AU2002249444A1 - Amplification process - Google Patents

Amplification process

Info

Publication number
AU2002249444A1
AU2002249444A1 AU2002249444A AU2002249444A AU2002249444A1 AU 2002249444 A1 AU2002249444 A1 AU 2002249444A1 AU 2002249444 A AU2002249444 A AU 2002249444A AU 2002249444 A AU2002249444 A AU 2002249444A AU 2002249444 A1 AU2002249444 A1 AU 2002249444A1
Authority
AU
Australia
Prior art keywords
ppase
polymerase
pyrophosphate
reaction mixture
pcr
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.)
Granted
Application number
AU2002249444A
Other versions
AU2002249444B2 (en
Inventor
Duncan Roy Clark
Suzanne Patricia Vincent
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.)
UK Secretary of State for Defence
Original Assignee
UK Secretary of State for Defence
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 GBGB0110501.4A external-priority patent/GB0110501D0/en
Application filed by UK Secretary of State for Defence filed Critical UK Secretary of State for Defence
Publication of AU2002249444A1 publication Critical patent/AU2002249444A1/en
Application granted granted Critical
Publication of AU2002249444B2 publication Critical patent/AU2002249444B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Description

Amplification Process
The present invention relates to processes for carrying out reactions in which nucleic acids are amplified, to means of controlling these reactions and kits and reagents, in particular enzymes, used for conducting them.
Amplification reactions such as the polymerase chain reaction (PCR) are very well known and widely used in the fields of biptechnological research, as well as in diagnostics and detection.
PCR is a procedure for generating large quantities of a particular nucleic acid sequence, in particular a DNA sequence, and is based upon DNA' s characteristics of base pairing and precise copying of complementary DNA strands. Typical PCR involves a cycling process of three basic steps.
Denaturation : A mixture containing the PCR reagents (including the nucleic acid to be copied, which may be DNA or RNA (the template), the individual nucleotide bases (A,T,G,C), suitable primers and polymerase enzyme) are heated to a predetermined temperature to separate the two strands of the target DNA.
Annealing : The mixture is then cooled to another predetermined temperature and the primers locate their complementary sequences on the DNA strands and bind to them.
Extension : The mixture is heated again to a further predetermined temperature. The polymerase enzyme (acting as a catalyst) joins the individual nucleotide bases to the end of the primer to form a new strand of DNA which is complementary to the sequence of the target DNA, the two strands being bound together .
Such reactions rely on the sequence of steps occurring in a very precise order and at the precise temperature required for the operation of that step. A problem arises when reagents are mixed together, even for short periods of time, at different temperatures, for example prior to the start of the reaction. Primers may interact with nucleic acid template, resulting in primer extension of the template. This can lead to a reduction in the overall yield of the desired product as well as the production of non-specific products.
Various means of overcoming this problem have been proposed previously. For example, initial attempts to overcome the problem used a wax barrier to separate the various PCR reagents from each other in a test tube (see for example USP 5,565,339). The wax melted as the reaction mixture was heated to the initial denaturation temperature, allowing the reagents to mix together at the last possible moment, so that the possibility of side-reactions was minimised. Such reactions are known as "Hot Start" reactions.
Other chemical methods for achieving the suppression of side- reactions have been attempted. For example, US Patent No. 5,677,152 describes a method in which the DNA polymerase is chemically modified to ensure that it only becomes active at elevated temperatures. In order to carry out this method however, it is necessary to incubate the reaction mixture at high temperatures for some time in order to generate active enzyme. Such delays, whilst not significant in some instances, can be detrimental where the results of PCR are required rapidly. For many applications of the PCR technique it is desirable to complete the sequence of cycles in the minimum possible time. In particular for example where respiratory air or fluids or foods for human and animal stock consumption are suspected of contamination rapid diagnostic methods may save considerable money if not health, even lives.
In other methods, a monoclonal antibody to Thermus aquaticus ( Taq) DNA polymerase such as the anti-Tag DNA polymerase antibody available from Sigma, is introduced into the reaction mixture. The antibody binds to the enzyme, so as to inactivate it, at ambient temperature. However, the antibody denatures and dissociates from the enzyme at elevated temperatures used during the amplification cycles and so the enzyme becomes active. The method however does not appear to eliminate nonspecific side-products in some cases.
