on a Fe (or equivalent metal in the case of magnetic materials other than iron oxides) weight basis based upon the overall dry weight of the coated particles.
  • Polymer particles according to the various aspects of the present invention may generally have sizes (i.e. diameters) that are in the micrometer range, e.g. 0.3 to 100 ⁇ m, especially 0.5 to 50 ⁇ m, more especially 0.8 to 8 ⁇ m, e.g. 0.8 to 1.2 ⁇ m. 1 ⁇ m beads are referred.
  • the porous particles used will have a surface area of at least 15 m 2 /g (measured by the BET nitrogen absorption method), and more preferably at least 30 m 2 /g, e.g. up to 700 m 2 /g, when corrected to a mean particle diameter of 2.7 ⁇ m (i.e. multiply surface area by 2.7/MD, where MD is the mean diameter in micrometers).
  • the particle pore volume is preferably at least 0.1 mL/g.
  • the polymer particles are spherical and substantially monodisperse before they are coated and especially preferably remain spherical and substantially monodisperse once they have been coated.
  • substantially monodisperse it is meant that for a majority of particles, the particles have a coefficient of variation (CV) of less than 20%, for example less than 15%, preferably less than 12%, more preferably less than 11%, still more preferably less than 10% and most preferably no more than about 8%, e.g. 2 to 5%.
  • CV is determined in percentage as
  • CV is preferably calculated on the main mode, i.e. by fitting a monomodal distribution curve to the detected particle size distribution. Thus some particles below or above mode size may be discounted in the calculation which may for example be based on about 90% of total particle number (of detectable particles that is). Such a determination of CV may be performable on a Coulter or other particle size analyzer.
  • Functionalization of the polymeric material may take place after polymerization by, for example, nitration and subsequent reduction of the thus-formed nitro groups to pendant amine groups; or direct amination, for example by treatment with amino ethanol.
  • polymeric particles prepared by the well-known Ugelstad two-step swelling process and the improvements thereto disclosed in WO 00/61647 may be used.
  • Porous polymer particles produced according to the processes described in this publication may have magnetic particles deposited in their pores by standard techniques.
  • porous polymer particles may be prepared from nitro styrene and DVB, and magnetic material introduced as taught in U.S. Pat. No. 4,654,267.
  • DYNABEADS are particularly advantageous since they remain in suspension and do not exhibit magnetic particle sedimentation often associated with other magnetic beads.
  • DYNABEADS also show excellent magnetic mobility compared to other magnetic particles in which high levels of iron are present.
  • DYNABEADS exhibit beneficial kinetics allowing shorter reaction times and higher throughputs. Their non-specific binding is lower than other magnetic beads and their proper use results in a concentration of the desired material taking place resulting in easier and more efficient washing procedures.
  • DYNABEADS MYONE beads are easy to automate and are monodisperse.
  • DYNABEADS MYONE carboxylic acid beads are used generally; however, other functional groups may also be utilized.
  • FIG. 1 Schematic is shown in FIG. 1 .
  • MYONE carboxylic acid beads 10 billion beads are placed in 1.5 ml microfuge tube. Beads are washed with 1000 ⁇ L 10 mM NaOH 3 times. Beads are further washed with 10000 ⁇ L deionized H 2 O, 4 times. pH is adjusted to about 6.5-7.
  • a reaction master mix is prepared in 1.5 mL tube, as follows:
  • 1M EDC is prepared in 5 mM aqueous Imidazole-HCl.
  • One mg/5.2 ⁇ L 18 mg/100 ⁇ L 5 mM aqueous Imidazole is prepared.
  • 192 ⁇ L (2 ⁇ ) of 0.5 mM aqueous anti-miRNA oligo is added to the master mix. Beads are suspended in reaction master mix and treated as follows: remove the H 2 O from the beads
  • the tube is rotated overnight at room temperature.
  • the beads are further washed as follows: Wash with 1000 ⁇ L DMSO. Wash 2 times with 1000 ⁇ L deionized water. Wash 2 times with 1000 ⁇ L 1 ⁇ TEX (Tris, EDTA, Triton X-100). Wash 3 times with 1000 ⁇ L 1 ⁇ TEX and heat for 8 min at 80° C. Beads are stored in 1.00 mL 1 ⁇ TEX at 4° C.
  • FIG. 2 Schematic is shown in FIG. 2 .
  • the reaction is incubated at 37° C. for 2 hrs.
  • the terminal transferase is inactivated by heating the reaction mix at 95° C. for 5 min.
  • Reagents SOLIDTM P1 DNA Beads (ABI), 5′P-Let-7a, b, c, d, e and f Probes, Ligation Buffer (EpiCentre Biotechnologies), Ligase (EpiCentre Biotechnologies), ATP (Ambion), MnCl 2 (Teknova), PEG6000 (Emerald Biosystems).
  • Denaturing Solution Added 200 ⁇ L Denaturant to 1800 ⁇ L, 1 ⁇ Denaturing buffer and mixed well. Placed the reaction tube in a magnetic rack for 1 min and remove the supernatant. Added 200 ⁇ L 1 ⁇ TEX buffer. Sonicated the beads using Covaris Covalent Declump 1. Placed the tube in a magnetic rack for 1 min and removed the supernatant. Added 200 ⁇ L of the Denaturing Buffer, vortexed, and let stand for 1 min. Placed the tube in a magnetic rack for 1 min and removed the supernatant. Added 200 ⁇ L 1 ⁇ TEX buffer, vortexed and pulsed down. Placed the tube in a magnetic rack for 1 min and removed the supernatant. Repeat twice. Beads are stored (in 200 ⁇ L TEX buffer, ⁇ 2.5 million beads/ ⁇ L) at 4° C.
  • Hybridization of ant-miRNA Beads Hybridization with Cy3-labeled miRNA
  • Fluorescent flow cytometry analysis is done to ensure that anti-miRNA probes are covalently attached to the beads and that Cy3-labeled miRNAs are effectively captured by the anti-miRNA beads.
  • Reagents Anti-ath-miR159 Beads, Cy-3-ath-miR159a, Control: non-anti-probe beads, Cy3-ath-miR159a.
  • RNAs may be eluted from the beads by de-hybridization using buffers that are known to one skilled in the art, e.g., 10 mM Tris:HCl or 10 mM Tris:HCl with 01 mM EDTA.
  • Blood Sample Collection 40 ⁇ L of fresh finger blood sample was collected in a tube containing 0.4 ⁇ L of 0.5M EDTA (pH 8.0) using a mini finger lancet.
  • Plasma and Red Blood Cells Separation and Lysis The blood sample is centrifuged and the plasma is carefully pipetted out. 20A plasma is mixed with 40 ⁇ L Tempus buffer. The red blood cells are washed with 100 ⁇ L 0.9% NaCl, centrifuged, and the supernatant is removed. Washing is repeated 2 more times. 10 ⁇ L red blood cells are mixed with 20 ⁇ L Tempus Buffer.
  • Hyb Sample Serial Dilution 6 ⁇ L of the lysate solution prepared above is diluted in 194 ⁇ L Oligo dT hybridization buffer and vortexed. This is Hyb sample A (1 ⁇ L blood in 1004 volume). The samples are serially diluted according to following table for the rest of Hyb samples.
  • Hybridization 5 ⁇ L each of Let-7a thru Let-7f beads ( ⁇ 2.5 million/ ⁇ L, 12.5 million total each) are combined in a LoBind tube, 200 ⁇ L TEX buffer is added, and the tubes are vortexed. Tubes/beads are magnetized to remove the supernatant. 6 ⁇ L of the mixed beads are added into each hybridization sample as prepared above. The beads are shaken at 1100 rpm for 20 min at room temperature. The beads are magnetized to remove the supernatants. The beads are washed according to the following table. Finally, 1 ⁇ TE buffer is added and the beads are sonicated with Covalent Declump 1.
  • RT Reverse Transcription
  • TaqMan PCR Reaction The reverse-transcribed cDNA from the captured miRNAs are quantitatively analyzed using Applied Biosystems miRNA TaqMan assays and TaqMan Universal PCR Master Mix following the recommended protocols. TaqMan real-time PCR reactions are run on Applied Biosystems 7500 instrument using the default cycling parameters. The results are listed in Table 1 and FIG. 4 .
  • a mixture of Let-7a-f miRNAs was spiked in plasma.
  • the spiked plasma sample was serially diluted 10 ⁇ with Hyb buffer.
  • the miRNAs were captured and quantified by the beads and TaqMan PCR. Results are shown in FIG. 5 .
  • Run 1 Run 2 Total miRNA Tested 96 96 Detectable miRNA (Ct ⁇ 35) 71 70 Undetectable miRNA 25 26 Detection Rate 74.0% 72.9%
  • miRNA capturing and quantification from saliva The following protocol was used.
  • A384C6 beads with 386 miRNA targets based on the ABI Megaplex A assay panel.
  • B384C6 beads with 385 miRNA targets based on the ABI Megaplex B assay panel.
  • Each panel contains 6 non-human miRNAs: ath-miR159a, cel-miR-2, cel-miR-39, cel-miR-54, cel-miR-238, cel-lin-4. They can be used as external controls at user's choice.
  • Reagents in the kit Lysis Buffer, ABC Buffer, Wash Buffer 1, Wash Buffer 2, Elution Buffer, LoBind Tubes (Eppendorf), Human Panel A Beads (1 million/microL). Or Human Panel Panel B Beads.
  • Reagent Preparation Add 6 mL of Lysis Buffer to the ABC Buffer bottle to make the final ABC Buffer for use. Add 7 mL of 100% Ethanol to the Wash Buffer 2 bottle to make the final Wash Buffer 2 for use.
  • Beads Preparation Vortex the beads thoroughly to suspend the beads in solution. Sonicate the beads by an ultrasonic water bath for 1 minute. Vortex and then aliquot 80 uL beads (80 million beads total) into a LoBind 1.5-mL microfuge tube. Magnet the beads for 1 minute and remove the supernatant.
  • Sample Preparation Select (check mark) the sample type you are going to test. Based on your selection, prepare the sample for hybridization according to following procedures:
  • Total volume ⁇ 150 uL Cell 5 ⁇ 10 ⁇ circumflex over ( ) ⁇ 4 Mix the 50 uL cell/PBS suspension with 150 uL Lysis Buffer and vortex Cultures cells in for ⁇ 30 seconds. Total volume ⁇ 200 uL 50 uL PBS Solid 1-10 mg If needed, grind or homogenize the tissue. Add 30 uL Lysis Buffer and Tissue vortex for ⁇ 30 seconds. Add 120 uL ABC Buffer and vortex for ⁇ 30 seconds. Total volume ⁇ 150 uL FFPE 1 Wash the FFPE sample with xylene (2 min x 2), 100%, 95% and 75% dissection ethanol (1 min each), H2O (1 min).
  • Hybridization Transfer all the prepared sample into the bead tube prepared above. Using a ThermoMixer, shake the vial at 1200 rpm at 30° C. for 40 minutes.
  • Wash Magnet the beads for 1 minute and remove the supernatant. Add 100 uL Wash Buffer 1. Vortex to resuspend the beads into solution then pulse briefly. Magnet the beads for 1 minute and remove the supernatant. Add 100 uL Wash Buffer 2. Vortex briefly to resuspend the beads into solution then pulse briefly. Magnet the beads for 1 minute and remove the supernatant. Repeat step c. Pulse the tube down to collect any residual liquid. Place the tube on magnetic rack for 20 sec, and remove any residual liquid using a fine pipette tip.
  • FFPE breast cancer formalin-fixed paraffin-embedded
  • the profiling of microRNA in human urine samples during pregnancy at various stages of pregnancy showed that of the 38 ⁇ L miRNAs tested, 200 were detected in Urine samples using a Ct cutoff of 35. 18 pregnancy related miRNAs consistently showed increased expression with pregnancy progression to second trimester.
  • miR143 and miR145 are significantly down regulated in balloon injured arteries in comparison to normal artery. The results are shown in the table below. A representative miRNA profiling is shown in FIG. 17 .

