WO2007146158A1 - Dna sequencing by nanopore using modified nucleotides - Google Patents

Dna sequencing by nanopore using modified nucleotides Download PDF

Info

Publication number
WO2007146158A1
WO2007146158A1 PCT/US2007/013559 US2007013559W WO2007146158A1 WO 2007146158 A1 WO2007146158 A1 WO 2007146158A1 US 2007013559 W US2007013559 W US 2007013559W WO 2007146158 A1 WO2007146158 A1 WO 2007146158A1
Authority
WO
WIPO (PCT)
Prior art keywords
nucleotide
dna
stranded
rna
nucleotides
Prior art date
Application number
PCT/US2007/013559
Other languages
French (fr)
Inventor
Jingyue Ju
Original Assignee
The Trustees Of Columbia University In The City Of New York
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 The Trustees Of Columbia University In The City Of New York filed Critical The Trustees Of Columbia University In The City Of New York
Priority to US12/308,091 priority Critical patent/US8889348B2/en
Priority to CN200780028545.1A priority patent/CN101495656B/en
Publication of WO2007146158A1 publication Critical patent/WO2007146158A1/en
Priority to US14/516,785 priority patent/US20150037788A1/en
Priority to US15/255,029 priority patent/US20170096704A1/en
Priority to US16/218,175 priority patent/US20190300947A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6869Methods for sequencing

Definitions

  • DNA sequencing is a fundamental technology for biology.
  • Several analytical methods have been developed to detect DNA or RNA at the single molecule level using chemical or physical microscopic technologies [15, 16, 21 and 23] .
  • the ion channel has been explored for detecting individual DNA or RNA strands, with nanopore being a candidate for high rate sequencing and analysis of DNA [9, 10, 4, 3 and 7] .
  • Kasianowicz efc al first demonstrated that the ⁇ -hemolysin channel, an exotoxin secreted by a bacterium, could be used to detect nucleic acids at the single molecule level [8] .
  • the monomeric polypeptide self-assembles in a lipid bilayer membrane to form a heptameric pore, with a 2.6 nm-diameter vestibule and 1.5 nm-diameter limiting aperture
  • the pore formed by the ⁇ -hemolysin channel conducts a sufficiently strong and steady ionic current when an appropriate voltage is applied across the membrane.
  • the limiting aperture of the nanbpore allows linear single-stranded but not double-stranded nucleic acid molecules (diameter -2.0 nm) to pass through.
  • the polyanionic nucleic acids are driven through the pore by the applied electric field, which blocks or reduces the ionic current that would be otherwise unimpeded.
  • This process of passage generates an electronic signature (Figure 1) [23 and 5J .
  • a particular nucleic acid molecule, when entering and passing through the nanopore, will generate a characteristic signature that distinguishes it from others.
  • the duration of the blockade is proportional to the length of the nucleic acid, and its signal strength is related to the steric and electronic properties of the nucleotides, namely the identity of the four bases (A, C, G and T) .
  • a specific event diagram is constructed which is the plot of translocation time versus blockade current.
  • This specific event diagram (also referred to as an electronic signature) is used to distinguish the lengths and the compositions of polynucleotides by single-channel recording techniques based on characteristic parameters such as translocation current, translocation duration, and their corresponding dispersions in the diagram [14] .
  • nanopore approach is known as a DNA detection method, this approach for base-to-base sequencing has not yet been achieved.
  • This invention provides a method for determining the nucleotide sequence of a single-stranded DNA comprising the steps of:
  • each A or each G residue and at least each C, each T or each U residue comprises a modifying group bound to its respective base so that each type of nucleotide in the DNA has an electronic signature which is distinguishable from the electronic signature of each other type of nucleotide in the DNA;
  • step (b) for each nucleotide of the DNA which passes through the pore, determining an electronic signature for such nucleotide; and (c) comparing each electronic signature determined in step (b) with electronic signatures corresponding to each of A, G, C and T modified as per the nucleotides in the single-stranded DNA, so as to determine the identity of each such nucleotide, thereby determining the nucleotide sequence of the single-stranded DNA.
  • This invention also provides a method for determining the nucleotide sequence of a single -stranded RNA comprising the steps of: (a) passing the single-stranded RNA through a pore of suitable diameter by applying an electric field to the RNA, wherein at least each A or each G residue and at least each C or each U residue comprises a modifying group bound to its respective base so that each type of nucleotide in the RNA has an electronic signature which is distinguishable from the electronic signature of each other type of nucleotide in the RNA;
  • step (c) comparing each electronic signature determined in step (b) with electronic signatures corresponding to each of A, G, C and U modified as per the nucleotides in the single-stranded RNA, so as to determine the identity of each such nucleotide, thereby determining the nucleotide sequence of the single-stranded RNA.
  • This invention also provides a nucleotide having an azido group covalently bound to its base.
  • This invention also provides a method for making a modified nucleotide comprising contacting the instant nucleotide with an alkyne-containing compound under conditions permitting reaction between the azido and the alkyne groups, thereby making the modified nucleotide.
  • Figure 2 Structures of nucleotides dATP, dGTP, dCTP and dTTP.
  • Figure 4 Structures of dCTP and dGTP, and modified nucleotides (dATP-NH 2 and dUTP-NH 2 ) .
  • Figure 7 Steps of verifying sequencing capacity via nanopore using various DNA sequences.
  • SEQ ID NOs. 3-6 for part (i), top to bottom, respectively; SEQ ID NO:7 for part (ii) ; SEQ ID NO : 8 for part (iii) ; and SEQ ID NO: 9 for part (iv) ) .
  • Figure 8. Structures of unmodified nucleotides (dCTP and dGTP) and hook-labeled nucleotides (dATP-NH 2 and dUTP-N 3 ) .
  • the amino and the azido groups function as hooks to conjugate with bulky groups after the nucleotides are incorporated into the DNA strand.
  • Figure 10 DNA-extension reaction using hook- labeled nucleotides (dATP-NH 2 and dUTP-N 3 ) to generate a modified single-stranded DNA chain, which will then react with large functional groups (Rl and R3) selectively for distinct detection by nanopore . (SEQ ID NOs. 1 and 2 for template and primer, respectively)
  • Electronic signature of a nucleotide passing through a pore via application of an electronic field shall include, for example, the duration of the nucleotide's passage through the pore together with the observed amplitude of current during that passage.
  • Electronic signatures can be visualized, for example, by a plot of current (e.g. pA) versus time.
  • Electronic signature for a DNA is also envisioned and can be, for example, a plot of current (e.g. pA) versus time for the DNA to pass through the pore via application of an electric field.
  • Nanopore includes, for example, a structure comprising (a) a first and a second compartment separated by a physical barrier, which barrier has at least one pore with a diameter, for example, of from about 1 to 10 run, and (b) a means for applying an electric field across the barrier so that a charged molecule such as DNA can pass from the first compartment through the pore to the second compartment .
  • the nanopore ideally further comprises a means for measuring the electronic signature of a molecule passing through its barrier.
  • the nanopore barrier may be synthetic or naturally occurring in part.
  • Barriers can include, for example, lipid bilayers having therein ⁇ -hemolysin, oligomeric protein channels such as porins, and synthetic peptides and the like. Barriers can also include inorganic plates having one or more holes of a suitable size.
  • nanopore , “nanopore barrier” and the "pore” in the nanopore barrier are sometimes used equivalentIy.
  • Nucleic acid shall mean any nucleic acid molecule, including, without limitation, DNA, RNA and hybrids thereof.
  • the nucleic acid bases that form nucleic acid molecules can be the bases A, C, G, T and U, as well as derivatives thereof. Derivatives of these bases are well known in the art, and are exemplified in PCR Systems, Reagents and Consumables (Perkin Elmer Catalogue 1996-1997, Roche Molecular Systems, Inc., Branchburg, New Jersey, USA) .
  • Type of nucleotide refers to A, G, C, T or U.
  • This invention provides a method for determining the nucleotide sequence of a single- stranded DNA comprising the steps of:
  • step (c) comparing each electronic signature determined in step (b) with electronic signatures corresponding to each of A, G, C and T modified as per the nucleotides in the single-stranded DNA, so as to determine the identity of each such nucleotide, thereby determining the nucleotide sequence of the single -stranded DNA.
  • the single- stranded DNA is obtained by (a) synthesizing double- stranded DNA using a single -stranded template, a DNA polymerase and nucleotides, wherein at least each A or each G residue and at least each C or each T residue comprises a modifying group bound to its respective base so that each type of nucleotide in the DNA has an electronic signature which is distinguishable from the electronic signature of each other type nucleotide in the DNA, and (b) removing from the resulting double- stranded DNA the single-stranded DNA containing modified nucleotides.
  • the single-stranded DNA is obtained by (a) synthesizing double-stranded DNA using a single-stranded template, a DNA polymerase and nucleotides, wherein at least each A, each G, each C, each U or each T residue comprises an azido group bound to its base, and at least each A, each G, each C, each U and each T comprises an amino group bound to its base, whereby the azido and amino groups do not reside on the same type of base, (b) removing from the resulting double- stranded DNA the single-stranded DNA containing the azido and amino group-containing nucleotides and (c) reacting the resulting single-stranded DNA with a first modifying group which forms a bond with the azido group and a second modifying group which forms a bond with the amino group so as to obtain the single- stranded DNA.
  • This invention also provides a method for determining the nucleotide sequence of a single-stranded RNA comprising the steps of:
  • step (c) comparing each electronic signature determined in step (b) with electronic signatures corresponding to each of A, G, C and U modified as per the nucleotides in the single-stranded
  • RNA so as to determine the identity of each such nucleotide, thereby determining the nucleotide sequence of the single-stranded RNA.
  • the single- stranded RNA is obtained by (a) synthesizing double- stranded RNA using a single-stranded template, an RNA polymerase and nucleotides, wherein at least each A, each G, each C or each U residue comprises an azido group bound to its base, and at least each A, each G, each C and each U comprises an amino group bound to its base, whereby the azido and amino groups do not reside on the same type of base, (b) removing from the resulting double- stranded RNA the single -stranded RNA containing the azido and amino group-containing nucleotides and (c) reacting the resulting single- stranded RNA with a first modifying group which forms a bond with the azido group and a second modifying group which forms a bond with the amino group so as to obtain the single-stranded RNA.
  • the single-stranded RNA is obtained by (a) synthesizing double-stranded RNA using a single-stranded template, an RNA polymerase and nucleotides, wherein at least each A or each G residue and at least each C or each U residue comprises a modifying group bound to its respective base so that each type of nucleotide in the RNA has an electronic signature which is distinguishable from the electronic signature of each other type nucleotide in the RNA, and (b) removing from the resulting double-stranded RNA the single- stranded RNA containing modified nucleotides.
  • the pore has a diameter of from about 1 nm to about 5 nm. In a further embodiment of the instant methods, the pore has a diameter of from about 1 nm to about 3 nm. In embodiments of the instant methods, the pore has a diameter of about 1 nm, 2 nm, 3 nm, 4 nm or 5 nm. In further embodiments, the pore is, for example, about
  • a single pore is employed. In another embodiment, multiple pores are employed.
  • Nanopore devices are known in the art. See, for example, references [24] through [34] . Nanopores and methods employing them are disclosed in U.S. Patent Nos. 7,005,264 B2 and 6,617,113 which are hereby incorporated by reference in their entirety.
  • each A and each T or each U residue comprises a modifying group; each A and each U residue comprises a modifying group; and/or each G and each C residue comprises a modifying group.
  • Moieties used to modify nucleotides can differ in size and/or charge, so long as each type of nucleotide in a nucleic acid whose sequence is being determined by the instant methods has an electronic signature which differs from each other type.
  • DNA polymerases which can be used in the instant invention include, for example E. CoIi DNA polymerase I, Bacteriophage T4 DNA polymerase, SequenaseTM, Taq DNA polymerase and 9°N polymerase (exo-) A485L/Y409V.
  • RNA polymerases which can be used in the instant invention include, for example, Bacteriophage SP6, T7 and T3 RNA polymerases .
  • This invention also provides a nucleotide having an azido group covalently bound to its base.
  • the nucleotide is dUTP and the azido group is bound Co the base at the 5-position.
  • the nucleotide is dATP and the azido group is bound to the base at the 8-position.
  • the nucleotide is dGTP and the azido group is bound to the base at the 8-position.
  • the azido and amino groups can also be any other groups which permit binding of a unique moiety to each type of nucleotide.
  • This invention also provides a method for making a modified nucleotide comprising contacting the instant nucleotide with an alkyne-containing compound under conditions permitting reaction between the azido and the alkyne groups, thereby making the modified nucleotide .
  • a and G are purines, while C and T are pyrimidines .
  • the overall molecular sizes of A and G are very similar, while the sizes of C and T are similar.
  • nanopore has been shown to be able to differentiate between purines and pyrimidines [1 and
  • a and T are bulky purines similar in size, they will generate similar blocking current signatures (also called electronic signatures) in the nanopore.
  • C and T both pyrimidines, will generate similar signatures.
  • the site selected for modification is on the 7-position of A and the 5- position of T nucleotide molecules.
  • the 7-position of A and the 5-position of T have been shown to be chemically modified with bulky groups while not affecting basic DNA properties, such as forming the double-stranded DNA structure and being able to carry out polymerase reactions [2, 13 and 17] .
  • nucleotide bulkiness will be: A* > U* > G > C, as purines (A and G) are larger than pyrimidines (C and U) and in general the modification group Rx is larger than R 2 .
  • the modified dATP and dUTP, and the unmodified dCTP and dGTP, are then be used in a polymerase reaction to generate single -stranded DNA.
  • the single-stranded DNA chain is obtained after being denatured from the template chain, which is composed of the modified dATP and dUTP as well as unmodified dCTP and dGTP.
  • the 5'- end of the primer chain is modified on the base by a biotin moiety to isolate only DNA product that has incorporated the modified nucleotides.
  • a series of tests are conducted as shown in Figure 7.
  • a polynucleotide stretch composed of only 50 identical nucleotides (i) is prepared by polymerase reaction as described above. Each DNA sequence is expected to generate different electronic blockade signatures due to the larger size difference of the nucleotides.
  • the modification effects of R 1 and R 2 for A and T can be compared for preliminary optimization.
  • a polynucleotide stretch composed of 30 modified A' s and 30 modified T's (ii) is prepared and then tested in nanopore to demonstrate that the electronic blockade signatures differ in magnitude between A and T and are easily distinguishable.
  • the candidates for R 1 and R 2 groups are selected to achieve the best discrimination in signal.
  • a shorter polynucleotide stretch composed of 10 A's, 10 Cs, 10 G' s and 10 T's are prepared and tested in nanopore for further confirmation on the electronic blockade signatures (also called electronic signatures) .
  • a polynucleotide stretch composed of three consecutive A-C-G-T sequence is prepared and tested in nanopore.
  • the detailed sequencing conditions can be optimized according to known methods. Based on these results, random DNA chain with modified A and T and unmodified C and G is evaluated for accurate detection and discrimination by the nanopore.
  • an alternative strategy is to introduce small 'hooks' to the nucleotides, then perform polymerase reaction to produce DNA products with hook-labeled nucleotides incorporated in them. The DNA products are then linked with the large functional groups through the hook for distinct detection by nanopore.
  • dUTP-N 3 is synthesized from 5-iodo-2* -deoxyuridine as shown in Figure 9.
  • 5-Iodo-2 ' -deoxyuridine is first coupled with propargylamine in the presence of palladium(0) and copper (I) catalysts. Then the amino group is converted into azido group by the diazo transfer method [11] . Finally triphosphate is introduced to the 5' -hydroxy group of the nucleoside to yield dUTP-N 3 [6] .
  • the dATP-NH 2 and dUTP-N 3/ and the unmodified dCTP and dGTP, are used in polymerase reaction on the single- stranded nucleic acid template to obtain hook-labeled DNA products. Due to the small sizes of the azido and amino groups, these nucleotides are expected to be good substrates of commonly used DNA polymerases . After isolation of the single stranded DNA carrying the hook, the azido groups on these modified DNA chains will be further modified by Huisgen 1,3 -dipolar cycloaddition with terminal alkynes (R 3 CsCH) in the presence of copper(I) catalyst ( Figure 10) [18 and 19].
  • nanopores are constructed with different configurations and modifications for characterizing DNA containing nucleotides of different sizes.
  • Synthetic nanopores are described in references [24] i. through [28] which are hereby incorporated by reference in their entirety.
  • the mechanics and kinetics of DNA passage through the pores are described in references [29] and [30] , respectively.