Primer extension of a template during a PCR reaction can be represented as :
Mg++ +
DNA polymerase DNA template + dNTP ► DNA template-dNMP + PPi
where dNTP is a deoxyribonucleic acid triphosphates, dNMP is the corresponding deoxyribonucleic acid monophosphate and PPi is an inorganic pyrophosphate. This reaction may also be represented as ( DNA) n resi ues + dNTP <-> (DNA) n+1 reSidues + PPi
The presence of increased levels of PPi, for example in a, DNA sequencing reaction is known to force the reaction shown above in reverse. This is known as pyrophosphorolysis and it is a recognised problem in DNA sequencing at 70°C using thermostable DNA polymerases . It has been overcome through the addition of a thermostable PPase to the DNA polymerase formulation used in DNA sequencing.
The applicants have found that this reaction can form the basis of an advantageous amplification reaction in which the production of non-specific products may be minimised.
According to the present invention there is provided a method for conducting a nucleic acid amplification reaction, said method comprising forming an amplification reaction mixture in the presence of sufficient of a pyrophosphate salt to prevent primer extension taking place, enzymatically digesting said pyrophosphate, and subjecting said reaction mixture to conditions such that an amplification reaction may proceed. Using the method of the invention, accurate amplification reactions, which may be carried out rapidly and with good specificity, can be carried out. It therefore represents a good alternative to existing "Hot Start" amplification technologies.
The initial amplification reaction mixture used in the method of the invention is broadly speaking, a conventional mixture, such as that used in the PCR reaction, to which pyrophosphate salt is added. Thus it will generally comprise: i) a sample which contains or is suspected of containing a target nucleic acid sequence, (ii) at least one primer which hybridises to an end region of said target sequence, iii) a source of magnesium ions, (iv) nucleotide or nucleoside bases which constitute the target sequence (i.e. A, T, C, G and/or U in the case of DNA amplification or A, U,C and G in the case of RNA amplification), and (v) a DNA polymerase which is thermostable at the temperatures at which the amplification reaction is effected. It will also comprise a buffer, as necessary in order to effect the reaction, as is known in the art.
In particular (iv) will comprise nucleotides A, T, G and C in respect of DNA amplification and nucleosides A, ϋ, C and G in respect of RNA amplification.
Other combinations may be used however, where other primer based amplifications reactions such as reverse transcriptase PCR (RT-PCR) are being conducted.
In addition, the reagents may include labelled probes or primers, and/or other labelling means such as intercalating dyes such as Sybr Green, Sybr Gold, ethidium bromide etc. or combinations of these, which might allow the application to be monitored, without the need to examine the product on a gel subsequently. The nature of these depends upon the type of assay being undertaken. Generic intercalator methods use intercalating dyes to monitor' the increase in double stranded DNA which occurs during an amplification process. These are only quasi-strand-specific and therefore other labels are required where strand specific detection is required.
Strand specific methods utilise additional nucleic acid reaction components to monitor the progress of amplification reactions . These methods often use fluorescence energy transfer (FET) as the basis of detection. One or more nucleic acid probes are labelled with fluorescent molecules, one of which is able to act as an energy donor and the other of which is an energy acceptor molecule. These are sometimes known as a reporter molecule and a quencher molecule respectively. The donor molecule is excited with a specific wavelength of light which falls within its excitation spectrum and subsequently it will emit light within its fluorescence emission wavelength. The acceptor molecule is also excited at this wavelength by accepting energy from the donor molecule by a variety of distance-dependent energy transfer mechanisms. A specific example of fluorescence energy transfer which can occur is Fluorescence Resonance Energy Transfer or "FRET". Generally, the acceptor molecule accepts the emission energy of the donor molecule when they are in close proximity (e.g. on the same, or a neighbouring molecule) . The basis of fluorescence energy transfer detection is to monitor the changes at donor and acceptor emission wavelengths.
There are two commonly used types of FET or FRET probes, those using hydrolysis of nucleic acid probes to separate donor from acceptor, and those using hybridisation to alter the spatial relationship of donor and acceptor molecules.
Hydrolysis probes are commercially available as TaqMan™ probes. These consist of DNA oligonucleotides that are labelled with donor and acceptor molecules . The probes are designed to bind to a specific region on one strand of a PCR product . Following annealing of the PCR primer to this strand, Taq enzyme extends the DNA with 5' to 3' polymerase activity. Tag enzyme also exhibits 5' to 3' exonuclease activity. TaqMan™ probes are protected at the 3' end by phosphorylation to prevent them from priming Taq extension. If the TaqMan™ probe is hybridised to the product strand, an extending Tag molecule may also hydrolyse the probe, liberating the donor from acceptor as the basis of detection. The signal in this instance is cumulative, the concentration of free donor and acceptor molecules increasing with each cycle of the amplification reaction.
