US20070020653A1 - DNA polymerase - Google Patents

DNA polymerase Download PDF

Info

Publication number
US20070020653A1
US20070020653A1 US11/417,403 US41740306A US2007020653A1 US 20070020653 A1 US20070020653 A1 US 20070020653A1 US 41740306 A US41740306 A US 41740306A US 2007020653 A1 US2007020653 A1 US 2007020653A1
Authority
US
United States
Prior art keywords
polymerase
dna polymerase
pol
dna
engineered
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/417,403
Other languages
English (en)
Inventor
Philipp Holliger
Farid Ghadessy
Marc D'Abbadie
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Medical Research Council
Original Assignee
Medical Research Council
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
Priority claimed from GB0325650A external-priority patent/GB0325650D0/en
Priority claimed from GB0410871A external-priority patent/GB0410871D0/en
Application filed by Medical Research Council filed Critical Medical Research Council
Assigned to MEDICAL RESEARCH COUNCIL reassignment MEDICAL RESEARCH COUNCIL ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HOLLIGER, PHILIPP, GHADESSY, FARID, D'ABBADIE, MARCV
Publication of US20070020653A1 publication Critical patent/US20070020653A1/en
Priority to US12/538,392 priority Critical patent/US20090305292A1/en
Priority to US13/527,342 priority patent/US9096835B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • C12N9/1252DNA-directed DNA polymerase (2.7.7.7), i.e. DNA replicase