Abstract

This invention provides a process for sequencing single-stranded DNA by employing a nanopore and modified nucleotides.

Description

DNA SEQUENCING BY NANOPORK USING MODIFIED NUCLEOTIDES
The invention disclosed herein was made with government support under grant no. 1R2IHGO03718-01 from the National Human Genome Research Institute. Accordingly, the U.S. Government has certain rights in this invention.
Throughout this application, various publications are referenced in parentheses by number. Full citations for these references may be found at the end of the specification immediately preceding the claims. The disclosures of these publications in their entireties are hereby incorporated by reference into this application to more fully describe the state of the art to which this invention pertains.
Background of the Invention
DNA sequencing is a fundamental technology for biology. Several analytical methods have been developed to detect DNA or RNA at the single molecule level using chemical or physical microscopic technologies [15, 16, 21 and 23] . In the past few years, the ion channel has been explored for detecting individual DNA or RNA strands, with nanopore being a candidate for high rate sequencing and analysis of DNA [9, 10, 4, 3 and 7] . In 1996, Kasianowicz efc al . first demonstrated that the α-hemolysin channel, an exotoxin secreted by a bacterium, could be used to detect nucleic acids at the single molecule level [8] . The monomeric polypeptide self-assembles in a lipid bilayer membrane to form a heptameric pore, with a 2.6 nm-diameter vestibule and 1.5 nm-diameter limiting aperture
(namely, the narrowest point of the pore) [1, 14 and
151. In an aqueous ionic salt solution such as KCl, the pore formed by the α-hemolysin channel conducts a sufficiently strong and steady ionic current when an appropriate voltage is applied across the membrane. The limiting aperture of the nanbpore allows linear single-stranded but not double-stranded nucleic acid molecules (diameter -2.0 nm) to pass through. The polyanionic nucleic acids are driven through the pore by the applied electric field, which blocks or reduces the ionic current that would be otherwise unimpeded. This process of passage generates an electronic signature (Figure 1) [23 and 5J . A particular nucleic acid molecule, when entering and passing through the nanopore, will generate a characteristic signature that distinguishes it from others. The duration of the blockade is proportional to the length of the nucleic acid, and its signal strength is related to the steric and electronic properties of the nucleotides, namely the identity of the four bases (A, C, G and T) .
A specific event diagram is constructed which is the plot of translocation time versus blockade current.
This specific event diagram (also referred to as an electronic signature) is used to distinguish the lengths and the compositions of polynucleotides by single-channel recording techniques based on characteristic parameters such as translocation current, translocation duration, and their corresponding dispersions in the diagram [14] .
Although the nanopore approach is known as a DNA detection method, this approach for base-to-base sequencing has not yet been achieved.
Summary of the Invention
This invention provides a method for determining the nucleotide sequence of a single-stranded DNA comprising the steps of:
(a) passing the single-stranded DNA through a pore of suitable diameter by applying an electric field to the DNA, wherein at least each A or each G residue and at least each C, each T or each U residue comprises a modifying group bound to its respective base so that each type of nucleotide in the DNA has an electronic signature which is distinguishable from the electronic signature of each other type of nucleotide in the DNA;
(b) for each nucleotide of the DNA which passes through the pore, determining an electronic signature for such nucleotide; and (c) comparing each electronic signature determined in step (b) with electronic signatures corresponding to each of A, G, C and T modified as per the nucleotides in the single-stranded DNA, so as to determine the identity of each such nucleotide, thereby determining the nucleotide sequence of the single-stranded DNA.
This invention also provides a method for determining the nucleotide sequence of a single -stranded RNA comprising the steps of: (a) passing the single-stranded RNA through a pore of suitable diameter by applying an electric field to the RNA, wherein at least each A or each G residue and at least each C or each U residue comprises a modifying group bound to its respective base so that each type of nucleotide in the RNA has an electronic signature which is distinguishable from the electronic signature of each other type of nucleotide in the RNA;
(b) for each nucleotide of the RNA which passes through the pore, determining an electronic signature for such nucleotide; and
(c) comparing each electronic signature determined in step (b) with electronic signatures corresponding to each of A, G, C and U modified as per the nucleotides in the single-stranded RNA, so as to determine the identity of each such nucleotide, thereby determining the nucleotide sequence of the single-stranded RNA.
This invention also provides a nucleotide having an azido group covalently bound to its base.
This invention also provides a method for making a modified nucleotide comprising contacting the instant nucleotide with an alkyne-containing compound under conditions permitting reaction between the azido and the alkyne groups, thereby making the modified nucleotide. Brief Description of the Figures
Figure 1. α-Hemolysin protein self-assembles in a lipid bilayer to form an ion channel and a nucleic acid stretch passes through it (left) , with the corresponding electronic signatures generated (right)
[23 and 5] .
Figure 2. Structures of nucleotides dATP, dGTP, dCTP and dTTP.
Figure 3. Nucleotide bulkiness in ascending order: (a) 5'-CG0T-3', (b) 5'-G*S0T-3', (c) 5'-A*60T-3', and (d)
Figure imgf000007_0001
Figure 4. Structures of dCTP and dGTP, and modified nucleotides (dATP-NH2 and dUTP-NH2) .
Figure 5. Modification of dATP-NH2 and dϋTP-NH2.
Figure 6. DNA-extension reaction using modified nucleotides (dATP-NHCORi and dUTP-NHCOR2) to generate a modified single-stranded DNA chain. (SEQ ID NOs. 1 and 2 for template and primer, respectively) .
Figure 7. Steps of verifying sequencing capacity via nanopore using various DNA sequences. (SEQ ID NOs. 3-6 for part (i), top to bottom, respectively; SEQ ID NO:7 for part (ii) ; SEQ ID NO : 8 for part (iii) ; and SEQ ID NO: 9 for part (iv) ) . Figure 8. Structures of unmodified nucleotides (dCTP and dGTP) and hook-labeled nucleotides (dATP-NH2 and dUTP-N3) . The amino and the azido groups function as hooks to conjugate with bulky groups after the nucleotides are incorporated into the DNA strand.
Figure 9. Synthesis of dUTP-N3.
Figure 10. DNA-extension reaction using hook- labeled nucleotides (dATP-NH2 and dUTP-N3) to generate a modified single-stranded DNA chain, which will then react with large functional groups (Rl and R3) selectively for distinct detection by nanopore . (SEQ ID NOs. 1 and 2 for template and primer, respectively)
Detailed Description of the Invention
Terms
As used herein, and unless stated otherwise, each of the following terms shall have the definition set forth below.
A - Adenine;
C - Cytosine;
DNA - Deoxyribonucleic acid;
G - Guanine; RNA - Ribonucleic acid;
T - Thymine; and
U - Uracil.
"Electronic signature" of a nucleotide passing through a pore via application of an electronic field shall include, for example, the duration of the nucleotide's passage through the pore together with the observed amplitude of current during that passage. Electronic signatures can be visualized, for example, by a plot of current (e.g. pA) versus time. Electronic signature for a DNA is also envisioned and can be, for example, a plot of current (e.g. pA) versus time for the DNA to pass through the pore via application of an electric field.
"Nanopore" includes, for example, a structure comprising (a) a first and a second compartment separated by a physical barrier, which barrier has at least one pore with a diameter, for example, of from about 1 to 10 run, and (b) a means for applying an electric field across the barrier so that a charged molecule such as DNA can pass from the first compartment through the pore to the second compartment . The nanopore ideally further comprises a means for measuring the electronic signature of a molecule passing through its barrier. The nanopore barrier may be synthetic or naturally occurring in part. Barriers can include, for example, lipid bilayers having therein α-hemolysin, oligomeric protein channels such as porins, and synthetic peptides and the like. Barriers can also include inorganic plates having one or more holes of a suitable size. Herein "nanopore" , "nanopore barrier" and the "pore" in the nanopore barrier are sometimes used equivalentIy.
"Nucleic acid" shall mean any nucleic acid molecule, including, without limitation, DNA, RNA and hybrids thereof. The nucleic acid bases that form nucleic acid molecules can be the bases A, C, G, T and U, as well as derivatives thereof. Derivatives of these bases are well known in the art, and are exemplified in PCR Systems, Reagents and Consumables (Perkin Elmer Catalogue 1996-1997, Roche Molecular Systems, Inc., Branchburg, New Jersey, USA) .
"Type" of nucleotide refers to A, G, C, T or U. Embodiments of the Invention
This invention provides a method for determining the nucleotide sequence of a single- stranded DNA comprising the steps of:
(a) passing the single- stranded DNA through a pore of suitable diameter by applying an electric field to the DNA, wherein at least each A or each G residue and at least each C, each T or each U residue comprises a modifying group bound to its respective base so that each type of nucleotide in the DNA has an electronic signature which is distinguishable from the electronic signature of each other type of nucleotide in the DNA;
(b) for each nucleotide of the DNA which passes through the pore, determining an electronic signature for such nucleotide; and
(c) comparing each electronic signature determined in step (b) with electronic signatures corresponding to each of A, G, C and T modified as per the nucleotides in the single-stranded DNA, so as to determine the identity of each such nucleotide, thereby determining the nucleotide sequence of the single -stranded DNA.
In an embodiment of the instant method, the single- stranded DNA is obtained by (a) synthesizing double- stranded DNA using a single -stranded template, a DNA polymerase and nucleotides, wherein at least each A or each G residue and at least each C or each T residue comprises a modifying group bound to its respective base so that each type of nucleotide in the DNA has an electronic signature which is distinguishable from the electronic signature of each other type nucleotide in the DNA, and (b) removing from the resulting double- stranded DNA the single-stranded DNA containing modified nucleotides.
In another embodiment of the instant method, the single-stranded DNA is obtained by (a) synthesizing double-stranded DNA using a single-stranded template, a DNA polymerase and nucleotides, wherein at least each A, each G, each C, each U or each T residue comprises an azido group bound to its base, and at least each A, each G, each C, each U and each T comprises an amino group bound to its base, whereby the azido and amino groups do not reside on the same type of base, (b) removing from the resulting double- stranded DNA the single-stranded DNA containing the azido and amino group-containing nucleotides and (c) reacting the resulting single-stranded DNA with a first modifying group which forms a bond with the azido group and a second modifying group which forms a bond with the amino group so as to obtain the single- stranded DNA.
This invention also provides a method for determining the nucleotide sequence of a single-stranded RNA comprising the steps of:
(a) passing the single-stranded RNA through a pore of suitable diameter by applying an electric field to the RNA, wherein at least each A or each G residue and at least each C or each U residue comprises a modifying group bound to its respective base so that each type of nucleotide in the RNA has an electronic signature which is distinguishable from the electronic signature of each other type of nucleotide in the RNA;
(b) for each nucleotide of the RNA which passes through the pore, determining an electronic signature for such nucleotide; and
(c) comparing each electronic signature determined in step (b) with electronic signatures corresponding to each of A, G, C and U modified as per the nucleotides in the single-stranded
RNA, so as to determine the identity of each such nucleotide, thereby determining the nucleotide sequence of the single-stranded RNA.
In an embodiment of the instant method, the single- stranded RNA is obtained by (a) synthesizing double- stranded RNA using a single-stranded template, an RNA polymerase and nucleotides, wherein at least each A, each G, each C or each U residue comprises an azido group bound to its base, and at least each A, each G, each C and each U comprises an amino group bound to its base, whereby the azido and amino groups do not reside on the same type of base, (b) removing from the resulting double- stranded RNA the single -stranded RNA containing the azido and amino group-containing nucleotides and (c) reacting the resulting single- stranded RNA with a first modifying group which forms a bond with the azido group and a second modifying group which forms a bond with the amino group so as to obtain the single-stranded RNA.
In another embodiment of the instant method, the single-stranded RNA is obtained by (a) synthesizing double-stranded RNA using a single-stranded template, an RNA polymerase and nucleotides, wherein at least each A or each G residue and at least each C or each U residue comprises a modifying group bound to its respective base so that each type of nucleotide in the RNA has an electronic signature which is distinguishable from the electronic signature of each other type nucleotide in the RNA, and (b) removing from the resulting double-stranded RNA the single- stranded RNA containing modified nucleotides.
In one embodiment of the instant methods, the pore has a diameter of from about 1 nm to about 5 nm. In a further embodiment of the instant methods, the pore has a diameter of from about 1 nm to about 3 nm. In embodiments of the instant methods, the pore has a diameter of about 1 nm, 2 nm, 3 nm, 4 nm or 5 nm. In further embodiments, the pore is, for example, about
1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0,
2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1,
3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.1, 4.2,
4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9 or 5.0 nm in diameter. In one embodiment, a single pore is employed. In another embodiment, multiple pores are employed.
Nanopore devices are known in the art. See, for example, references [24] through [34] . Nanopores and methods employing them are disclosed in U.S. Patent Nos. 7,005,264 B2 and 6,617,113 which are hereby incorporated by reference in their entirety.
In one embodiment of the instant methods, each A and each T or each U residue comprises a modifying group; each A and each U residue comprises a modifying group; and/or each G and each C residue comprises a modifying group.
Moieties used to modify nucleotides can differ in size and/or charge, so long as each type of nucleotide in a nucleic acid whose sequence is being determined by the instant methods has an electronic signature which differs from each other type.
DNA polymerases which can be used in the instant invention include, for example E. CoIi DNA polymerase I, Bacteriophage T4 DNA polymerase, Sequenase™, Taq DNA polymerase and 9°N polymerase (exo-) A485L/Y409V.RNA polymerases which can be used in the instant invention include, for example, Bacteriophage SP6, T7 and T3 RNA polymerases .