US Patent No. 5,491,063 describes a method for in-solution quenching of fluorescently labelled probes which relies on modification of the signal from a labelled single stranded oligonucleotide by a DNA binding agent. The difference in this signal which occurs as a result of a reduced chain length of the probe following probe cleavage (hydrolysis) during a polymerase chain reaction is suggested for providing a means for detecting the presence of a target nucleic acid.
Hybridisation probes are available in a number of forms. Molecular beacons are oligonucleotides that have complementary 5' and 3' sequences such that they form hairpin loops. Terminal fluorescent labels are in close proximity for FRET to occur when the hairpin structure is formed. Following hybridisation of molecular beacons to a complementary sequence the fluorescent labels are separated, so FRET does not occur, and this forms the basis of detection.
Pairs of labelled oligonucleotides may also be used. These hybridise in close proximity on a PCR product strand bringing donor and acceptor molecules together so that FRET can occur. Enhanced FRET is the basis of detection. Variants of this type include using a labelled amplification primer with a single adjacent probe. US Patent No. 4,868,103 describes in general terms, a FRET system for detecting the presence of an analyte, which utilises an intercalating dye as the donor molecule. The process does not involve an amplification stage.
Other examples of assays which utilise FET or FRET detection are described in WO 99/28500, which utilises a combination of an intercalating dye and a single labelled probe as a signalling system, WO 99/28501 which utilises a combination of a labelled primer and an enzyme to generate a detectable fluorescent signal, and WO 99/42611 which uses a combination of an intercalating dye and a fluorescently labelled nucleotide as the basis of the signal. Yet further assays which utilise complex primers including labels and chemical blocking agents and which are complementary are described for example in WO 99/66071.
Reaction mixtures used in the method of the invention may indlude any of the labelling reagents necessary to conduct assays as described above. In particular, such reaction mixtures may advantageously be used in genotyping and, more especially, in SNP evaluation. In these instances, the method of the present invention is used in combination with dual Taqman™ probes, one specific for the basic sequence and one specific for the mutant. Each probe preferably contains a different flurophore and therefore different signals are generated depending on the amount of the various forms of the gene present. A single signal is generated from a homozygote and a mixed signal is generated from a heterozygote.
Examples of suitable DNA polymerases which may be used in the context of the invention are thermostable polymerases such as Thermus aquaticus polymerase { Taq) , Thermus thermophilus polymerase { Tth) , Thermus species NH polymerase ( TspΝH) , Thermus brockianus polymerase {Tbr) (all obtainable for example from GeneSys Limited, Farnborough, U. K. ) , Pyrococcus furiosus polymerase { Pfu) (obtainable from Stratagene) , 9°Ν7 exo-DNA polymerase, and Thermococcus litoralis DNA polymerase (obtainable from New England Biolabs as VENT™ DNA polymerase) .
The pyrophosphate used in the method of the invention may be any soluble pyrophosphate including soluble metal and non-metal (e.g. ammonium salts) . Such compounds are often generically known as "inorganic pyrophosphate" or PPi and this nomenclature is used in the present application. In particular, the pyrophosphate will be an alkali metal pyrophosphate, such as sodium or potassium pyrophosphates including disodium pyrophosphate (Na2H2P2θ7) , anhydrous tetrasodium pyrophosphate (Na4P2θ7) , tetrasodium pyrophosphate decahydrate ( a4P2O7.10H2O) and tetrapotassium pyrophosphate (anhydrous) . Other soluble pyrophosphates which may be used include iron pyrophosphates such as ferric pyrophosphate (Fe (P207)3) , and soluble ammonium salts such as anhydrous tributylammonium pyrophosphate. Other soluble pyrophosphates are available from commercial sources .
A preferred inorganic pyrophosphate is tetrasodium pyrophosphate of formula NaP207.
The concentration of pyrophosphate used in the reaction mixture should be sufficient to prevent primer extension taking place. This will depend to a large extent upon the particular nature and concentration of the sequences being amplified, the primers and the polymerase enzymes being used, as well as their concentrations, and may be determined in any particular case by routine methods .
The reaction mixture formed initially suitably contains pyrophosphate at a concentration of at least 0.5mM, suitably at a concentration of least ImM, for example from 1-lOmM and preferably from l-5mM.
Enzymatic digestion of the inorganic pyrophosphate is suitably effected immediately prior to or during first phase of the amplification reaction. This may be achieved by addition of an pyrophosphatase enzyme (PPase) (which may be known as an inorganic pyrophosphatase enzyme - PPiase) immediately prior to the start of the amplification reaction.