Abstract

Method and compositions and kits for isolation, identification, and quantification of miRNAs and other small RNAs, including but not limited to, siRNAs, mRNAs, and snRNAs are disclosed. Methods of diagnosing a disease or its progression are also disclosed.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of priority under 35 U.S.C. §119(e) to U.S. Provisional Patent Application No. 61/432,874, filed 14 Jan. 2011, which is herein incorporated by reference in its entirety.
  • FIELD OF THE INVENTION
  • This application relates to method and compositions and kits for isolation, identification, and quantification of miRNAs and other RNAs, including but not limited to, siRNAs, mRNAs, and snRNAs.
  • BACKGROUND
  • MicroRNAs regulate virtually every aspect of biology, including developmental timing, differentiation, proliferation, antiviral defense and metabolism. MicroRNAs are ˜22-nucleotide-long RNAs that are generated by sequential processing from longer transcripts that contain a stem-loop. One strand is loaded into the miRNA-induced silencing complex (miRISC), which contains the proteins argonaute (Ago) and Tnrc6 (trinucleotide repeat—containing 6; GW182). The other strand is usually degraded. The mature miRNA guides the miRISC to partially complementary sequences, termed miRNA recognition elements (MREs), in target mRNAs to repress mRNA translation, promote transcript decay or both. MicroRNAs probably regulate the expression of most coding genes. miRNAs have been reported from all living organisms, including humans, bacteria, viruses, plants, worms, and others. About 3% of human genes encode for miRNAs and about 30% of genes are believed to be regulated by miRNAs. It has also been widely reported that an abnormal level of miRNA expression or presence is associated with diseases, such as cancer, heart attack, diabetes, etc.
  • Quantitative and qualitative isolation of miRNAs from various biological samples has been hampered for several reasons, including, but not limited to, labor-intensive and time consuming protocols; the nature of small size of miRNA leads to easy loss of the targets during extraction; miRNAs, like other RNAs, are not stable and can be degraded easily during processing and storage; the miRNA detection rate from real patient samples is usually low or undetectable, i.e., not quantitative; and further the extracted miRNA targets normally are poorly correlated between related study objects (e.g. placenta vs. blood).
  • For example, most miRNA (or RNA), isolation reagents or kits that are commercially available (from QIAGEN—PAXGENE blood RNA kit, AMBION—LEUKOLOCK total RNA kit, and others), suffer from one or more of the following drawbacks: low sensitivity and detectability; low yield; not reproducible; large sample volume requirement; miRNA extraction does not scale well; time-consuming workflows, 4-5 hours workflow in general; low throughput and not automatable. Thus, there is a need in the field for better methods for isolating, identifying, and quantifying miRNAs.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1: Schematic for making anti-miRNA probe beads using chemical synthesis.
  • FIG. 2: Schematic for enzymatic synthesis of anti-miRNA probe beads.
  • FIG. 3A: Non-anti-probe beads hybridization with Cy3-laneled Let-7a.
  • FIG. 3B: Anti-Let-7a-probe beads hybridization with Cy3-labeled Let-7a.
  • FIG. 3C: FACS after de-hybridization of Cy3-labeled Let-7a bond from anti-Let-7a-probe beads.
  • FIG. 4: Let-7 family miRNA profiling by Ct values.
  • FIG. 5: miRNA detection after addition (spiking) to plasma.
  • FIG. 6A: Quantitative real-time PCR Ct profiles of 96 miRNAs from blood.
  • FIG. 6B: Quantitative real-time PCR Ct profiles of 95 miRNAs from blood.
  • FIG. 7: Saliva miRNA profiling using anti-miRNA probe beads.
  • FIG. 8: A384C6 Beads showing Sensitivity and Dynamic Range of anti-miRNA probe beads.
  • FIG. 9: miRNA Detectability and reproducibility.
  • FIG. 10A: Let7a—miRNA profiling from human bladder LCM samples; LCM area and Ct correlation.
  • FIG. 10B: Let7c—miRNA profiling from human bladder LCM samples; LCM area and Ct correlation.
  • FIG. 10C: miR-16—miRNA profiling from human bladder LCM samples; LCM area and Ct correlation.
  • FIG. 11: Breast cancer FFPE sample miRNA profiling.
  • FIG. 12: Difference in expression of miRNA levels between breast cancer lines: MBA-MB-21 versus MCF-7 (delta Ct is shown).
  • FIG. 13: miRNA profiling from raw cow milk.
  • FIG. 14: miRNA profiling from urine from three donors.
  • FIG. 15: Megaplex A miRNA profiling from 25 microL serum.
  • FIG. 16: Megaplex B miRNA profiling from 25 microL serum.
  • FIG. 17: miRNA expression before and at 15-week of pregnancy.
  • SUMMARY OF THE INVENTION
  • As described above, the isolation, detection and quantification of miRNAs have been a daunting problem. Applicants have now developed a simple and elegant solution to the problem. In one embodiment, the invention relates to a method for isolating, identifying, or quantifying an miRNA from a sample of interest, comprising: contacting the sample of interest with an anti-miRNA probe covalently attached to a bead; incubating the sample of interest—bead mixture under suitable conditions to form hybridized complexes between the miRNA in the sample of interest and the anti-miRNA probes on the beads; washing the beads under suitable conditions to remove the unbound sample material; and isolating, identifying, or quantifying the miRNA bound to the anti-miRNA probes.
  • In a further embodiment of the invention, the miRNA is isolated from the anti-miRNA probe beads before identifying or quantifying the miRNA.
  • In another embodiment of the invention, the identifying or quantifying the miRNA is comprises using reverse transcription followed by conventional or real-time or quantitative polymerase chain reaction (q-PCR) or other methods of amplification. In another embodiment, the identifying or quantifying a miRNA involves non-qPCR techniques.
  • In one embodiment of the invention, the beads are magnetic beads. In another embodiment of the invention, the beads are non-magnetic beads. In one embodiment of the invention, the beads are carboxylic acid functionalized beads. In another embodiment of the invention, the beads are DNA beads. In another embodiment of the invention, the beads are P1 DNA beads.
  • In one embodiment of the invention, the anti-miRNA probe is attached to the beads using chemical synthesis.
  • In another embodiment of the invention, the anti-miRNA probe is attached to the beads using enzymatic synthesis. In another embodiment of the invention, the enzymatic synthesis comprises: taking an oligonucleotide that has a sequence complementary to an miRNA of interest (anti-miRNA probe); adding a terminal didexoy nucleotide to the 3′ end using a terminal transferase; adding a 5′-phosphate group to the oligonucleotide using a kinase; and ligating the oligonucleotide to DNA beads using a single-strand DNA ligase. In one embodiment, the kinase is a T4 polynucleotide kinase.
  • In one embodiment of the invention, a single species of ant-miRNA probe is attached to an individual bead. In another embodiment of the invention more than one species of anti-miRNA probes are attached to an individual bead.
  • In one embodiment of the invention, sample of interest is a biological sample. In another embodiment of the invention, the biological sample is selected from the group consisting of blood, serum, plasma, urine, saliva, cerebrospinal fluid, wound exudates, biopsies, autopsies, tissues, formalin-fixed, paraffin-embedded (FFPE) samples, or organs. In another embodiment of the invention, the biological sample is a biological fluid. In another embodiment of the invention, the biological fluid is blood. In another embodiment of the invention, the biological fluid is plasma. In another embodiment of the invention, the biological fluid is serum. In another embodiment of the invention, the biological fluid is saliva. In another embodiment, the sample could be a fossil or a fossilized rock or sediment.
  • In another embodiment, a method of diagnosing a disease or disease progression is disclosed, comprising: identifying a set of miRNA markers that are differentially regulated during a causation or progression of a disease; isolating and measuring the levels of such miRNA markers from a suitable biological sample from a patient in need thereof, at different time points if necessary; and diagnosing the disease or its progression over time in the patient.
  • In another embodiment of the invention, a kit for isolating, identifying, or quantifying an miRNA from a sample of interest is provided, comprising: beads comprising anti-miRNA probe molecules covalently attached thereto; a wash buffer; and an elution buffer.
  • In another embodiment of the kit an instruction leaflet may be included describing the product and protocol to carry out the inventive method.
  • DETAILED DESCRIPTION OF THE INVENTION
  • It is to be understood that the particular methods, reaction mixtures, and/or systems described herein may vary. The terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting. Further, unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention pertains. As used in this specification and the appended claims, the singular forms “a”, “an”, and “the” also include plural referents unless the context clearly provides otherwise. All numerical ranges are intended to encompass each individual value within the range as if each were separately listed (e.g., 10-20 may include one or more of 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 and/or 20). In terms of concentration ranges, these encompass fractional ranges, e.g., all values between 10-20, represents as if each were individually written out (e.g., 10.8, 11.5). The term “approximately”, when used to modify a group of numerical values, is meant to apply to each value individually unless otherwise indicated.
  • An “amplicon” typically refers to a molecule made by copying or transcribing another molecule, e.g., as occurs in transcription, cloning, and/or in a polymerase chain reaction (“PCR”) (e.g., strand displacement PCR amplification (SDA), duplex PCR amplification, etc.) or other nucleic acid amplification technique. Typically, an amplicon is a copy of a selected nucleic acid or a portion thereof (e.g., a template or target nucleic acid) or is complementary thereto. The term “amplifying” or “amplification” in the context of nucleic acids typically refers to the production of multiple copies of a polynucleotide, or a portion of the polynucleotide, typically starting from a small amount of the polynucleotide (e.g., a single polynucleotide molecule), where the amplification products or amplicons are generally detectable. Any of several methods may be used to amplify the target nucleic acid from the sample. The term “amplifying” which typically refers to an “exponential” increase in target nucleic acid is being used herein to describe both linear and exponential increases in the numbers of a select target sequence of nucleic acid. The term “amplification reaction mixture” refers to an aqueous solution comprising the various reagents used to amplify a target nucleic acid. These include enzymes, aqueous buffers, salts, amplification primers, target nucleic acid, and nucleoside triphosphates. Depending upon the context, the mixture can be either a complete or incomplete amplification reaction mixture. The method used to amplify the target nucleic acid may be any available to one of skill in the art. Any in vitro means for multiplying the copies of a target sequence of nucleic acid may be utilized. These include linear, logarithmic, and/or any other amplification method. Exemplary methods include polymerase chain reaction (PCR; see, e.g., U.S. Pat. Nos. 4,683,202; 4,683,195; 4,965,188; and/or 5,035,996), isothermal procedures (using one or more RNA polymerases (see, e.g., WO 2006/081222), strand displacement (see, e.g., U.S. Pat. No. RE39007E), partial destruction of primer molecules (see, e.g., WO2006087574)), ligase chain reaction (LCR) (see, e.g., Wu, et al., Genomics 4: 560-569 (1990) and/or Barany, et al. PNAS USA 88:189-193 (1991)), Qβ RNA replicase systems (see, e.g., WO/1994/016108), RNA transcription-based systems (e.g., TAS, 3SR), rolling circle amplification (RCA) (see, e.g., U.S. Pat. No. 5,854,033; U.S. Pub. No. 2004/265897; Lizardi et al. Nat. Genet. 19: 225-232 (1998); and/or Banér al. Nucleic Acid Res., 26: 5073-5078 (1998)), and strand displacement amplification (SDA) (Little, et al. Clin Chem 45:777-784 (1999)), among others. Many systems are suitable for use in amplifying target nucleic acids and are contemplated herein as would be understood by one of skill in the art.
  • Any of several methods may be used to detect target nucleic acids using various primers and/or probes. Many different reagents, systems, and/or detectable labels may be used in the methods described herein. These include, for example, TaqMan® systems, detectable label-quencher systems (e.g., FRET, salicylate/DTPA ligand systems (see, e.g., Oser et al. Angew. Chem. Int. Engl. 29(10):1167 (1990), displacement hybridization, homologous probes, assays described in EP 070685), molecular beacons (e.g., NASBA), Scorpion, locked nucleic acid (LNA) bases (Singh, et al. Chem Commun 4:455-456 (1998)), peptide nucleic acid (PNA) probes (Pellestor, et al. European J. Human Gen. 12:694-700 (2004)), Eclipse probes (Afonina, et al. Biotechniques 32:940-949 (2002)), light-up probes (Svanvik, et al. Anal Biochem 281:26-35 (2001)), molecular beacons (Tyagi, et al. Nat. Biotechnol. 14:303-308 (1996)), tripartite molecular beacons (Nutiu, et al. Nucleic Acids Res. 30:e94 (2002)), QuantiProbes (www.qiagen.com), HyBeacons (French, et al. Mol. Cell. Probes 15:363-374 (2001)), displacement probes (Li, et al. Nucleic Acids Res. 30:e5 (2002)), HybProbes (Cardullo, et al. PNAS 85:8790-8794 (1988)), MGB Alert (www.nanogen.com), Q-PNA (Fiandaca, et al. Genome Res. 11:609-611 (2001)), Plexor (www.Promega.com), LUX primers (Nazarenko, et al. Nucleic Acids Res. 30:e37 (2002)), Scorpion primers (Whitcombe, et al. Nat Biotechnol 17:804-807 (1999)), AmpliFluor (Sunrise) primers (Nazarenko, et al. Nucleic Acids Res. 25:2516-2521 (1997)), DzyNA primers (Todd, et al. Clin. Chem. 46:625-630 (2000)), and the like. In each of these assays, the generation of amplification products may be monitored while the reaction is in progress. An apparatus for detecting the signal generated by the detectable label may be used to detect, measure, and quantify the signal before, during, and/or after amplification. The particular type of signal may dictate the choice of detection method. For example, in some embodiments, fluorescent dyes are used to label probes and/or amplified products. The probes bind to single-stranded and/or double-stranded amplified products, and/or the dyes intercalate into the double-stranded amplified products, and consequently, the resulting fluorescence increases as the amount of amplified product increases. In some embodiments, the Tm is ascertained by observing a fluorescence decrease as the double-stranded amplified product dissociates and the intercalating dye is released therefrom. The amount of fluorescence may be quantitated using standard equipment such as a spectra-fluorometer, for example. The use of other methods and/or reagents is also contemplated herein.
  • One exemplary method for amplifying and detecting target nucleic acids is commercially available as TaqMan® (see, e.g., U.S. Pat. Nos. 4,889,818; 5,079,352; 5,210,015; 5,436,134; 5,487,972; 5,658,751; 5,210,015; 5,487,972; 5,538,848; 5,618,711; 5,677,152; 5,723,591; 5,773,258; 5,789,224; 5,801,155; 5,804,375; 5,876,930; 5,994,056; 6,030,787; 6,084,102; 6,127,155; 6,171,785; 6,214,979; 6,258,569; 6,814,934; 6,821,727; 7,141,377; and/or 7,445,900). TaqMan® assays are typically carried out by performing nucleic acid amplification on a target polynucleotide using a nucleic acid polymerase having 5′-3′ nuclease activity, a primer capable of hybridizing to said target polynucleotide, and an oligonucleotide probe capable of hybridizing to said target polynucleotide 3′ relative to said primer. The oligonucleotide probe typically includes a detectable label (e.g., a fluorescent reporter molecule) and a quencher molecule capable of quenching the fluorescence of said reporter molecule. Typically, the detectable label and quencher molecule are part of a single probe. As amplification proceeds, the polymerase digests the probe to separate the detectable label from the quencher molecule. The detectable label (e.g., fluorescence) is monitored during the reaction, where detection of the label corresponds to the occurrence of nucleic acid amplification (e.g., the higher the signal the greater the amount of amplification). Variations of TaqMan® assays (e.g., LNA™ spiked TaqMan® assay) are known in the art and would be suitable for use in the methods described herein.
  • Another exemplary system utilizes double-stranded probes in displacement hybridization methods (see, e.g., Morrison et al. Anal. Biochem., 18:231-244 (1989); and/or Li, et al. Nucleic Acids Res., 30(2,e5) (2002)). In such methods, the probe typically includes two complementary oligonucleotides of different lengths where one includes a detectable label and the other includes a quencher molecule. When not bound to a target nucleic acid, the quencher suppresses the signal from the detectable label. The probe becomes detectable upon displacement hybridization with a target nucleic acid. Multiple probes may be used, each containing different detectable labels, such that multiple target nucleic acids may be queried in a single reaction.
  • Additional exemplary methods for amplifying and detecting target nucleic acids involve “molecular beacons”, which are single-stranded hairpin shaped oligonucleotide probes. In the presence of the target sequence, the probe unfolds, binds and emits a signal (e.g., fluoresces). A molecular beacon typically includes at least four components: 1) the “loop”, an 18-30 nucleotide region which is complementary to the target sequence; 2) two 5-7 nucleotide “stems” found on either end of the loop and being complementary to one another; 3) at the 5′ end, a detectable label; and 4) at the 3′ end, a quencher dye that prevents the detectable label from emitting a single when the probe is in the closed loop shape (e.g., not bound to a target nucleic acid). Thus, in the presence of a complementary target, the “stem” portion of the beacon separates out resulting in the probe hybridizing to the target. Other types of molecular beacons are also known and may be suitable for use in the methods described herein. Molecular beacons may be used in a variety of assay systems. One such system is nucleic acid sequence-based amplification (NASBA®), a single step isothermal process for amplifying RNA to double stranded DNA without temperature cycling. A NASBA reaction typically requires avian myeloblastosis virus reverse transcriptase (AMV-RT or AMV), T7 RNA polymerase, RNase H, and two oligonucleotide primers. After amplification, the amplified target nucleic acid may be detected using a molecular beacon. Other uses for molecular beacons are known in the art and would be suitable for use in the methods described herein.
  • The Scorpion system is another exemplary assay format that may be used in the methods described herein. Scorpion primers are bi-functional molecules in which a primer is covalently linked to the probe, along with a detectable label (e.g., a fluorophore) and a quencher. In the presence of a target nucleic acid, the detectable label and the quencher separate which leads to an increase in signal emitted from the detectable label. Typically, a primer used in the amplification reaction includes a probe element at the 5′ end along with a “PCR blocker” element (e.g., HEG monomer) at the start of the hairpin loop. The probe typically includes a self-complementary stem sequence with a detectable label at one end and a quencher at the other. In the initial amplification cycles (e.g., PCR), the primer hybridizes to the target and extension occurs due to the action of polymerase. The Scorpion system may be used to examine and identify point mutations using multiple probes that may be differently tagged to distinguish between the probes. Using PCR as an example, after one extension cycle is complete, the newly synthesized target region will be attached to the same strand as the probe. Following the second cycle of denaturation and annealing, the probe and the target hybridize. The hairpin sequence then hybridizes to a part of the newly produced PCR product. This results in the separation of the detectable label from the quencher and causes emission of the signal. Other uses for molecular beacons are known in the art and would be suitable for use in the methods described herein.
  • One or more detectable labels and/or quenching agents are typically attached to an oligonucleotide primer and/or probe. The detectable label may emit a signal when free or when bound to the target nucleic acid. The detectable label may also emit a signal when in proximity to another detectable label. Detectable labels may also be used with quencher molecules such that the signal is only detectable when not in sufficiently close proximity to the quencher molecule. For instance, in some embodiments, the assay system may cause the detectable label to be liberated from the quenching molecule. Any of several detectable labels may be used to label the primers and probes used in the methods described herein. As mentioned above, in some embodiments the detectable label may be attached to a probe, which may be incorporated into a primer, or may otherwise bind to amplified target nucleic acid (e.g., a detectable nucleic acid binding agent such as an intercalating or non-intercalating dye). When using more than one detectable label, each should differ in their spectral properties such that the labels may be distinguished from each other, or such that together the detectable labels emit a signal that is not emitted by either detectable label alone. Exemplary detectable labels include, for instance, a fluorescent dye or fluorophore (e.g., a chemical group that can be excited by light to emit fluorescence or phosphorescence), “acceptor dyes” capable of quenching a fluorescent signal from a fluorescent donor dye, and the like. Suitable detectable labels may include, for example, fluorosceins (e.g., 5-carboxy-2,7-dichlorofluorescein; 5-Carboxyfluorescein (5-FAM); 5-HAT (Hydroxy Tryptamine); 5-Hydroxy Tryptamine (HAT); 6-JOE; 6-carboxyfluorescein (6-FAM); FITC); Alexa fluors (e.g., 350, 405, 430, 488, 500, 514, 532, 546, 555, 568, 594, 610, 633, 635, 647, 660, 680, 700, 750); BODIPY fluorophores (e.g., 492/515, 493/503, 500/510, 505/515, 530/550, 542/563, 558/568, 564/570, 576/589, 581/591, 630/650-X, 650/665-X, 665/676, FL, FL ATP, FI-Ceramide, R6G SE, TMR, TMR-X conjugate, TMR-X, SE, TR, TR ATP, TR-X SE), coumarins (e.g., 7-amino-4-methylcoumarin, AMC, AMCA, AMCA-S, AMCA-X, ABQ, CPM methylcoumarin, coumarin phalloidin, hydroxycoumarin, CMFDA, methoxycoumarin), calcein, calcein AM, calcein blue, calcium dyes (e.g., calcium crimson, calcium green, calcium orange, calcofluor white), Cascade Blue, Cascade Yellow; Cy™ dyes (e.g., 3, 3.18, 3.5, 5, 5.18, 5.5, 7), cyan GFP, cyclic AMP Fluorosensor (FiCRhR), fluorescent proteins (e.g., green fluorescent protein (e.g., GFP. EGFP), blue fluorescent protein (e.g., BFP, EBFP, EBFP2, Azurite, mKalama1), cyan fluorescent protein (e.g., ECFP, Cerulean, CyPet), yellow fluorescent protein (e.g., YFP, Citrine, Venus, YPet), FRET donor/acceptor pairs (e.g., fluorescein/tetramethylrhodamine, IAEDANS/fluorescein, EDANS/dabcyl, fluorescein/fluorescein, BODIPY FL/BODIPY FL, Fluorescein/QSY7 and QSY9), LysoTracker and LysoSensor (e.g., LysoTracker Blue DND-22, LysoTracker Blue-White DPX, LysoTracker Yellow HCK-123, LysoTracker Green DND-26, LysoTracker Red DND-99, LysoSensor Blue DND-167, LysoSensor Green DND-189, LysoSensor Green DND-153, LysoSensor Yellow/Blue DND-160, LysoSensor Yellow/Blue 10,000 MW dextran), Oregon Green (e.g., 488, 488-X, 500, 514); rhodamines (e.g., 110, 123, B, B 200, BB, BG, B extra, 5-carboxytetramethylrhodamine (5-TAMRA), 5 GLD, 6-Carboxyrhodamine 6G, Lissamine, Lissamine Rhodamine B, Phallicidine, Phalloidine, Red, Rhod-2,5-ROX (carboxy-X-rhodamine), Sulphorhodamine B can C, Sulphorhodamine G Extra, Tetramethylrhodamine (TRITC), WT), Texas Red, Texas Red-X, VIC and other labels described in, e.g., US Pub. No. 2009/0197254), among others as would be known to those of skill in the art. Other detectable labels may also be used (see, e.g., US Pub. No. 2009/0197254), as would be known to those of skill in the art.
  • Nucleic acid binding agents may also be used to detect nucleic acids amplified using the methods described herein. Many suitable detectable nucleic acid binding agents are available to one of skill in the art and may be used alone or in combination with other agents and/or components of an assay system. Exemplary DNA binding agents may include, for example, acridines (e.g., acridine orange, acriflavine), actinomycin D (Jain, et al. J. Mol. Biol. 68:21 (1972)), anthramycin, BOBO™-1, BOBO™-3, BO-PRO™-1, cbromomycin, DAPI (Kapuseinski, et al. Nuc. Acids Res. 6(112): 3519 (1979)), daunomycin, distamycin (e.g., distamycin D), dyes described in U.S. Pat. No. 7,387,887, ellipticine, ethidium salts (e.g., ethidium bromide), fluorcoumanin, fluorescent intercalators as described in U.S. Pat. No. 4,257,774, GelStar® (Cambrex Bio Science Rockland Inc., Rockland, Me.), Hoechst 33258 (Searle and Embrey, 1990, Nuc. Acids Res. 18:3753-3762), Hoechst 33342, homidium, JO-PRO™-1, LIZ dyes, LO-PRO™-1, mepacrine, mithramycin, NED dyes, netropsin, 4′6-diamidino-α-phenylindole, proflavine, POPO™-1, POPO™-3, PO-PRO™-1, propidium iodide, ruthenium polypyridyls, S5, SYBR® Gold, SYBR® Green I (U.S. Pat. Nos. 5,436,134 and 5,658,751), SYBR® Green II, SYTOX blue, SYTOX green, SYTO® 43, SYTO® 44, SYTO® 45, SYTOX® Blue, TO-PRO®-1, SYTO® 11, SYTO® 13, SYTO® 15, SYTO® 16, SYTO® 20, SYTO® 23, thiazole orange (Aldrich Chemical Co., Milwaukee, Wis.), TOTO™-3, YO-PRO®-1, and YOYO®-3 (Molecular Probes, Inc., Eugene, Oreg.), among others. SYBR® Green I (see, e.g., U.S. Pat. Nos. 5,436,134; 5,658,751; and/or 6,569,927), for example, has been used to monitor a PCR reaction by amplifying the target sequence in the presence of the dye, exciting the biological sample with light at a wavelength absorbed by the dye and detecting the emission therefrom; and, determining a melting profile of the amplified target sequence. The presence of amplified products and, therefore, the target sequence in the sample, may thereafter be determined by, for example, performing a melting curve analysis (e.g., non-linear least squares regression of the sum of multiple gaussians). It is to be understood that the use of the SYBR® Green dye is presented as an example and that many such dyes may be used in the methods described herein. Other nucleic acid binding agents may also be suitable as would be understood by one of skill in the art.