Definitions

  • the present invention relates to DNA polymerases.
  • the invention relates to a method for the generation of DNA polymerases which exhibit a relaxed substrate specificity. Uses of engineered polymerases produced using the methods of the invention are also described.
  • Some other naturally occurring polymerases are less stringent with regard to their substrate specificity.
  • viral reverse transcriptases like HIV-1 reverse transcriptase or AMV reverse transcriptase and polymerases capable of translesion synthesis such as polY-family polymerases, pol X (Vaisman et al, 2001, JBC) or pol X (Washington (2002), PNAS; or the unusual polB-family polymerase pol X (Johnson, Nature), all extend 3′ mismatches with elevated efficiency compared to high fidelity polymerases.
  • the disadvantage of the use of translesion synthesis polymerases for biotechnological uses is that they depend on cellular processivity factors for their activity, such as PCNA. Moreover such polymerases are not stable at the temperatures at which certain biotechnological techniques are performed, such as PCR. Furthermore most Translesion synthesis polymerases have a much reduced fidelity, which would severely compromise their utility for cloning.
  • CSR compartmentalized self-replication
  • the present inventors modified the principles of directed evolution, (in particular compartmentalised self replication) described in GB97143002, 986063936 and GB 01275643 in the name of the present inventors, to relax the steric control of high fidelity DNA polymerases and consequently to expand the substrate range of such polymerases. All of the documents listed above are herein incorporated by reference.
  • mutants were generated which not only exhibited the ability to extend the A*G and C*C tranversion mismatches used in the CSR selection, but also surprisingly exhibited a generic ability to extend mispaired 3′ termini. This finding is especially significant since Taq polymerase is not able to extend 3′ mismatches (Kwok wt al, (1990), Huang (1992).
  • mutant polymerases generated also exhibit high catalytic turnover, concomitant with other high fidelity polymerases and are capable of efficient amplification of DNA fragments up to 26 kb.
  • the present invention provides a method for the generation of an engineered DNA polymerase with an expanded substrate range which comprises the step of preparing and expressing nucleic acid encoding an engineered DNA polymerase utilising template nucleic acid and flanking primers which bear one or more distorting 3′ termini/ends.
  • flanking primers which bear a 3′ distorting terminus/end refer to those primers which possess at their 3′ ends one or more group/s, preferably nucleotide group/s which deviate from cognate base-pairing geometry.
  • Such deviations from cognate base-pairing geometry includes but is not limited to: nucleotide mismatches, base lesions (i.e. modified or damaged bases) or entirely unnatural, synthetic base substitutes.
  • the flanking primer/s bear one or more nucleotide mismatches at their 3′ end/terminus.
  • the flanking primers may have one, two, three, four, or five or more nucleotide mismatches at the 3′ primer end. More advantageously, the one or more nucleotide mismatches are consecutive mismatches. More advantageously, according to the above aspects of the invention, the flanking primers have one or two nucleotide mismatches at the 3′ primer end. Most preferably according to the above aspects of the invention, the flanking primers have one nucleotide mismatch at their 3′ primer end.
  • mismatches are transversion mismatches i.e. apposing purines with purines and pyrimidines with pyrimidines.
  • transversion mismatches are G.A and C.C. This type of primer terminus distortion is referred to herein as ‘primer mismatch distortion’.
  • flanking primers bearing distorting 3′ termini/ends includes within its scope flanking primers bearing one or more unatural base analogues at the 3′ termini/end of the one or more flanking primers so that distortion of the cognate DNA duplex geometry is created.
  • the method of the invention may be used to expand the substrate range of any DNA polymerase which lacks an intrinsic 3-5′ exonuclease proofreading activity or where a 3-5′ exonuclease proofreading activity has been disabled, e.g. through mutation.
  • Suitable DNA polymerases include polA, polB (see e.g. Patrel & Loeb, Nature Struc Biol 2001) polC, polD, polY, polX and reverse transcriptases (RT) but preferably are processive, high-fidelity polymerases.
  • an engineered DNA polymerase with an expanded substrate range according to the invention is generated from a pol A-family DNA polymerase.
  • the DNA polymerase is generated from a repertoire of pol A DNA polymerase nucleic acid as template nucleic acid.
  • the pol A polymerase is Taq polymerase and the flanking primers used in the generation of the polymerase are one or more of those primers selected from the group consisting of the following: 5′-CAG GAA ACA GCT ATG ACA AAA ATC TAG ATA ACG AGG GA-3′; A•G mismatch; 5′GTA AAA CGA CGG CCA GTA CCA CCG AAC TGC GGG TGA CGC CAA GCC-3° C.*C mismatch
  • the nucleic acid encoding the engineered polymerase according to the invention is generated using PCR using one or more flanking primers listed herein.
  • the method of the present invention involves the use of compartmentalised self replication, and consists of the steps listed below:
  • the method of the invention comprises the use of one or more DNA polymerases and flanking primers which bears one or more nucleotide mismatches at their 3′primer ends.
  • engineered DNA polymerase refers to a DNA polymerase which has a nucleic acid sequence which is not 100% identical at the nucleic acid level to the one or more DNA polymerase/s or fragments thereof, from which it is derived, and which is synthetic.
  • an engineered DNA polymerase may belong to any family of DNA polymerase.
  • an engineered DNA polymerase according to the invention is a pol A DNA polymerase.
  • the term ‘engineered DNA polymerase’ also includes within its scope fragments, derivatives and homologues of an ‘engineered DNA polymerase’ as herein defined so long as it exhibits the requisite property of possessing an expanded substrate range as defined herein.
  • an engineered DNA polymerase according to the invention does not include a polymerase with a 3-5′ exonuclease activity under the conditions used for the polymerisation reaction.
  • the term ‘expanded substrate range’ (of an engineered DNA polymerase) means that substrate range of an engineered DNA polymerase according to the present invention is broader than that of the one or more DNA polymerases, or fragments thereof from which it is derived.
  • the term ‘a broader substrate range’ refers to the ability of an engineered polymerase according to the present invention to extend one or more 3′distorting ends, advantageously transversion mismatches (purine*purine, pyrimidine*pyrimidine) for example A*A, C*C, G*G, T*T and G*A, which the one or more polymerase/s from which it is derived cannot extend.
  • a DNA polymerase which exhibits a relaxed substrate range as herein defined has the ability not only to extend the 3′ distorting endsused in its generation, IE those of the flanking primers) but also exhibits a generic ability to extend 3′ distorting ends (for example A*G, A*A, G*G mismatches).
  • ‘expanded substrate range’ (of an engineered DNA polymerase) includes a wider spectrum of unnatural nucleotide substrates including ⁇ S dNTPs, dye-labelled nucleotides, damaged DNA templates and so on. More details are given in the Examples.
  • the DNA polymerase generated using CSR technology is a pol A polymerase and it is generated using flanking primers selected from the group consisting of the following: 5′-CAG GAA ACA GCT ATG ACA AAA ATC TAG ATA ACG AGG GA-3′; A•G mismatch 5′GTA AAA CGA CGG CCA GTA CCA CCG AAC TGC GGG TGA CGC CAA GCC-3′C*C mismatch.
  • any DNA polymerase flanking primer which incorporates a 3′ mismatch will work with any suitable repertoire.
  • the process of mismatch extension will vary in characteristics from polymerase to polymerase, and will also vary according to the experimental conditions. For example, G*A and C*C are the most disfavoured mismatches for extension by Taq polymerase (Huang et al, 92). Other mismatches are favoured for extension by other polymerases and this can be routinely determined by the skilled person.
  • the present inventors generated a number of pol A polymerase mutants.
  • Two of the mutants named M1 and M4 not only exhibit the ability to extend the G*A and C*C transversion mismatches used in the CSR selection, but also surprisingly exhibit a generically enhanced ability to extend 3′ mismatched termini.
  • the present invention provides an engineered DNA polymerase which exhibits an expanded substrate range.
  • an engineered polymerase is obtainable using one or more method/s of the present invention.
  • the DNA polymerase is a pol A polymerase.
  • the engineered DNA polymerase is obtained using the method of the invention.
  • the present invention provides a pol A DNA polymerase with an expanded substrate range, or the nucleic acid encoding it, wherein the DNA polymerase is designated M1 or M4 as shown in FIG. 1 and FIG. 2 respectively and depicted as SEQ No 1 and SEQ No 2 respectively.
  • the engineered DNA polymerase as herein defined is that polymerase designated M1 in FIG. 1 and depicted SEQ No 1.
  • the invention provides a pol A DNA polymerase with an expanded substrate range, wherein the polymerase exhibits at least 95% identity to one or more of the amino acid sequences designated M1 and M4 as shown in FIG. 1 and FIG. 2 respectively and depicted SEQ No 1 and SEQ No 2 respectively and which comprises any one or more of the following mutations: E520G, D144G, L254P, E520G, E524G, N583S, 1.1-D144G, L254P, E520G, E524G, N583S, V113I, A129V, L245R, E315K, G364D, G403R, E432D, P481A, I614M, R704W, D144G, G370D, E742G, K56E, 163T, K127R, M317I, Q680R, R343G, G370D, E520G, G12A, A109T, D251E, P3
  • the invention provides a pol A DNA polymerase with an expanded substrate range, or the nucleic acid encoding it, wherein the polymerase exhibits at least 95% identity to one or more of the amino acid sequences designated M1 and M4 as shown in FIG. 1 and FIG. 2 respectively and depicted SEQ 1 and 2 respectively and which comprises any one or more of the following mutations: G84A, D144G, K314R, E520G, F598L, A608V, E742G, D58G, R74P, A109T, L245R, R343G, G370D, E520G, N583S, E694K, A743P.
  • the invention provides a pol A DNA polymerase with an expanded substrate range, or the nucleic acid encoding it, wherein the polymerase exhibits at least 95% identity to one or more of the amino acid sequences designated M1 and M4 as shown in FIG. 1 and FIG. 2 respectively and depicted SEQ 1 and 2 respectively and which comprises any one or more of the following mutations: G84A, D144G, K314R, E520G, F598L, A608V, E742G.
  • the mutation ‘E520G’ describes a DNA polymerase according to the invention in which glycine is present at position 520 of the amino acid sequence.
  • the present inventors were surprised to find that E520, which is located at the tip of the thumb domain at a distance 20A from the 3′OH of the mismatched primer terminus, would be involved in mismatch recognition or extension.
  • the mutation of E520 to G520 is clearly important in such roles however as the present inventors have demonstrated. This aspect of the invention is described further in the detailed description of the invention.
  • a ‘blend’ of more than one polymerase refers to a mixture of 2 or more, 3 or more 4 or more, 5 or more engineered polymerases.
  • the term ‘blends’ refers to a mixture of 6, 7, 8, 9 or 10 or more ‘engineered polymerases’.
  • mismatched 3′ primer termini is a feature of naturally occurring polymerases.
  • Viral reverse transcriptases like HIV-1 RT or AMV RT and polymerases capable of translesion synthesis (TLS) such as the polY-family polymerases pol ⁇ (Vaisman 2001JBC) or pol ⁇ (Washington 2002 PNAS) or the unusual polB-family polymerase pol ⁇ (Johnson Nature), all extend 3′ mismatches with elevated efficiency compared to high-fidelity polymerases.
  • mutant polA polymerases according to the present invention share significant functional similarities with other polymerases found in nature but so far represent, the only known member of the polA-family polymerases that are proficient in mismatch extension (ME) and translesion synthesis (TLS).
  • ME mismatch extension
  • TLS translesion synthesis
  • M1 and M4 combine mismatch extension (ME) and translesion synthesis (TLS) with high processivity and in the case of M1 are capable of efficient amplification of DNA fragments of up to 26 kb.
  • ME mismatch extension
  • TLS translesion synthesis
  • the present invention provides a nucleic acid construct which is capable of encoding a pol A DNA polymerase which exhibits an expanded substrate range, wherein said pol A DNA polymerase is depicted in FIG. 1 and FIG. 2 as SEQ No 1 or SEQ No 2 and is designated M1 and M4 respectively.
  • the nucleic acid construct encodes the M1 pol A polymerase as described herein.
  • the invention provides a pol A DNA polymerase with an expanded substrate range, in particular which is capable of mismatch extension, wherein the DNA polymerase comprises, preferably consists of the amino acid sequence of any one or more of the clones designated herein as 3B5, 3B8, 3C12 and 3D1.
  • the invention provides a pol A DNA polymerase with an expanded substrate range, in particular which is capable of abasic site bypass, wherein the DNA polymerase comprises, preferably consists of the amino acid sequence of any one or more of the clones designated herein as 3A10, 3B6 and 3B11.
  • the invention provides a pol A DNA polymerase with an expanded substrate range, in particular which is capable of DNA replication involving the incorporation of unatural base analogues into the newly replicated DNA, wherein the pol A DNA polymerase comprises, preferably consists of the amino acid sequence of any one or more of the clones designated herein as 4D11 and 5D4.
  • the present invention provides a pol A DNA polymerase with an expanded substrate range, wherein the polymerase exhibits at least 95% identity to one or more of the amino acid sequences designated 3B5, 3B8, 3C12, 3D1, 3A10, 3B6, 3B11, 4D11 and 5D4. which comprises any one or more of the mutations (with respect to either of the three parent genes Taq, Tth, Tfl) or gene segments found in clones 3B5, 3B8, 3C12, 3D1, 3A10, 3B6, 3B11, 4D11 and 5D4.
  • the present invention provides a vector comprising a nucleic acid construct according to the present invention.
  • the present invention provides the use of a DNA polymerase according to the present invention in any one or more of the following applications selected from the group consisting of the following: PCR amplification, sequencing of damaged DNA templates, the incorporation of unnatural base analogues into DNA and the creation of novel polymerase activities.
  • the use is of a ‘blend’ of DNA polymerases according to the invention or selected according to the method of the invention.
  • the DNA polymerase is a pol A DNA polymerase.
  • it is generated using CSR technology using flanking primers bearing one or more 3′ mismatch pairs of interest as described herein.
  • Other suitable methods include screening after activity preselection (see Patel & Loeb 01) and phage display with proximity coupled template-primer duplex substrate (Jestin 01, Xue, 02. CST is also ideally suited as the present inventors have demonstrated.
  • a polymerase according to the invention is in PCR amplification and the polymerase is M1 as herein described.
  • engineered DNA polymerase refers to a DNA polymerase which has a nucleic acid sequence which is not 100% identical at the nucleic acid level to the one or more DNA polymerase/s or fragments thereof, from which it is derived, and which has been generated using one or more biotechnological methods.
  • an engineered DNA polymerase according to the invention is a pol-A family DNA polymerase or a pol-B family DNA polymerase. More advantageously, an engineered DNA polymerase according to the invention is a pol-A family DNA polymerase.
  • engineered DNA polymerase also includes within its scope fragments, derivatives and homologues of an ‘engineered DNA polymerase’ as herein defined so long as it exhibits the requisite property of possessing an expanded substrate range as defined herein.
  • an engineered DNA polymerase according to the invention does not include a polymerase with a 3-5′ exonuclease activity under the conditions used for the polymerisation reaction. Such a proofreading activity would remove any 3′ mismatches incorporated according to the method of the invention, and thus would prevent a polymerase according to the invention possessing an expanded substrate range as defined herein.
  • flanking primers which bear a 3′ distorting terminus refer to those DNA polymerase primers which possess at their 3′ ends one or more group/s, preferably nucleotide group/s which deviate from cognate base-pairing geometry.
  • Such deviations from cognate base-pairing geometry includes but is not limited to: nucleotide mismatches, base lesions (i.e. modified or damaged bases) or entirely unnatural, synthetic base substitutes at the 3 end of a flanking primer used according to the methods of the invention.