This invention also provides a nucleotide having an azido group covalently bound to its base. In one embodiment, the nucleotide is dUTP and the azido group is bound Co the base at the 5-position. In one embodiment, the nucleotide is dATP and the azido group is bound to the base at the 8-position. In another embodiment, the nucleotide is dGTP and the azido group is bound to the base at the 8-position. The azido and amino groups can also be any other groups which permit binding of a unique moiety to each type of nucleotide.
This invention also provides a method for making a modified nucleotide comprising contacting the instant nucleotide with an alkyne-containing compound under conditions permitting reaction between the azido and the alkyne groups, thereby making the modified nucleotide .
This invention will be better understood by reference to the Experimental Details which follow, but those skilled in the art will readily appreciate that the specific experiments detailed are only illustrative of the invention as described more fully in the claims which follow thereafter.
Experimental Details
The structures of the four nucleotides are shown in
Figure 2. A and G are purines, while C and T are pyrimidines . The overall molecular sizes of A and G are very similar, while the sizes of C and T are similar. Thus, nanopore has been shown to be able to differentiate between purines and pyrimidines [1 and
14] , but not to be able to distinguish between individual purines, A and G, or between individual pyrimidines, C and T.
Disclosed here is the design of modified nucleotides to enhance discrimination of each nucleotide by modifying A and T. Since A and G are bulky purines similar in size, they will generate similar blocking current signatures (also called electronic signatures) in the nanopore. Likewise C and T, both pyrimidines, will generate similar signatures. The site selected for modification is on the 7-position of A and the 5- position of T nucleotide molecules. The 7-position of A and the 5-position of T have been shown to be chemically modified with bulky groups while not affecting basic DNA properties, such as forming the double-stranded DNA structure and being able to carry out polymerase reactions [2, 13 and 17] . These modifications will enlarge the discrimination of the bases by nanopore due to the increased size differences between the four nucleotides (A, G, C and T) . In addition, the DNA translocation rate through the nanopore is expected to slow down due to the bulkiness of the modified nucleotides. Thus, achieving the accuracy and reliability required for the base-to- base sequencing is envisioned. The overall analytical parameters in the nanopore sequencing, such as concentration of the polynucleotide, magnitude of applied voltage, temperature and pH value of the solution, are optimized in order to get the most accurate and reliable results for the detection and analysis of the DNA chain.
Use Of Synthetic DNA Carrying Bulky Groups For Detection By Nanopore
In order to investigate the effect of nucleotide bulkiness on electronic blockade signals generated by the nanopore, various polynucleotides are synthesized with different bulky groups attached to the base of the nucleotide by a DNA synthesizer. Initially, regular C s and G' s are used to synthesize a series of polynucleotides (Figure 3a and 3b) . In addition, a series of polynucleotides using modified A's (6-amino- hexylamino attached to the 8-position of the base) and modified T' s (BIOTIN attached to the 5-position of the base) (Figure 3c and 3d) , which increase the bulkiness of the nucleotides, are synthesized. The order of the bulkiness of the nucleotides in Figure 3 is as follows: T* > A* > G > C. These polynucleotides are then passed through the nanopore to identify the relationship between the bulky groups attached to the base and the difference in electronic blockade signal between the different bases. Attachment Of Bulky Groups To Nucleotides For Nanopore Detection
(1) Design and synthesis of modified nucleotides (dATP-NHCORi and dUTP-NHCOR2) .
Synthesized dATP-NIfc and dUTP-NH2 are used as starting materials for further nucleotide modification while unmodified dCTP and dGTP are used directly (Figure 4) . The routes of nucleotide modification are shown in Figure 5. The commercially available carboxylic acids 1-10 will be converted into the corresponding N- hydroxysuccinimidyl (NHS) esters conveniently using N- hydroxysuccinimide and DCC [20 and 22] . Then the nucleotides for modification (dATP-NH2 and dUTP-NH2) will be connected with the modification groups R1 and
R2 respectively in DMF and NaHCO3/Na2CO3 buffer solution
[13 and 17] . After modification, the order of nucleotide bulkiness will be: A* > U* > G > C, as purines (A and G) are larger than pyrimidines (C and U) and in general the modification group Rx is larger than R2.
(2) DMA-extension reaction using modified nucleotides CdATP-NHCOR1 and dUl1P-NHCOR2) .
The modified dATP and dUTP, and the unmodified dCTP and dGTP, are then be used in a polymerase reaction to generate single -stranded DNA. As shown in Figure 6, after the polymerase reaction, the single-stranded DNA chain is obtained after being denatured from the template chain, which is composed of the modified dATP and dUTP as well as unmodified dCTP and dGTP. The 5'- end of the primer chain is modified on the base by a biotin moiety to isolate only DNA product that has incorporated the modified nucleotides. These modified single-stranded chains are then used in the nanopore by single-channel recording techniques for sequencing sensitivity and accuracy evaluation.
DNA-Sequencing Study By Nanopore
To validate nanopore' s ability to distinguish the four different nucleotides in DNA, a series of tests are conducted as shown in Figure 7. First, a polynucleotide stretch composed of only 50 identical nucleotides (i) is prepared by polymerase reaction as described above. Each DNA sequence is expected to generate different electronic blockade signatures due to the larger size difference of the nucleotides. The modification effects of R1 and R2 for A and T can be compared for preliminary optimization. Next, a polynucleotide stretch composed of 30 modified A' s and 30 modified T's (ii) is prepared and then tested in nanopore to demonstrate that the electronic blockade signatures differ in magnitude between A and T and are easily distinguishable.
Based on the signatures generated, the candidates for R1 and R2 groups are selected to achieve the best discrimination in signal. Third, a shorter polynucleotide stretch composed of 10 A's, 10 Cs, 10 G' s and 10 T's (iii) are prepared and tested in nanopore for further confirmation on the electronic blockade signatures (also called electronic signatures) . Finally, a polynucleotide stretch composed of three consecutive A-C-G-T sequence (iv) is prepared and tested in nanopore. The detailed sequencing conditions can be optimized according to known methods. Based on these results, random DNA chain with modified A and T and unmodified C and G is evaluated for accurate detection and discrimination by the nanopore. These procedures allow characterization of the signals from each of the - nucleotides and the transitions between nucleotides of different identities. The magnitude and duration of the blockade signatures on the event diagram are then analyzed and compared with known diagrams for validation. The schematic of the predicted blockade signals from DNA molecules (ii) , (iii) and (iv) are shown in Figure 7. Thus, with these rational chemical designs and modifications of the building blocks of DNA, this invention envisions using nanopore to decipher DNA sequences at the single molecule level with single base resolution.
Attach small hooks to the nucleotides for synthesis of DNA in polymerase reaction for nanopore detection
If a DNA polymerase is not able to synthesize a long strand of DNA due to the bulkiness of the functional groups introduced, an alternative strategy is to introduce small 'hooks' to the nucleotides, then perform polymerase reaction to produce DNA products with hook-labeled nucleotides incorporated in them. The DNA products are then linked with the large functional groups through the hook for distinct detection by nanopore.
(1) Design and synthesis of hook-labeled nucleotide dUTP-N3.
The available dCTP, dGTP and dATP-NH3 are used as starting materials directly (Figure 8) , while dUTP-N3 is synthesized from 5-iodo-2* -deoxyuridine as shown in Figure 9. 5-Iodo-2 ' -deoxyuridine is first coupled with propargylamine in the presence of palladium(0) and copper (I) catalysts. Then the amino group is converted into azido group by the diazo transfer method [11] . Finally triphosphate is introduced to the 5' -hydroxy group of the nucleoside to yield dUTP-N3 [6] .
(2) DNA-extension reaction using hook-labeled nucleotides (dATP-NH2 and dUTP-N3) .
The dATP-NH2 and dUTP-N3/ and the unmodified dCTP and dGTP, are used in polymerase reaction on the single- stranded nucleic acid template to obtain hook-labeled DNA products. Due to the small sizes of the azido and amino groups, these nucleotides are expected to be good substrates of commonly used DNA polymerases . After isolation of the single stranded DNA carrying the hook, the azido groups on these modified DNA chains will be further modified by Huisgen 1,3 -dipolar cycloaddition with terminal alkynes (R3CsCH) in the presence of copper(I) catalyst (Figure 10) [18 and 19]. The amino groups on the "A" nucleotides of these modified DNA chains are connected with the modification groups R1 in DMF and NaHCOaZNa2CO3 buffer solution [13 and 17] . After modification, the order of nucleotides bulkiness on the chain will be: A* > U* > G > C since in general the modification group R1 is larger than R3.
Nanopore Contruction and Detection of DNA
Based on information in the art, nanopores are constructed with different configurations and modifications for characterizing DNA containing nucleotides of different sizes.
Synthetic nanopores are described in references [24] i. through [28] which are hereby incorporated by reference in their entirety. The mechanics and kinetics of DNA passage through the pores are described in references [29] and [30] , respectively.
Natural nanopores are described in references [31] through [34] which are hereby incorporated by reference in their entirety.
References
1. Akeson, M., Branton, D., Kasianowicz, J.J., Brandin, E. and Deamer, D.W. Microsecond time- scale discrimination between polycytidylic acid and polyadenylic acid segments within single RNA molecules. Biophys. J. 1999, 77, 3227-3233.
2. Bai, X., Kim, S., Li, Z., Turro, N.J. and Ju, J. Design and synthesis of a photocleavable biotinylated nucleotide for DNA analysis by mass spectrometry. Nucleic Acids Research 2004, 32(2), 535-541.
3. Bezrukov, S.M., and Kasianowicz, J.J. Neutral polymers in the nanopores of alamethicin and alpha-hemolysin. Biologicheskie Membrany 2001, 18, 453-457.
4. Chandler, E.L. , Smith, A.L., Burden, L.M., Kasianowicz and Burden, D. L. Membrane Surface Dynamics of DNA-Threaded Nanopores Revealed by Simultaneous Single-Molecule Optical and Ensemble Electrical Recording. Langmuir 2004, 20, 898-905.
5. Deamer, D.W. and Branton, D. Characterization of nucleic acids by nanopore analysis. Ace. Cheπt. Res. 2002, 35(10), 817-825.
6. Lee S.E., Sidorov A., Gourlain T., Mignet N., Thorpe S.J., Brazier J.A. , Dickman M.J. , Hornby D. P., Grasby, J.A. and Williams, D.M. Enhancing the catalytic repertoire of nucleic acids: a systematic study of linker length and rigidity. Nucleic Acida Research 2001, 29(7), 1565-1573.
7. (' Henrickson, S. E., Misakian, M., Robertson, B. and Kasianowicz, J.J. Driven asymmetric DNA transport in a nanometer-scale pore. Physical Review Letters 2000, 85, 3057-3060.
8. Kasianowicz, J.J., Brandin, B., Branton, D. and Deamer, D.W. Characterization of individual polynucleotide molecules using a membrane channel . Proc. Natl. Acad. Sci. USA 1996, 93, 13770-13773.
9. Kasianowicz, J.J. Nanometer-scale pores: potential applications for DNA characterization and analyte detection. Disease Markers 2003, 18, 185-191.
10. Kasianowicz, J.J. Nanopore. Flossing with DNA. Nature Materials 2004, 3, 355-356.
11. Lundquist, J. T. and Pelletier, J. C. A New Tri- Orthogonal Strategy for Peptide Cyclization. Org. Lett. 2002, 4(19), 3219-3221.
12. Li, L., Stein, D., McMullan, C, Branton, D., Aziz, M.J. and Golovchenko, J.A. Ion-beam sculpting at nanometre length scales. Nature 2001, 412, 166-169. 13. Li, Z., Bai, X., Ruparel, H., Kim, S., Turro, N.J. and Ju, J. A photocleavable fluorescent nucleotide for DNA sequencing and analysis- Proc. Natl. Acad. Sci. USA 2003, 100, 414-419.
14. Meller, A., Nivon, L., Brandin, E., Golovchenko, J. and Branton, D. Rapid nanopore discrimination between single polynucleotide molecules. Proc. Natl. Acad. Sci. USA 2000, 97, 1079-1084.
15. Perkins, T.T., Quake, S.R., Smith, D. E. and Chu, S. Relaxation of a single DNA molecule observed by optical microscopy. Science 1994, 264, 822-826.
16. Rief, M., Clausen-Schaumann, H. and Gaub, H. E. Sequence-dependent mechanics of single DNA molecules. Mat. Struct. Biol. 1999, 6, 346-349.
17. Rosenblum, B.B., Lee, L. G., Spurgeon, S. L., Khan, S.H., Menchen, S.M., Heiner, CR. and Chen, S.M. New dye-labeled terminators for improved DNA sequencing patterns. Nucleic Acida Research 1997, 25(22), 4500-4504.
18. Rostovtsev, V.V. , Green, L.G. , Fokin, V.V. and
Sharpless, K. B. A stepwise huisgen cycloaddition process: copper (I) -catalyzed regioselective
"ligation" of azides and terminal alkynes. Angew.
Chem. Int. Ed. 2002, 41(14), 2596-2599.
19. Seo, T. S., Bai, X., Ruparel, H., Li, Z., Turro,
N.J. and Ju, J. Photocleavable fluorescent nucleotides for DNA sequencing on a chip constructed by site-specific coupling chemistry. Proc. Natl. Acad. Sci . USA 2004, 101, 5488-5493.
20. Singh, S. B. and Tomassini, J. E. Synthesis of natural flutimide and analogous fully substituted pyrazine-2 , 6-diones, endonuclease inhibitors of influenza virus. J". Org. Chem. 2001, 66(16), 5504-5516.
21. Smith, S.B., Cui, Y. and Bustamante, C. Overstretching B-DNA: the elastic response of individual double-stranded and single-stranded DNA molecules. Science 1996, 271, 795-799.
22. Streater, M., Taylor, P. D., Hider, R. C, and Porter, J. Novel 3-hydroxy-2 (IH) -pyridinones. Synthesis, iron (III) -chelating properties, and biological activity. J. Medicinal Chem. 1990, 33(6), 1749-1755.
23. Vercoutere, W., Winters-Hilt, S., Olsen, H., Deamer, D., Haussler, D. and Akeson, M. Rapid discrimination among individual DNA hairpin molecules at single-nucleotide resolution using an ion channel. Nat. Biotech 2001, 19, 248-252.
24. Heng, J. B. et al . , The Electromechanics of DNA in a synthetic Nanopore . Biophysical Journal 2006, 90, 1098-1106. 21
25. Fologea, D. et al . , Detecting Single Stranded DNA with a Solid State Nanopore. Nano Letters 2005 5(10), 1905-1909.
26. Heng, J. B. et al . , Stretching DNA Using the Electric Field in a Synthetic Nanopore. Nano Letters 2005 5(10), 1883-1888.
27. Fologea, D. et al . , Slowing DNA Translocation in a Solid State Nanopore. Nano Letters 2005 5(9),
1734-1737.
28. Bokhari, S. H. and Sauer, J. R., A Parallel Graph Decomposition Algorithm for DNA Sequencing with Nanopores. Bioinformαtics 2005 21(7), 889-896.
29. Mathe, J. et al . , Nanopore Unzipping of Individual Hairpin Molecules. Biophysical Journal 2004 87, 3205-3212.
30. Aksimentiev, A. et al . , Microscopic Kinetics of DNA Translocation through Synthetic Nanopores. Biophysical Journal 2004 87, 2086-2097.
31. Wang, H. et al . , DNA heterogeneity and Phosphorylation unveiled by Single-Molecule Electrophoresis. PWAS 2004 101(37), 13472-13477.
32. Sauer-Budge, A. F. et al . , Unzipping Kinetics of Doubel Stranded DNA in a Nanopore. Physical Review Letters 2003 90(23), 238101-1 - 238101-4. 33. Vercoutere, W.A. et al . , Discrimination Among Individual Watson-Crick Base Pairs at the Terminin of Single DNA Hairpin Molecules. Nucleic Acids Research 2003 31(4), 1311-1318.
34. Meller, A. et al . , Single Molecule Measurements of DNA Transport Through a Nanopore.
, Electrophoresis 2002 23, 2583-2591.