  • Nucleic acids “hybridize” or “anneal” in a base-pairing interaction of one polynucleotide with another polynucleotide (typically an antiparallel polynucleotide) that results in formation of a duplex or other higher-ordered structure, typically termed a hybridization complex. The primary interaction between the antiparallel polynucleotides is typically base specific, e.g., A/T and G/C, by Watson/Crick and/or Hoogsteen-type interactions. It is not a requirement that two polynucleotides have 100% complementarity over their full length to achieve hybridization. In some aspects, a hybridization complex can form from intermolecular interactions, or alternatively, can form from intramolecular interactions. Hybridization occurs due to a variety of well-characterized forces, including hydrogen bonding, solvent exclusion, and base stacking. An extensive guide to nucleic hybridization may be found in, Laboratory Techniques in Biochemistry and Molecular Biology-Hybridization with Nucleic Acid Probes, part I, chapter 2, “Overview of principles of hybridization and the strategy of nucleic acid probe assays,” Elsevier (1993).
  • A “mixture” refers to a combination of two or more different components. A “reaction mixture” refers a mixture that comprises molecules that can participate in and/or facilitate a given reaction or assay. To illustrate, an amplification reaction mixture generally includes a solution containing reagents necessary to carry out an amplification reaction, and typically contains primers, a biocatalyst (e.g., a nucleic acid polymerase, a ligase, etc.), dNTPs, and a divalent metal cation in a suitable buffer. A reaction mixture is referred to as complete if it contains all reagents necessary to carry out the reaction, and incomplete if it contains only a subset of the necessary reagents. It will be understood by one of skill in the art that reaction components are routinely stored as separate solutions, each containing a subset of the total components, for reasons of convenience, storage stability, or to allow for application-dependent adjustment of the component concentrations, and that reaction components are combined prior to the reaction to create a complete reaction mixture. Furthermore, it will be understood by one of skill in the art that reaction components are packaged separately for commercialization and that useful commercial kits may contain any subset of the reaction or assay components, which includes the biomolecules of the invention.
  • A “moiety” or “group” refers to one of the portions into which something, such as a molecule, is or can be divided (e.g., a functional group, substituent group, or the like). For example, an oligonucleotide described herein includes at least one donor moiety and/or at least one acceptor moiety in certain embodiments.
  • The term “mutation” refers to a nucleic acid that has been altered in its nucleic acid sequence or an encoded protein product of a nucleic acid that has been altered in its amino acid sequence relative to an unaltered or native form of the nucleic acid or encoded protein product. Such alterations include, for example, point mutations or substitutions, deletions and insertions.
  • The term “nucleic acid” or “polynucleotide” refers to a polymer that can be corresponded to a ribose nucleic acid (RNA) or deoxyribose nucleic acid (DNA) polymer, or an analog thereof. This includes polymers of nucleotides such as RNA and DNA, as well as modified forms thereof, peptide nucleic acids (PNAs), locked nucleic acids (LNA™), and the like. In certain embodiments, a nucleic acid can be a polymer that includes multiple monomer types, e.g., both RNA and DNA subunits. A nucleic acid can be or can include, e.g., a chromosome or chromosomal segment, a vector (e.g., an expression vector), an expression cassette, a naked DNA or RNA polymer, the product of a polymerase chain reaction (PCR), an oligonucleotide, a probe, a primer, etc. A nucleic acid may be, e.g., single-stranded, double-stranded, triple-stranded (and the like) and is not limited to any particular length. Unless otherwise indicated, a particular nucleic acid sequence optionally comprises or encodes complementary sequences, in addition to any sequence explicitly indicated.
  • Nucleic acids are not limited to molecules having naturally occurring polynucleotide sequences or structures, naturally occurring backbones, and/or naturally occurring internucleotide linkages. For example, nucleic acids containing one or more carbocyclic sugars are also included within this definition (Jenkins et al. (1995) Chem. Soc. Rev. pp 169-176, which is incorporated by reference). To further illustrate, although a nucleic acid will generally contain phosphodiester bonds, in some cases nucleic acid analogs are included that have alternate backbones. These may include, without limitation, phosphoramide (Beaucage et al. (1993) Tetrahedron 49(10): 1925 and the references therein; Letsinger (1970) J. Org. Chem. 35:3800; Sprinzl et al. (1977) Eur. J. Biochem. 81:579; Letsinger et al. (1986) Nucl. Acids Res. 14: 3487; Sawai et al. (1984) Chem. Lett. 805; Letsinger et al. (1988) J. Am. Chem. Soc. 110:4470; and Pauwels et al. (1986) Chemica Scripta 26:1419), phosphorothioate (Mag et al. (1991) Nucleic Acids Res. 19:1437 and U.S. Pat. No. 5,644,048), phosphorodithioate (Brill et al. (1989) J. Am. Chem. Soc. 111:2321), O-methylphosphoroamidite linkages (Eckstein, Oligonucleotides and Analogues: A Practical Approach, Oxford University Press (1992)), and peptide nucleic acid backbones and linkages (Egholm (1992) J. Am. Chem. Soc. 114:1895; Meier et al. (1992) Chem. Int. Ed. Engl. 31:1008; Nielsen (1993) Nature 365:566; and Carlsson et al. (1996) Nature 380:207). Other analog nucleic acids include those with positively charged backbones (Denpcy et al. (1995) Proc. Natl. Acad. Sci. USA 92:6097); non-ionic backbones (U.S. Pat. Nos. 5,386,023, 5,637,684, 5,602,240, 5,216,141 and 4,469,863; Angew (1991) Chem. Intl. Ed. English 30: 423; Letsinger et al. (1988) J. Am. Chem. Soc. 110:4470; Letsinger et al. (1994) Nucleoside & Nucleotide 13:1597; Chapters 2 and 3, ASC Symposium Series 580, “Carbohydrate Modifications in Antisense Research”, Ed. Y. S. Sanghvi and P. Dan Cook; Mesmaeker et al. (1994) Bioorganic & Medicinal Chem. Lett. 4: 395; Jeffs et al. (1994) J. Biomolecular NMR 34:17; Tetrahedron Lett. 37:743 (1996)) and non-ribose backbones, including those described in U.S. Pat. Nos. 5,235,033 and 5,034,506, and Chapters 6 and 7, ASC Symposium Series 580, Carbohydrate Modifications in Antisense Research, Ed. Y. S. Sanghvi and P. Dan Cook, which are each incorporated by reference. Several nucleic acid analogs are also described in, e.g., Rawls, C & E News Jun. 2, 1997 page 35. Modifications of the ribose-phosphate backbone may be done to facilitate the addition of additional moieties, such as labeling moieties, or to alter the stability and half-life of such molecules in physiological environments.
  • In addition to naturally occurring heterocyclic bases that are typically found in nucleic acids (e.g., adenine, guanine, thymine, cytosine, and uracil), nucleic acid analogs also include those having non-naturally occurring heterocyclic or other modified bases. For instance, certain bases used in nucleotides that act as melting temperature (Tm) modifiers are optionally included. Exemplary of these are 7-deazapurines (e.g., 7-deazaguanine, 7-deazaadenine, and the like); pyrazolo[3,4-d]pyrimidines, propynyl-dN (e.g., propynyl-dU, propynyl-dC, and the like); hypoxanthine; inosine; xanthine; 8-aza derivatives of 2-aminopurine, 2,6-diaminopurine, 2-amino-6-chloropurine, hypoxanthine, inosine and xanthine; 7-deaza-8-aza derivatives of adenine, guanine, 2-aminopurine, 2,6-diaminopurine, 2-amino-6-chloropurine, hypoxanthine, inosine and xanthine; 6-azacytosine; 5-fluorocytosine; 5-chlorocytosine; 5-iodocytosine; 5-bromocytosine; 5-methylcytosine; 5-propynylcytosine; 5-bromovinyluracil; 5-fluorouracil; 5-chlorouracil; 5-iodouracil; 5-bromouracil; 5-trifluoromethyluracil; 5-methoxymethyluracil; 5-ethynyluracil; 5-propynyluracil; non-naturally occurring bases as described by, for example, Seela et al. ((1991) Helv. Chim. Acta 74:1790; (1999) Helv. Chim. Acta 82:1640); Grein et al. ((1994) Bioorg. Med. Chem. Lett. 4:971-976), U.S. Pat. Nos. 5,484,908, 5,645,985, 5,990,303, 5,830,653, 6,639,059, 6,303,315, U.S. Pat. Appln. No. 2003/0092905, and the like.
  • “Nucleoside” typically refers to a nucleic acid component that comprises a base or basic group (e.g., comprising at least one homocyclic ring, at least one heterocyclic ring, at least one aryl group, and/or the like) covalently linked to a sugar moiety (e.g., a ribose sugar, etc.), a derivative of a sugar moiety, or a functional equivalent of a sugar moiety (e.g., an analog, such as carbocyclic ring). For example, when a nucleoside includes a sugar moiety, the base is typically linked to a 1′-position of that sugar moiety. As described above, a base can be naturally occurring (e.g., a purine base, such as adenine (A) or guanine (G), a pyrimidine base, such as thymine (T), cytosine (C), or uracil (U)), or non-naturally occurring (e.g., a 7-deazapurine base, a pyrazolo[3,4-d]pyrimidine base, a propynyl-dN base, etc.). Exemplary nucleosides include ribonucleosides, deoxyribonucleosides, dideoxyribonucleosides, carbocyclic nucleosides, and the like. A “nucleotide” refers to an ester of a nucleoside, e.g., a phosphate ester of a nucleoside. To illustrate, a nucleotide can include 1, 2, 3, or more phosphate groups covalently linked to a 5′ position of a sugar moiety of the nucleoside. Nucleoside triphosphates and 2′-deoxynucleoside triphosphates or their chemically modified versions may be used as substrates for multiple-nucleotide extension by APP where, for example, one nucleotide is incorporated the extending strand can be further extended. 2′,3′-dideoxynucleoside triphosphates, chemically modified versions thereof, or other suitable compounds (e.g., as in US 2005/0037398A1, acycloNMP) may be used as terminators for further extension may be used for single-nucleotide extension. 2′,3′-dideoxynucleoside triphosphates may be labeled with radioactivity or fluorescence dye for differentiation from the 3′ terminal dideoxynucleotide of oligonucleotide P*. Mixtures of nucleoside triphosphates or 2′-deoxynucleoside triphosphates and 2′,3′-dideoxynucleoside triphosphates may also be used.
  • A “nucleotide incorporating biocatalyst” refers to a catalyst that catalyzes the incorporation of nucleotides into a nucleic acid. Nucleotide incorporating biocatalysts are typically enzymes. An “enzyme” is a protein- and/or nucleic acid-based catalyst that acts to reduce the activation energy of a chemical reaction involving other compounds or “substrates.” A “nucleotide incorporating enzyme” refers to an enzyme that catalyzes the incorporation of nucleotides into a nucleic acid, e.g., during nucleic acid amplification or the like. Exemplary nucleotide incorporating enzymes include, e.g., polymerases, terminal transferases, reverse transcriptases (e.g., RQY polymerase described above), telomerases, polynucleotide phosphorylases, and the like. A “thermostable enzyme” refers to an enzyme that is stable to heat, is heat resistant, and retains sufficient catalytic activity when subjected to elevated temperatures for selected periods of time. For example, a thermostable polymerase retains sufficient activity to effect subsequent primer extension reactions when subjected to elevated temperatures for the time necessary to effect denaturation of double-stranded nucleic acids. Heating conditions necessary for nucleic acid denaturation are well known to persons skilled in the art and are exemplified in, for example, U.S. Pat. Nos. 4,683,202, 4,683,195, and 4,965,188. To further illustrate, a “thermostable polymerase” refers to an enzyme that is suitable for use in a temperature cycling reaction, such as a polymerase chain reaction (“PCR”). For a thermostable polymerase, enzymatic activity refers to the catalysis of the combination of the nucleotides in the proper manner to form primer extension products that are complementary to a template nucleic acid. Exemplary thermostable polymerases are described herein, and others may available to the skilled artisan may also be suitable.
  • TdT terminal deoxynucleotidyl transferase) catalyses the addition of nucleotides to the 3′ terminus of a DNA molecule. Unlike most DNA polymerases it does not require a template. The preferred substrate of this enzyme is a 3′-overhang, but it can also add nucleotides to blunt or recessed 3′ ends. Cobalt is a necessary cofactor; however the enzyme catalyzes reaction upon Mg and Mn administration in vitro.
  • CircLigase™ (Epicentre Biotechnologies), ssDNA Ligase is a thermostable ATP-dependent ligase that catalyzes intramolecular ligation (i.e. circularization) of ssDNA templates having a 5′-phosphate and a 3′-hydroxyl group. In contrast to T4 DNA Ligase and Ampligase® DNA Ligase, which ligate DNA ends that are annealed adjacent to each other on a complementary DNA sequence, CircLigase ssDNA Ligase ligates ends of ssDNA in the absence of a complementary sequence. The enzyme is therefore useful for making circular ssDNA molecules from linear ssDNA. Circular ssDNA molecules can be used as substrates for rolling-circle replication or rolling-circle transcription. Linear ssDNA of >30 bases is circularized by CircLigase enzyme. Under standard reaction conditions, virtually no linear concatamers or circular concatamers are produced. In addition to its activity on ssDNA, CircLigase enzyme also has activity in ligating a single-stranded nucleic acid having a 3′-hydroxyl ribonucleotide and a 5′-phosphorylated ribonucleotide or deoxyribonucleotide.
  • An “oligonucleotide” refers to a nucleic acid that includes at least two nucleic acid monomer units (e.g., nucleotides), typically more than three monomer units, and more typically greater than ten monomer units. The exact size of an oligonucleotide generally depends on various factors, including the ultimate function or use of the oligonucleotide. Typically, the nucleoside monomers are linked by phosphodiester bonds or analogs thereof, including phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phosphoranilidate, phosphoramidate, and the like, including associated counterions, e.g., H+, NH4 +, Na+, and the like, if such counterions are present. Typically, the 3′ end linkage of P* is a phosphodiester bond. Oligonucleotides may be prepared by any suitable method, including, but not limited to, isolation of an existing or natural sequence, DNA replication or amplification, reverse transcription, cloning and restriction digestion of appropriate sequences, or direct chemical synthesis by a method such as the phosphotriester method of Narang et al. ((1979) Meth. Enzymol. 68:90-99); the phosphodiester method of Brown et al. ((1979) Meth. Enzymol. 68:109-151); the diethylphosphoramidite method of Beaucage et al. ((1981) Tetrahedron Lett. 22:1859-1862); the triester method of Matteucci et al. ((1981) J. Am. Chem. Soc. 103:3185-3191); automated synthesis methods; or the solid support method described in U.S. Pat. No. 4,458,066, and/or other methods known to those skilled in the art.
  • A “primer nucleic acid” or “primer” is a nucleic acid that can hybridize to a target or template nucleic acid and permit chain extension or elongation using, e.g., a nucleotide incorporating biocatalyst, such as a polymerase under appropriate reaction conditions. A primer nucleic acid is typically a natural or synthetic oligonucleotide (e.g., a single-stranded oligodeoxyribonucleotide). Although other primer nucleic acid lengths are optionally utilized, they typically comprise hybridizing regions that range from about 8 to about 100 nucleotides in length. Shorter primer nucleic acids generally require lower temperatures to form sufficiently stable hybrid complexes with template nucleic acids. A primer nucleic acid that is at least partially complementary to a subsequence of a template nucleic acid is typically sufficient to hybridize with the template for extension to occur. A primer nucleic acid may be labeled, if desired, by incorporating a detectable label as described herein.
  • The term “probe nucleic acid” or “probe” refers to a labeled or unlabeled oligonucleotide capable of selectively hybridizing to a target or template nucleic acid under suitable conditions. Typically, a probe is sufficiently complementary to a specific target sequence contained in a nucleic acid sample to form a stable hybridization duplex with the target sequence under selected hybridization conditions. A hybridization assay carried out using a probe under sufficiently stringent hybridization conditions permits the selective detection of a specific target sequence. The term “hybridizing region” refers to that region of a nucleic acid that is exactly or substantially complementary to, and therefore capable of hybridizing to, the target sequence. For use in a hybridization assay for the discrimination of single nucleotide differences in sequence, the hybridizing region is typically from about 8 to about 100 nucleotides in length. Although the hybridizing region generally refers to the entire oligonucleotide, the probe may include additional nucleotide sequences that function, for example, as linker binding sites to provide a site for attaching the probe sequence to a solid support. A probe of the invention may be generally included in a nucleic acid that comprises one or more labels (e.g., donor moieties, acceptor moieties, and/or quencher moieties), such as a 5′-nuclease probe, a hybridization probe, a fluorescent resonance energy transfer (FRET) probe, a hairpin probe, or a molecular beacon, which can also be utilized to detect hybridization between the probe and target nucleic acids in a sample. In some embodiments, the hybridizing region of the probe is completely complementary to the target sequence. However, in general, complete complementarity is not necessary (e.g., nucleic acids can be partially complementary to one another); stable hybridization complexes may contain mismatched bases or unmatched bases. Modification of the stringent conditions may be necessary to permit a stable hybridization complex with one or more base pair mismatches or unmatched bases. Sambrook et al., Molecular Cloning: A Laboratory Manual, 3rd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (2001) provides guidance for suitable modification. Stability of the target/probe hybridization complex depends on a number of variables including length of the oligonucleotide, base composition and sequence of the oligonucleotide, temperature, and ionic conditions. One of skill in the art will recognize that, in general, the exact complement of a given probe is similarly useful as a probe. One of skill in the art will also recognize that, in certain embodiments, probe nucleic acids can also be used as primer nucleic acids.
  • The methods described herein may be useful for detecting and/or quantifying a variety of target nucleic acids from a test sample. A target nucleic acid is any nucleic acid for which an assay system is designed to identify or detect as present (or not), and/or quantify in a test sample. Such nucleic acids may include, for example, those of infectious agents (e.g., virus, bacteria, parasite, and the like), a disease process such as cancer, diabetes, or the like, or to measure an immune response. Exemplary “test samples” include various types of samples, such as biological samples. Exemplary biological samples include, for instance, a bodily fluid (e.g., blood, saliva, or spinal fluid), a tissue sample, a food (e.g., meat) or beverage (e.g., milk) product, or the like. Expressed nucleic acids may include, for example, genes for which expression (or lack thereof) is associated with medical conditions such as infectious disease (e.g., bacterial, viral, fungal, protozoal infections) or cancer. The methods described herein may also be used to detect contaminants (e.g., bacteria, virus, fungus, and/or protozoan) in pharmaceutical, food, or beverage products. The methods may be useful to, for example, detect minimal residual disease (e.g., rare remaining cancer cells during remission, especially mutations in the p53 gene or other tumor suppressor genes previously identified within the tumors), and/or measure mutation load (e.g., the frequency of specific somatic mutations present in normal tissues, such as blood or urine).
  • Kits for performing the methods described herein are also provided. The kit typically includes at least a pair of oligonucleotides (e.g., at least one of the pair being a P* oligonucleotide) for amplifying at least one target nucleic acid from a sample, one or more polyphosphorolyzing agents described herein, a biocatalyst (e.g., DNA polymerase) and/or corresponding one or more probes labeled with a detectable label. The kit may also include samples containing pre-defined target nucleic acids to be used in control reactions. The kit may also optionally include stock solutions, buffers, enzymes, detectable labels or reagents required for detection, tubes, membranes, and the like that may be used to complete the amplification reaction. In some embodiments, multiple primer sets are included. Other embodiments of particular systems and kits are also contemplated.
  • Magnetic polymer particles are of general utility in various medical and biochemical fields, for example as transport vehicles for the delivery of pharmaceutical products, for diagnostic purposes, for separation and for synthetic purposes. Such particles rely upon their magnetic properties in order to perform these functions: in diagnostic assay applications, for example, application of a magnetic field to a sample containing an analyte bound to magnetic polymer particles allows the isolation of the analyte without the use of centrifugation or filtration; and in therapeutic applications, for example, application of a magnetic field to the patient may serve to target drug-carrying magnetic polymer particles to a desired body site.
  • By magnetic is meant herein that the polymer particles contain superparamagnetic crystals. Thus the magnetic polymer particles are magnetically displaceable but are not permanently magnetizable. Many processes for preparing magnetic polymer particles are known, a large number of which involve preparing maghemite- or magnetite-containing polymer particles from pre-formed magnetic iron oxides, e.g. magnetite. Some of processes involved are described in U.S. Pat. No. 4,654,267 (Ugelstad).
  • The magnetic polymer particles used in the process of the invention may be any magnetic polymer particle e.g. as described in U.S. Pat. No. 4,654,267. The particles are preferably porous to allow the presence of the superparamagnetic crystals in the pores thereof. The surface of the magnetic particles is normally functionalized to allow coupling of a ligand to the polymer particle, e.g. it may be functionalized to carry any known surface structure such as carboxyl groups, tosyl groups, amino groups, epoxy groups, maleamido groups, thiol groups etc. Hence, the surface may be amine functionalized. Alternatively, an amine functionalized surface can itself be further functionalized to carry other functional groups, e.g. COOH groups.
  • The polymer particle is preferably made from combinations of vinylic polymers (e.g. styrene), acrylates and/or methacrylates. The polymeric material may optionally be cross-linked, for example by incorporation of cross-linking agents, for example as comonomers, e.g. divinylbenzene (DVB) or ethyleneglycol dimethacrylate. Appropriate quantities of the cross-linking agents (e.g. comonomers) required will be well known to the skilled person. Preferably the polymer is a cross-linked styrenic polymer (e.g. a styrene-divinylbenzene polymer, surface functionalized by the use of a nitro-group containing comonomer, e.g. nitro-styrene, and subsequent reduction) or a cross-linked (meth)acrylic polymer surface functionalized by the use of an epoxy-group containing co-monomer (e.g. glycidylmethacrylate) and subsequent amination (e.g. by reaction with ethylene diamine).
  • The superparamagnetic crystals in the polymer particles used in the process of the invention may be of any material capable of being deposited in superparamagnetic crystalline form in the porous polymer particles. Magnetic iron oxides, e.g. magnetite or maghemite are preferred; however the crystals may be of mixed metal oxides or other magnetic material if desired. The total quantity of crystalline magnetic material present is generally more than 1%, preferably more than 3%, desirably more than or equal to 5% (by weight, e.g. up to 40% wt. The percentage is calculated on a Fe (or equivalent metal in the case of magnetic materials other than iron oxides) weight basis based upon the overall dry weight of the coated particles.
  • Polymer particles according to the various aspects of the present invention may generally have sizes (i.e. diameters) that are in the micrometer range, e.g. 0.3 to 100 μm, especially 0.5 to 50 μm, more especially 0.8 to 8 μm, e.g. 0.8 to 1.2 μm. 1 μm beads are referred.
  • Typically the porous particles used will have a surface area of at least 15 m2/g (measured by the BET nitrogen absorption method), and more preferably at least 30 m2/g, e.g. up to 700 m2/g, when corrected to a mean particle diameter of 2.7 μm (i.e. multiply surface area by 2.7/MD, where MD is the mean diameter in micrometers). Similarly scaled, the particle pore volume is preferably at least 0.1 mL/g.
  • Typically, the polymer particles are spherical and substantially monodisperse before they are coated and especially preferably remain spherical and substantially monodisperse once they have been coated.
  • By substantially monodisperse, it is meant that for a majority of particles, the particles have a coefficient of variation (CV) of less than 20%, for example less than 15%, preferably less than 12%, more preferably less than 11%, still more preferably less than 10% and most preferably no more than about 8%, e.g. 2 to 5%. CV is determined in percentage as