  • the flanking primer/s bear one or more nucleotide mismatches at their 3′ end.
  • the flanking primers may have one, two, three, four, or five or more nucleotide mismatches at the 3′ primer end.
  • the flanking primers Preferably according to the above aspects of the invention, the flanking primers have one or two nucleotide mismatches at the 3′ primer end.
  • the flanking primers have one nucleotide mismatch at their 3′ primer end.
  • the term ‘expanded substrate range’ (of an engineered DNA polymerase) means that substrate range of an engineered DNA polymerase according to the present invention is broader than that of the one or more DNA polymerases, or fragments thereof from which it is derived.
  • the term ‘a broader substrate range’ refers to the ability of an engineered polymerase according to the present invention to extend one or more 3′distorting ends, advantageously transversion mismatches (purine*purine, pyrimidine*pyrimidine) for example A*A, C*C, G*G, T*T and G*A, which the one or more polymerase/s from which it is derived cannot extend.
  • a DNA polymerase which exhibits a relaxed substrate range as herein defined has the ability not only to extend the 3′ distorting ends used in its generation, IE those of the flanking primers) but also exhibits a generic ability to extend 3′ distorting ends (for example A*G, A*A, G*G mismatches).
  • FIG. 1 shows the M1 nucleic acid (a and amino acid sequence (b
  • FIG. 2 shows the M4 nucleic acid (a amino acid sequence (b).
  • FIG. 3 shows the general scheme of mismatch extension CSR selection.
  • Self-replication of the pol gene by the encoded polymerase requires extension of flanking primers bearing G ⁇ A and C ⁇ C. 3′ mismatches.
  • Polymerases capable of mismatch extension (Pol*) replicate their own encoding gene (pol*), while pol x cannot extend mismatches and fails to self-replicate. Black bars denote incorporation of the mismatch into replication products.
  • FIG. 4 Mismatch extension properties of selected polymerases.
  • A Polymerase activity in PCR for matched 3′ ends and mismatches. Only mutant polymerases M4 and M1 (not shown) generate amplification products using primers with 3′ transversion mismatches.
  • B Mismatch extension PCR assay. Mismatch extension capability is expressed as arbitrary mismatch extension units (ratio of polymerase activity in PCR with matched vs. mismatched flanking primers). Different polymerases (black diamonds) and derivatives (open squares, triangles) are shown in separate columns.
  • FIG. 6 Polymerase activity on unnatural substrates.
  • A Polymerase activity in PCR using all ⁇ S dNTPs. ⁇ S DNA amplification products of 0.4 kb, 0.8 kb and 2 kb, are obtained with M1 but not with wtTaq (wt).
  • ⁇ X HaeIII-digested phage ⁇ X174 DNA marker.
  • ⁇ H HindIII-digested phage ⁇ DNA marker.
  • B Polymerase activity in PCR with complete replacement of dATP with FITC-12-dATP (left) or dTTP with Biotin-16-dUTP (right). Only M1 yields amplification products. M, 1 kb DNA ladder (Invitrogen).
  • C Bypass of a 5-nitroindol template (5NI) base. Polymerase activity was assayed over time for its ability to extend a radiolabeled primer annealed to a template containing a 5NI template base.
  • 5NI 5-
  • FIG. 7 Long range PCR. PCR amplification of fragments of increasing length from a phage ⁇ DNA template. WtTaq (wt) fails to generate amplification products larger than 8.8 kb while M1 is able to amplify fragments of >25 kb. ⁇ H, HindIII-digested phage ⁇ DNA marker.
  • FIG. 8 Hairpin-ELISAs to test nucleotide analogue incorporation by mismatch extension clones
  • FIG. 9 Clones 3B5. 3B8, 3C12 and 3D1 (where 3 indicates that these are third round clones) were able to extend primers containing four mismatches.
  • the 292 base pair product is indicated with an arrow and was produced after 50 cycles of PCR. It is noteworthy that significant amount of non-specific products are produced in all cases, although the amount of non-specific product varies from polymerase to polymerase.
  • the C12 lane has been appended from another gel. Lane M: markers, Hae III digest of ⁇ X174.
  • FIG. 10 A list of polymerases selected to extend four mismatches were assayed for their ability to extend abasic sites in PCR. Primers with an abasic site seven bases from their 3′ end were designed. Such primers will prevent exponential amplification of the target sequence, restriciting it to geometric amplification, unless the abasic site is bypassed. 20 cycles of PCR were sufficient to produce the 176 bp product with the selected polymerases but not with the wild type. (A) Screen which identified clone A10. (B) A further 4 polymerases that display good abasic site bypass. Lane M: markers, Hae III digest of ⁇ X174.
  • FIG. 11 Seven polymerases were assayed for their ability to bypass abasic sites in a primer extension assay. Translesion synthesis activity on an undamaged template, on a template containing an abasic site or a cis-syn cyclobutane thymine-thymine dimer (CPD) tend a radiolabelled primer (pr) annealed to template. The c site or a CPD located immediately downstream of the primer.
  • CPD radiolabelled primer
  • FIG. 12 Several samples of cave hyena ( Crocuta spelaea ) were extracted and analysed. The seven samples were from Teufelslucke cave (Austria, 40 000 years old), Aufhausener Höhle (Germany, no date determined (2 samples)); Irpfelhöhle (Germany, no date determined); Kiskevelyi (Romania 48 500 years old); Miskolc III (Hungary, 44 000 years old); Mala ladnica (Slovakia, no date determined). The target was a 215 bp fragment from the cytochrome B gene in the mitochondrial genome. The amplification was only successful in the presence of sspDNA.
  • FIG. 13 Appropriate primers for use in the method of the invention. See example 15 for details.
  • Target sequences in the cave bear mitochondrial D loop. Outer primer sequences are underlined, Inner primer sequences are in red.
  • FIG. 14 Polymerases selected for replication of 5NI were tested for activity with a range of substrates using the hairpin ELISA assay described in example 8. See example 16 for details. Sample 366 is from the Herdengel cave (Austria) and is 60 000 years old.
  • Sample GS 3-7 is from the Gamsulzen cave (Austria) and is between 25 000 and 45 000 years old.
  • FIG. 15 Polymerases selected for replication of 5NI were tested for activity with a range of substrates. Polymerase 4D11. P is primer, Ch is the chase reaction. Reaction times in minutes. See example 16 for details.
  • FIG. 16 Polymerases selected for replication of 5NI were tested for activity with a range of substrates Polymerase 5D4. P is primer, Ch is the chase reaction. Reaction times in minutes. See example 16 for details.
  • FIG. 17 Polymerases selected for replication of 5NI were tested for activity with a range of substrates Polymerase 4D11.
  • P primer
  • Ch is the chase reaction. Reaction times in minutes. See example 16 for details.
  • FIG. 18 Polymerases selected for replication of 5NI were tested for activity with a range of substrates Polymerase 5D4.
  • P primer
  • Ch is the chase reaction. Reaction times in minutes. See example 16 for details.
  • FIG. 19 Microarray hybridisations of FITC-labelled probes.
  • Microarrays contained replicate features of serial dilutions of Taq, RT and genomic salmon sperm DNA target sequences, as indicated. Labelled randomers were used to visualise the microarray and assess the availability of target sequences for hybridisation.
  • Array co-hybridisations were performed with a Cy5-labelled Taq probe (Cy5 Taq ), as a reference, and equivalent unlabelled or FITC-labelled probes (FITC10 Taq , FITC10 M1 , FITC100 M1 ). Single examples from 3 replicate experiments are displayed for each co-hybridisation.
  • FIG. 20 , FIG. 21 Microarray signals from FITC-labelled probes.
  • Mean FITC fluorescence signal of FITC-labelled probes (FITC10 Taq , FITC10 M1 , FITC100 M1 ) for each co-hybridisation is plotted against the Cy5 fluorescence signal of the reference probe (Cy5 Taq ) for A) Taq, B) RT and C) genomic salmon sperm DNA target sequences, as indicated.
  • Microarray background signals from FITC-labelled probes are determined using 3 replicate microarrays for each co-hybridisation experiment of a Cy5-labelled Taq probe (Cy5 Taq ), as a reference, and unlabeled or FITC-labelled probes (FITC10 Taq , FITC10 M1 , FITC100 M1 ). Background information was generated by measuring fluorescence signal from 12 non-feature areas of each microarray. Mean pixel intensities were generated and used to derive a ratiometric value for each non-feature area. A mean of the mean ratio +/ ⁇ 1 standard deviation is displayed for each co-hybridisation experiment.
  • FIG. 22 Fidelity.
  • A MutS ELISA. Relative replication fidelity of wtTaq, M1 and M4 was determined using mutS ELISA of two different DNA fragments (either a 0.4 kb or 2.5 kb region of the cloned Taq gene) obtained by PCR and probed at two different concentrations.
  • B Spectra of nucleotide substitutions observed in PCR fragments amplified with either wtTaq or M1. Types of substitutions are given as % of total substitutions (wtTaq: 48, M1: 74). Equivalent substitutions on either strand (e.g. G->A, C->T) were added together (GC->AT). Observed ⁇ 1 detections (wtTaq: 3, M1: 1) are not shown.
  • FIG. 23 Processivity of wtTaq, M1 and M4 was measured at three different polymerase concentrations in the absence (A) or presence (B) of trap DNA.
  • the processivity for nucleotide incorporation at each position was variable but essentially identical for all three polymerases. For example, the probability of enzyme dissociation is higher at positions 2-5 compared to positions 6 and 7 for all three polymerases.
  • trap DNA to ensure all primer extension is the result of a single DNA binding event
  • the inventors modified the methods of compartmentalised self replication and surprisingly generated DNA polymerases which exhibited an expanded substrate range as herein defined.
  • Example 1 The present inventors used recently developed oil in water emulsions but modified the composition of the surfactant as well as the oil to water ratio. Details are given in Example 1. These modifications greatly increased the heat stability of the compartments and allowed PCR yields in the emulsion to approach those of PCR in solution. Further details of the method of compartmentalised self replication are given below.
  • microcapsules used according to the method of the invention require appropriate physical properties to allow the working of the invention.
  • the contents of each microcapsule must be isolated from the contents of the surrounding microcapsules, so that there is no or little exchange of the nucleic acids and gene products between the microcapsules over the timescale of the experiment.
  • the method of the present invention requires that there are only a limited number of nucleic acids per microcapsule. This ensures that the gene product of an individual nucleic acid will be isolated from other nucleic acids. Thus, coupling between nucleic acid and gene product will be highly specific. The enrichment factor is greatest with on average one or fewer nucleic acids per microcapsule, the linkage between nucleic acid and the activity of the encoded gene product being as tight as is possible, since the gene product of an individual nucleic acid will be isolated from the products of all other nucleic acids.
  • nucleic acid there is a single nucleic acid, or fewer, per microcapsule.
  • the formation and the composition of the microcapsules must not abolish the function of the machinery the expression of the nucleic acids and the activity of the gene products.
  • any microencapsulation system used must fulfil these three requirements.
  • the appropriate system(s) may vary depending on the precise nature of the requirements in each application of the invention, as will be apparent to the skilled person.
  • microencapsulation procedures are available (see Benita, 1996) and may be used to create the microcapsules used in accordance with the present invention. Indeed, more than 200 microencapsulation methods have been identified in the literature (Finch, 1993).
  • lipid vesicles liposomes
  • non-ionic surfactant vesicles van Hal et al., 1996.
  • lipid vesicles liposomes
  • van Hal et al., 1996 closed-membranous capsules of single or multiple bilayers of non-covalently assembled molecules, with each bilayer separated from its neighbour by an aqueous compartment.
  • liposomes the membrane is composed of lipid molecules; these are usually phospholipids but sterols such as cholesterol may also be incorporated into the membranes (New, 1990).
  • RNA and DNA polymerisation can be performed within liposomes (Chakrabarti et al., 1994; Oberholzer et al., 1995a; Oberholzer et al., 1995b; Walde et al., 1994; Wick & Luisi, 1996).
  • aqueous phase With a membrane-enveloped vesicle system much of the aqueous phase is outside the vesicles and is therefore non-compartmentalised. This continuous, aqueous phase should be removed or the biological systems in it inhibited or destroyed (for example, by digestion of nucleic acids with DNase or RNase) in order that the reactions are limited to the microcapsules (Luisi et al., 1987).
  • Enzyme-catalysed biochemical reactions have also been demonstrated in microcapsules generated by a variety of other methods. Many enzymes are active in reverse micellar solutions (Bru & Walde, 1991; Bru & Walde, 1993; Creagh et al., 1993; Haber et al., 1993; Kumar et al., 1989; Luisi & B., 1987; Mao & Walde, 1991; Mao et al., 1992; Perez et al., 1992; Walde et al., 1994; Walde et al., 1993; Walde et al., 1988) such as the AOT-isooctane-water system (Menger & Yamada, 1979).
  • Microcapsules can also be generated by interfacial polymerisation and interfacial complexation (Whateley, 1996). Microcapsules of this sort can have rigid, nonpermeable membranes, or semipermeable membranes. Semipermeable microcapsules bordered by cellulose nitrate membranes, polyamide membranes and lipid-polyamide membranes can all support biochemical reactions, including multienzyme systems (Chang, 1987; Chang, 1992; Lim, 1984). Alginate/polylysine microcapsules (Lim & Sun, 1980), which can be formed under very mild conditions, have also proven to be very biocompatible, providing, for example, an effective method of encapsulating living cells and tissues (Chang, 1992; Sun et al., 1992).
  • Non-membranous microencapsulation systems based on phase partitioning of an aqueous environment in a colloidal system, such as an emulsion, may also be used.
  • the microcapsules of the present invention are formed from emulsions; heterogeneous systems of two immiscible liquid phases with one of the phases dispersed in the other as droplets of microscopic or colloidal size (Becher, 1957; Sherman, 1968; Lissant, 1974; Lissant, 1984).
  • Emulsions may be produced from any suitable combination of immiscible liquids.
  • the emulsion of the present invention has water (containing the biochemical components) as the phase present in the form of finely divided droplets (the disperse, internal or discontinuous phase) and a hydrophobic, immiscible liquid (an ‘oil’) as the matrix in which these droplets are suspended (the nondisperse, continuous or external phase).
  • water containing the biochemical components
  • an ‘oil’ hydrophobic, immiscible liquid
  • Such emulsions are termed ‘water-in-oil’ (W/O). This has the advantage that the entire aqueous phase containing the biochemical components is compartmentalised in discreet droplets (the internal phase).
  • the external phase being a hydrophobic oil, generally contains none of the biochemical components and hence is inert.
  • the emulsion may be stabilised by addition of one or more surface-active agents (surfactants).
  • surfactants are termed emulsifying agents and act at the water/oil interface to prevent (or at least delay) separation of the phases.
  • Many oils and many emulsifiers can be used for the generation of water-in-oil emulsions; a recent compilation listed over 16,000 surfactants, many of which are used as emulsifying agents (Ash and Ash, 1993). Suitable oils include light white mineral oil and non-ionic surfactants (Schick, 1966) such as sorbitan monooleate (SpanTM80; ICI) and polyoxyethylenesorbitan monooleate (TweenTM 80; ICI) and Triton-X-100.
  • anionic surfactants may also be beneficial.
  • Suitable surfactants include sodium cholate and sodium taurocholate. Particularly preferred is sodium deoxycholate, preferably at a concentration of 0.5% w/v, or below. Inclusion of such surfactants can in some cases increase the expression of the nucleic acids and/or the activity of the gene products. Addition of some anionic surfactants to a non-emulsified reaction mixture completely abolishes translation. During emulsification, however, the surfactant is transferred from the aqueous phase into the interface and activity is restored. Addition of an anionic surfactant to the mixtures to be emulsified ensures that reactions proceed only after compartmentalisation.
  • stirrers such as magnetic stir-bars, propeller and turbine stirrers, paddle devices and whisks
  • homogenisers including rotor-stator homogenisers, high-pressure valve homogenisers and jet homogenisers