Preferably, however, the enzymatic digestion is effected using a thermostable PPase, which is active at elevated temperatures, for example at temperatures in excess of 50°C. Preferably the enzyme is only significantly active at these elevated temperatures. This means that the PPase may be included in the reaction mixture on formation, but it will not or not significantly digest the inhibitory pyrophosphate at ambient temperature. It will only become properly active when the reaction mixture is heated as will be necessary for example during the initial denaturation phase of a PCR reaction. However, a short preliminary incubation at elevated temperature, for example at from 50 to 100°C, and, preferably, at from 80 to 95°C, may be carried out.
Examples of thermostable PPase include Sulfolbus acidicaldarius pyrophosphatase, (Sac PPase - Meyer et al. Achives of Biochem. and Biophys. (1995) 319, 1, 149-156) obtainable from GeneSys Limited, Farnborough UK., or Thermococcus litoralis pyrophosphatase, available from New England Biolabs (Catalogue nos #M0296S and #M0296L) . Preferably the thermostable PPase is Aeropyrum pernix inorganic pyrophosphatase obtainable from Genesys Limited, Farnborough UK.
Aeropyrum pernix Kl, the first strictly aerobic hyperthermophilic archeaon, was isolated in 1993 from a coastal solfataric thermal vent at Kodaka ra-Jima Island, Japan, (Sako et al, Int. J. Syst. Bacteriol. 46 (1996): 1070-1077. It is deposited in the Japan Collection of Microorganisms, JCM 9820.
The applicants have for the first time isolated a thermostable PPase from Aeropyrum pernix and this forms a further aspect of the present invention. The genomic sequence comprising this pyrophosphatase is shown in SEQ ID NO. 1 and the corresponding amino acid sequence is shown in SEQ ID NO. 2 (Figure 11 hereinafter) . In particular the enzyme of the invention has the amino acid sequence as shown as SEQ ID NO 25, . which is encoded by the region of SEQ ID NO 1 shown in bold type in Figure 11, and represented also as SEQ ID NO 26.
The present invention, therefore, includes a polynucleotide comprising SEQ ID NO 26 and variants or fragments thereof. For example, the invention provides a polynucleotide of SEQ ID NO l .
The present invention further includes an amino acid sequence comprising SEQ ID NO 25 and variants or fragments thereof. For example, the amino acid sequence may comprise SEQ ID NO 2.
The term "fragment thereof" as used herein in relation to a polynucleotide sequence refers to any portion of the given polynucleotide sequence which has the same activity as the complete polynucleotide sequence. Fragments will suitably comprise at least 300 and preferably at least 450 consecutive bases from the basic sequence.
The term "variant thereof" in relation to a polynucleotide sequences means any substitution of, variation of, modification of, replacement of deletion of, or the addition of one or more nucleic acid(s) from or to a polynucleotide sequence providing the resultant protein sequence encoded by the polynucleotide exhibits the same properties as the protein encoded by the basic sequence. The term therefore includes allelic variants and also includes a polynucleotide which substantially hybridises to the polynucleotide sequence of the present invention. Preferably, such hybridisation occurs at, or between low and high stringency conditions. In general terms, low stringency conditions can be defined as 3 x SSC at about ambient temperature to about 55°C and high stringency condition as 0.1 x SSC at about 65°C. SSC is the name of the buffer of 0.15M NaCl. 0.015M tri-sodium citrate. 3 x SSC is three times as strong as SSC and so on.
Typically, variants have 62% or more of the nucleotides in common with the polynucleotide sequence of the present invention, more typically 65%, preferably 70%, even more preferably 80% or 85% and, especially preferred are 90%, 95%, 98% or 99% or more identity.
When comparing nucleic acid sequences for the purposes of determining the degree of identity, programs such as BESTFIT and GAP (both from Wisconsin Genetics Computer Group (GCG) software package) . BESTFIT, for example, compares two sequences and produces an optimal alignment of the most similar segments. GAP enables sequences to be aligned along their whole length and fins the optimal alignment by inserting spaces in either sequence as appropriate. Suitably, in the context of the present invention when discussing identity of nucleic acid sequences, the comparison is made by alignment of the sequences along their whole length.
The term "fragment thereof" as used herein in relation to an amino acid sequence refers to any portion of the given amino acid sequence which has the same activity as the complete amino acid sequence. Fragments will suitably comprise at least 100 and preferably at least 150 consecutive amino acids from the basic sequence.