  • CV=(100×standard deviation)/mean
  • where mean is the mean particle diameter and standard deviation is the standard deviation in particle size. CV is preferably calculated on the main mode, i.e. by fitting a monomodal distribution curve to the detected particle size distribution. Thus some particles below or above mode size may be discounted in the calculation which may for example be based on about 90% of total particle number (of detectable particles that is). Such a determination of CV may be performable on a Coulter or other particle size analyzer.
  • Functionalization of the polymeric material may take place after polymerization by, for example, nitration and subsequent reduction of the thus-formed nitro groups to pendant amine groups; or direct amination, for example by treatment with amino ethanol. As further alternatives, polymeric particles prepared by the well-known Ugelstad two-step swelling process and the improvements thereto disclosed in WO 00/61647 may be used. Porous polymer particles produced according to the processes described in this publication may have magnetic particles deposited in their pores by standard techniques.
  • As a further possibility, porous polymer particles may be prepared from nitro styrene and DVB, and magnetic material introduced as taught in U.S. Pat. No. 4,654,267.
  • The superparamagnetic polymer beads sold by Life Technologies (Invitrogen Dynal Biotech ASA), under the trade names DYNABEADS, especially DYNABEADS MYONE are especially preferred. DYNABEADS are particularly advantageous since they remain in suspension and do not exhibit magnetic particle sedimentation often associated with other magnetic beads. DYNABEADS also show excellent magnetic mobility compared to other magnetic particles in which high levels of iron are present. DYNABEADS exhibit beneficial kinetics allowing shorter reaction times and higher throughputs. Their non-specific binding is lower than other magnetic beads and their proper use results in a concentration of the desired material taking place resulting in easier and more efficient washing procedures. DYNABEADS MYONE beads are easy to automate and are monodisperse. DYNABEADS MYONE carboxylic acid beads are used generally; however, other functional groups may also be utilized.
  • Certain embodiments are further described in the following examples. These embodiments are provided as examples only and are not intended to limit the scope of the claims in any way. All references, including US patents, patent applications, PCT applications and articles cited within this application are incorporated by reference in their entirety into this application.
  • EXAMPLES Example 1
  • Covalent Bead—anti-miRNA Probe bead—Chemical Preparation.
  • Schematic is shown in FIG. 1.
  • Reagents: EDC, miRNA oligo, and MYONE beads (Beads: 1 mL=10 mg=10 billion).
  • One ml of MYONE carboxylic acid beads (10 billion beads) are placed in 1.5 ml microfuge tube. Beads are washed with 1000 μL 10 mM NaOH 3 times. Beads are further washed with 10000 μL deionized H2O, 4 times. pH is adjusted to about 6.5-7. A reaction master mix is prepared in 1.5 mL tube, as follows:
  • add H2O 80 μL add 20 μL 5M NaCl add 248 μL DMSO
  • 1M EDC is prepared in 5 mM aqueous Imidazole-HCl. One mg/5.2 μL=18 mg/100 μL 5 mM aqueous Imidazole is prepared. 192 μL (2×) of 0.5 mM aqueous anti-miRNA oligo is added to the master mix. Beads are suspended in reaction master mix and treated as follows: remove the H2O from the beads
  • add 100 μL μM EDC solution to the reaction master Mix
    add the anti-miRNA oligo/Master Mix/EDC to the beads
    immediately vortex and sonicate 3 times.
    place the tube on a rotator.
  • The tube is rotated overnight at room temperature. The beads are further washed as follows: Wash with 1000 μL DMSO. Wash 2 times with 1000 μL deionized water. Wash 2 times with 1000 μL 1× TEX (Tris, EDTA, Triton X-100). Wash 3 times with 1000 μL 1× TEX and heat for 8 min at 80° C. Beads are stored in 1.00 mL 1× TEX at 4° C.
  • Example 2
  • Covalent anti-miRNA bead—Enzymatic Preparation
  • Schematic is shown in FIG. 2.
  • 3′-Modification and 5′-Phosphorylation of anti-miRNA Probes
  • Reagents: Terminal Transferase, ddTTP (TriLink),
  • mlRNA-Probe Sequence Tm
    Let-7aP 5′AACTATACAACCTACTACCTCA 46.41
    Let-7bP 5′AACCACACAACCTACTACCTCA 54.8
  • 3′-Modification Procedure
  • Component Let-7aP Let-7bP Final Concentration
    10X Terminal Transferase 10 10 1X
    Buffer
    10X Cobalt Chloride 10 10 1X
    20 U/μL Terminal 10 10 0.4 U/μL
    Transferase
    1 mM ddTTP 10 10 100 μM
    5′P-Probe 30 30 ~20 μM
    H2O 30 30
    Total 100 100
    Input 19.3 OD × 15.5 OD ×
    Yield Output 30 μL 30 μL
  • The reaction is incubated at 37° C. for 2 hrs. The terminal transferase is inactivated by heating the reaction mix at 95° C. for 5 min.
  • Phosphorylation Procedure
  • Component Let-7aPddT Let-7bPddT
    Carryover from above 100 100
    10X Kinase Buffer 20 20
    10 mM ATP 20 20
    10 U/μL T4 Kinase 15 15
    DI Water 45 45
    Total (μL) 200 200
    Oligo Output (OD) F3 1.5 × 50 μL F3 2.7 × 50 μL
    F4 25.9 × 50 μL F4 26.9 × 50 μL