  • colloid mills ultrasound and ‘membrane emulsification’ devices
  • Aqueous microcapsules formed in water-in-oil emulsions are generally stable with little if any exchange of nucleic acids or gene products between microcapsules. Additionally, we have demonstrated that several biochemical reactions proceed in emulsion microcapsules. Moreover, complicated biochemical processes, notably gene transcription and translation are also active in emulsion microcapsules. The technology exists to create emulsions with volumes all the way up to industrial scales of thousands of litres (Becher, 1957; Sherman, 1968; Lissant, 1974; Lissant, 1984).
  • the preferred microcapsule size will vary depending upon the precise requirements of any individual selection process that is to be performed according to the present invention. In all cases, there will be an optimal balance between gene library size, the required enrichment and the required concentration of components in the individual microcapsules to achieve efficient expression and reactivity of the gene products.
  • Example 1 Details of one example of an emulsion used when performing the method of the present invention are given in Example 1.
  • the processes of expression must occur within each individual microcapsule provided by the present invention. Both in vitro transcription and coupled transcription-translation become less efficient at sub-nanomolar DNA concentrations. Because of the requirement for only a limited number of DNA molecules to be present in each microcapsule, this therefore sets a practical upper limit on the possible microcapsule size.
  • the mean volume of the microcapsules is less that 5.2 ⁇ 10 ⁇ 16 m 3 , (corresponding to a spherical microcapsule of diameter less than 10 ⁇ m, more preferably less than 6.5 ⁇ 10 ⁇ 17 m 3 (5 ⁇ m), more preferably about 4.2 ⁇ 10 ⁇ 18 m 3 (2 ⁇ m) and ideally about 9 ⁇ 10 ⁇ 18 m 3 (2.6 ⁇ m).
  • the effective DNA or RNA concentration in the microcapsules may be artificially increased by various methods that will be well-known to those versed in the art. These include, for example, the addition of volume excluding chemicals such as polyethylene glycols (PEG) and a variety of gene amplification techniques, including transcription using RNA polymerases including those from bacteria such as E. coli (Roberts, 1969; Blattner and Dahlberg, 1972; Roberts et al., 1975; Rosenberg et al., 1975), eukaryotes e.g.
  • PEG polyethylene glycols
  • RNA polymerases including those from bacteria such as E. coli (Roberts, 1969; Blattner and Dahlberg, 1972; Roberts et al., 1975; Rosenberg et al., 1975), eukaryotes e.g.
  • thermostable for example, the coupled transcription-translation systems could be made from a thermostable organism such as Thermus aquaticus ).
  • microcapsule volume 5.2 ⁇ 10 ⁇ 16 m 3 (corresponding to a sphere of diameter 10 um).
  • microcapsule size must be sufficiently large to accommodate all of the required components of the biochemical reactions that are needed to occur within the microcapsule.
  • both transcription reactions and coupled transcription-translation reactions require a total nucleoside triphosphate concentration of about 2 mM.
  • RNA molecules of nucleoside triphosphate per microcapsule 8.33 ⁇ 10 ⁇ 22 moles.
  • this number of molecules must be contained within a microcapsule of volume 4.17 ⁇ 10 ⁇ 19 litres (4.17 ⁇ 10 ⁇ 22 m 3 which if spherical would have a diameter of 93 nm.
  • the ribosomes necessary for the translation to occur are themselves approximately 20 nm in diameter.
  • the preferred lower limit for microcapsules is a diameter of approximately 100 nm.
  • the microcapsule volume is preferably of the order of between 5.2 ⁇ 10 ⁇ 22 m 3 and 5.2 ⁇ 10 ⁇ 16 m 3 corresponding to a sphere of diameter between 0.1 um and 10 um, more preferably of between about 5.2 ⁇ 10 ⁇ 19 m 3 and 6.5 ⁇ 10 ⁇ 17 m 3 (1 um and 5 um). Sphere diameters of about 2.6 um are most advantageous.
  • compartments droplets of 2.6 um mean diameter
  • Escherichia are 1.1-1.5 ⁇ 2.0-6.0 um rods
  • Azotobacter are 1.5-2.0 um diameter ovoid cells.
  • Darwinian evolution is based on a ‘one genotype one phenotype’ mechanism.
  • the concentration of a single compartmentalised gene, or genome drops from 0.4 nM in a compartment of 2 um diameter, to 25 pM in a compartment of 5 um diameter.
  • the prokaryotic transcription/translation machinery has evolved to operate in compartments of ⁇ 1-2 um diameter, where single genes are at approximately nanomolar concentrations.
  • a single gene, in a compartment of 2.6 um diameter is at a concentration of 0.2 nM. This gene concentration is high enough for efficient translation. Compartmentalisation in such a volume also ensures that even if only a single molecule of the gene product is formed it is present at about 0.2 nM, which is important if the gene product is to have a modifying activity of the nucleic acid itself.
  • the volume of the microcapsule should thus be selected bearing in mind not only the requirements for transcription and translation of the nucleic acid/nucleic acid, but also the modifying activity required of the gene product in the method of the invention.
  • the size of emulsion microcapsules may be varied simply by tailoring the emulsion conditions used to form the emulsion according to requirements of the selection system.
  • the size of the microcapsules is selected not only having regard to the requirements of the transcription/translation system, but also those of the selection system employed for the nucleic acid/nucleic acid construct.
  • the components of the selection system such as a chemical modification system, may require reaction volumes and/or reagent concentrations which are not optimal for transcription/translation.
  • such requirements may be accommodated by a secondary re-encapsulation step; moreover, they may be accommodated by selecting the microcapsule size in order to maximise transcription/translation and selection as a whole.
  • Empirical determination of optimal microcapsule volume and reagent concentration for example as set forth herein, is preferred.
  • nucleic acid/nucleic acid in accordance with the present invention is as described above.
  • a nucleic acid is a molecule or construct selected from the group consisting of a DNA molecule, an RNA molecule, a partially or wholly artificial nucleic acid molecule consisting of exclusively synthetic or a mixture of naturally-occurring and synthetic bases, any one of the foregoing linked to a polypeptide, and any one of the foregoing linked to any other molecular group or construct.
  • the other molecular group or construct may be selected from the group consisting of nucleic acids, polymeric substances, particularly beads, for example polystyrene beads, magnetic substances such as magnetic beads, labels, such as fluorophores or isotopic labels, chemical reagents, binding agents such as macrocycles and the like.
  • the nucleic acid portion of the nucleic acid may comprise suitable regulatory sequences, such as those required for efficient expression of the gene product, for example promoters, enhancers, translational initiation sequences, polyadenylation sequences, splice sites and the like.
  • Example 1 Details of a preferred method of performing the method of the invention are given in Example 1. However, those skilled in the art will appreciate that the examples given are non-limiting and methods for product selection are discussed in more general terms below.
  • a ligand or substrate can be connected to the nucleic acid by a variety of means that will be apparent to those skilled in the art (see, for example, Hermanson, 1996). Any tag will suffice that allows for the subsequent selection of the nucleic acid. Sorting can be by any method which allows the preferential separation, amplification or survival of the tagged nucleic acid. Examples include selection by binding (including techniques based on magnetic separation, for example using DynabeadsTM), and by resistance to degradation (for example by nucleases, including restriction endonucleases).
  • nucleic acid molecule may be linked to a ligand or substrate is through biotinylation. This can be done by PCR amplification with a 5′-biotinylation primer such that the biotin and nucleic acid are covalently linked.
  • the ligand or substrate to be selected can be attached to the modified nucleic acid by a variety of means that will be apparent to those of skill in the art.
  • a biotinylated nucleic acid may be coupled to a polystyrene microbead (0.035 to 0.2 um in diameter) that is coated with avidin or streptavidin, that will therefore bind the nucleic acid with very high affinity.
  • This bead can be derivatised with substrate or ligand by any suitable method such as by adding biotinylated substrate or by covalent coupling.
  • a biotinylated nucleic acid may be coupled to avidin or streptavidin complexed to a large protein molecule such as thyroglobulin (669 Kd) or ferritin (440 Kd).
  • This complex can be derivatised with substrate or ligand, for example by covalent coupling to the alpha-amino group of lysines or through a non-covalent interaction such as biotin-avidin.
  • the substrate may be present in a form unlinked to the nucleic acid but containing an inactive “tag” that requires a further step to activate it such as photoactivation (e.g. of a “caged” biotin analogue, (Sundberg et al., 1995; Pirrung and Huang, 1996)).
  • the catalyst to be selected then converts the substrate to product.
  • the “tag” could then be activated and the “tagged” substrate and/or product bound by a tag-binding molecule (e.g. avidin or streptavidin) complexed with the nucleic acid.
  • a tag-binding molecule e.g. avidin or streptavidin
  • the nucleic acids encoding active enzymes can be enriched using an antibody or other molecule which binds, or reacts specifically with the “tag”. Although both substrates and product have the molecular tag, only the nucleic acids encoding active gene product will co-purify.
  • Isolation refers to the process of separating an entity from a heterogeneous population, for example a mixture, such that it is free of at least one substance with which it was associated before the isolation process.
  • isolation refers to purification of an entity essentially to homogeneity.
  • Sorting of an entity refers to the process of preferentially isolating desired entities over undesired entities. In as far as this relates to isolation of the desired entities, the terms “isolating” and “sorting” are equivalent.
  • the method of the present invention permits the sorting of desired nucleic acids from pools (libraries or repertoires) of nucleic acids which contain the desired nucleic acid. Selecting is used to refer to the process (including the sorting process) of isolating an entity according to a particular property thereof.
  • nucleic acid/nucleic acid from a nucleic acid library for example a mutant taq library
  • Libraries of nucleic acids can be created in a variety of different ways, including the following.
  • genes can also be made by encoding all (see for example Smith, 1985; Parmley and Smith, 1988) or part of genes (see for example Lowman et al., 1991) or pools of genes (see for example Nissim et al., 1994) by a randomised or doped synthetic oligonucleotide.
  • Libraries can also be made by introducing mutations into a nucleic acid or pool of nucleic acids ‘randomly’ by a variety of techniques in vivo, including; using ‘mutator strains’, of bacteria such as E. coli mutD5 (Liao et al., 1986; Yamagishi et al., 1990; Low et al., 1996). Random mutations can also be introduced both in vivo and in vitro by chemical mutagens, and ionising or UV irradiation (see Friedberg et al., 1995), or incorporation of mutagenic base analogues (Freese, 1959; Zaccolo et al., 1996).
  • ‘Random’ mutations can also be introduced into genes in vitro during polymerisation for example by using error-prone polymerases (Leung et al., 1989).
  • the repertoire of nucleic fragments used is a mutant Taq repertoire which has been mutated using error prone PCR. Details are given in Examples 1.
  • the term ‘random’ may be in terms of random positions with random repertoire of amino acids at those positions or it may be selected (predetermined) positions with random repertoire of amino acids at those selected positions.
  • the microcapsules according to the invention will comprise further components required for the sorting process to take place.
  • Other components of the system will for example comprise those necessary for transcription and/or translation of the nucleic acid. These are selected for the requirements of a specific system from the following; a suitable buffer, an in vitro transcription/replication system and/or an in vitro translation system containing all the necessary ingredients, enzymes and cofactors, RNA polymerase, nucleotides, nucleic acids (natural or synthetic), transfer RNAs, ribosomes and amino acids, and the substrates of the reaction of interest in order to allow selection of the modified gene product.
  • a suitable buffer will be one in which all of the desired components of the biological system are active and will therefore depend upon the requirements of each specific reaction system. Buffers suitable for biological and/or chemical reactions are known in the art and recipes provided in various laboratory texts, such as Sambrook et al., 1989.
  • the in vitro translation system will usually comprise a cell extract, typically from bacteria (Zubay, 1973; Zubay, 1980; Lesley et al., 1991; Lesley, 1995), rabbit reticulocytes (Pelham and Jackson, 1976), or wheat germ (Anderson et al., 1983).
  • a cell extract typically from bacteria (Zubay, 1973; Zubay, 1980; Lesley et al., 1991; Lesley, 1995), rabbit reticulocytes (Pelham and Jackson, 1976), or wheat germ (Anderson et al., 1983).
  • Many suitable systems are commercially available (for example from Promega) including some which will allow coupled transcription/translation (all the bacterial systems and the reticulocyte and wheat germ TNTTM extract systems from Promega).
  • the mixture of amino acids used may include synthetic amino acids if desired, to increase the possible number or variety of proteins produced in the library. This can be accomplished by charging tRNAs with artificial amino acids and using these tRNAs for the in vitro translation of the proteins to
  • the enrichment of the pool of nucleic acids for those encoding the molecules of interest can be assayed by non-compartmentalised in vitro transcription/replication or coupled transcription-translation reactions.
  • the selected pool is cloned into a suitable plasmid vector and RNA or recombinant protein is produced from the individual clones for further purification and assay.
  • Microcapsules may be identified by virtue of a change induced by the desired gene product which either occurs or manifests itself at the surface of the microcapsule or is detectable from the outside as described in section iii (Microcapsule Sorting). This change, when identified, is used to trigger the modification of the gene within the compartment.
  • microcapsule identification relies on a change in the optical properties of the microcapsule resulting from a reaction leading to luminescence, phosphorescence or fluorescence within the microcapsule. Modification of the gene within the microcapsules would be triggered by identification of luminescence, phosphorescence or fluorescence.
  • identification of luminescence, phosphorescence or fluorescence can trigger bombardment of the compartment with photons (or other particles or waves) which leads to modification of the nucleic acid.
  • Modification of the nucleic acid may result, for example, from coupling a molecular “tag”, caged by a photolabile protecting group to the nucleic acids: bombardment with photons of an appropriate wavelength leads to the removal of the cage. Afterwards, all microcapsules are combined and the nucleic acids pooled together in one environment. Nucleic acids encoding gene products exhibiting the desired activity can be selected by affinity purification using a molecule that specifically binds to, or reacts specifically with, the “tag”.
  • the selection procedure may comprise two or more steps. First, transcription/replication and/or translation of each nucleic acid of a nucleic acid library may take place in a first microcapsule. Each gene product is then linked to the nucleic acid which encoded it (which resides in the same microcapsule). The microcapsules are then broken, and the nucleic acids attached to their respective gene products optionally purified. Alternatively, nucleic acids can be attached to their respective gene products using methods which do not rely on encapsulation. For example phage display (Smith, G.
  • each purified nucleic acid attached to its gene product is put into a second microcapsule containing components of the reaction to be selected. This reaction is then initiated. After completion of the reactions, the microcapsules are again broken and the modified nucleic acids are selected. In the case of complicated multistep reactions in which many individual components and reaction steps are involved, one or more intervening steps may be performed between the initial step of creation and linking of gene product to nucleic acid, and the final step of generating the selectable change in the nucleic acid.