Claims

What is claimed is:
1. A method for determining the nucleotide sequence of a single-stranded DNA comprising the steps of:
(a) passing the single-stranded ONA through a pore of suitable diameter by applying an electric field to the DNA, wherein at least each A or each G residue and at least each C, each T or each U residue comprises a modifying group bound to its respective base so that each type of nucleotide in the DNA has an electronic signature which is distinguishable from the electronic signature of each other type of nucleotide in the DNA;
(b) for each nucleotide of the DNA which passes through the pore, determining an electronic signature for such nucleotide; and
(c) comparing each electronic signature determined in step (b) with electronic signatures corresponding to each of A, G, C and T modified as per the nucleotides in the single-stranded DNA, so as to determine the identity of each such nucleotide, thereby determining the nucleotide sequence of the single-stranded DNA.
2. A method for determining the nucleotide sequence of a single-stranded RNA comprising the steps of:
(a) passing the single-stranded RNA through a pore of suitable diameter by applying an electric field to the RNA, wherein at least each A or each G residue and at least each C or each U residue comprises a modifying group bound to its respective base so that each type of nucleotide in the RNA has an electronic signature which is distinguishable from the electronic signature of each other type of nucleotide in the RNA;
(b) for each nucleotide of the RNA which passes through the pore, determining an electronic signature for such nucleotide; and
(c) comparing each electronic signature determined in step (b) with electronic signatures corresponding to each of A, G, C and U modified as per the nucleotides in the single-stranded RNA, so as to determine the identity of each such nucleotide, thereby determining the nucleotide sequence of the single-stranded RNA.
3. The method of claim 1 or 2, wherein the pore has a diameter of from about 1 nm to about 5 nm.
4. The method of claim 1 or 2, wherein the pore has a diameter of from about 1 nm to about 3 nm.
5. The method of claim 1 or 2, wherein the pore has a diameter of about 1 nm, 2 nm, 3 nm, 4 nm or 5 nm.
6. The method of claim 1, wherein each A and each T or each U residue comprises a modifying group.
7. The method of claim 2 , wherein each A and each U residue comprises a modifying group.
8. The method of claim 1 or 2, wherein each G and each C residue comprises a modifying group.
9. The method of claim 1, wherein the single-stranded DNA is obtained by (a) synthesizing double-stranded DNA using a single-stranded template, a DNA polymerase and nucleotides, wherein at least each A or each G residue and at least each C or each T residue comprises a modifying group bound to its respective base so that each type of nucleotide in the DNA has an electronic signature which is distinguishable from the electronic signature of each other type nucleotide in the DNA, and (b) removing from the resulting double-stranded DNA the single-stranded DNA containing modified nucleotides .
10. The method of claim 2, wherein the single-stranded RNA is obtained by (a) synthesizing double-stranded RNA using a single-stranded template, an RNA polymerase and nucleotides, wherein at least each A or each G residue and at least each C or each U residue comprises a modifying group bound to its respective base so that each type of nucleotide in the RNA has an electronic signature which is distinguishable from the electronic signature of each other type nucleotide in the RNA, and (b) removing from the resulting double- stranded RNA the single-stranded RNA containing modified nucleotides.
11. The method of claim 1, wherein the single-stranded DNA is obtained by (a) synthesizing double-stranded DNA using a single-stranded template, a DNA polymerase and nucleotides, wherein at least each A, each G, each C, each U or each T residue comprises an azido group bound to its base, and at least each A, each G, each C, each U and each T comprises an amino group bound to its base, whereby the azido and amino groups do not reside on the same type of base, (b) removing from the resulting double-stranded DNA the single-stranded DNA containing the azido and amino group-containing nucleotides and (c) reacting the resulting single- stranded DNA with a first modifying group which forms a bond with the azido group and a second modifying group which forms a bond with the amino group so as to obtain the single-stranded DNA.
12. The method of claim 2, wherein the single-stranded RNA is obtained by (a) synthesizing double-stranded RNA using a single-stranded template, an RNA polymerase and nucleotides, wherein at least each A, each G, each C or each U residue comprises an azido group bound to its base, and at least each A, each G, each C and each U comprises an amino group bound to its base, whereby the azido and amino groups do not reside on the same type of base, (b) removing from the resulting double- stranded RNA the single-stranded RNA containing the azido and amino group-containing nucleotides and (c) reacting the resulting single-stranded RNA with a first modifying group which forms a bond with the azido group and a second modifying group which forms a bond with the amino group so as to obtain the single- stranded RNA.
13. A nucleotide having an azido group covalently bound to its base.
14. The nucleotide of claim 13, wherein the nucleotide is dUTP and the azido group is bound to the base at the 5-position.
15. The nucleotide of claim 13, wherein the nucleotide is dATP and the azido group is bound to the base at the 8-position.
16. The nucleotide of claim 13, wherein the nucleotide is dGTP and the azido group is bound to the base at the 8-position.
17. A method for making a modified nucleotide comprising contacting the nucleotide of claim 13 with an alkyne- containing compound under conditions permitting reaction between the azido and the alJcyne groups, thereby making the modified nucleotide.
PCT/US2007/013559 2006-06-07 2007-06-07 Dna sequencing by nanopore using modified nucleotides WO2007146158A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US12/308,091 US8889348B2 (en) 2006-06-07 2007-06-07 DNA sequencing by nanopore using modified nucleotides
CN200780028545.1A CN101495656B (en) 2006-06-07 2007-06-07 DNA sequencing by nanopore using modified nucleotides
US14/516,785 US20150037788A1 (en) 2006-06-07 2014-10-17 Dna sequencing by nanopore using modified nucleotides
US15/255,029 US20170096704A1 (en) 2006-06-07 2016-09-01 Dna sequencing by nanopore using modified nucleotides
US16/218,175 US20190300947A1 (en) 2006-06-07 2018-12-12 Dna sequencing by nanopore using modified nucleotides

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US81191206P 2006-06-07 2006-06-07
US60/811,912 2006-06-07