    The reaction mix is incubated at 37° C. for 1 hr. Purification is done using PD-10 column by desalting.
  • Bead Ligation with 5′P-Let-7a, b, c, d, e and f Probe-3′ddT
  • Reagents: SOLID™ P1 DNA Beads (ABI), 5′P-Let-7a, b, c, d, e and f Probes, Ligation Buffer (EpiCentre Biotechnologies), Ligase (EpiCentre Biotechnologies), ATP (Ambion), MnCl2 (Teknova), PEG6000 (Emerald Biosystems).
  • Stock Volume for Final Concentration
    Component Concentration 2X (μL) (1X)
    10X Ligation Buffer 10X 160 1X
    50% PEG6000 50% 240 7.5%
    25 mM MnCl2 25 mM 160 2.5 mM
    250 μM ATP 250 uM 160 25 μM
    H2O 80
    Total 800
  • Pre-Treatment of Beads
  • Transferred 300 μL beads (10 million/A x 50=500 million beads x 6) to a Lo-Bind tube. Placed the Lo-Bind tube in a magnetic rack for 1 min and removed the supernatant. Added 200 μL 1× TEX (Tris-EDTA-Triton X-100) buffer. Sonicated the beads using Covaris Covalent Declump 1. Placed the Lo-Bind tube in a magnetic rack for 1 min and remove the supernatant. Added 150 μL the Buffer mix and 150 μL H2O. Vortexed. Divided the beads into 6 tubes, 50 μL each. Placed the Lo-Bind tubes in a magnetic rack for 1 min and removed the supernatant.
  • Added following reaction mix into the tube: Ligation Reaction
  • Component 7a 7b 7c 7d 7e 7f
    Solid P1 Beads 500 500 500 500 500 500
    (500 million beads) million million million million million million
    2X Buffer Mix 100 100 100 100 100 100
    5′P-Let-7P -3′-ddT A260 11.8 A260 10.5 A260 4.92 A260 4.44 A260 4.59 A260 4.55 A260
    5′P-Let-7P -3′-ddT 40 40 80 80 80 80
    H2O 40 40 0 0 0 0
    100 U/μL CircLigase I 20 20 20 20 20 20
    Total 200 200 200 200 200 200