The term "variant thereof" as used herein in relation to an amino acid sequence means sequences of amino acids which differ from the base sequence from which they are derived in that one or more amino acids within the sequence are substituted for other amino acids. Amino acid substitutions may be regarded as "conservative" where an amino acid is replaced with a different amino acid with broadly similar properties. Non-conservative substitutions are where amino acids are replaced with amino acids of a different type. Broadly speaking, fewer non- conservative substitutions will be possible without altering the biological activity of the polypeptide. Suitably variants will be at least 60% identical, preferably at least 75% identical, and more preferably at least 90% identical to the base sequence.
Homology in this instance can be judged for example using the algorithm of Lipman-Pearson, with Ktuple:2, gap penalty: 4, Gap Length Penalty: 12, standard PAM scoring matrix (Lipman, D.J. and Pearson, W.R., Rapid and Sensitive Protein Similarity Searches, Science, 1985, vol. 227, 1435-1441).
Preferably, the polynucleotide of the present invention comprises SEQ ID NO 26 and sequences having greater than 62% identity thereto.
These enzymes may be obtained from the natural source, or may be expressed in recombinant host cells, such as E. coli cells, using conventional methods.
Removal of pyrophosphate for example, at >50°C by the action of a thermostable pyrophosphatase enzyme (PPase) then allows primer extension (and therefore amplification) to proceed as normal. During this process, lmole of pyrophosphate is converted to 2moles of inorganic phosphate (Pi) , which does not interfere with the amplification reaction.
The amount of pyrophosphatase included should be sufficient to digest excess pyrophosphate salt present in the reaction mixture. Generally speaking, this will be greater than the amounts of these enzymes used conventionally in an equivalent cycle reaction to prevent pyrophosphorolysis, for example some 5 fold more. The precise amounts will depend upon various factors including the particular enzyme being used, the concentration of the pyrophosphate etc. Typically, PPase and particularly thermostable PPase enzymes will be included in the amplification reaction mixture at concentrations of at least 0.04 units per 50μL PCR reaction mixture, preferably at least 0.08 units per 50μL PCR reaction mixture and more preferably from about 0.2-10 units per 50μL PCR reaction mixture. In this case, one unit is defined as the amount of enzyme catalysing the conversion of lμmol pyrophosphate into 2μmol orthophosphate in one minute at 75°C under the following reaction conditions: ImM K4P207, 2mM MgCl2, 50mM Tris-HCl, pH 9.0 (25°C) .
Enzymes used in the method of the invention can result in rapid removal of inorganic pyrophosphate, depending upon the temperature being used. Generally complete removal can be achieved in less than 5 minutes, more often, in less than 2 minutes and as little as 15 seconds if required.
Once the inorganic pyrophosphate has been enzymatically removed from the reaction mixture, the amplification reaction can proceed, for example using a conventional thermal cycling procedure .
The mechanism by which the method of the invention achieves the desired result is not clear. It is probable that the 'presence of excess pyrophosphate inhibits the primer extension reaction. There appears, however, to be no noticeable decrease in PCR sensitivity or product yield.
The method of the invention can be conducted in any conventional apparatus for conducting application reactions . These include conventional block heating devices as described for example in EP-A-0810030 and supplied by The Perkin-Elmer Corporation, or rapid hot air thermal cyclers such as the RapidCycler™ and LightCycler™ from Idaho Technologies Inc. or other types of thermal cycler such as those described in W098/24548.
According to a further aspect, the invention provides a kit for conducting an amplification reaction,' said kit comprising an inorganic pyrophosphate, an inorganic pyrophosphatase enzyme, and optionally one or more reagents required for use in an amplification reaction. The inorganic pyrophosphate is suitably present in a sufficient amount to inhibit an amplification reaction, as described above. Preferably the amount of inorganic pyrophosphatase enzyme present in the kit is sufficient to digest all of the said inorganic pyrophosphate .
The one or more reagents include any one of reagents (ii) to (v) listed above, and may also include buffers. Particular examples of inorganic pyrophosphatase enzymes are thermostable inorganic pyrophosphatase enzymes as described above.
In particular, the kits may suitably comprise as an optional additional reagent, one or more primers required to conduct amplification of a particular target DNA sequence, for example, a sequence, which is diagnostic of a particular disease condition or the presence of a particular pathogen in a sample. The methods may also be used in the detection of polymorphisms or allelic variations in genetic analysis.
Furthermore, the kits may comprise one or more labelled reagents such as intercalating dyes, or fluorescently labelled probes, primers or nucleotides, which may be useful in detecting or monitoring the amplification reaction in situ.