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/308,091 A-371-Of-International US8889348B2 (en) 2006-06-07 2007-06-07 DNA sequencing by nanopore using modified nucleotides
US14/516,785 Continuation US20150037788A1 (en) 2006-06-07 2014-10-17 Dna sequencing by nanopore using modified nucleotides

Publications (1)

Publication Number Publication Date
WO2007146158A1 true WO2007146158A1 (en) 2007-12-21

Family

ID=38832077

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/013559 WO2007146158A1 (en) 2006-06-07 2007-06-07 Dna sequencing by nanopore using modified nucleotides

Country Status (3)

Country Link
US (4) US8889348B2 (en)
CN (1) CN101495656B (en)
WO (1) WO2007146158A1 (en)

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009007743A1 (en) * 2007-07-06 2009-01-15 Ucl Business Plc Nucleic acid detection method
GB2498021A (en) * 2011-09-07 2013-07-03 Univ Utah Res Found Detection of Nucleic acid lesions and adducts using nanopores
EP2652153A2 (en) * 2010-12-17 2013-10-23 The Trustees of Columbia University in the City of New York Dna sequencing by synthesis using modified nucleotides and nanopore detection
US20140051069A1 (en) * 2008-07-07 2014-02-20 Oxford Nanopore Technologies Limited Enzyme-pore constructs
US8845880B2 (en) 2010-12-22 2014-09-30 Genia Technologies, Inc. Nanopore-based single DNA molecule characterization, identification and isolation using speed bumps
US8889348B2 (en) 2006-06-07 2014-11-18 The Trustees Of Columbia University In The City Of New York DNA sequencing by nanopore using modified nucleotides
US8962242B2 (en) 2011-01-24 2015-02-24 Genia Technologies, Inc. System for detecting electrical properties of a molecular complex
US8986629B2 (en) 2012-02-27 2015-03-24 Genia Technologies, Inc. Sensor circuit for controlling, detecting, and measuring a molecular complex
US9005425B2 (en) 2010-03-05 2015-04-14 University Of Utah Research Foundation Detection of nucleic acid lesions and adducts using nanopores
US9041420B2 (en) 2010-02-08 2015-05-26 Genia Technologies, Inc. Systems and methods for characterizing a molecule
US9110478B2 (en) 2011-01-27 2015-08-18 Genia Technologies, Inc. Temperature regulation of measurement arrays
WO2015124935A1 (en) * 2014-02-21 2015-08-27 Oxford Nanopore Technologies Limited Sample preparation method
US9322062B2 (en) 2013-10-23 2016-04-26 Genia Technologies, Inc. Process for biosensor well formation
US9494554B2 (en) 2012-06-15 2016-11-15 Genia Technologies, Inc. Chip set-up and high-accuracy nucleic acid sequencing
US9551697B2 (en) 2013-10-17 2017-01-24 Genia Technologies, Inc. Non-faradaic, capacitively coupled measurement in a nanopore cell array
US9588079B2 (en) 2010-02-23 2017-03-07 University Of Washington Analyte sequencing with nanopores
US9605309B2 (en) 2012-11-09 2017-03-28 Genia Technologies, Inc. Nucleic acid sequencing using tags
US9605307B2 (en) 2010-02-08 2017-03-28 Genia Technologies, Inc. Systems and methods for forming a nanopore in a lipid bilayer
US9678055B2 (en) 2010-02-08 2017-06-13 Genia Technologies, Inc. Methods for forming a nanopore in a lipid bilayer
US9759711B2 (en) 2013-02-05 2017-09-12 Genia Technologies, Inc. Nanopore arrays
US9890426B2 (en) 2015-03-09 2018-02-13 The Trustees Of Columbia University In The City Of New York Pore-forming protein conjugate compositions and methods
US9957560B2 (en) 2011-07-25 2018-05-01 Oxford Nanopore Technologies Ltd. Hairpin loop method for double strand polynucleotide sequencing using transmembrane pores
WO2018069484A3 (en) * 2016-10-13 2018-05-24 F. Hoffmann-La Roche Ag Molecular detection and counting using nanopores
EP3274293A4 (en) * 2015-03-23 2018-08-22 The University of North Carolina at Chapel Hill Method for identification and enumeration of nucleic acid sequences, expression, splice variant, translocation, copy, or dna methylation changes using combined nuclease, ligase, polymerase, terminal transferase, and sequencing reactions
EP3415901A1 (en) 2012-01-20 2018-12-19 Genia Technologies, Inc. Nanopore based molecular detection and sequencing
US10221450B2 (en) 2013-03-08 2019-03-05 Oxford Nanopore Technologies Ltd. Enzyme stalling method
US10421995B2 (en) 2013-10-23 2019-09-24 Genia Technologies, Inc. High speed molecular sensing with nanopores
US10501767B2 (en) 2013-08-16 2019-12-10 Oxford Nanopore Technologies Ltd. Polynucleotide modification methods
KR20190140447A (en) * 2017-04-12 2019-12-19 에프. 호프만-라 로슈 아게 Methods for Labeling Aldehyde-containing Target Molecules
US10570440B2 (en) 2014-10-14 2020-02-25 Oxford Nanopore Technologies Ltd. Method for modifying a template double stranded polynucleotide using a MuA transposase
US11155860B2 (en) 2012-07-19 2021-10-26 Oxford Nanopore Technologies Ltd. SSB method
WO2022002251A1 (en) * 2020-07-02 2022-01-06 深圳今是科技有限公司 Nucleic acid sequencing method and apparatus
US11352664B2 (en) 2009-01-30 2022-06-07 Oxford Nanopore Technologies Plc Adaptors for nucleic acid constructs in transmembrane sequencing
US11649480B2 (en) 2016-05-25 2023-05-16 Oxford Nanopore Technologies Plc Method for modifying a template double stranded polynucleotide
US11725205B2 (en) 2018-05-14 2023-08-15 Oxford Nanopore Technologies Plc Methods and polynucleotides for amplifying a target polynucleotide

Families Citing this family (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9708358B2 (en) 2000-10-06 2017-07-18 The Trustees Of Columbia University In The City Of New York Massive parallel method for decoding DNA and RNA
AU2001296645A1 (en) 2000-10-06 2002-04-15 The Trustees Of Columbia University In The City Of New York Massive parallel method for decoding dna and rna
JP4107269B2 (en) * 2004-02-23 2008-06-25 ソニー株式会社 Solid-state imaging device
WO2007002204A2 (en) 2005-06-21 2007-01-04 The Trustees Of Columbia University In The City Of New York Pyrosequencing methods and related compostions
GB2446084B (en) 2005-10-31 2011-03-02 Univ Columbia Synthesis of four color 3-o-allyl modified photocleavable fluorescent nucleotides and related methods
WO2007053719A2 (en) 2005-10-31 2007-05-10 The Trustees Of Columbia University In The City Of New York Chemically cleavable 3'-o-allyl-dntp-allyl-fluorophore fluorescent nucleotide analogues and related methods
US7883869B2 (en) 2006-12-01 2011-02-08 The Trustees Of Columbia University In The City Of New York Four-color DNA sequencing by synthesis using cleavable fluorescent nucleotide reversible terminators
WO2008124706A2 (en) 2007-04-06 2008-10-16 Arizona Board Of Regents Acting For And On Behalf Of Arizona State University Devices and methods for target molecule characterization
EP2940029B1 (en) 2007-10-19 2023-11-29 The Trustees of Columbia University in the City of New York Design and synthesis of cleavable fluorescent nucleotides as reversible terminators for dna sequencing by synthesis
EP2725107B1 (en) 2007-10-19 2018-08-29 The Trustees of Columbia University in the City of New York DNA sequencing with non-fluorescent nucleotide reversible terminators and cleavable label modified ddNTPs and nucleic acid comprising inosine with reversible terminators
WO2010034018A2 (en) 2008-09-22 2010-03-25 University Of Washington Msp nanopores and related methods
US8968540B2 (en) 2008-10-06 2015-03-03 Arizona Board Of Regents, A Body Corporate Of The State Of Arizona Acting For And On Behalf Of Arizona State University Trans-base tunnel reader for sequencing
WO2010117470A2 (en) 2009-04-10 2010-10-14 Pacific Biosciences Of California, Inc. Nanopore sequencing devices and methods
US8926904B2 (en) 2009-05-12 2015-01-06 Daniel Wai-Cheong So Method and apparatus for the analysis and identification of molecules
EP2507387B1 (en) 2009-12-01 2017-01-25 Oxford Nanopore Technologies Limited Biochemical analysis instrument and method
KR20120045922A (en) * 2010-11-01 2012-05-09 엘지전자 주식회사 Apparatus for deterimining sequences of nucleic acids including nanopore
GB201021499D0 (en) 2010-12-20 2011-02-02 Loxbridge Res Llp Detection of quantative genetic differnces
WO2012162429A2 (en) 2011-05-23 2012-11-29 The Trustees Of Columbia University In The City Of New York Dna sequencing by synthesis using raman and infrared spectroscopy detection
EP2836604B1 (en) * 2012-04-09 2021-09-15 The Trustees of Columbia University in the City of New York Method of preparation of nanopore and uses thereof
EP3674412A1 (en) 2012-06-20 2020-07-01 The Trustees of Columbia University in the City of New York Nucleic acid sequencing by nanopore detection of tag molecules
US9274430B2 (en) 2012-10-10 2016-03-01 Arizona Board Of Regents On Behalf Of Arizona State University Systems and devices for molecule sensing and method of manufacturing thereof
EP2965073B1 (en) * 2013-03-05 2018-10-31 Arizona Board Of Regents Acting For And On Behalf Of State Arizona University Translocation of a polymer through a nanopore
US10648026B2 (en) 2013-03-15 2020-05-12 The Trustees Of Columbia University In The City Of New York Raman cluster tagged molecules for biological imaging
CN105102627B (en) 2013-03-15 2018-10-19 纽约哥伦比亚大学理事会 Method for detecting a variety of predetermined compounds in sample
WO2015031909A1 (en) * 2013-08-30 2015-03-05 University Of Washington Through Its Center For Commercialization Selective modification of polymer subunits to improve nanopore-based analysis
US10336713B2 (en) 2014-02-27 2019-07-02 Arizona Board Of Regents, Acting For And On Behalf Of, Arizona State University Triazole-based reader molecules and methods for synthesizing and use thereof
CA2943952A1 (en) 2014-03-24 2015-10-01 The Trustees Of Columbia University In The City Of New York Chemical methods for producing tagged nucleotides
EP3131920B1 (en) 2014-04-16 2021-09-01 The UAB Research Foundation Msp nanopores and uses thereof
DK3152320T3 (en) 2014-06-03 2021-01-11 Illumina Inc Compositions, systems and methods for detecting events using tethers anchored to or adjacent to nanopores
EP3253889A4 (en) 2015-02-02 2018-09-26 Two Pore Guys, Inc. Nanopore detection of target polynucleotides from sample background
ES2856691T3 (en) 2015-03-23 2021-09-28 Univ North Carolina Chapel Hill Universal molecular processor for precision medicine
IL293410B2 (en) 2015-06-03 2024-01-01 Illumina Inc Compositions, Systems, and Methods for Sequencing Polynucleotides using Tethers Anchored to Polymerases Adjacent to Nanopores
CN108779138B (en) 2015-09-28 2022-06-17 哥伦比亚大学董事会 Design and synthesis of nucleotides based on novel disulfide linkers for use as reversible terminators for DNA sequencing by synthesis
US11266673B2 (en) 2016-05-23 2022-03-08 The Trustees Of Columbia University In The City Of New York Nucleotide derivatives and methods of use thereof
WO2017202917A1 (en) 2016-05-27 2017-11-30 F. Hoffmann-La Roche Ag Tagged multi-nucleotides useful for nucleic acid sequencing
US10669580B2 (en) 2016-08-26 2020-06-02 Roche Sequencing Solutions, Inc. Tagged nucleotides useful for nanopore detection
WO2018067457A1 (en) 2016-10-03 2018-04-12 So Daniel Wai Cheong Method and apparatus for the analysis and identification of molecules
US10689684B2 (en) 2017-02-14 2020-06-23 Microsoft Technology Licensing, Llc Modifications to polynucleotides for sequencing
US10930370B2 (en) 2017-03-03 2021-02-23 Microsoft Technology Licensing, Llc Polynucleotide sequencer tuned to artificial polynucleotides
WO2018165207A1 (en) 2017-03-06 2018-09-13 Singular Genomic Systems, Inc. Nucleic acid sequencing-by-synthesis (sbs) methods that combine sbs cycle steps
WO2018183380A1 (en) * 2017-03-28 2018-10-04 Two Pore Guys, Inc. Target polynucleotide detection and sequencing by incorporation of modified nucleotides for nanopore analysis
WO2020086834A1 (en) 2018-10-25 2020-04-30 Singular Genomics Systems, Inc. Nucleotide analogues
US20220332752A2 (en) 2019-01-08 2022-10-20 Singular Genomics Systems, Inc. Nucleotide cleavable linkers and uses thereof
JP2023513128A (en) 2020-02-06 2023-03-30 エフ. ホフマン-ラ ロシュ アーゲー Compositions that reduce penetration of templates into nanopores