    Incubated reaction tubes at 60° C. overnight on ThermoMixer at 1100 rpm.
  • Bead Clean-up:
  • Denaturing Solution: Added 200 μL Denaturant to 1800 μL, 1× Denaturing buffer and mixed well. Placed the reaction tube in a magnetic rack for 1 min and remove the supernatant. Added 200 μL 1× TEX buffer. Sonicated the beads using Covaris Covalent Declump 1. Placed the tube in a magnetic rack for 1 min and removed the supernatant. Added 200 μL of the Denaturing Buffer, vortexed, and let stand for 1 min. Placed the tube in a magnetic rack for 1 min and removed the supernatant. Added 200 μL 1× TEX buffer, vortexed and pulsed down. Placed the tube in a magnetic rack for 1 min and removed the supernatant. Repeat twice. Beads are stored (in 200 μL TEX buffer, ˜2.5 million beads/μL) at 4° C.
  • Example 3
  • Hybridization of ant-miRNA Beads (Hybridization with Cy3-labeled miRNA)
  • Fluorescent flow cytometry analysis is done to ensure that anti-miRNA probes are covalently attached to the beads and that Cy3-labeled miRNAs are effectively captured by the anti-miRNA beads.
  • Reagents: Anti-ath-miR159 Beads, Cy-3-ath-miR159a, Control: non-anti-probe beads, Cy3-ath-miR159a.
  • Hybridization is carried out using the following protocol:
  • Sonicate the beads with Covalent Declump 1 on Covaris. Aliquot 1 μL of ath-miR159a beads and 1 μL Solid P1beads into 1.5 mL LoBind tubes separately. Add 100 μL hybridization buffer. Add 5 μL 100 μM Cy3-ath-miR159a and vortex. Shake tubes at 1200 rpm for 20 min at room temperature. Magnet the beads for 1 mins and remove the supernatant. Add 200 μL RNA Purification Wash Solution 1 (ABI). Vortex and incubate for 1 min before placing tubes on magnetic rack. Magnet and after 1 min, remove the supernatant. Add 200 μL RNA Purification Wash Solution 2 (ABI). Vortex and incubate for 1 min before placing tubes on magnetic rack. Add 200 μL 0.5× SSPE/T buffer (ABI). Vortex and incubate for 1 min before placing tubes on magnetic rack. Briefly spin the tubes, magnet, and after 1 min, remove the supernatant. Add 100 μL 0.5× SSPE/T Buffer, vortex, and sonicate with an ultrasonic bath briefly. ABI is Applied BioSystems, Inc. miRNAs may be eluted from the beads by de-hybridization using buffers that are known to one skilled in the art, e.g., 10 mM Tris:HCl or 10 mM Tris:HCl with 01 mM EDTA.
  • Fluorescent Flow Cytometry (FACS) (BD Bioscience) Analysis
  • Add about 250 μL FACS read buffer. Add 10 μL beads. Read. The beads are fluorescent and the center of the distribution peak of the beads is distinctively different from the control beads. Representative FACS profiles are shown in FIGS. 3A-3C.
  • Example 4
  • Let-7 miRNA Profiling from Red Blood Cells and Plasma
  • Reagents: P1-Let-7a-f Beads, Tempus Buffer (ABI), Finger Tip Fresh Blood
  • Blood Sample Collection: 40 μL of fresh finger blood sample was collected in a tube containing 0.4 μL of 0.5M EDTA (pH 8.0) using a mini finger lancet.
  • Plasma and Red Blood Cells Separation and Lysis: The blood sample is centrifuged and the plasma is carefully pipetted out. 20A plasma is mixed with 40 μL Tempus buffer. The red blood cells are washed with 100 μL 0.9% NaCl, centrifuged, and the supernatant is removed. Washing is repeated 2 more times. 10 μL red blood cells are mixed with 20 μL Tempus Buffer.
  • Hyb Sample Serial Dilution: 6 μL of the lysate solution prepared above is diluted in 194 μL Oligo dT hybridization buffer and vortexed. This is Hyb sample A (1 μL blood in 1004 volume). The samples are serially diluted according to following table for the rest of Hyb samples.
  • Oligo dT Lys/ Sample and Total Volume,
    Hyb Sample Bind Buffer Volume μL Blood, nL
    A 1000
    B 135 A: 15 μL 150 100
    C 135 B: 15 μL 150 10
    D 135 C: 15 μL 150 1
    E 135 D: 15 μL 150 0.1
  • Hybridization: 5 μL each of Let-7a thru Let-7f beads (˜2.5 million/μL, 12.5 million total each) are combined in a LoBind tube, 200 μL TEX buffer is added, and the tubes are vortexed. Tubes/beads are magnetized to remove the supernatant. 6 μL of the mixed beads are added into each hybridization sample as prepared above. The beads are shaken at 1100 rpm for 20 min at room temperature. The beads are magnetized to remove the supernatants. The beads are washed according to the following table. Finally, 1× TE buffer is added and the beads are sonicated with Covalent Declump 1.
  • Wash 1, Wash 2,
    Test Beads 100 μL 100 μL Suspension, 100 μL 1X TE
    A Let-7a-f Buffer A Buffer B 1X TE (0.125 million/μL)
    B Let-7a-f Buffer A Buffer B 1X TE (0.125 million/μL)
    C Let-7a-f Buffer A Buffer B 1X TE (0.125 million/μL)
    D Let-7a-f Buffer A Buffer B 1X TE (0.125 million/μL)
    E Let-7a-f Buffer A Buffer B 1X TE (0.125 million/μL)
  • Reverse Transcription (RT): The captured miRNAs (in suspension, above Table) are reverse transcribed to cDNA using Taq Man® MicroRNA Reverse Transcription Kit (ABI) following the recommended protocol. The reactions are incubated at 16° C./30 min, 42° C./30 min, and 85° C./5 min.
  • TaqMan PCR Reaction: The reverse-transcribed cDNA from the captured miRNAs are quantitatively analyzed using Applied Biosystems miRNA TaqMan assays and TaqMan Universal PCR Master Mix following the recommended protocols. TaqMan real-time PCR reactions are run on Applied Biosystems 7500 instrument using the default cycling parameters. The results are listed in Table 1 and FIG. 4.
  • TABLE 1
    Blood Volume Let-7a Let-7b Let-7c Let-7d Let-7e Let-7f
    NTC 40.00 40.00 40.00 40.00 40.00 39.00
      1 nL 33.21 32.89 34.48 36.17 39.03 34.34
      10 nL 31.01 30.99 36.54 32.01 36.08 34.33
     100 nL 27.74 27.68 34.12 28.24 37.28 30.27
     1000 nL 24.25 24.52 31.46 25.19 35.32 27.92
    10000 nL 21.78 21.03 27.50 22.46 32.57 24.19
    Linear Data Points 4 4 3 5 2 4
    Slope 3.12 3.30 3.31 3.42 3.28
    R2 0.99 0.99 0.99 0.99 0.99
    (Ct values for each let-7 species are shown)
  • Example 5 miRNA Spiking and Recovery Test from a Plasma Sample
  • A mixture of Let-7a-f miRNAs was spiked in plasma. The spiked plasma sample was serially diluted 10× with Hyb buffer. The miRNAs were captured and quantified by the beads and TaqMan PCR. Results are shown in FIG. 5.
  • Example 6
  • Capturing and quantification of a panel of 96 miRNAs using anti miRNA beads
  • The following protocol was used. Dilute 5 μL fresh blood in 490 μL Hybridization Dilution buffer (ABI). Spike in 5 μL of 1 nM ath-miR159a RNA Artificial Template. Add anti miRNA beads prepared according to Example 2 with 96 anti miRNA oligonucleotides and shake at 1200 rpm for 20 min at room temperature to hybridize. Wash the beads with RNA Purification Wash Solution I and RNA Purification Wash Solution II as described previously. Add elution buffer and vortex to mix. Heat bead elution sample in 70° C. for 2 minutes and then place immediately on ice. Perform reverse transcription and TaqMan real-time PCR as described in Example 4. The quantitative real-time PCR Ct profiles are shown in FIG. 6A. A similar experiment is conducted on 50 microL of human plasma. The results are presented in FIG. 6B. miRNA detectability and reproducibility was further confirmed as shown in FIG. 9 and the table below:
  • Run 1 Run 2
    Total miRNA Tested 96 96
    Detectable miRNA (Ct < 35) 71 70
    Undetectable miRNA 25 26
    Detection Rate 74.0% 72.9%
  • Example 7
  • miRNA capturing and quantification from saliva. The following protocol was used.
  • Collect about 200 μL saliva and centrifuge briefly. Take 100 μL of the centrifuged saliva and add to 196 μL Hybridization buffer. Vortex vigorously for 30 seconds. Spike in 4 μL of 1 nM ath-miR159a RNA Artificial Template. This is Sample E. Dilute Sample E serially according to following table using Hybridization buffer.
  • Hyb Hyb Dilution Stock Sample Total Volume Saliva Input
    Sample buffer μL and Volume μL nL/100 μL
    E Prepared in step 1 33300
    D 135 E: 15 μL 150 3330
    C 135 D: 15 μL 150 333
    B 135 C: 15 μL 150 33.3
    A 135 B: 15 μL 150 3.33
  • Add anti miRNA beads to Hyb Samples A, B, C, D, and E prepared above. Shake the hybridization samples in tubes at 1200 rpm for 20 min at room temperature. Wash the beads with RNA Purification Wash Solution I and RNA Purification Wash Solution II as described previously. Add elution buffer and vortex to mix. Heat bead elution sample in 70° C. for 2 minutes and then place immediately on ice. Perform reverse transcription and TaqMan real-time PCR as described in Example 4. The quantitative real-time PCR Ct profiles are shown in table below and in FIG. 7.
  • Let- Let- Let- Let- Let- Let- ath-
    7a 7b 7c 7d 7e 7f 159a miR320
    NTC 40.00 36.58 37.30 40.00 40.00 40.00 37.13 40.00
      3 nL 34.65 33.11 35.49 38.60 40.00 37.15 29.54 34.09
      30 nL 31.84 32.13 34.83 38.60 40.00 35.43 27.25 34.24
     300 nL 29.44 29.77 31.80 34.09 40.00 34.13 23.75 34.04
     3000 nL 26.97 26.93 29.93 30.52 36.23 30.08 20.33 31.45
    30000 nL 24.03, 24.07 26.96 27.67 33.80 27.50 17.04 28.66
  • Example 8 Protocols for miRNA Detection and Profiling Using Anti-Probe Beads
  • Two anti—miRNA bead Panels: A384C6: beads with 386 miRNA targets based on the ABI Megaplex A assay panel. B384C6: beads with 385 miRNA targets based on the ABI Megaplex B assay panel.
  • None-Human miRNA Controls: Each panel contains 6 non-human miRNAs: ath-miR159a, cel-miR-2, cel-miR-39, cel-miR-54, cel-miR-238, cel-lin-4. They can be used as external controls at user's choice.
  • Reagents in the kit: Lysis Buffer, ABC Buffer, Wash Buffer 1, Wash Buffer 2, Elution Buffer, LoBind Tubes (Eppendorf), Human Panel A Beads (1 million/microL). Or Human Panel Panel B Beads.
  • Reagent Preparation: Add 6 mL of Lysis Buffer to the ABC Buffer bottle to make the final ABC Buffer for use. Add 7 mL of 100% Ethanol to the Wash Buffer 2 bottle to make the final Wash Buffer 2 for use.
  • Reagents required: TaqMan® MicroRNA Reverse Transcription Kit (ABI PN: 4366597), TaqMan® MicroRNA Assays (ABI PN: 4427975), TaqMan® Universal Master Mix II (ABI PN: 4440040)
  • miRNA Purification
  • Beads Preparation: Vortex the beads thoroughly to suspend the beads in solution. Sonicate the beads by an ultrasonic water bath for 1 minute. Vortex and then aliquot 80 uL beads (80 million beads total) into a LoBind 1.5-mL microfuge tube. Magnet the beads for 1 minute and remove the supernatant.
  • Sample Preparation: Select (check mark) the sample type you are going to test. Based on your selection, prepare the sample for hybridization according to following procedures:
  • Sample Sample
    Type Amount Preparation
    Whole 10 uL Mix 10 uL whole blood with 20 uL Lysis Buffer and vortex for ~30
    Blood seconds. Add 120 uL ABC Buffer and vortex for ~30 seconds. Total
    volume ~150 uL
    Blood 30 uL Dilute 30 uL blood lysate with 120 uL ABC Buffer and vortex. Total
    Lysate volume ~150 uL
    Plasma 50 uL Mix 50 uL plasma with 100 uL ABC Buffer and vortex for ~30 seconds.
    Total volume ~150 uL
    Serum 50 uL Mix 50 uL serum with 100 uL ABC Buffer and vortex for ~30 seconds.
    Total volume ~150 uL
    Cell 5 × 10{circumflex over ( )}4 Mix the 50 uL cell/PBS suspension with 150 uL Lysis Buffer and vortex
    Cultures cells in for ~30 seconds. Total volume ~200 uL
    50 uL PBS
    Solid 1-10 mg If needed, grind or homogenize the tissue. Add 30 uL Lysis Buffer and
    Tissue vortex for ~30 seconds. Add 120 uL ABC Buffer and vortex for ~30
    seconds. Total volume ~150 uL
    FFPE 1 Wash the FFPE sample with xylene (2 min x 2), 100%, 95% and 75%
    dissection ethanol (1 min each), H2O (1 min). Add 30 uL Lysis Buffer and vortex
    (5-10 um) for ~30 seconds. Add 120 uL ABC Buffer and vortex for ~30 seconds. Total
    volume ~150 uL
    Raw Milk 100 uL Mix 100 uL raw milk with 200 uL Lysis Buffer and vortex for ~30 seconds.
    Total volume ~300 uL
    Saliva 50 uL Centrifuge 15 min at 2000 rpm to remove debris. Mix 50 uL saliva with
    100 uL Lysis Buffer and vortex for ~30 seconds. Total volume ~150 uL
    Urine 50 uL Centrifuge 15 min at 2000 rpm to remove debris. Mix 50 uL urine with
    100 uL Lysis Buffer and vortex for ~30 seconds. Total volume ~150 uL
  • (Optional) Spike in 2 uL of 1 nM external control miRNA (following six can be captured by the beads: ath-miR-159a, cel-miR-2, cel-miR-39, cel-miR-54, cel-miR-238, cel-Lin4) and vortex.
  • Hybridization: Transfer all the prepared sample into the bead tube prepared above. Using a ThermoMixer, shake the vial at 1200 rpm at 30° C. for 40 minutes.
  • Wash: Magnet the beads for 1 minute and remove the supernatant. Add 100 uL Wash Buffer 1. Vortex to resuspend the beads into solution then pulse briefly. Magnet the beads for 1 minute and remove the supernatant. Add 100 uL Wash Buffer 2. Vortex briefly to resuspend the beads into solution then pulse briefly. Magnet the beads for 1 minute and remove the supernatant. Repeat step c. Pulse the tube down to collect any residual liquid. Place the tube on magnetic rack for 20 sec, and remove any residual liquid using a fine pipette tip.
  • Elution: Add 100 uL Elution Buffer and vortex to mix then pulse briefly. Using a ThermoMixer, shake the tube at 1200 rpm at 70° C. for 3 minutes then place it immediately on the magnetic stand for 1 minute. Carefully transfer the supernatant (miRNA sample) into a clean LoBind tube. (Optional—this dilution step is only for a full 96-well plate run. The concentrated sample can be directly used). Add 500 uL H2O to dilute the sample (total volume ˜600 uL). Place the sample vial on ice. Store the miRNA sample at −80° C. if not used immediately.
  • Reverse Transcription Reaction
  • Following operations are for a panel of 96 miRNA assays in a 96-well plate. Aliquot each 5×RT primer into a 96-well PCR plate, 3 uL per assay per well. Prepare miRNA RT mix:
  • Volume: 1 Volume: 100
    Component Stock Reaction Reactions
    Nuclease-free dH2O 4.16 416
    25 mM each (100 mM total) 25 mM 0.15 15
    dNTPs
    50X MultiScribe Reverse 50X 1.0 100
    Transcriptase
    10X RT Buffer 10X 1.5 150
    20 U/UL AB RNase Inhibitor 20 U/uL 0.19 19
    Total 7.0 700
  • Pipette-7 uL miRNA RT mix to each well of the RT plate prepared above. Add 5 uL miRNA sample to each well (final total RT volume 15 uL/well). Cover the plate with Heavy-Duty sealing film, mix, and centrifuge. Incubate at 16° C./30 min, 42° C./30 min, 85° C./5 min, 4° C. hold. Store at −20° C.
  • TAQMAN PCR Reaction
  • Following operations are for a panel of 96 miRNA assays in a 96-well plate. Aliquot 20× TaqMan miRNA assays into a 96-well PCR plate, 1 uL per assay per well. Each miRNA assay location should match the corresponding RT plate location. Prepare TaqMan PCR mix.
  • Volume (uL): 1 Volume (uL):
    Component Stock reaction 100 reaction
    Nuclease-free dH2O 7 700
    2X Universal Master Mix II 2X 10 1000
    Total 17 1700
  • Pipette the PCR mix to each well, 17 uL/well. Transfer 2 uL RT reaction solution obtained from the RT step into corresponding well location of the TaqMan plate. Total reaction volume 20 uL. Run PCR: 95° C./10 min, 95° C./15 sec & 60° C./60 sec for 40 cycles, Set FAM as reporter dye with ROX as Reference.
  • Representative results from various tissues, body fluids, cancer cell lines, breast cancer formalin-fixed paraffin-embedded (FFPE) samples, among others are shown in figures XX-XX and tables below. The miRNAs are isolated from various tissues using Laser Capture Microdissection (LCM) technique.
  • miRNA profiling from human bladder LCM samples
  • LCM Diameter Area, um2 Log(Area) hsa-Let-7a hsa-Let-7c hsa-miR-16 hsa-miR-320 hsa-miR-92a
    NTC 0 40.00 40.00 38.27 37.39 40.00
     100 um 7850 3.8949 35.34 37.56 34.20 35.61 40.00
     200 um 31400 4.4969 33.19 35.52 31.68 35.50 40.00
    1000 um 785000 5.8949 27.16 31.17 28.03 33.28 40.00
  • Example 9 microRNA Profiling from Urine
  • The profiling of microRNA in human urine samples during pregnancy at various stages of pregnancy showed that of the 38 μL miRNAs tested, 200 were detected in Urine samples using a Ct cutoff of 35. 18 pregnancy related miRNAs consistently showed increased expression with pregnancy progression to second trimester.
  • Expression of miR143 and miR145 in urine for patients with balloon injured artery and normal individuals was compared. miR143 and miR145 are significantly down regulated in balloon injured arteries in comparison to normal artery. The results are shown in the table below. A representative miRNA profiling is shown in FIG. 17.
  • Assay ID Workflow Sample ID# Avg. CT StDev CT
    miR-143 Indy Normal 35.72 1.06
    miR-143 Indy Stent Artery 48.66 1.9
    miR-143 TiLDA Normal 33.08 0.06
    miR-143 TiLDA Stent Artery 39.61 0.55
    miR-145 Indy Normal 36.5 1.63
    miR-145 Indy Stent Artery 48.09 1.69
    miR-145 TiLDA Normal 30.73 0.32
    miR-145 TiLDA Stent Artery 36.84 0.53
  • All references cited within this disclosure are hereby incorporated by reference in their entirety. While certain embodiments have been described in terms of the preferred embodiments, it is understood that variations and modifications will occur to those skilled in the art. Therefore, it is intended that the appended claims cover all such equivalent variations that come within the scope of the following claims.