  • genetic material comprised in the nucleic acids may be amplified and the process repeated in iterative steps.
  • Amplification may be by the polymerase chain reaction (Saiki et al., 1988) or by using one of a variety of other gene amplification techniques including; Q ⁇ replicase amplification (Cahill, Foster and Mahan, 1991; Chetverin and Spirin, 1995; Katanaev, Kumasov and Spirin, 1995); the ligase chain reaction (LCR) (Landegren et al., 1988; Barany, 1991); the self-sustained sequence replication system (Fahy, Kwoh and Gingeras, 1991) and strand displacement amplification (Walker et al., 1992).
  • LCR ligase chain reaction
  • High fidelity DNA polymerases such as Pol A (like Taq polymerase) and Pol-B family polymerases which lack a 3′-5′ exonuclease proofreading capability show a strict blockage to the extension of distorted or mismatched 3′ primer termini to avoid propagation of misincorporations. While the degree of blockage varies considerably depending on the nature of the mismatch, some transversion (purine•purine/pyrimidine•pyrimidine) mismatches are extended up to 10 6 -fold less efficiently than matched termini (Huang 92). Likewise, many unnatural base analogues, while incorporated efficiently, act as strong terminators (Kool, Loakes).
  • the present inventors have modified the principles described in Ghadessy, F. G et al (2001) Proc. Nat. Acad. Sci, USA, 93, 4552-4557 (compartmentalised self replication) and Ghadessy 2003, and outlined above. Both these documents are herein incorporated by reference.
  • the present inventors have used these modified techniques to develop a method by which the substrates specificity of high fidelity DNA polymerases may be expanded in a generic way.
  • the inventors have exemplified the technique by expanding the substrate specificity of the high-fidelity pol-A family polymerases.
  • the present inventors created two repertoires of randomly mutated Taq genes, as described in Ghadessy, F. G et al (2001) referred to above. Three cycles of mismatch extension CSR was performed using flanking primers bearing the mismatches A*G and C*C at their 3′ ends. Selected clones were ranked using a PCR extension assay described herein.
  • the inventors have generated DNA polymerases which exhibit a relaxed substrate specificity/expanded substrate range.
  • the term ‘expanded substrate range’ (of an engineered DNA polymerase) means that substrate range of an engineered DNA polymerase according to the present invention is broader than that of the one or more DNA polymerases, or fragments thereof from which it is derived.
  • the term ‘a broader substrate range’ refers to the ability of an engineered polymerase according to the present invention to extend one or more 3′ mismatches, for example A*A, G*A, G*G, T*T, C*C, which the one or more polymerase/s from which it is derived cannot extend.
  • a DNA polymerase which exhibits a relaxed substrate range as herein defined has the ability not only to extend the 3′ mismatches used in its generation, (IE those of the flanking primers), but also exhibits a generic ability to extend 3′ mismatches (for example A*G, A*A, G*G).
  • the two best mutants M1 (G84A, D144G, K314R, E520G, F598L, A608V, E742G) and M4 (D58G, R74P, A109T, L245R, R343G, G370D, E520G, N583S, E694K, A743P) were chosen for further investigation.
  • M1 and M4 not only had greatly increased ability to extend the G•A and C•C transversion mismatches used in the CSR selection, but appeared to have acquired a more generic ability to extend 3′ mispaired termini, including other strongly disfavoured transversion mismatches (such as A•G, A•A, G•G) ( FIG. 1B ), which wtTaq polymerase was unable to extend, as previously reported (Kwok et al 1990, Huang 92).
  • Nucleic acid sequences encoding M1 and M4 pol A DNA polymerase mutants are depicted SEQ No 1 and SEQ No 2 respectively and are shown in FIG. 1 and FIG. 2 respectively.
  • E520G a mutation that is shared by all but one of the four best clones of the final selection. Curiously, E520 is located at the very tip of the thumb domain at a distance of 20 ⁇ from the 3′ OH of the mismatched primer terminus and its involvement in mismatch recognition or extension is unclear. However, E520G is clearly important for mismatch extension as backmutation reduces mismatch extension in both M1 and M4 to near wt levels ( FIG. 2 ).
  • mutations in the N-terminal 5′-3′ exonuclease domain also appear to be contributing to mismatch extension as suggested by the 2-4 fold increased mismatch extension ability of chimeras of the 53exoD of M1, M4 and polD of wtTaq ( FIG. 4 ). How they promote mismatch extension is unclear but given the apparent distance of the 53exoD from the active site (Utz 99, Eom 96) is unlikely to involve direct effects on extension catalysis.
  • Viral reverse transcriptases like HIV-1 RT or AMV RT and polymerases capable of translesion synthesis (TLS) such as the polY-family polymerases pol ⁇ (Vaisman 2001JBC) or pol ⁇ (Washington 2002 PNAS) or the unusual polB-family polymerase pol ⁇ (Johnson Nature), all extend 3′ mismatches with elevated efficiency compared to high-fidelity polymerases.
  • TLS translesion synthesis
  • the selected polymerases share significant functional similarities with preexisting polymerases but represent, to our knowledge, the only known polA-family polymerases that are proficient in mismatch extension (ME) and translesion synthesis (TLS).
  • M1 and M4 combine ME and TLS with high processivity and in the case of M1 are capable of efficient amplification of DNA fragments of up to 26 kb.
  • ME may play a crucial role in allowing error-prone yet processive replication of a multi-kb viral genome.
  • proficient mismatch extension is also a necessary prerequisite for their biological function as unpaired and distorted primer termini necessarily occur opposite lesions in the DNA template strand.
  • the ability of TLS polymerases to traverse replication blocking lesions in DNA is thought to arise from a relaxed geometric selection in the active site (Goodman 02).
  • the ability of M1 and M4 to process both bulky mispairs and a distorting CPD (cys-syn thymidine-thymidine dimer) dimer makes it plausible that, in analogy to TLS polymerases, they also have acquired a more open active site. Indeed, modelling showed that a CPD dimer can not be accommodated in the wtTaq polymerase active site without mayor steric clashes (Trincao01).
  • M1 (and to a lesser degree M4) also display a much increased ability to incorporate extend and replicate different types of unnatural nucleotide substrates that deviate to varying degrees from the canonical nucleobase structure.
  • ⁇ S substitution is the most conservative.
  • the sulfur anion is significantly larger than oxygen anion and coordinates cations poorly, which may be among the reasons why the wt enzyme will not tolerate full ⁇ S substitution.
  • Fluorescently-labelled nucleotides like ⁇ S nucleotides retain base-pairing potential but include a bulky and hydrophobic substituent that must be accomodated by the polymerase active site. Steric clashes in the active site are allievated by the presence of a long, flexible linker.
  • the hydrophobic analogue 5NI represents the most drastic departure from standard nucleotide chemistry we investigated. Comparable in size to a purine base, 5NI competely lacks any hydrogen bonding potential but like the natural bases, favours the anti-position with respect to the ribose sugar as judged by NMR (J. Gallego, D. L. and P. H., unpublished results). Therefore, a 5NI•A or 5NI•G basepair would closely resemble a purine-purine transversion mismatch and may cause similar distortions to the canonical DNA duplex geometry.
  • the 53exoD is too distant from the active site to have direct effects on mismatch extension. It is, however, thought to be crucial for polymerase processivity and may thus influence mismatch extension (24). Indeed, the Stoffel fragment of Taq polymerase (26), which lacks the 53exoD, displays both reduced processivity and more stringent mismatch discrimination (27). Mutations in the 53exoD of M1 and M4 may therefore contribute to mismatch extension by enhancing polymerase processivity. Together with the ability to bypass abasic sites (generated in large DNA fragments during thermocycling) this may also contribute to the proficiency of M1 at long PCR ( FIG. 5 ).
  • E520 is located at the very tip of the thumb domain at the end of the H2 helix at a distance of 20A from the 3′ OH of the mismatched primer terminal base (P1) (2). Mechanistic aspects of the involvement of the E520G mutation in mismatch recognition or extension are therefore not obvious either. It is worth noting though that adjacent regions, especially the preceding loop connecting helices H1 and H2 and parts of helix I, make extensive contacts with the template-primer duplex between P3-P7 (2). It has previously been observed that mismatch incorporation affects extension kinetics up to the P4 position (24). E520G may modify the structure of these regions to ease passage of mismatches and increase elongation efficiency post incorporation. Base flipping, i.e.
  • a change in the mutation spectrum towards a more even distribution of transition and transversion mutations may be an effective solution to accelerate adaptation, while maintaining a healthy distance from the error threshold.
  • This may also make M1 a useful tool for protein engineering as the bias of Taq (and other DNA polymerases) for transition mutations limits the regions of sequence space that can be accessed effectively using PCR mutagenesis TABLE 1 Mutation spectrum of wtTaq and M1 in PCR Transitions Transversions AT ⁇ > GC GC ⁇ > AT AT ⁇ > TA AT ⁇ > CG GC ⁇ > TA GC ⁇ > CG Deletions WtTaq* 25 9 8 2 3 1 3 M1* 25 16 15 4 5 10 1 *Mutations derived from sequencing of 40 clones (800 bp) each.
  • DNA polymerases according to the present invention in particular M1 and M4 respectively as depicted in SEQ No 1 and SEQ No 2 possess the following properties:
  • M1 has the ability to efficiently amplify DNA targets up to 26 kb.
  • the unusual properties of the DNA polymerases according to the present invention may have immediate uses for example for the improved incorporation of dye-modified nucleotides in sequencing and array labelling and/or the amplification of ultra-long DNA targets. They may prove useful in the amplification of damaged DNA templates in forensics or paelobiology, may permit an expansion of the chemical repertoire of aptamers or deoxi-ribozymes (Benner, Barbas, ribozyme review) and may aid efforts to expand the genetic alphabet (Benner, Schultz).
  • the altered mutation spectrum of M1 may make a useful tool in random mutagenesis experiments as the strong bias of Taq and other polymerases towards (A->G, T->C) transitions limits the combinatorial diversity accessible through PCR mutagenesis. Furthermore, the ability of M1 & M4 to extend 3′ ends in which the last base is mismatched with the template strand and the ability of H10 (see example 6) to extend 3′ ends in which the last two bases are mismatched with the template strand may extend the scope of DNA shuffling methods (Stemmer) by allowing to recombine more distantly related sequences.
  • DNA polymerases according to the invention in particular pol A polymerases, for example M1 and M4 pol A polymerases as herein described may serve as a useful framework for mutagenesis and evolution towards polymerases capable of utilizing an ever wider array of modified nucleotide substrates.
  • the inventors anticipate that directed evolution may ultimately permit modification of polymerase chemistry itself, allowing the creation of amplifiable DNA-like polymers of defined sequence thus extending molecular evolution to material science.
  • Template affinity assays An equilibrium binding assay (12) was used to determine relative affinity of polymerases for the mismatched primer-templates used in the kinetics assays. Polymerases were preincubated at 60° C. in 1 ⁇ Taq buffer with 50 nM 32 P-labeled matched primer-template and 50 nM unlabeled mismatched competitor primer-templates. Reactions were initiated by simultaneous addition of dCTP (200 ⁇ M) and trap DNA (XbaI/SalI-restricted sheared salmon sperm DNA, 4.5 mg/ml). Prior experiments demonstrated trap-effectiveness over the time period used (15 seconds).
  • Template primers 22 (undamaged) or 23 (containing a synthetic abasic site) were synthesized by Lofstrand Laboratories (Gaithersburg, Md.).
  • Template primer 24 (containing a single cis-syn thymine dimer), was synthesized as described (34).
  • Primer 25 was 32 P-labeled and annealed to one of the three templates 22, 23, 24 (at a primer template ratio of molar 1:1.5) and extended in 40 mM Tris•HCl at pH 8.0, 5 mM MgCl 2 , 100 ⁇ M of each dNTP, 10 mM DTT, 250 ⁇ g/ml BSA, 2.5% glycerol, 10 nM primer-template DNA and 0.1 Unit of polymerase at 60° C. for various times.
  • Primer 26 was 32 P-labeled and annealed to template primer 27 (containing a single 5-nitroindole) in 1 ⁇ Taq buffer, 0.1 or 0.5 U of the polymerase was added and reactions incubated at 60° C. for 15 mins, after which 40 ⁇ M of each dNTP were added and incubation at 60° C. continued for various times.
  • Mutation rates were determined using the mutS ELISA assay (Genecheck, Ft. Collins, Colo.) or by performing 2 ⁇ 50 cycles of PCR on three different templates and sequencing the cloned products.
  • Translesion synthesis Transversion mispairs represent distorting deviations from the cognate duplex structure.
  • M1 and M4 were capable of processing other deviations of the DNA structure such as lesions in the template strand.
  • Using a gel-extension assay we investigated their ability to traverse an abasic site and a cis-syn thymine pyrmidine dimer (CPD) template strand lesion.
  • CPD cis-syn thymine pyrmidine dimer
  • both M1 and M4 are able to extend past the lesion and to the end of the template.
  • the size of the final product is similar to that observed on the undamaged template indicating that bypass occurred without deletions.
  • Perhaps the most striking example of the proficiency of M1 and M4 to bypass template lesions is observed on the CPD-containing template ( FIG. 5 ).
  • wtTaq utilizes a fraction of the available template and is only able to insert a base opposite the 3′T of the dimer after prolonged reaction conditions.
  • both M1 and M4 are able to readily extend all of the primer to the 3′T of the dimer.
  • TLS trans-lesion synthesis
  • Unnatural substrates We reasoned that relaxed geometric selection might also aid the incorporation of unnatural base analogues, some of which inhibit or arrest polymerase activity due to poor geometric fit or lack of interaction with either polymerase or template strand.
  • a first, conservative example are phosphothioate nucleotide triphosphates ( ⁇ S dNTPs), in which one of the oxygen atoms in the ⁇ phosphate group is replaced by sulfur.
  • ⁇ S dNTPs are generally well accepted as substrates by DNA polymerases but when we replaced all four dNTPs with their ⁇ S counterparts in PCR wtTaq failed to generate any amplification products, while M1 (and to lesser extent M4) were able to generate PCR products of up to 2 kbp, indicating that they could utilize ⁇ S dNTPs with much increased efficiency compared to the wt enzyme ( FIG. 6 ). As expected, the resulting ⁇ S DNA was completely resistant to cleavage by DNA endonucleases (not shown). Nucleotides bearing bulky adducts such as fluorescent dyes are widely used in applications such as dye terminator sequencing or array labelling.
  • Amplification product size with wtTaq is generally limited to fragments a few kb long but can be extended to much longer targets by inclusion of a proofreading polymerase (Barnes 92).
  • a proofreading polymerase Barnes 92.
  • M1 was able to efficiently amplify of targets up to 26 kb ( FIG. 7 ), using standard PCR conditions in the absence of auxiliary polymerases or other processivity factors.
  • wtTaq enzyme failed to amplify targets >9 kb.
  • the molecular basis for the product size limitation in the wt enzyme is thought to be premature termination due to an inability to extend mismatches following nucleotide misincorporation. These are thought to be removed by the proofreading polymerase allowing extension to resume.
  • This technique allows two or more genes of interest from different species to be randomly recombined to produce chimeras, the sequence of which contains parts of the original input parent genes.
  • Thermus aquaticus (Taq) wild type and T8 (a previously selected 11 fold more thermostable Taq variant (Ghadessy, Ong et al. 2001)), Thermus thermophilus (Tth) and Thermus flavus (Tfl) polymerases had previously been amplified from genomic DNA and cloned into pASK75 (Skerra 1994) and tested for activity. These genes were then shuffled using the staggered extension protocol (StEP) as described (Zhao, Giver et al.
  • StEP staggered extension protocol
  • the library size was scored by dilution assays and determining the ratio of clones containing insert using PCR screening and was approximately 10 8 .
  • a diagnostic restriction digest of 20 clones produced 20 unique restriction patterns, indicating that the library was diverse.
  • Subsequent sequencing of selected chimeras showed an average of 4 to 6 crossovers per gene.