In a further aspect, the invention provides the use of an inorganic pyrophosphate as described above, in a method for carrying oμt amplification reactions as described above. Preferably, the inorganic pyrophosphatase enzyme is from Aeropyrum pernix.
Finally, in yet a further aspect, the invention provides the use of an inorganic pyrophosphatase enzyme as described above, in a method for carrying out amplification reactions as described above. The invention will now be particularly described by way of example with reference to the accompanying diagrammatic drawings in which
Figure 1 shows the results of conducting a PCR in the presence of various amounts of PPi where PPi is tetrasodium pyrophosphate;
Figure 2 shows the effect of increasing MgCl2 in the absence and presence of 3mM PPi;
Figure 3 shows the results obtained using the method of the invention and conventional PCR reaction;
Figure 4 shows the results obtained using the method of the invention in an assay compared to a conventional PCR assay;
Figure 5 shows the results of an experiment to test the storage stability of PCR reaction mixtures used in the method of the invention, as compared to conventional mixtures;
Figure 6 shows the results of the use of a different PPase in the method of the invention;
Figure 7a and 7b and Figures 8a and 8b show the results of PCR experiments using the method of the invention and a variety of different DNA polymerases;
Figure 9 shows the results of an experiment comparing a conventional "Hot Start" PCR with the method of the invention;
Figure 10 shows the results obtained by carrying out a similar assay but using an alternative conventional PCR;
Figure 11 shows the genomic sequence of Aeropyrum pernix shown as SEQ ID NO. 1 (Referenced as NC 000854 in GenBank BA000002) , the corresponding amino acid sequence SEQ ID NO.2 and the sequence of the enzyme (SEQ ID NO 25) ;
Figure 12 shows an alignment of different PPase sequences (SEQ ID NOS 2 to 9) , including the protein sequence of Aeropyrum pernix shown as SEQ ID NO. 2;
Figure 13 shows the 686 base pair PCR product (SEQ ID NO 10) produced during isolation of the pyrophosphatase enzyme from Aeropyrum pernix;
Figure 14 shows the polylinker sequence (SEQ ID NO 11) used in the isolation of the pyrophosphatase from Aeropyrum pernix;
Figure 15 shows the sequence of the pTTQ18NHK vector (SEQ ID NO 12) used in the isolation of the pyrophosphatase from Aeropyrum pernix;
Figure 16 shows the sequence (Z = stop) of the pTTQlθNHK vector including the PPase sequence used in the isolation of the pyrophosphatase from Aeropyrum pernix (SEQ ID NO 13) ; and
Figure 17 shows the results of the method of the invention using the inorganic pyrophosphatase from Aeropyrum pernix.
Example 1
Effect of PPi on PCR
Using Taq DNA polymerase, a standard 500bp lambda template PCR using the following reagents, was conducted in the presence of differing quantities of the inorganic pyrorphosphate, tetrasodium pyrophosphate decahydrate (PPi) .
Lambda 500bp Primer sequences
5' Primer GAT GAG TTC GTG TCC GTA CAA CTG G (SEQ ID NO 14) 3' Primer GGT TAT CGA AAT CAG CCA CAG CGC C (SEQ ID NO 15)
1 x Reaction Buffer: lOmM Tris. pH 8.0, 50mM KCl. PCR conditions for the assay were as follows: i) 94°C 3.00 min ii) 20 cycles of 94°C for 10 sees
50°C for 10 sees 72°C for 30 sees iii) 72°C for 7 mins iv) 25°C hold,
The PPi was added such that the final concentration in the reaction mixture was 0, 1, 2, 3, 4 and 5mM. The results are shown in Figure 1. In this Figure, the lanes correspond to the following concentrations of PPi
Lanes
1 + 2 0 PPi
3 + 4 ImM PPi
5 + 6 2mM PPi
7 + 8 3mM PPi
9 +10 4mM PPi
11 5mM PPi
At all levels of PPi tested, no PCR product was produced. Example 2
Effect of increasing magnesium ion concentration Mg binds to PPi and therefore it is possible that the observations of Example 1 are due to chelation of Mg by excess PPi. This would lead to insufficient Mg being present to allow primer extension to proceed. In order to eliminate this possibility, the procedure of Example 1 with 3mM PPi was repeated in the presence of various concentrations of magnesium ions .