Claims (20)

1. A method for isolating, identifying, or quantifying an miRNA from a sample of interest, comprising:
a) contacting the sample of interest with an anti-miRNA probe covalently attached to a bead;
b) incubating the sample of interest—bead mixture under suitable conditions to form hybridized complexes between the miRNA in the sample of interest and the anti-miRNA probes on the beads;
c) washing the beads under suitable conditions to remove the unbound sample material; and
d) isolating, identifying, or quantifying the miRNA bound to the anti-miRNA probes.
2. The method according to claim 1, wherein the miRNA is isolated from the anti-miRNA probe beads before identifying or quantifying the miRNA.
3. The method according to claim 1, wherein the identifying or quantifying the miRNA comprises using reverse transcription followed by real-time (RT) or quantitative polymerase chain reaction (q-PCR).
4. The method according to claim 1, wherein the beads are magnetic beads.
5. The method according to claim 4, wherein the beads are carboxylic acid beads.
6. The method according to claim 4, wherein the beads are DNA beads.
7. The method according to claim 6, wherein the beads are P1 DNA beads.
8. The method according to claim 1, wherein the anti-miRNA probe is attached to the beads using chemical synthesis.
9. The method according to claim 1, wherein the anti-miRNA probe is attached to the beads using enzymatic synthesis.
10. The method according to claim 9, wherein the enzymatic synthesis comprises:
a) taking an oligonucleotide that has a sequence complementary to an miRNA of interest (anti-miRNA probe);
b) adding a terminal didexoynucleotide to the 3′ end using a terminal transferase;
c) adding a 5′-phosphate group to the oligonucleotide using a kinase; and
d) ligating the oligonucleotide to DNA beads using a single-strand DNA ligase.
11. The method according to claim 1, wherein only one species of anti-miRNA probe is attached to an individual bead.
12. The method according to claim 1, wherein more than one species of anti-miRNA probes are attached to an individual bead.
13. The method according to claim 1, wherein the sample of interest is a biological sample.
14. The method according to claim 13, wherein the biological sample is selected from the group consisting of blood, serum, plasma, urine, saliva, cerebrospinal fluid, wound exudates, biopsies, autopsies, tissues, formalin-fixed, paraffin-embedded (PPFE) samples, and organs.
15. A method according to claim 14, wherein the biological sample is a biological fluid.
16. A method according to claim 15, wherein the biological fluid is blood.
17. A method according to claim 15, wherein the biological fluid is serum.
18. A method according to claim 15, wherein the biological fluid is saliva.
19. A method of diagnosing a disease or disease progression, comprising:
a) identifying a set of miRNA markers that are differentially regulated during a causation or progression of a disease;
b) isolating and measuring the levels of such miRNA markers from a suitable biological sample from a patient in need thereof, at different time points if necessary; and
c) diagnosing the disease or its progression over time in the patient.
20. A kit for isolating, identifying, or quantifying an miRNA from a sample of interest, comprising:
a) beads comprising anti-miRNA probe molecules covalently attached thereto;
b) a wash buffer; and
c) an elution buffer.
US13/350,277 2011-01-14 2012-01-13 Methods for isolation, identification, and quantification of mirnas Abandoned US20120208189A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US13/350,277 US20120208189A1 (en) 2011-01-14 2012-01-13 Methods for isolation, identification, and quantification of mirnas
US15/682,103 US11046727B2 (en) 2011-01-14 2017-08-21 Methods for isolation, identification, and quantification of miRNAs
US17/304,883 US20210323994A1 (en) 2011-01-14 2021-06-28 Methods for Isolation, Identification, and Quantification of miRNAs

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161432874P 2011-01-14 2011-01-14
US13/350,277 US20120208189A1 (en) 2011-01-14 2012-01-13 Methods for isolation, identification, and quantification of mirnas

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/682,103 Division US11046727B2 (en) 2011-01-14 2017-08-21 Methods for isolation, identification, and quantification of miRNAs

Publications (1)

Publication Number Publication Date
US20120208189A1 true US20120208189A1 (en) 2012-08-16

Family

ID=46637177

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/350,277 Abandoned US20120208189A1 (en) 2011-01-14 2012-01-13 Methods for isolation, identification, and quantification of mirnas
US15/682,103 Active US11046727B2 (en) 2011-01-14 2017-08-21 Methods for isolation, identification, and quantification of miRNAs
US17/304,883 Pending US20210323994A1 (en) 2011-01-14 2021-06-28 Methods for Isolation, Identification, and Quantification of miRNAs

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/682,103 Active US11046727B2 (en) 2011-01-14 2017-08-21 Methods for isolation, identification, and quantification of miRNAs
US17/304,883 Pending US20210323994A1 (en) 2011-01-14 2021-06-28 Methods for Isolation, Identification, and Quantification of miRNAs