  • the aqueous phase was ether extracted, PCR purified (Qiagen, Chatsworth, Calif.) with an additional 35% GnHCl, digested with DpnI to remove methylated plasmid DNA, treated with ExoSAP (USB) to remove residual primers, reamplified with outnested primers and recloned and transformed into E. coli as above.
  • clone H10 has significant activity on the primers with two mismatches.
  • H10 is a chimera of T. aquaticus wild type (residues 4 to 20 and 221 to 640), T8 (residues 1 to 3 and 641 to 834) and T. thermophilus (residues 21 to 220).
  • H10 has five detectable crossover sites and 13 point mutations, of which 4 are silent (F74 ⁇ I, F280 ⁇ L, P300 ⁇ S, T387 ⁇ A, A441 ⁇ V, A519 ⁇ V Q536 ⁇ R, R679 ⁇ G, F699 ⁇ L).
  • CSR emulsification and selection was performed on the StEP Taq, Tth and Tfl library essentially as described (Ghadessy, Ong et al. 2001).
  • the library had previously been cloned into pASK75 (see example 6).
  • the aqueous phase was ether extracted and replication products were purified using a PCR purification kit (Qiagen, Chatsworth, Calif.) including a wash with an 35% GnHCl. 7 ⁇ l of purified replication products (from 48) were digested with 1 ⁇ l DpnI (20 Units) to remove plasmid DNA and treated with 2 ⁇ l ExoSAP (USB) to remove residual primers for 1 h at 37° C.
  • Reamplification products were digested with XbaI and SalI, recloned into pASK75 and transformed into E. coli as above.
  • the induced library was emulsified as above with the additional presence of biotinylated dUTP and incubated at 94° C. 5 minutes, 50° C. 1 minute and 72° C. 1 minute.
  • the aqueous phase was ether extracted, the DNA in the aqueous phase was precipitated by addition of 1/10 volume of 3M NaAc, 1 ⁇ l glycogen and 2.5 volumes of 100% ethanol. This was then incubated for 1 hour at ⁇ 20° C., spun for at 13000 rpm for 30 minutes in a benchtop microcentrifuge, washed with 70% ethanol and resuspended in 50 ⁇ l buffer EB (Qiagen).
  • the plasmid DNA of the ten best clones was then purified and shuffled as described above (StEP, (Zhao, Giver et al. 1998)). This was then purified, cut and cloned and the resultant library was subjected to another round of CSR as described (Ghadessy, Ong et al. 2001). The same two sets of mismatch primers with four mismatches at their 3′ end were used in the emulsion as two separate sources source of selective pressure. This was then dealt with as above and the resultant clones were screened and ranked by PCR assay (as above).
  • the best clones from the second round were combined with the best clones from the first round on a 96 well plate and were subjected to further screening.
  • A1 is Tth polymerase; A2 Tfl; A3 Taq; A4 M1; A5 M4; A6H10 (see previous example.
  • 1A7 to 1D12 are first round clones (where 1 indicates that these are first round clones), 2E1 to 2H12 are second round clones (where 2 indicates that these are second round clones)
  • the below protocol is a sensitive method to measure polymerase activity both for the incorporation of unnatural nucleotide substrates (added to the reaction mixture) or the extension or replication of unnatural nucleotide substrates (incorporated as part of the hairpin oligo).
  • the assay comprises a hairpin oligonucleotide which constitutes both primer and template in one.
  • a hairpin oligonucleotide which constitutes both primer and template in one.
  • a biotinylated dU residue which allows capture of the hairpin oligonucleotide on streptavidin-coated surfaces.
  • Extension reactions are carried out in the presence of small amounts of a labelled nucleotide typically DIG-16-dUTP.
  • Product is captured (for example on a streptavidin coated ELISA plate) and incorporation of labelled nucleotide into the product strand is measured (using for example an anti-DIG antibody) and taken as a measure of polymerase activity.
  • Extension reactions are carried out in 1 ⁇ Taq buffer including 1-100 nM of hairpin primer and 100 ⁇ M dNTP mixture (comprising 0.3-30% dUTP-DIG), typically incubated at 94° C. for 1-5 min, followed by incubation at 50° C. for 1-5 min, followed by incubation at 72° C. for 1-5 min. (1-10 ⁇ l) Reaction products are added to Streptavidin coated ELISA plates (Streptawell, Roche) in 200 ⁇ l PBS, 0.2% Tween20 (PBST) and incubated at room temperature for 10 min to 1 h.
  • Streptavidin coated ELISA plates Streptawell, Roche
  • PBST 0.2% Tween20
  • ELISA plates are washed 3 ⁇ in PBST and 200 ⁇ l of anti-DIG-POD Fab2 fragment (Roche) diluted 1/2000 in PBST is added and the plate is incubated at room temperature for 10 min to 1 h. The plate is washed 3-4 ⁇ in PBST and developed with an appropriate POD substrate.
  • Clones previously selected for their ability to extend from a 4 basepair mismatch were assayed for their ability to incorporate a variety of nucleotide analogues.
  • Clones were grown at 30° C. overnight in 200 ⁇ l 2 ⁇ TY+ampicillin (100 ⁇ g/ml). A 150 ⁇ l (2 ⁇ TY+ampicillin 100 ⁇ g/ml) overday culture was started from the overnight and grown for 3 hours at 37° C. After 3 hours protein expression was induced by the addition of 50 ⁇ l of 2 ⁇ TY+anhydrous tetracycline (8 ng/ml) to the culture which was then allowed to grow for a further 3 h at 37° C.
  • the cells were pelleted at 2254 ⁇ g for 5 minutes and the growth medium removed by aspiration after which the cell pellet was resuspended in 100 ⁇ l 1 ⁇ Taq buffer (10 mM Tris-HCl, pH 9.0, 1.5 mM MgCl 2 , 50 mM KCl, 0.1% Triton X-100, 0.01% (w/v) stabiliser; HT Biotechnology Ltd). Resuspended cells were lysed by incubation at 85° C. for 10 minutes and the cell debris was pelleted at 2254 ⁇ g for 5 minutes.
  • 1 ⁇ Taq buffer 10 mM Tris-HCl, pH 9.0, 1.5 mM MgCl 2 , 50 mM KCl, 0.1% Triton X-100, 0.01% (w/v) stabiliser; HT Biotechnology Ltd.
  • reaction conditions were:
  • the plate was washed 3 ⁇ in PBS-Tween and 200 ⁇ l of the substrate added (per ml 100 ⁇ l of 1M NaAc pH 6.0, 10 ⁇ L of DAB, 1 ⁇ l of H 2 O 2 , the reaction was allowed to develop after which it was stopped by adding 100 ⁇ l of 1M H 2 SO 4 .
  • Clones selected for 4-mismatch extension were assayed for activity with different substrates using an ELISA assay.
  • Polymerases 3A10 and 3D1 were investigated further for their ability to bypass abasic sites and 5-hydroxy hydantoins, which are both known to exist in damaged DNA such as found in ancient samples, using the ELISA based activity screen as described above. Both polymerases were more proficient at lesion bypass than wild type Taq by up to two orders of magnitude.
  • the hydantion phosphoramidite was synthesised by standard procedures starting from the hydantoin free base. Glycosylation of the silylated hydantoin base in the presence of tin (IV) chloride with the ditoluoyl (alpha) chlorosugar gave rise to two N-glycosylated products which were separated and characterised by 2D-NMR experiments. The tolyl groups were removed with ammonia to yield the free nucleoside which was dimethoxytritylated and phosphytylated in the usual manner.
  • the sequences of the clones referred to in Examples are shown below: For the avoidance of any doubt, the first sequence provided in each section is the nucleic acid sequence.
  • the second sequence provided is the corresponding amino acid sequence of the clone. 2F3: (SEQ ID NO: 55) ATGGCGATGCTTCCCCTCTTTGAGCCCAAGGGCCGCGTCCTCCTGGTGGACGGCCACCACCTGGCCTACCGCACCTTCTT CGCCCTGAAGGGCCCCACCACGAGCCGGGGCGAACCGGTGCAGGTGGTCTACGGCTTCGCCAAGAGCCTCCTCAAGGCC CTGAAGGAGGACGGGTACAAGGCCGTCTTCGTGGTCTTTGACGCCAAGGCCCCCTCATTCCGCCACAAGGCCTACGAGG CCTACAGGGCGGGGAGGGCCCCGACCCCCGAGGACTTCCCCCGGCAGCTCGCCCTCATCAAGGAGCTGGTGGACCTCCT GGGGTTTACCCGCCTCGAGGTCCCCGGCTACGAGGCGGACGACGTTCTCGCC
  • a list of polymerases selected to extend four mismatches were assayed for their ability to extend abasic sites in PCR ( FIG. 10 ).
  • C12 and D1 which can also extend four mismatched primers in PCR, as well as A10, B6 and B8, which cannot, all produced an amplification product.
  • a list of polymerases selected to extend four mismatches were assayed for their ability to extend abasic sites in PCR ( FIG. 10 ).
  • C12 and D1 which can also extend four mismatched primers in PCR, as well as A10, B6 and B8, which cannot, all produced an amplification product.
  • Primer extension assays were essentially as described in (Ghadessy et al., 2004). Briefly, undamaged oligonucleotides and a 51mer containing a synthetic abasic site were synthesized by Lofstrand Laboratories (Gaithersburg, Md.) using standard techniques and were gel purified prior to use.
  • Radiolabeled primer-template DNAs were prepared by annealing the 5′[ 32 P] labeled 20mer primer to one of the two following 51mer templates (at a primer template ratio of molar 1:1.5).
  • 1) undamaged DNA (UNDT51T); 5′-AGC TAC CAT GCC TGC ACG AAT TCG GCA TCC GTC GCG ACC ACG GTC GCA GCG-3′; 2) an oligo (LABA51T) containing a synthetic abasic site (indicated as an X in bold font); 5′-AGC TAC CAT GCC TGC ACG ACA XCG GCA TCC GTC GCG ACC ACG GTC GCA GCG-3′.
  • Standard replication reactions of 10 ⁇ l contained 40 mM Tris•HCl at pH 8.0, 5 mM MgCl 2 , 100 ⁇ M of each ultrapure dNTP (Amersham Pharmacia Biotech, NJ), 10 mM DTT, 250 ⁇ g/ml BSA, 2.5% glycerol, 10 nM 5′[32P] primer-template DNA and 0.1 Unit of polymerase. After incubation at 60° C. for various times reactions were terminated by the addition of 10 ⁇ l of 95% formamide/10 mM EDTA and the samples heated to 100° C. for 5 min. Reaction mixtures (5 ⁇ l) were subjected to 20% polyacrylamide/7 M Urea gel electrophoresis and replication products visualized by PhosphorImager analysis.
  • Polymerases A10 was the most active and was chosen for further analysis ( FIG. 26J RF nomenclature) on abasic sites and cyclobutane thymine-thymine dimers (CPD). A10 was clearly better at both abasic site and CPD extension and bypass than both wild type and M1.
  • MutS is an E. coli derived mismatch binding protein that binds single base pair mismatches or small (1-4 base) additions or deletions. It can be used to monitor PCR fidelity in an ELISA based assay (Debbie et al., 1997).
  • Immobilised Mismatch Binding protein plates (Genecheck, Ft Collins, USA) were used for fidelity measurements as per manufacturer's instructions, essentially as described in (Debbie et al., 1997).
  • the mutation rate of D1 was compared that of wtTaq and M1 M1 was already known to have a modestly increased mutation rate (approximately 2 fold) (Ghadessy et al., 2004).
  • the data presented here suggests that D1 has a 2 fold increased error rate compared to M1 and a four fold increased error rate compared to wtTaq. This corresponds approximately to a 1 in 2500 error ratio and is sufficiently low to not be problematic for many applications.
  • DNA recovered from ancient samples is invariably damaged, limiting the information it can yield.
  • Polymerases that can bypass damage might therefore be useful in increasing the information that can be recovered from ancient samples of DNA.
  • M1 has a slightly reduced kcat/Km, 14% of Taq wild type, and is hence slightly less efficient in PCR. Therefore, M1 was blended with a commercial preparation of Taq (SuperTaq (HT biotechnology Ltd)) in a ratio of 1 unit to 10 and compared to Taq in the absence of M1. It was hoped that if M1 could bypass the blocking lesions, then the wild type Taq would amplify the resulting translesion synthesis product. On two separate occasions, the M1/SuperTaq mix was able to produce an amplification product whereas SuperTaq alone did not (see FIG. 12 for one example)
  • the DNA was cloned and sequence and found to differ in two positions (A71 ⁇ G, 77A ⁇ G) from the expected sequence. This could either be a miscoding lesion resulting from a deamination of C or a population variant sequence not seen previously in aDNA. Indeed, both mutations exist in modern spotted hyena ( Crocuta crocuta ), arguing for the second interpretation. Of the 10 sequences obtained from the same successful PCR, two each had a further unique single mutation, an A to G in different places. These are most likely errors incurred during amplification. Such errors are frequently seen in aDNA PCR and are one reason why multiple sequences need to be obtained from the same PCR product.
  • the activity of the blend was checked against the activity of SuperTaq by a PCR activity dilution series. By this measure, the blend was less active than SuperTaq, by a factor of two.
  • the aDNA is amplified over 28 cycles with either SuperTaq or the blend.
  • the first PCR is diluted 20 fold in a secondary clean room and amplified with SuperTaq using in-nested primers. This is the approach subsequently used to compare SuperTaq and the blend
  • flanking primers (5′-CAG GAA ACA GCT ATG ACA AAA ATC TAG ATA 5NICG AGG GCA 5NI-3′, 5′-GTA AAA CGA CGG CCA GTA CCA C5NIG AAC TGC GGG TGA CGC CAA GC5NI-3′) comprising internal 5NI (or a derivative) as well as 3′ terminal 5NI (or a derivative) to increase selection pressure for 5NI replication.
  • Round 5 polymerases selected for replication of 5NI were tested for activity with a range of substrates using the hairpin ELISA assay described in example 8.
  • tUTP and ceATP were kind gifts from the laboratory of P. Herdewijin, Rega Institute, Katholieke Universiteit Leuven, Belgium. Results are shown in FIG. 14
  • ELISA protocol was a described except that The DIG labelled dUTP in the extension reaction was replaced with Fluorescein 12-dATP (Perkin-Elmer) (at 3% of dATP) and the incorporation of Fluorescein 12-dATP was detected by anti-Fluorescein-POD Fab fragments (Roche).
  • 5NITP 5-nitroindole-5′-triphosphate
  • 5NITP ⁇ Primer 5′-TAATACGACTCACTATAGGGAGA (SEQ ID NO: 114) Template 3′-ATTATGCTGAGTGATATCCCTCTXGTCA (SEQ ID NO: 116)
  • X A, T, C, G
  • the NI-NI self-pair is also formed exceptionally well, though further extension is reduced (data not shown). Similar reactions using Taq, Tth and Tfl wild-type polymerases under identical conditions leads to almost undetectable extension reactions (data not shown).
  • Targets were prepared by PCR amplification of 2.5 kb Taq gene using primers 29, 28 or 2 kb of the HIV pol gene using primers 30, 31.
  • Salmon sperm DNA (Invitrogen) was prepared at 100 ng/ul in 50% DMSO.
  • FITC and Cy5 probes were prepared by PCR amplification of 0.4 kb fragment of Taq using primers 8, 28 with either 100% (FITC100 M1 ) or 10% of dATP (FITC10 M1 , FITC10 Taq ) replaced by FITC-12-dATP or 10% of dCTP replaced by Cy5-dCTP (Cy5 Taq ).
  • Cy5 and Cy3 random 20mers were used at 250 nM.
  • Targets were purified using PCR purification kit (Qiagen) and prepared in 50% DMSO and spotted onto GAPSII aminosilane-coated glass slides (Corning) using a MicroGrid (BioRobotics). Array hybridizations were performed according to standard protocols:
  • Printed slides were baked for 2 hr at 80° C., incubated with agitation for 30 min at 42° C. in 5 ⁇ SSC/0.1% BSA Fraction V (Roche)/0.1% SDS, boiled for 2 min in ultrapure water, washed 20 ⁇ in ultrapure water at room temperature (RT), rinsed in propan-2-ol and dried in a clean airstream.
  • 50 ng of FITC- and Cy5-labelled probes were prepared in 20 ⁇ l of hybridization buffer (1 mM Tris-HCl pH7.4, 50 mM tetrasodium pyrophosphate, 1 ⁇ Denhardts solution, 40% deionised formamide, 0.1% SDS, 100 ⁇ g/ml sheared salmon sperm DNA).
  • HybriSlip (Sigma). Hybridizations were performed at 48° C. for 16 hr in a hybridization chamber (Corning). Arrays were washed once with 2 ⁇ SSC/0.1% SDS at 65° C. for 5 min once with 0.2 ⁇ SSC at RT for 5 min and twice with 0.05 ⁇ SSC at RT for 5 min. Slides were dried in a clean airstream, scanned with an ArrayWoRx autoloader (Applied Precision Instruments) and the array images analysed using SoftWoRx tracker (Molecularware).
  • the frequency of fluorophore incorporation per 1000 nucleotides (FOI) is commonly used to specify the fluorescence intensity of a probe.
  • FOIs of microarray probes commonly range from 10-50, while FITC100 M1 has an FOI of 295.
  • Mutation rates were determined using the mutS ELISA assay 26 (Genecheck, Ft. Collins, Colo.) according to manufacturers instructions. Alternatively, amplification products derived from 2 ⁇ 50 cycles of PCR of 2 targets with different GC content (HIV pol (38% GC), Taq (68% GC)) were cloned, 40 clones (800 bp each) were sequenced and mutations (wtTaq (51), M1 (75)) analyzed.
  • HAV pol 38% GC
  • Taq 68% GC
  • Termination probabilities were calculated according to the method of Kokoska et al.
  • Oligonucleotide primer 32 (5′-GCG GTG TAG AGA CGA GTG CGG AG-3′) was 32 P-labelled and annealed to the template 33 (5′-CTC TCA CAA GCA GCC AGG CAA GCT CCG CAC TCG TCT CTA CAC CGC TCC GC-3′) (at a primer/template ratio of molar 1/1.5).
  • wtTaq (0.0025 nM; 0.025 nM; 0.25 nM), M1(0.05 nM; 0.5 nM; 5 nM), and M4 (0.05 nM; 0.5 nM; 5 nM) were preincubated with the primer-template DNA substrates (10 nM) in 10 mM Tris-HCl at pH 9.0, 5 mM MgCl 2 , 50 mM KCl, 0.1% Triton X 100 at 25° C. for 15 min. Reactions were initiated by addition of 100 ⁇ M dNTPs with or without trap DNA (1000-fold excess of unlabeled primer-templates). Reactions were performed at 60° C. for 2 min.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
US11/417,403 2003-11-03 2006-05-03 DNA polymerase Abandoned US20070020653A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/538,392 US20090305292A1 (en) 2003-11-03 2009-08-10 Dna polymerase
US13/527,342 US9096835B2 (en) 2003-11-03 2012-06-19 DNA polymerase