The results are shown in Figure 2. In that Figure the lanes represent the following reactions:
Lanes
1 + 2 1 . 5mM MgCl2
3 + 4 5mM MgCl2
5 + 6 7 . 5mM MgCl2
7 + 8 lOmM MgCl2
9 + 10 1.5mM MgCl2 + 3mM PPi
11 + 12 5mM MgCl2 + 3mM PPi
13 + 14 7.5mM MgCl2 + 3mM PPi
15 lOmM MgCl2 + 3mM PPi
The results show that the addition of Mg++ up to lOmM final concentration (1.5mM is standard in a PCR) does not allow PCR to occur, suggesting that it is the PPi which is blocking primer extension.
Example 3 PCR reactions in the presence of Ppi and PPase
The 500bp lambda PCR of Example 1 was repeated, but this time, 0.2u of Sulfolobus acidocaldarius PPase { Sac PPase) was included in reactions containing pyrophosphate (PPi) . Incubating the reaction at 95°C for 5 mins in the presence of 0.2u of Sac PPase was sufficient to destroy the- pyrophosphate so that the PCR reaction could proceed. The results are shown in Figure 3 where the lanes represent the following reactions:
Lanes
Top Row
1 + 2 ImM PPi + 0.2u PPase
3 + 4 2mM PPi + 0.2u PPase
5 + 6 3mM PPi + 0.2u PPase
7 + 8 4mM PPi + 0.2u PPase
9 + 10 5mM PPi + 0.2u PPase
Bottom Row
1 + 2 ImM PPi 3 + 4 2mM PPi 5 + 6 3mM PPi 7 + 8 4mM PPi 9 + 10 5mM PPi 11 + 12 OmM PPi
A comparable level of PCR product was generated when compared to the reaction without both PPi and PPase.
The example was repeated using concentrations of PPi of less than ImM. Results (not shown) indicated that 0.4mM PPi did not completely suppress the PCR, but no PCR occurred at concentrations of O.δmM
Example 4 PCR Assay
The method of the invention was then applied to an assay system that requires a "HotStart" reaction in order to generate a PCR product of the correct size.
The assay is based around the amplification of a 321bp fragment of the human angiotensin gene. It has been recognised that the assay will only generate the correct amplification product in the presence of betaine (EP-A-0962526 - see in particular Example 8) .
Without betaine a HotStart DNA polymerase generates few non- specific amplification products or no products at all whereas a non-HotStart DNA polymerase PCR generates a large number of non-specific amplification products.
The PCR conditions used in the Angiotensin assay can be summarised as follows.
Reagent Volume Final concn. lOx Reaction Buffer 5μl lx
25mM MgCl2 3μl 1.5mM
5mM dNTPs 2μl 200μM
5' primer (lOOμm) 0.25 0.5/μM
3' primer (lOOμm) 0.25 0.5/μM
Template lOOng/μl 50ng Human xso al DNA
5M Betaine 10.0 IM
DNA polymerase (5u/μl) 0.25 1.25u
Water to Total volume 50. Oμl
Angiotensin primer sequences
5' Primer GCA ACG CCC CTC ACT ATA AA (SEQ ID NO 16) 3' Primer GCA CCC CGC CCT TGA AGT CC (SEQ ID NO 17)
1 x Reaction Buffer: lOmM Tris. pH 8.0, 50mM KCl. PCR conditions for the assay were as follows: i) 95°C 2.00 min or less ii) 35 cycles of 95°C for 15 sees
50°C for 30 sees 72°C for 30 sees iii) 72°C for 7 mins iv) 25°C hold The reaction was conducted using a PE9700 Instrument in the presence of 3mM PPi and 0.2u PPase as described in Example 3.
The results are shown in Figure 4 in which the Lanes shown represent the following reactions .
Lanes
1 Standard Taq polymerase PCR - without betaine - lots of false priming
2 Standard Taq polymerase PCR - with betaine - bright band is correct product - with some false priming
3 Standard Taq polymerase PCR - without betaine but plus 3mM PPi and 0.2u Sac PPase - No false priming at all - 5mins denaturation at 95°C
4 Standard Taq polymerase PCR - with betaine but plus 3mM PPi and 0.2u Sac PPase - only correct product - 5mins denaturation at 95°C
5 + 6 As per 3 but only 2 mins denaturation at 95°C 7 + 8 As per 4 but only 2 mins denaturation at 95°C
It is clear that using the method of the invention, an effective "HotStart" reaction is achieved. A clear single product band was obtained using PPi - and Sac PPase in the presence of betaine. In addition, no false priming was seen, even in the absence of betaine.