Country Status (3)

Country Link
US (3) US20120208189A1 (en)
EP (3) EP3733870A3 (en)
WO (1) WO2012125220A2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014113614A1 (en) * 2013-01-16 2014-07-24 The Regents Of The University Of California Label free molecular detection methods, systems and devices
US9994839B2 (en) 2013-01-16 2018-06-12 The Regents Of The University Of California Microfluidic devices to extract, concentrate and isolate molecules
US10144951B2 (en) * 2014-05-18 2018-12-04 Glc Biotechnology, Inc. Method and materials to deplete impurities for extraction and purification of nucleic acids from stool
US10160965B2 (en) * 2014-05-18 2018-12-25 Glc Biotechnology, Inc. Method and materials for nucleic acids extraction and purification
US11014957B2 (en) 2015-12-21 2021-05-25 Realseq Biosciences, Inc. Methods of library construction for polynucleotide sequencing
US11072819B2 (en) 2011-12-22 2021-07-27 Realseq Biosciences, Inc. Methods of constructing small RNA libraries and their use for expression profiling of target RNAs
US11674133B2 (en) * 2017-09-13 2023-06-13 Becton, Dickinson And Company Methods and compositions for extracting nucleic acids using ferric oxide particles

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3733870A3 (en) * 2011-01-14 2021-01-27 Life Technologies Corporation Methods for identification and quantification of mirnas

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5750341A (en) * 1995-04-17 1998-05-12 Lynx Therapeutics, Inc. DNA sequencing by parallel oligonucleotide extensions
US20070065844A1 (en) * 2005-06-08 2007-03-22 Massachusetts Institute Of Technology Solution-based methods for RNA expression profiling
US20080076674A1 (en) * 2006-07-06 2008-03-27 Thomas Litman Novel oligonucleotide compositions and probe sequences useful for detection and analysis of non coding RNAs associated with cancer
US20080274904A1 (en) * 2006-08-10 2008-11-06 Niall Anthony Gormley Method of target enrichment

Family Cites Families (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4257774A (en) 1979-07-16 1981-03-24 Meloy Laboratories, Inc. Intercalation inhibition assay for compounds that interact with DNA or RNA
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4469863A (en) 1980-11-12 1984-09-04 Ts O Paul O P Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
CA1190838A (en) 1981-07-17 1985-07-23 Cavit Akin Homogeneous nucleic acid hybridization diagnostics by non-radiative energy transfer
NO155316C (en) 1982-04-23 1987-03-11 Sintef PROCEDURE FOR MAKING MAGNETIC POLYMER PARTICLES.
US5235033A (en) 1985-03-15 1993-08-10 Anti-Gene Development Group Alpha-morpholino ribonucleoside derivatives and polymers thereof
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4965188A (en) 1986-08-22 1990-10-23 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences using a thermostable enzyme
DE3529478A1 (en) 1985-08-16 1987-02-19 Boehringer Mannheim Gmbh 7-DESAZA-2'DESOXYGUANOSINE NUCLEOTIDES, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE FOR NUCLEIC ACID SEQUENCING
US5079352A (en) 1986-08-22 1992-01-07 Cetus Corporation Purified thermostable enzyme
US5618711A (en) 1986-08-22 1997-04-08 Hoffmann-La Roche Inc. Recombinant expression vectors and purification methods for Thermus thermophilus DNA polymerase
US4889818A (en) 1986-08-22 1989-12-26 Cetus Corporation Purified thermostable enzyme
US6127155A (en) 1986-08-22 2000-10-03 Roche Molecular Systems, Inc. Stabilized thermostable nucleic acid polymerase compositions containing non-ionic polymeric detergents
US5216141A (en) 1988-06-06 1993-06-01 Benner Steven A Oligonucleotide analogs containing sulfur linkages
US5512439A (en) * 1988-11-21 1996-04-30 Dynal As Oligonucleotide-linked magnetic particles and uses thereof
US5035996A (en) 1989-06-01 1991-07-30 Life Technologies, Inc. Process for controlling contamination of nucleic acid amplification reactions
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5386023A (en) 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
US5210015A (en) 1990-08-06 1993-05-11 Hoffman-La Roche Inc. Homogeneous assay system using the nuclease activity of a nucleic acid polymerase
US5994056A (en) 1991-05-02 1999-11-30 Roche Molecular Systems, Inc. Homogeneous methods for nucleic acid amplification and detection
AU3222793A (en) 1991-11-26 1993-06-28 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
US5484908A (en) 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
TW393513B (en) 1991-11-26 2000-06-11 Isis Pharmaceuticals Inc Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
US5644048A (en) 1992-01-10 1997-07-01 Isis Pharmaceuticals, Inc. Process for preparing phosphorothioate oligonucleotides
EP0688366B1 (en) 1993-01-15 2002-05-22 The Public Health Research Institute Of The City Of New York, Inc. Sensitive nucleic acid sandwich hybridization assays and kits
US5658751A (en) 1993-04-13 1997-08-19 Molecular Probes, Inc. Substituted unsymmetrical cyanine dyes with selected permeability
US5436134A (en) 1993-04-13 1995-07-25 Molecular Probes, Inc. Cyclic-substituted unsymmetrical cyanine dyes
US6821727B1 (en) 1993-11-15 2004-11-23 Applera Corporation Hybridization assay using self-quenching fluorescence probe
US5538848A (en) 1994-11-16 1996-07-23 Applied Biosystems Division, Perkin-Elmer Corp. Method for detecting nucleic acid amplification using self-quenching fluorescence probe
US5637684A (en) 1994-02-23 1997-06-10 Isis Pharmaceuticals, Inc. Phosphoramidate and phosphorothioamidate oligomeric compounds
CA2185239C (en) 1994-03-16 2002-12-17 Nanibhushan Dattagupta Isothermal strand displacement nucleic acid amplification
US5801155A (en) 1995-04-03 1998-09-01 Epoch Pharmaceuticals, Inc. Covalently linked oligonucleotide minor grove binder conjugates
US5773258A (en) 1995-08-25 1998-06-30 Roche Molecular Systems, Inc. Nucleic acid amplification using a reversibly inactivated thermostable enzyme
US5854033A (en) 1995-11-21 1998-12-29 Yale University Rolling circle replication reporter systems
WO2000056920A1 (en) 1999-03-18 2000-09-28 Exiqon A/S One step sample preparation and detection of nucleic acids in complex biological samples
US6639059B1 (en) 1999-03-24 2003-10-28 Exiqon A/S Synthesis of [2.2.1]bicyclo nucleosides
US6734291B2 (en) 1999-03-24 2004-05-11 Exiqon A/S Synthesis of [2.2.1]bicyclo nucleosides
CA2369293C (en) 1999-04-09 2010-06-08 Dynal Particles As Process for the preparation of monodisperse polymer particles
US6569927B1 (en) 2000-10-06 2003-05-27 Uniroyal Chemical Company, Inc. Thermoplastic resins stabilized by blends of sterically hindered phenols, secondary amines, and lactones
US7572581B2 (en) 2003-06-30 2009-08-11 Roche Molecular Systems, Inc. 2′-terminator nucleotide-related methods and systems
US20050059024A1 (en) * 2003-07-25 2005-03-17 Ambion, Inc. Methods and compositions for isolating small RNA molecules
US20050037362A1 (en) * 2003-08-11 2005-02-17 Eppendorf Array Technologies, S.A. Detection and quantification of siRNA on microarrays
US7387887B2 (en) 2004-04-20 2008-06-17 University Of Utah Research Foundation Nucleic acid melting analysis with saturation dyes
US20060057595A1 (en) * 2004-09-16 2006-03-16 Applera Corporation Compositions, methods, and kits for identifying and quantitating small RNA molecules
EP1841879A4 (en) 2005-01-25 2009-05-27 Population Genetics Technologi Isothermal dna amplification
WO2006087574A2 (en) 2005-02-19 2006-08-24 Geneform Technologies Limited Isothermal nucleic acid amplification
US20070031865A1 (en) * 2005-07-07 2007-02-08 David Willoughby Novel Process for Construction of a DNA Library
JP2009504168A (en) * 2005-08-17 2009-02-05 メディクシス エス.エー. Composition and method for confirming CK19 expression
US20100209933A1 (en) * 2007-10-29 2010-08-19 Mcreynolds Larry A Methods and Compositions for Detection and Enrichment of Target Small RNAs
US20090197254A1 (en) 2007-12-14 2009-08-06 Ming-Chou Lee Variant scorpion primers for nucleic acid amplification and detection
EP3733870A3 (en) * 2011-01-14 2021-01-27 Life Technologies Corporation Methods for identification and quantification of mirnas

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5750341A (en) * 1995-04-17 1998-05-12 Lynx Therapeutics, Inc. DNA sequencing by parallel oligonucleotide extensions
US20070065844A1 (en) * 2005-06-08 2007-03-22 Massachusetts Institute Of Technology Solution-based methods for RNA expression profiling
US20080076674A1 (en) * 2006-07-06 2008-03-27 Thomas Litman Novel oligonucleotide compositions and probe sequences useful for detection and analysis of non coding RNAs associated with cancer
US20080274904A1 (en) * 2006-08-10 2008-11-06 Niall Anthony Gormley Method of target enrichment

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
Flagella et al. (2006) A multiplex branched DNA assay for parallel quantitative gene expression profiling. Analytical Biochemistry, 352:50-60 *
Gilad et al. (2008) Serum MicroRNAs Are Promising Novel Biomarkers. PLoS ONE, 3(9):e3148, pages 1-7). *
Kroh et al. (2010) Analysis of circulating microRNA biomarkers in plasma and serum using quantitative reverse transcription-PCR (qRT-PCR). Methods, 298-301 *
Kuhn et al. (2005) Template-independent ligation of single-stranded DNA by T4 DNA ligase. FEBS Journal, 272:5991-6000 *
Sambrook et al. (2001) Molecular Cloning: A Laboratory manual,. 3rd ed. Cold Spring Harbor Laboratory Press, New York, pages 9.55-9.56. *
Siva et al. (2009) Molecular assays for the detection of microRNAs in prostate cancer. Molecular Cancer, 8(17):pages 1-12 *
Thompson et al. (1989) Enzymatic Amplification of RNA Purified from Crude Cell Lysate by Reversible Target Capture. Clinical Chemistry, 35(9):1878-1881 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11072819B2 (en) 2011-12-22 2021-07-27 Realseq Biosciences, Inc. Methods of constructing small RNA libraries and their use for expression profiling of target RNAs
WO2014113614A1 (en) * 2013-01-16 2014-07-24 The Regents Of The University Of California Label free molecular detection methods, systems and devices
US9862987B2 (en) 2013-01-16 2018-01-09 The Regents Of The University Of California Label free molecular detection methods, systems and devices
US9994839B2 (en) 2013-01-16 2018-06-12 The Regents Of The University Of California Microfluidic devices to extract, concentrate and isolate molecules
US10144951B2 (en) * 2014-05-18 2018-12-04 Glc Biotechnology, Inc. Method and materials to deplete impurities for extraction and purification of nucleic acids from stool
US10160965B2 (en) * 2014-05-18 2018-12-25 Glc Biotechnology, Inc. Method and materials for nucleic acids extraction and purification
US11014957B2 (en) 2015-12-21 2021-05-25 Realseq Biosciences, Inc. Methods of library construction for polynucleotide sequencing
US11674133B2 (en) * 2017-09-13 2023-06-13 Becton, Dickinson And Company Methods and compositions for extracting nucleic acids using ferric oxide particles

Also Published As

Publication number Publication date
EP2663639A4 (en) 2015-06-17
EP3733870A3 (en) 2021-01-27
US20210323994A1 (en) 2021-10-21
US20180044371A1 (en) 2018-02-15
WO2012125220A3 (en) 2012-11-01
EP3272885A3 (en) 2018-04-18
EP3272885B1 (en) 2020-04-22
WO2012125220A2 (en) 2012-09-20
EP2663639B1 (en) 2017-09-13
EP3272885A2 (en) 2018-01-24
EP3733870A2 (en) 2020-11-04
EP2663639A2 (en) 2013-11-20
US11046727B2 (en) 2021-06-29

Similar Documents

Publication Publication Date Title
US20210323994A1 (en) Methods for Isolation, Identification, and Quantification of miRNAs
US11274340B2 (en) Methods, compositions and kits for small RNA capture, detection and quantification
US20220119875A1 (en) Small RNA Capture, Detection and Quantification
WO2019068880A1 (en) Rna templated ligation
EP2900836B1 (en) Two-primer pcr for microrna multiplex assay
US20170253921A1 (en) Methods, kits &amp; compositions for determining gene copy numbers
EP3174998B1 (en) Method for selecting a target nucleic acid sequence
JP2023175685A (en) Compositions, methods, and kits for urinary tract microorganism detection
US11680285B2 (en) Hooked probe, method for ligating nucleic acid and method for constructing sequencing library
JP5537940B2 (en) Novel labeled oligonucleotide
CN115175985A (en) Method for extracting single-stranded DNA and RNA from untreated biological sample and sequencing

Legal Events

Date Code Title Description
AS Assignment

Owner name: LIFE TECHNOLOGIES CORPORATION, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:XU, TOM;TRINH, CHRISTOPHER;BORNARTH, CAROLE;AND OTHERS;REEL/FRAME:028492/0045

Effective date: 20120430

STCB Information on status: application discontinuation

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