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
GB0325650A GB0325650D0 (en) 2003-11-03 2003-11-03 Polymerase
GB0325650.0 2003-11-03
GB041087.8 2004-05-14
GB0410871A GB0410871D0 (en) 2004-05-14 2004-05-14 Polymerase
PCT/GB2004/004643 WO2005045015A2 (en) 2003-11-03 2004-11-03 Polymerase

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2004/004643 Continuation WO2005045015A2 (en) 2003-11-03 2004-11-03 Polymerase

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/538,392 Division US20090305292A1 (en) 2003-11-03 2009-08-10 Dna polymerase

Publications (1)

Publication Number Publication Date
US20070020653A1 true US20070020653A1 (en) 2007-01-25

Family

ID=34575755

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/417,403 Abandoned US20070020653A1 (en) 2003-11-03 2006-05-03 DNA polymerase
US12/538,392 Abandoned US20090305292A1 (en) 2003-11-03 2009-08-10 Dna polymerase
US13/527,342 Active 2025-01-09 US9096835B2 (en) 2003-11-03 2012-06-19 DNA polymerase

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/538,392 Abandoned US20090305292A1 (en) 2003-11-03 2009-08-10 Dna polymerase
US13/527,342 Active 2025-01-09 US9096835B2 (en) 2003-11-03 2012-06-19 DNA polymerase

Country Status (7)

Country Link
US (3) US20070020653A1 (de)
EP (3) EP1801213A3 (de)
AU (2) AU2004288017B2 (de)
CA (1) CA2544841C (de)
DK (1) DK2145956T3 (de)
ES (1) ES2546945T3 (de)
WO (1) WO2005045015A2 (de)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070048748A1 (en) * 2004-09-24 2007-03-01 Li-Cor, Inc. Mutant polymerases for sequencing and genotyping
US20080050738A1 (en) * 2006-05-31 2008-02-28 Human Genetic Signatures Pty Ltd. Detection of target nucleic acid
US20090226906A1 (en) * 2008-03-05 2009-09-10 Helicos Biosciences Corporation Methods and compositions for reducing nucleotide impurities
US20100041013A1 (en) * 2005-09-14 2010-02-18 Human Genetic Signatures Pty Ltd. Assay for a health state
US20100221785A1 (en) * 2005-05-26 2010-09-02 Human Genetic Signatures Pty Ltd Isothermal Strand Displacement Amplification Using Primers Containing a Non-Regular Base
US7833942B2 (en) 2004-12-03 2010-11-16 Human Genetic Signatures Pty. Ltd. Methods for simplifying microbial nucleic acids by chemical modification of cytosines
US7846693B2 (en) 2003-09-04 2010-12-07 Human Genetic Signatures Pty. Ltd. Nucleic acid detection assay
US20110003700A1 (en) * 2007-12-20 2011-01-06 Human Genetic Signatures Pty Ltd. Elimination of contaminants associated with nucleic acid amplification
EP2554665A2 (de) 2011-08-03 2013-02-06 Fermentas UAB DNA-Polymerasen
US8685675B2 (en) 2007-11-27 2014-04-01 Human Genetic Signatures Pty. Ltd. Enzymes for amplification and copying bisulphite modified nucleic acids
WO2017019617A1 (en) * 2015-07-24 2017-02-02 Manel Camps System for continuous mutagenesis in vivo to facilitate directed evolution
US20170029792A1 (en) * 2011-12-08 2017-02-02 Roche Molecular Systems, Inc. Dna polymerases with improved activity
US9732375B2 (en) 2011-09-07 2017-08-15 Human Genetic Signatures Pty. Ltd. Molecular detection assay using direct treatment with a bisulphite reagent
US10323243B2 (en) 2016-01-19 2019-06-18 Board Of Regents, The University Of Texas System Thermostable reverse transcriptase
US10633641B2 (en) 2011-08-10 2020-04-28 Life Technologies Corporation Polymerase compositions, methods of making and using same
US10865441B2 (en) * 2019-03-13 2020-12-15 Abclonal Science, Inc. Mutant Taq polymerase for faster amplification
US10982273B2 (en) 2016-08-09 2021-04-20 Integrated Dna Technologies, Inc. RNase H mutants in an emulsion
CN113383086A (zh) * 2019-03-10 2021-09-10 武汉爱博泰克生物科技有限公司 用于在提高的盐浓度或体液中扩增的突变型Taq聚合酶
US11208636B2 (en) 2011-08-10 2021-12-28 Life Technologies Corporation Polymerase compositions, methods of making and using same
US11613738B2 (en) 2019-05-14 2023-03-28 Abclonal Science, Inc. Mutant Taq polymerase resistant to inhibition of amplification in the presence of cyanine dye
US11866785B2 (en) 2017-10-27 2024-01-09 Board Of Regents, The University Of Texas System Tumor specific antibodies and T-cell receptors and methods of identifying the same