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030054360A1 (en) * 1999-01-19 2003-03-20 Larry Gold Method and apparatus for the automated generation of nucleic acid ligands
US20030166282A1 (en) * 2002-02-01 2003-09-04 David Brown High potency siRNAS for reducing the expression of target genes
US20030198982A1 (en) * 2000-08-03 2003-10-23 Frank Seela Nucleic acid binding compounds containing pyrazolo[3,4-d]pyrimidine analogues of purin-2,6-diamine and their uses
US20050239194A1 (en) * 2004-03-02 2005-10-27 Koji Takahashi Biosensor
US20060105461A1 (en) * 2004-10-22 2006-05-18 May Tom-Moy Nanopore analysis system

Family Cites Families (165)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4121192A (en) 1974-01-31 1978-10-17 Gte Sylvania Incorporated System and method for determining position and velocity of an intruder from an array of sensors
US4859945A (en) 1988-05-03 1989-08-22 Elscint Ltd. Optimized signal to noise ratio
US5198543A (en) * 1989-03-24 1993-03-30 Consejo Superior Investigaciones Cientificas PHI29 DNA polymerase
US5302509A (en) * 1989-08-14 1994-04-12 Beckman Instruments, Inc. Method for sequencing polynucleotides
WO1991006678A1 (en) 1989-10-26 1991-05-16 Sri International Dna sequencing
JPH04236295A (en) * 1991-01-18 1992-08-25 Sharp Corp Ferromagnetic liquid crystal composition and liquid crystal element
GB9208733D0 (en) 1992-04-22 1992-06-10 Medical Res Council Dna sequencing method
ATE143730T1 (en) * 1992-04-22 1996-10-15 Ecole Polytech LIPID MEMBRANES FOR BIOSENSOR APPLICATIONS
GB9315847D0 (en) * 1993-07-30 1993-09-15 Isis Innovation Tag reagent and assay method
US5457342A (en) 1994-03-30 1995-10-10 Herbst, Ii; Gerhardt G. Integrated circuit cooling apparatus
JP2986381B2 (en) 1994-08-16 1999-12-06 インターナショナル・ビジネス・マシーンズ・コーポレイション Electronic chip temperature control device and method
US5912155A (en) 1994-09-30 1999-06-15 Life Technologies, Inc. Cloned DNA polymerases from Thermotoga neapolitana
US6014213A (en) 1994-12-12 2000-01-11 Visible Genetics Inc. High dynamic range apparatus for separation and detection of polynucleotide fragments
US5795782A (en) 1995-03-17 1998-08-18 President & Fellows Of Harvard College Characterization of individual polymer molecules based on monomer-interface interactions
US6362002B1 (en) * 1995-03-17 2002-03-26 President And Fellows Of Harvard College Characterization of individual polymer molecules based on monomer-interface interactions
US20120160687A1 (en) * 1995-03-17 2012-06-28 President And Fellows Of Harvard College Characterization of individual polymer molecules based on monomer-interface interactions
US5780231A (en) 1995-11-17 1998-07-14 Lynx Therapeutics, Inc. DNA extension and analysis with rolling primers
US6261797B1 (en) * 1996-01-29 2001-07-17 Stratagene Primer-mediated polynucleotide synthesis and manipulation techniques
EP0923650B1 (en) 1996-06-06 2007-03-07 Solexa, Inc Sequencing by ligation of encoded adaptors
JPH10187253A (en) * 1996-12-27 1998-07-14 Ando Electric Co Ltd Temperature control device for optical semiconductor element
US5876936A (en) 1997-01-15 1999-03-02 Incyte Pharmaceuticals, Inc. Nucleic acid sequencing with solid phase capturable terminators
US6046005A (en) 1997-01-15 2000-04-04 Incyte Pharmaceuticals, Inc. Nucleic acid sequencing with solid phase capturable terminators comprising a cleavable linking group
US5804386A (en) 1997-01-15 1998-09-08 Incyte Pharmaceuticals, Inc. Sets of labeled energy transfer fluorescent primers and their use in multi component analysis
EP2202312B1 (en) * 1997-03-12 2016-01-06 Applied Biosystems, LLC DNA polymerases having improved labelled nucleotide incorporation properties
SE9703958D0 (en) 1997-10-29 1997-10-29 Pacesetter Ab Method and device for determination of concentration
EP0921196A1 (en) * 1997-12-02 1999-06-09 Roche Diagnostics GmbH Modified DNA-polymerase from carboxydothermus hydrogenoformans and its use for coupled reverse transcription and polymerase chain reaction
DE19810879A1 (en) * 1998-03-13 1999-09-16 Roche Diagnostics Gmbh New chimeric polymerase with 5'-3'-polymerase activity, and optionally proofreading activity, used for polymerase chain reactions and sequencing
AU3199699A (en) * 1998-03-23 1999-10-18 Invitrogen Corporation Modified nucleotides and methods useful for nucleic acid sequencing
US6485703B1 (en) 1998-07-31 2002-11-26 The Texas A&M University System Compositions and methods for analyte detection
US6210896B1 (en) * 1998-08-13 2001-04-03 Us Genomics Molecular motors
US6217731B1 (en) 1998-10-21 2001-04-17 Spectrumedix Corporation Method and apparatus for monitoring and displaying the status of a parallel capillary electrophoresis device
US6267872B1 (en) * 1998-11-06 2001-07-31 The Regents Of The University Of California Miniature support for thin films containing single channels or nanopores and methods for using same
JP2002532717A (en) * 1998-12-11 2002-10-02 サイミックス テクノロジーズ、インク Sensor array based system and method for rapid material characterization
DE69930310T3 (en) * 1998-12-14 2009-12-17 Pacific Biosciences of California, Inc. (n. d. Ges. d. Staates Delaware), Menlo Park KIT AND METHOD FOR THE NUCLEIC ACID SEQUENCING OF INDIVIDUAL MOLECULES BY POLYMERASE SYNTHESIS
US6082115A (en) * 1998-12-18 2000-07-04 National Semiconductor Corporation Temperature regulator circuit and precision voltage reference for integrated circuit
NO986133D0 (en) * 1998-12-23 1998-12-23 Preben Lexow Method of DNA Sequencing
US6399335B1 (en) * 1999-11-16 2002-06-04 Advanced Research And Technology Institute, Inc. γ-phosphoester nucleoside triphosphates
US6383749B2 (en) * 1999-12-02 2002-05-07 Clontech Laboratories, Inc. Methods of labeling nucleic acids for use in array based hybridization assays
US6916665B2 (en) * 2000-02-11 2005-07-12 The Texas A&M University System Biosensor compositions and methods of use
US6616895B2 (en) 2000-03-23 2003-09-09 Advanced Research Corporation Solid state membrane channel device for the measurement and characterization of atomic and molecular sized samples
US6413792B1 (en) * 2000-04-24 2002-07-02 Eagle Research Development, Llc Ultra-fast nucleic acid sequencing device and a method for making and using the same
US6936702B2 (en) 2000-06-07 2005-08-30 Li-Cor, Inc. Charge-switch nucleotides
EP1320596A4 (en) 2000-08-30 2005-01-12 Univ Rochester Method of performing reverse transcription reaction using reverse transcriptase encoded by non-ltr retrotransposable element
US20060057565A1 (en) 2000-09-11 2006-03-16 Jingyue Ju Combinatorial fluorescence energy transfer tags and uses thereof
US6627748B1 (en) 2000-09-11 2003-09-30 The Trustees Of Columbia University In The City Of New York Combinatorial fluorescence energy transfer tags and their applications for multiplex genetic analyses
EP1322785A4 (en) 2000-09-11 2005-11-09 Univ Columbia Combinatorial fluorescence energy transfer tags and uses thereof
AU2001296645A1 (en) * 2000-10-06 2002-04-15 The Trustees Of Columbia University In The City Of New York Massive parallel method for decoding dna and rna
US20030027140A1 (en) 2001-03-30 2003-02-06 Jingyue Ju High-fidelity DNA sequencing using solid phase capturable dideoxynucleotides and mass spectrometry
US6800933B1 (en) 2001-04-23 2004-10-05 Advanced Micro Devices, Inc. Integrated circuit cooling device
US6686997B1 (en) * 2001-08-27 2004-02-03 Raytheon Company Apparatus and a method for pulse detection and characterization
US7256019B2 (en) 2001-08-29 2007-08-14 Ge Healthcare Bio-Sciences Corp. Terminal phosphate blocked nucleoside polyphosphates
US7033762B2 (en) * 2001-08-29 2006-04-25 Amersham Biosciences Corp Single nucleotide amplification and detection by polymerase
US7041812B2 (en) * 2001-08-29 2006-05-09 Amersham Biosciences Corp Labeled nucleoside polyphosphates
US7223541B2 (en) * 2001-08-29 2007-05-29 Ge Healthcare Bio-Sciences Corp. Terminal-phosphate-labeled nucleotides and methods of use
US7727722B2 (en) * 2001-08-29 2010-06-01 General Electric Company Ligation amplification
US7052839B2 (en) * 2001-08-29 2006-05-30 Amersham Biosciences Corp Terminal-phosphate-labeled nucleotides and methods of use
CA2460546A1 (en) * 2001-09-14 2003-03-27 Invitrogen Corporation Dna polymerases and mutants thereof
US7057026B2 (en) * 2001-12-04 2006-06-06 Solexa Limited Labelled nucleotides
US6952651B2 (en) 2002-06-17 2005-10-04 Intel Corporation Methods and apparatus for nucleic acid sequencing by signal stretching and data integration
US7074597B2 (en) 2002-07-12 2006-07-11 The Trustees Of Columbia University In The City Of New York Multiplex genotyping using solid phase capturable dideoxynucleotides and mass spectrometry
DE10238843B8 (en) * 2002-08-20 2008-01-03 Infineon Technologies Ag Semiconductor component
US7373199B2 (en) * 2002-08-27 2008-05-13 University Of Florida Research Foundation, Inc. Optimization of multi-dimensional time series processing for seizure warning and prediction
US7355216B2 (en) 2002-12-09 2008-04-08 The Regents Of The University Of California Fluidic nanotubes and devices
AU2003297859A1 (en) 2002-12-13 2004-07-09 The Trustees Of Columbia University In The City Of New York Biomolecular coupling methods using 1,3-dipolar cycloaddition chemistry
EP1592779A4 (en) 2003-02-05 2007-12-12 Ge Healthcare Bio Sciences Terminal-phosphate-labeled nucleotides with new linkers
AU2004211920B2 (en) 2003-02-05 2009-05-14 Global Life Sciences Solutions Usa Llc Solid phase sequencing
US7745116B2 (en) * 2003-04-08 2010-06-29 Pacific Biosciences Of California, Inc. Composition and method for nucleic acid sequencing
GB0308851D0 (en) * 2003-04-16 2003-05-21 Lingvitae As Method
US8233959B2 (en) 2003-08-22 2012-07-31 Dexcom, Inc. Systems and methods for processing analyte sensor data
US6880345B1 (en) * 2003-11-04 2005-04-19 Intel Corporation Cooling system for an electronic component
US7019346B2 (en) 2003-12-23 2006-03-28 Intel Corporation Capacitor having an anodic metal oxide substrate
US20050186576A1 (en) 2004-02-19 2005-08-25 Intel Corporation Polymer sequencing using selectively labeled monomers and data integration
JP4107269B2 (en) * 2004-02-23 2008-06-25 ソニー株式会社 Solid-state imaging device
EP2436778A3 (en) 2004-03-03 2012-07-11 The Trustees of Columbia University in the City of New York Photocleavable fluorescent nucleotides for DNA sequencing on chip constructed by site-specific coupling chemistry
US7279337B2 (en) 2004-03-10 2007-10-09 Agilent Technologies, Inc. Method and apparatus for sequencing polymers through tunneling conductance variation detection
WO2006028508A2 (en) * 2004-03-23 2006-03-16 President And Fellows Of Harvard College Methods and apparatus for characterizing polynucleotides
US20050221351A1 (en) 2004-04-06 2005-10-06 Affymetrix, Inc. Methods and devices for microarray image analysis
US20050239134A1 (en) 2004-04-21 2005-10-27 Board Of Regents, The University Of Texas System Combinatorial selection of phosphorothioate single-stranded DNA aptamers for TGF-beta-1 protein
US6880346B1 (en) * 2004-07-08 2005-04-19 Giga-Byte Technology Co., Ltd. Two stage radiation thermoelectric cooling apparatus
AT501510B1 (en) * 2004-07-19 2009-05-15 Univ Wien Tech DECENTRALIZED ERROR TOLERANT CLOCK GENERATION IN VLSI CHIPS
GB2431013B (en) 2004-07-23 2008-05-21 Electronic Bio Sciences Llc Method and apparatus for sensing a time varying current passing through an ion channel
AU2005272823B2 (en) * 2004-08-13 2012-04-12 President And Fellows Of Harvard College An ultra high-throughput opti-nanopore DNA readout platform
GB0422733D0 (en) 2004-10-13 2004-11-17 Lingvitae As Method
US7867716B2 (en) * 2004-12-21 2011-01-11 The Texas A&M University System High temperature ion channels and pores
GB0505971D0 (en) * 2005-03-23 2005-04-27 Isis Innovation Delivery of molecules to a lipid bilayer