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2546945T3 (es) 2003-11-03 2015-09-30 Medical Research Council Polimerasa
GB0325653D0 (en) 2003-11-03 2003-12-10 Medical Res Council CST emulsions
US8962293B2 (en) * 2006-10-18 2015-02-24 Roche Molecular Systems, Inc. DNA polymerases and related methods
DK2076592T3 (en) 2006-10-23 2017-07-24 Medical Res Council POLYMERASE
US8026091B2 (en) * 2007-07-13 2011-09-27 Roche Molecular Systems, Inc. DNA polymerases and related methods
EP2524036B1 (de) * 2009-07-31 2019-06-12 Agilent Technologies, Inc. Wärmestabile dna-polymerasemutanten vom typ a mit erhöhter polymerisationsrate und resistenz gegenüber inhibitoren
ES2561885T3 (es) 2011-04-11 2016-03-01 F. Hoffmann-La Roche Ag ADN polimerasas de actividad mejorada
CN103687943B (zh) 2011-07-28 2015-11-25 霍夫曼-拉罗奇有限公司 具有改进活性的dna聚合酶
ES2569723T3 (es) 2011-12-08 2016-05-12 F. Hoffmann-La Roche Ag Polimerasas de DNA con actividad mejorada
EP2788480B1 (de) 2011-12-08 2019-01-16 Roche Diagnostics GmbH Dna-polymerasen mit verbesserter aktivität
WO2013128281A1 (en) 2012-02-28 2013-09-06 Population Genetics Technologies Ltd Method for attaching a counter sequence to a nucleic acid sample
CA2888148C (en) 2012-12-13 2019-01-15 F. Hoffmann-La Roche Ag Dna polymerases with improved activity
RS59991B1 (sr) 2013-08-08 2020-04-30 Scripps Research Inst Metoda enzimskog in vitro obeležavanja specifičnog za položaj nukleinskih kiselina uvođenjem neprirodnih nukleotida
US9758773B2 (en) * 2014-02-14 2017-09-12 Agilent Technologies, Inc. Thermostable type-A DNA polymerase mutant with increased resistance to inhibitors in blood
ES2891738T3 (es) 2014-04-09 2022-01-31 Scripps Research Inst Incorporación de nucleósidos trifosfato no naturales o modificados a células a través de transportadores de ácido nucleico trifosfato
US11091745B2 (en) 2015-05-12 2021-08-17 Dna Polymerase Technology, Inc. Mutant polymerases and uses thereof
US10280411B2 (en) 2015-10-27 2019-05-07 Pacific Biosciences of California, In.c Methods, systems, and reagents for direct RNA sequencing
ES2846949T3 (es) 2015-11-16 2021-07-30 Stratos Genomics Inc Variantes de la polimerasa DPO4
WO2017106767A1 (en) 2015-12-18 2017-06-22 The Scripps Research Institute Production of unnatural nucleotides using a crispr/cas9 system
US20190218257A1 (en) 2016-06-24 2019-07-18 The Scripps Research Institute Novel nucleoside triphosphate transporter and uses thereof
CA3061651A1 (en) * 2017-05-04 2018-11-08 Stratos Genomics, Inc. Dp04 polymerase variants
AU2018300069A1 (en) 2017-07-11 2020-02-27 Synthorx, Inc. Incorporation of unnatural nucleotides and methods thereof
MX2020001336A (es) 2017-08-03 2020-08-20 Synthorx Inc Conjugados de citoquina para el tratamiento de enfermedades autoinmunes.
CA3084812A1 (en) 2017-12-11 2019-06-20 Stratos Genomics, Inc. Dpo4 polymerase variants with improved accuracy
WO2019143622A1 (en) 2018-01-19 2019-07-25 Bio-Rad Laboratories, Inc. Mutant dna polymerases
MA54952A (fr) 2019-02-06 2022-05-11 Synthorx Inc Conjugués d'il-2 et méthodes d'utilisation de ceux-ci
US11980083B2 (en) * 2019-05-10 2024-05-07 Samsung Display Co., Ltd. Method of manufacturing thin film transistor, method of manufacturing display apparatus and thin film transistor substrate
US11879145B2 (en) 2019-06-14 2024-01-23 The Scripps Research Institute Reagents and methods for replication, transcription, and translation in semi-synthetic organisms

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030134349A1 (en) * 2000-05-24 2003-07-17 Third Wave Technologies, Inc. Enzymes for the detection of nucleic acid sequences

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1275643A (en) 1969-02-13 1972-05-24 Scragg & Sons Spin-finish condenser
JP2004521606A (ja) 2000-05-24 2004-07-22 サード・ウェーブ・テクノロジーズ・インク Rnaの検出法
GB0022458D0 (en) * 2000-09-13 2000-11-01 Medical Res Council Directed evolution method
JP2005516611A (ja) * 2001-09-14 2005-06-09 アプレラ コーポレイション Thermusscotoductus核酸ポリメラーゼ
US20030134292A1 (en) * 2001-10-30 2003-07-17 Farchaus Joseph W. Thermostable DNA polymerases and methods of making same
GB0127564D0 (en) * 2001-11-16 2002-01-09 Medical Res Council Emulsion compositions
ES2546945T3 (es) 2003-11-03 2015-09-30 Medical Research Council Polimerasa

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030134349A1 (en) * 2000-05-24 2003-07-17 Third Wave Technologies, Inc. Enzymes for the detection of nucleic acid sequences

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7846693B2 (en) 2003-09-04 2010-12-07 Human Genetic Signatures Pty. Ltd. Nucleic acid detection assay
US20070048748A1 (en) * 2004-09-24 2007-03-01 Li-Cor, Inc. Mutant polymerases for sequencing and genotyping
US8598088B2 (en) 2004-12-03 2013-12-03 Human Genetic Signatures Pty. Ltd. Methods for simplifying microbial nucleic acids by chemical modification of cytosines
US7833942B2 (en) 2004-12-03 2010-11-16 Human Genetic Signatures Pty. Ltd. Methods for simplifying microbial nucleic acids by chemical modification of cytosines
US20110136098A1 (en) * 2004-12-03 2011-06-09 Human Genetic Signatures Pty. Ltd. Methods for simplifying microbial nucleic acids by chemical modification of cytosines
US8431347B2 (en) 2005-05-26 2013-04-30 Human Genetic Signatures Pty Ltd Isothermal strand displacement amplification using primers containing a non-regular base
US20100221785A1 (en) * 2005-05-26 2010-09-02 Human Genetic Signatures Pty Ltd Isothermal Strand Displacement Amplification Using Primers Containing a Non-Regular Base
US20100041013A1 (en) * 2005-09-14 2010-02-18 Human Genetic Signatures Pty Ltd. Assay for a health state
US8343738B2 (en) 2005-09-14 2013-01-01 Human Genetic Signatures Pty. Ltd. Assay for screening for potential cervical cancer
US20080050738A1 (en) * 2006-05-31 2008-02-28 Human Genetic Signatures Pty Ltd. Detection of target nucleic acid
US8685675B2 (en) 2007-11-27 2014-04-01 Human Genetic Signatures Pty. Ltd. Enzymes for amplification and copying bisulphite modified nucleic acids
US20110003700A1 (en) * 2007-12-20 2011-01-06 Human Genetic Signatures Pty Ltd. Elimination of contaminants associated with nucleic acid amplification
US20090226906A1 (en) * 2008-03-05 2009-09-10 Helicos Biosciences Corporation Methods and compositions for reducing nucleotide impurities
US10590399B2 (en) 2011-08-03 2020-03-17 Thermo Fisher Scientific Baltics Uab DNA polymerases
EP2554665A2 (de) 2011-08-03 2013-02-06 Fermentas UAB DNA-Polymerasen
US9493848B2 (en) 2011-08-03 2016-11-15 Thermo Fisher Scientific Baltics Uab DNA polymerases
US11208636B2 (en) 2011-08-10 2021-12-28 Life Technologies Corporation Polymerase compositions, methods of making and using same
US10633641B2 (en) 2011-08-10 2020-04-28 Life Technologies Corporation Polymerase compositions, methods of making and using same
US9732375B2 (en) 2011-09-07 2017-08-15 Human Genetic Signatures Pty. Ltd. Molecular detection assay using direct treatment with a bisulphite reagent
US9890366B2 (en) * 2011-12-08 2018-02-13 Roche Molecular Systems, Inc. DNA polymerases with improved activity
US10294462B2 (en) 2011-12-08 2019-05-21 Roche Molecular Systems, Inc. DNA polymerases with improved activity
US20170029792A1 (en) * 2011-12-08 2017-02-02 Roche Molecular Systems, Inc. Dna polymerases with improved activity
WO2017019617A1 (en) * 2015-07-24 2017-02-02 Manel Camps System for continuous mutagenesis in vivo to facilitate directed evolution
US10760071B2 (en) * 2015-07-24 2020-09-01 The Regents Of The University Of California System for continuous mutagenesis in vivo to facilitate directed evolution
US10858652B2 (en) 2016-01-19 2020-12-08 Board Of Regents, The University Of Texas System Thermostable reverse transcriptase
US11912991B2 (en) 2016-01-19 2024-02-27 Board Of Regents, The University Of Texas System Thermostable reverse transcriptase
US10323243B2 (en) 2016-01-19 2019-06-18 Board Of Regents, The University Of Texas System Thermostable reverse transcriptase
US10982273B2 (en) 2016-08-09 2021-04-20 Integrated Dna Technologies, Inc. RNase H mutants in an emulsion
US11866785B2 (en) 2017-10-27 2024-01-09 Board Of Regents, The University Of Texas System Tumor specific antibodies and T-cell receptors and methods of identifying the same
CN113383086A (zh) * 2019-03-10 2021-09-10 武汉爱博泰克生物科技有限公司 用于在提高的盐浓度或体液中扩增的突变型Taq聚合酶
US11168312B2 (en) 2019-03-10 2021-11-09 Abclonal Science, Inc. Mutant taq polymerase for amplification in increased salt concentration or body fluids
CN113597468A (zh) * 2019-03-13 2021-11-02 武汉爱博泰克生物科技有限公司 用于快速扩增的突变型Taq聚合酶
US11821010B2 (en) * 2019-03-13 2023-11-21 Abclonal Science, Inc. Mutant Taq polymerase for faster amplification
US20210079365A1 (en) * 2019-03-13 2021-03-18 Abclonal Science, Inc. Mutant Taq Polymerase for Faster Amplification
US10865441B2 (en) * 2019-03-13 2020-12-15 Abclonal Science, Inc. Mutant Taq polymerase for faster amplification
US11613738B2 (en) 2019-05-14 2023-03-28 Abclonal Science, Inc. Mutant Taq polymerase resistant to inhibition of amplification in the presence of cyanine dye

Also Published As

Publication number Publication date
AU2004288017B2 (en) 2009-10-08
EP2145956B1 (de) 2015-06-17
CA2544841C (en) 2014-03-25
EP1801213A3 (de) 2007-10-24
US9096835B2 (en) 2015-08-04
CA2544841A1 (en) 2005-05-19
EP1801213A2 (de) 2007-06-27
WO2005045015A2 (en) 2005-05-19
US20140127694A1 (en) 2014-05-08
EP1685246A2 (de) 2006-08-02
ES2546945T3 (es) 2015-09-30
US20090305292A1 (en) 2009-12-10
AU2010200031B2 (en) 2013-03-07
EP2145956A2 (de) 2010-01-20
WO2005045015A3 (en) 2005-12-22
EP2145956A3 (de) 2010-02-10
DK2145956T3 (da) 2015-09-21
AU2004288017A1 (en) 2005-05-19
AU2010200031A1 (en) 2010-01-28

Similar Documents

Publication Publication Date Title
US9096835B2 (en) DNA polymerase
US7514210B2 (en) Compartmentalised self replication method for in vitro evolution of molecular libraries
US8435775B2 (en) Mutant Pfu DNA polymerase
US9783791B2 (en) Mutant reverse transcriptase and methods of use
US9228179B2 (en) Polymerase
AU2001286103A1 (en) Directed evolution method
JPH11501801A (ja) 耐熱性が向上し、かつ、プライマーエクステンションの長さと効率が向上したdnaポリメラーゼ
CA2617790A1 (en) Mutant reverse transcriptase and methods of use
JP2024028958A (ja) 不連続な複数の鋳型から相補的DNA(cDNA)を順序だてて連続的に合成するための組成物およびその方法
ZA200603524B (en) Polymerase

Legal Events

Date Code Title Description
AS Assignment

Owner name: MEDICAL RESEARCH COUNCIL, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HOLLIGER, PHILIPP;GHADESSY, FARID;D'ABBADIE, MARCV;REEL/FRAME:018659/0221;SIGNING DATES FROM 20060711 TO 20060809

STCB Information on status: application discontinuation

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