WO2007002204A2 (en) 2005-06-21 2007-01-04 The Trustees Of Columbia University In The City Of New York Pyrosequencing methods and related compostions
US7645866B2 (en) 2005-06-28 2010-01-12 Life Technologies Corporation Methods of producing and sequencing modified polynucleotides
JP4524652B2 (en) * 2005-07-06 2010-08-18 ソニー株式会社 AD converter and semiconductor device
US7405281B2 (en) 2005-09-29 2008-07-29 Pacific Biosciences Of California, Inc. Fluorescent nucleotide analogs and uses therefor
US20070190542A1 (en) 2005-10-03 2007-08-16 Ling Xinsheng S Hybridization assisted nanopore sequencing
US7397232B2 (en) 2005-10-21 2008-07-08 The University Of Akron Coulter counter having a plurality of channels
GB2446084B (en) * 2005-10-31 2011-03-02 Univ Columbia Synthesis of four color 3-o-allyl modified photocleavable fluorescent nucleotides and related methods
WO2007053719A2 (en) 2005-10-31 2007-05-10 The Trustees Of Columbia University In The City Of New York Chemically cleavable 3'-o-allyl-dntp-allyl-fluorophore fluorescent nucleotide analogues and related methods
US20090088332A1 (en) 2005-11-21 2009-04-02 Jingyue Ju Multiplex Digital Immuno-Sensing Using a Library of Photocleavable Mass Tags
WO2007070572A2 (en) * 2005-12-12 2007-06-21 The Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services Probe for nucleic acid sequencing and methods of use
ATE518010T1 (en) 2005-12-22 2011-08-15 Pacific Biosciences California ACTIVE SURFACE COUPLED POLYMERASES
ES2648313T3 (en) * 2005-12-22 2017-12-29 Pacific Biosciences Of California, Inc. Polymerases for the incorporation of nucleotide analogs
WO2007087625A2 (en) * 2006-01-26 2007-08-02 Euliano Neil R Breath and breath condensate analysis system and associated methods
US7368668B2 (en) 2006-02-03 2008-05-06 Freescale Semiconductor Inc. Ground shields for semiconductors
US20070298511A1 (en) 2006-04-27 2007-12-27 The Texas A&M University System Nanopore sensor system
US7777505B2 (en) 2006-05-05 2010-08-17 University Of Utah Research Foundation Nanopore platforms for ion channel recordings and single molecule detection and analysis
US7446017B2 (en) 2006-05-31 2008-11-04 Freescale Semiconductor, Inc. Methods and apparatus for RF shielding in vertically-integrated semiconductor devices
WO2007146158A1 (en) 2006-06-07 2007-12-21 The Trustees Of Columbia University In The City Of New York Dna sequencing by nanopore using modified nucleotides
BRPI0603825A (en) 2006-09-19 2008-05-06 Syngenta Protecao De Cultivos sprout growth regulator composition in tobacco plantations, method of controlling sprout growth in tobacco plantations and use of an effective amount of flumetralin, at least one solvent and at least one surfactant
US8343746B2 (en) * 2006-10-23 2013-01-01 Pacific Biosciences Of California, Inc. Polymerase enzymes and reagents for enhanced nucleic acid sequencing
US7883869B2 (en) 2006-12-01 2011-02-08 The Trustees Of Columbia University In The City Of New York Four-color DNA sequencing by synthesis using cleavable fluorescent nucleotide reversible terminators
US7902345B2 (en) * 2006-12-05 2011-03-08 Sequenom, Inc. Detection and quantification of biomolecules using mass spectrometry
CA2672315A1 (en) * 2006-12-14 2008-06-26 Ion Torrent Systems Incorporated Methods and apparatus for measuring analytes using large scale fet arrays
US8003319B2 (en) * 2007-02-02 2011-08-23 International Business Machines Corporation Systems and methods for controlling position of charged polymer inside nanopore
US20100196203A1 (en) 2007-02-20 2010-08-05 Gurdial Singh Sanghera Formation of Lipid Bilayers
US20110005918A1 (en) * 2007-04-04 2011-01-13 Akeson Mark A Compositions, devices, systems, and methods for using a nanopore
ES2327886B1 (en) 2007-04-19 2010-08-30 Universitat Politecnica De Catalunya FLEXIBLE ROD FOR AERODYNAMIC PLATFORM SCALE.
US9121843B2 (en) 2007-05-08 2015-09-01 Trustees Of Boston University Chemical functionalization of solid-state nanopores and nanopore arrays and applications thereof
US7939259B2 (en) * 2007-06-19 2011-05-10 Stratos Genomics, Inc. High throughput nucleic acid sequencing by expansion
US20090073293A1 (en) * 2007-06-27 2009-03-19 Yoel Yaffe CMOS image sensors with increased dynamic range and methods of operating the same
JP4929090B2 (en) * 2007-07-26 2012-05-09 パナソニック株式会社 Solid-state imaging device and driving method thereof
EP2201136B1 (en) * 2007-10-01 2017-12-06 Nabsys 2.0 LLC Nanopore sequencing by hybridization of probes to form ternary complexes and variable range alignment
JP5309145B2 (en) * 2007-10-02 2013-10-09 プレジデント アンド フェロウズ オブ ハーバード カレッジ Molecular capture, recapture and trapping by nanopores
EP2725107B1 (en) * 2007-10-19 2018-08-29 The Trustees of Columbia University in the City of New York DNA sequencing with non-fluorescent nucleotide reversible terminators and cleavable label modified ddNTPs and nucleic acid comprising inosine with reversible terminators
EP2940029B1 (en) * 2007-10-19 2023-11-29 The Trustees of Columbia University in the City of New York Design and synthesis of cleavable fluorescent nucleotides as reversible terminators for dna sequencing by synthesis
US8592182B2 (en) 2007-10-23 2013-11-26 Stratos Genomics Inc. High throughput nucleic acid sequencing by spacing
GB0724736D0 (en) 2007-12-19 2008-01-30 Oxford Nanolabs Ltd Formation of layers of amphiphilic molecules
US20110165652A1 (en) * 2008-01-14 2011-07-07 Life Technologies Corporation Compositions, methods and systems for single molecule sequencing
US7989928B2 (en) 2008-02-05 2011-08-02 Advanced Semiconductor Engineering Inc. Semiconductor device packages with electromagnetic interference shielding
US8022511B2 (en) 2008-02-05 2011-09-20 Advanced Semiconductor Engineering, Inc. Semiconductor device packages with electromagnetic interference shielding
US8252911B2 (en) 2008-02-12 2012-08-28 Pacific Biosciences Of California, Inc. Compositions and methods for use in analytical reactions
US8652781B2 (en) 2008-02-12 2014-02-18 Pacific Biosciences Of California, Inc. Cognate sampling kinetics
US20090215050A1 (en) 2008-02-22 2009-08-27 Robert Delmar Jenison Systems and methods for point-of-care amplification and detection of polynucleotides
US7973146B2 (en) * 2008-03-26 2011-07-05 Pacific Biosciences Of California, Inc. Engineered fluorescent dye labeled nucleotide analogs for DNA sequencing
US8420366B2 (en) 2008-03-31 2013-04-16 Pacific Biosciences Of California, Inc. Generation of modified polymerases for improved accuracy in single molecule sequencing
ATE530497T1 (en) * 2008-03-31 2011-11-15 Sony Deutschland Gmbh METHOD FOR PRODUCING A MEMBRANE WITH A CONICAL PORE
US9127259B2 (en) * 2008-03-31 2015-09-08 Pacific Biosciences Of California, Inc. Enzymes resistant to photodamage
EP2274446B1 (en) * 2008-03-31 2015-09-09 Pacific Biosciences of California, Inc. Two slow-step polymerase enzyme systems and methods
US8999676B2 (en) * 2008-03-31 2015-04-07 Pacific Biosciences Of California, Inc. Recombinant polymerases for improved single molecule sequencing
ES2614078T3 (en) 2008-03-31 2017-05-29 Pacific Biosciences Of California, Inc. Generation of modified polymerases for improved accuracy in single molecule sequencing
WO2009134469A1 (en) 2008-04-29 2009-11-05 Life Technologies Unnatural polymerase substrates that can sustain enzymatic synthesis of double stranded nucleic acids from a nucleic acid template and methods of use
US8940142B2 (en) 2008-05-05 2015-01-27 The Regents Of The University Of California Functionalized nanopipette biosensor
US7906371B2 (en) 2008-05-28 2011-03-15 Stats Chippac, Ltd. Semiconductor device and method of forming holes in substrate to interconnect top shield and ground shield
US20100025238A1 (en) 2008-07-31 2010-02-04 Medtronic Minimed, Inc. Analyte sensor apparatuses having improved electrode configurations and methods for making and using them
AU2009288696A1 (en) * 2008-09-05 2010-03-11 Pacific Biosciences Of California, Inc. Sequencing by cognate sampling
US8481264B2 (en) * 2008-09-19 2013-07-09 Pacific Biosciences Of California, Inc. Immobilized nucleic acid complexes for sequence analysis
US8921046B2 (en) * 2008-09-19 2014-12-30 Pacific Biosciences Of California, Inc. Nucleic acid sequence analysis
US8063469B2 (en) 2008-09-30 2011-11-22 Infineon Technologies Ag On-chip radio frequency shield with interconnect metallization
US20100301398A1 (en) 2009-05-29 2010-12-02 Ion Torrent Systems Incorporated Methods and apparatus for measuring analytes
US20100148126A1 (en) * 2008-11-26 2010-06-17 Board Of Regents, The University Of Texas System Genomic sequencing using modified protein pores and ionic liquids
GB0905140D0 (en) 2009-03-25 2009-05-06 Isis Innovation Method
WO2010111605A2 (en) 2009-03-27 2010-09-30 Nabsys, Inc. Devices and methods for analyzing biomolecules and probes bound thereto
WO2010117470A2 (en) 2009-04-10 2010-10-14 Pacific Biosciences Of California, Inc. Nanopore sequencing devices and methods
US20130053252A1 (en) 2009-09-25 2013-02-28 President & Fellows Of Harvard College Nucleic acid amplification and sequencing by synthesis with fluorogenic nucleotides
WO2011040996A1 (en) 2009-09-30 2011-04-07 Quantapore, Inc. Ultrafast sequencing of biological polymers using a labeled nanopore
US20110192723A1 (en) 2010-02-08 2011-08-11 Genia Technologies, Inc. Systems and methods for manipulating a molecule in a nanopore
US9605307B2 (en) 2010-02-08 2017-03-28 Genia Technologies, Inc. Systems and methods for forming a nanopore in a lipid bilayer
US8324914B2 (en) 2010-02-08 2012-12-04 Genia Technologies, Inc. Systems and methods for characterizing a molecule
US20120052188A1 (en) * 2010-02-08 2012-03-01 Genia Technologies, Inc. Systems and methods for assembling a lipid bilayer on a substantially planar solid surface
CN105273991B (en) 2010-02-08 2019-05-10 吉尼亚科技公司 System and method for operating molecule in nano-pore
US20110287414A1 (en) 2010-02-08 2011-11-24 Genia Technologies, Inc. Systems and methods for identifying a portion of a molecule
DK2539707T3 (en) 2010-02-23 2021-07-26 Univ Washington Artificial mycolic acid membranes
WO2012009578A2 (en) 2010-07-14 2012-01-19 The Curators Of The University Of Missouri Nanopore-facilitated single molecule detection of nucleic acids
CN103282518B (en) 2010-12-17 2016-11-16 纽约哥伦比亚大学理事会 Use synthesis limit, the DNA limit order-checking of modified nucleotide and nano-pore detection
US8845880B2 (en) * 2010-12-22 2014-09-30 Genia Technologies, Inc. Nanopore-based single DNA molecule characterization, identification and isolation using speed bumps
US8962242B2 (en) * 2011-01-24 2015-02-24 Genia Technologies, Inc. System for detecting electrical properties of a molecular complex
US9110478B2 (en) 2011-01-27 2015-08-18 Genia Technologies, Inc. Temperature regulation of measurement arrays
WO2012121756A1 (en) 2011-03-04 2012-09-13 Quantapore, Inc. Apparatus and methods for performing optical nanopore detection or sequencing
EP2836604B1 (en) 2012-04-09 2021-09-15 The Trustees of Columbia University in the City of New York Method of preparation of nanopore and uses thereof
EP3674412A1 (en) 2012-06-20 2020-07-01 The Trustees of Columbia University in the City of New York Nucleic acid sequencing by nanopore detection of tag molecules

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030054360A1 (en) * 1999-01-19 2003-03-20 Larry Gold Method and apparatus for the automated generation of nucleic acid ligands
US20030198982A1 (en) * 2000-08-03 2003-10-23 Frank Seela Nucleic acid binding compounds containing pyrazolo[3,4-d]pyrimidine analogues of purin-2,6-diamine and their uses
US20030166282A1 (en) * 2002-02-01 2003-09-04 David Brown High potency siRNAS for reducing the expression of target genes
US20050239194A1 (en) * 2004-03-02 2005-10-27 Koji Takahashi Biosensor
US20060105461A1 (en) * 2004-10-22 2006-05-18 May Tom-Moy Nanopore analysis system

Cited By (78)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8889348B2 (en) 2006-06-07 2014-11-18 The Trustees Of Columbia University In The City Of New York DNA sequencing by nanopore using modified nucleotides
WO2009007743A1 (en) * 2007-07-06 2009-01-15 Ucl Business Plc Nucleic acid detection method
US9885078B2 (en) 2008-07-07 2018-02-06 Oxford Nanopore Technologies Limited Enzyme-pore constructs
US11859247B2 (en) 2008-07-07 2024-01-02 Oxford Nanopore Technologies Plc Enzyme-pore constructs
US20140051069A1 (en) * 2008-07-07 2014-02-20 Oxford Nanopore Technologies Limited Enzyme-pore constructs
US11078530B2 (en) 2008-07-07 2021-08-03 Oxford Nanopore Technologies Ltd. Enzyme-pore constructs
US10077471B2 (en) 2008-07-07 2018-09-18 Oxford Nanopore Technologies Ltd. Enzyme-pore constructs
US11459606B2 (en) 2009-01-30 2022-10-04 Oxford Nanopore Technologies Plc Adaptors for nucleic acid constructs in transmembrane sequencing
US11352664B2 (en) 2009-01-30 2022-06-07 Oxford Nanopore Technologies Plc Adaptors for nucleic acid constructs in transmembrane sequencing
US11027502B2 (en) 2010-02-08 2021-06-08 Roche Sequencing Solutions, Inc. Systems and methods for forming a nanopore in a lipid bilayer
US9678055B2 (en) 2010-02-08 2017-06-13 Genia Technologies, Inc. Methods for forming a nanopore in a lipid bilayer
US10343350B2 (en) 2010-02-08 2019-07-09 Genia Technologies, Inc. Systems and methods for forming a nanopore in a lipid bilayer
US9041420B2 (en) 2010-02-08 2015-05-26 Genia Technologies, Inc. Systems and methods for characterizing a molecule
US9605307B2 (en) 2010-02-08 2017-03-28 Genia Technologies, Inc. Systems and methods for forming a nanopore in a lipid bilayer
US10926486B2 (en) 2010-02-08 2021-02-23 Roche Sequencing Solutions, Inc. Systems and methods for forming a nanopore in a lipid bilayer
US9377437B2 (en) 2010-02-08 2016-06-28 Genia Technologies, Inc. Systems and methods for characterizing a molecule
US10371692B2 (en) 2010-02-08 2019-08-06 Genia Technologies, Inc. Systems for forming a nanopore in a lipid bilayer
US11913905B2 (en) 2010-02-23 2024-02-27 University Of Washington Analyte sequencing with nanopores
US9588079B2 (en) 2010-02-23 2017-03-07 University Of Washington Analyte sequencing with nanopores
US11187675B2 (en) 2010-02-23 2021-11-30 University Of Washington Analyte sequencing with nanopores
US9429561B2 (en) 2010-03-05 2016-08-30 University Of Utah Research Foundation Detection of nucleic acid lesions and adducts using nanopores
US9005425B2 (en) 2010-03-05 2015-04-14 University Of Utah Research Foundation Detection of nucleic acid lesions and adducts using nanopores
US10443096B2 (en) 2010-12-17 2019-10-15 The Trustees Of Columbia University In The City Of New York DNA sequencing by synthesis using modified nucleotides and nanopore detection
US11499186B2 (en) 2010-12-17 2022-11-15 The Trustees Of Columbia University In The City Of New York DNA sequencing by synthesis using modified nucleotides and nanopore detection
EP2652153A4 (en) * 2010-12-17 2014-05-14 Univ Columbia Dna sequencing by synthesis using modified nucleotides and nanopore detection
EP2652153A2 (en) * 2010-12-17 2013-10-23 The Trustees of Columbia University in the City of New York Dna sequencing by synthesis using modified nucleotides and nanopore detection
US10400278B2 (en) 2010-12-22 2019-09-03 Genia Technologies, Inc. Nanopore-based single DNA molecule characterization, identification and isolation using speed bumps
US9617593B2 (en) 2010-12-22 2017-04-11 Genia Technologies, Inc. Nanopore-based single DNA molecule characterization, identification and isolation using speed bumps
US9121059B2 (en) 2010-12-22 2015-09-01 Genia Technologies, Inc. Nanopore-based single molecule characterization
US10920271B2 (en) 2010-12-22 2021-02-16 Roche Sequencing Solutions, Inc. Nanopore-based single DNA molecule characterization, identification and isolation using speed bumps
US8845880B2 (en) 2010-12-22 2014-09-30 Genia Technologies, Inc. Nanopore-based single DNA molecule characterization, identification and isolation using speed bumps
US9581563B2 (en) 2011-01-24 2017-02-28 Genia Technologies, Inc. System for communicating information from an array of sensors
US8962242B2 (en) 2011-01-24 2015-02-24 Genia Technologies, Inc. System for detecting electrical properties of a molecular complex
US10156541B2 (en) 2011-01-24 2018-12-18 Genia Technologies, Inc. System for detecting electrical properties of a molecular complex
US9110478B2 (en) 2011-01-27 2015-08-18 Genia Technologies, Inc. Temperature regulation of measurement arrays
US10010852B2 (en) 2011-01-27 2018-07-03 Genia Technologies, Inc. Temperature regulation of measurement arrays
US11261487B2 (en) 2011-07-25 2022-03-01 Oxford Nanopore Technologies Plc Hairpin loop method for double strand polynucleotide sequencing using transmembrane pores
US10851409B2 (en) 2011-07-25 2020-12-01 Oxford Nanopore Technologies Ltd. Hairpin loop method for double strand polynucleotide sequencing using transmembrane pores
US9957560B2 (en) 2011-07-25 2018-05-01 Oxford Nanopore Technologies Ltd. Hairpin loop method for double strand polynucleotide sequencing using transmembrane pores
US10597713B2 (en) 2011-07-25 2020-03-24 Oxford Nanopore Technologies Ltd. Hairpin loop method for double strand polynucleotide sequencing using transmembrane pores
US11168363B2 (en) 2011-07-25 2021-11-09 Oxford Nanopore Technologies Ltd. Hairpin loop method for double strand polynucleotide sequencing using transmembrane pores
GB2498021A (en) * 2011-09-07 2013-07-03 Univ Utah Res Found Detection of Nucleic acid lesions and adducts using nanopores
EP3415901A1 (en) 2012-01-20 2018-12-19 Genia Technologies, Inc. Nanopore based molecular detection and sequencing
US11275052B2 (en) 2012-02-27 2022-03-15 Roche Sequencing Solutions, Inc. Sensor circuit for controlling, detecting, and measuring a molecular complex
US8986629B2 (en) 2012-02-27 2015-03-24 Genia Technologies, Inc. Sensor circuit for controlling, detecting, and measuring a molecular complex
US9494554B2 (en) 2012-06-15 2016-11-15 Genia Technologies, Inc. Chip set-up and high-accuracy nucleic acid sequencing
US11155860B2 (en) 2012-07-19 2021-10-26 Oxford Nanopore Technologies Ltd. SSB method
US11674174B2 (en) 2012-11-09 2023-06-13 The Trustees Of Columbia University In The City Of New York Nucleic acid sequences using tags
US10526647B2 (en) 2012-11-09 2020-01-07 The Trustees Of Columbia University In The City Of New York Nucleic acid sequences using tags
US10822650B2 (en) 2012-11-09 2020-11-03 Roche Sequencing Solutions, Inc. Nucleic acid sequencing using tags
US9605309B2 (en) 2012-11-09 2017-03-28 Genia Technologies, Inc. Nucleic acid sequencing using tags
US9759711B2 (en) 2013-02-05 2017-09-12 Genia Technologies, Inc. Nanopore arrays
US10012637B2 (en) 2013-02-05 2018-07-03 Genia Technologies, Inc. Nanopore arrays
US10809244B2 (en) 2013-02-05 2020-10-20 Roche Sequencing Solutions, Inc. Nanopore arrays
US11560589B2 (en) 2013-03-08 2023-01-24 Oxford Nanopore Technologies Plc Enzyme stalling method
US10221450B2 (en) 2013-03-08 2019-03-05 Oxford Nanopore Technologies Ltd. Enzyme stalling method
US10501767B2 (en) 2013-08-16 2019-12-10 Oxford Nanopore Technologies Ltd. Polynucleotide modification methods
US11186857B2 (en) 2013-08-16 2021-11-30 Oxford Nanopore Technologies Plc Polynucleotide modification methods
US9551697B2 (en) 2013-10-17 2017-01-24 Genia Technologies, Inc. Non-faradaic, capacitively coupled measurement in a nanopore cell array
US10393700B2 (en) 2013-10-17 2019-08-27 Roche Sequencing Solutions, Inc. Non-faradaic, capacitively coupled measurement in a nanopore cell array
US9322062B2 (en) 2013-10-23 2016-04-26 Genia Technologies, Inc. Process for biosensor well formation
US11021745B2 (en) 2013-10-23 2021-06-01 Roche Sequencing Solutions, Inc. Methods for forming lipid bilayers on biochips
US10421995B2 (en) 2013-10-23 2019-09-24 Genia Technologies, Inc. High speed molecular sensing with nanopores
US9567630B2 (en) 2013-10-23 2017-02-14 Genia Technologies, Inc. Methods for forming lipid bilayers on biochips
US11542551B2 (en) 2014-02-21 2023-01-03 Oxford Nanopore Technologies Plc Sample preparation method
WO2015124935A1 (en) * 2014-02-21 2015-08-27 Oxford Nanopore Technologies Limited Sample preparation method
US10669578B2 (en) 2014-02-21 2020-06-02 Oxford Nanopore Technologies Ltd. Sample preparation method
US10570440B2 (en) 2014-10-14 2020-02-25 Oxford Nanopore Technologies Ltd. Method for modifying a template double stranded polynucleotide using a MuA transposase
US11390904B2 (en) 2014-10-14 2022-07-19 Oxford Nanopore Technologies Plc Nanopore-based method and double stranded nucleic acid construct therefor
US9890426B2 (en) 2015-03-09 2018-02-13 The Trustees Of Columbia University In The City Of New York Pore-forming protein conjugate compositions and methods
EP3274293A4 (en) * 2015-03-23 2018-08-22 The University of North Carolina at Chapel Hill Method for identification and enumeration of nucleic acid sequences, expression, splice variant, translocation, copy, or dna methylation changes using combined nuclease, ligase, polymerase, terminal transferase, and sequencing reactions
US11649480B2 (en) 2016-05-25 2023-05-16 Oxford Nanopore Technologies Plc Method for modifying a template double stranded polynucleotide
US11041845B2 (en) 2016-10-13 2021-06-22 Roche Sequencing Solutions, Inc. Molecular detection and counting using nanopores
WO2018069484A3 (en) * 2016-10-13 2018-05-24 F. Hoffmann-La Roche Ag Molecular detection and counting using nanopores
KR20190140447A (en) * 2017-04-12 2019-12-19 에프. 호프만-라 로슈 아게 Methods for Labeling Aldehyde-containing Target Molecules
KR102495650B1 (en) 2017-04-12 2023-02-02 에프. 호프만-라 로슈 아게 Methods for Labeling Aldehyde-Containing Target Molecules
US11725205B2 (en) 2018-05-14 2023-08-15 Oxford Nanopore Technologies Plc Methods and polynucleotides for amplifying a target polynucleotide
WO2022002251A1 (en) * 2020-07-02 2022-01-06 深圳今是科技有限公司 Nucleic acid sequencing method and apparatus

Also Published As

Publication number Publication date
CN101495656B (en) 2017-02-08
CN101495656A (en) 2009-07-29
US8889348B2 (en) 2014-11-18
US20170096704A1 (en) 2017-04-06
US20150037788A1 (en) 2015-02-05
US20090298072A1 (en) 2009-12-03
US20190300947A1 (en) 2019-10-03

Similar Documents

Publication Publication Date Title
US20190300947A1 (en) Dna sequencing by nanopore using modified nucleotides
US10273527B2 (en) Nanopore-facilitated single molecule detection of nucleic acids
US20230392157A1 (en) Aptamer method
CA2906007C (en) Encoded nanopore sensor for multiplex nucleic acids detection
EP3380607B1 (en) Purification of polymerase complexes
CN114457145A (en) Linkers, constructs, methods, and uses for characterizing sequencing of target polynucleotides
US20180164280A1 (en) Modified nucleic acids for nanopore analysis
EP4177331A1 (en) Nucleic acid sequencing method and apparatus

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200780028545.1

Country of ref document: CN

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

Ref document number: 07777449

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 12308091

Country of ref document: US

122 Ep: pct application non-entry in european phase

Ref document number: 07777449

Country of ref document: EP

Kind code of ref document: A1