Example 5 Effects of Storage at Ambient Temperature
The effect of leaving a PCR mixture containing 0.2u Sac PPase and 3mM PPi at room temperature 20°C for various lengths of time prior to conducting the Angiotensin assay, was investigated. Although Sac PPase is a thermostable enzyme, it was possible that there would be a small level of enzyme activity at ambient temperatures. This might lead to insufficient PPi in the reaction to inhibit/stop the DNA polymerase leading to primer extension and lack of "HotStart" functionality. The method of Example 4 was repeated but the reaction mixtures were stored at ambient temperature for various lengths of time up to 2 hours prior to conducting the assay.
The results are shown in Figure 5 in which:
The Top Row - shows the results of a conventional Taq polymerase PCR of angiotensin (with and without betaine present) following incubation of reagents at room temperature for the time shown; and
The Bottom Row shows the results of a similar set of assays in accordance with the method of the invention where, in all cases, the assay mix contained 3mM PPi and 0.2u PPase per 50μl PCR.
Lanes Presence of betaine Time at 22°C (Room Temp)
1 + 2 - 0
3 + 4 + 0
5 + 6 - 30 mins
7 + 8 + 30 mins
9 + 10 - 60 mins
11 + 12 + 60 mins
13 + 14 - 120 mins
15 + 16 + 120 mins
Even after two hours, assay conducted in accordance with the present invention functioned as expected, suggesting there is insufficient ambient temperature digestion of the PPi by the Sac PPase.
The result shown in Figure 5 showed that a 2 hour incubation of the PCR mix at room temperature, prior to PCR, had no effect on the specificity providing PPi and Sac PPase was used. Example 6
Use of other thermostable PPase enzymes in the method of the invention
The assay described in Example 4 was repeated alongside a similar reaction using a different commercially available thermostable PPase (with different unit definition of activity) in place of Sac PPase. The results are shown in Figure 6 in which the lanes represent the following reactions:
Lanes
1 + 2 Standard Taq polymerase PCR - without betaine
3 + 4 Standard Taq polymerase PCR - with betaine
5 + 6 Standard Taq polymerase PCR - without betaine but plus
3mM PPi and 0.2u Sac PPase 7 + 8 Standard Taq polymerase PCR - with betaine plus 3mM PPi and 0.2u Sac PPase 9 + 10 Standard Taq polymerase PCR - without betaine but plus
3mM PPi and lOu* Thermococcus litoralis PPase 11 + 12 Standard Tag polymerase PCR - with betaine plus 3mM PPi and lOu* Thermococcus litoralis PPase
* Units used in this case were as supplied by the manufacturer and are defined as the amount of enzyme that will generate 40nmoles of phosphate per minute under standard reaction conditions (10 minute reaction at 75°C in 50mM Tricine [pH 8.5], ImM MgCl2, 0.32mM PPi, reaction volume of 0.5ml).
Thermococcus litoralis PPase (available from New England
Biolabs) appears to have the same effect as Sac PPase in this assay.
Example 7
Use of different thermostable DNA polymerases in the method of the invention
A variety of thermostable DNA polymerases were employed in the method of the invention and some comparative assays. These included several non-proofreading Thermus sp. DNA polymerases, proof-reading hyperthermophilic archael DNA polymerases and mixes of non-proofreading and proofreading DNA polymerases.
They were all tested using the 500bp lambda PCR as described in Example 1 (Figure 7a and 7b) , and several using the Angiotensin assay as described in Example 4 (Figure 8a and 8b) .
Details of the assay conditions are summarised as follows:
Figure 7a - Thermus DNA polymerases
Lanes
Top Row
1 + 2 Taq polymerase OmM PPi and no PPase
3 + 4 Taq polymerase 3mM PPi and no PPase
5 + 6 Taq polymerase 3mM PPi and 0.2u Sac PPase
7 + 8 TJr polymerase OmM PPi and no PPase
9 + 10 Tjr polymerase 3mM PPi and no PPase
11 + 12 TJr polymerase 3mM PPi and 0.2u Sac PPase
Bottom Row
I + 2 Tth polymerase OmM PPi and no PPase 3 + 4 Tth polymerase 3mM PPi and no PPase 5 + 6 Tth polymerase 3mM PPi and 0.2u Sac PPase 7 + 8 TspNH polymerase OmM PPi and no PPase
9 + 10 TspNH polymerase 3mM PPi and no PPase
II + 12 TspNH polymerase 3mM PPi and 0.2u Sac PPase
Figure 7b - Archael Proof-reading DNA polymerases
Lanes
Top Row
1 + 2 Pfu polymerase OmM PPi and no PPase
3 + 4 Pfu polymerase 3mM PPi and no PPase
5 + 6 Pfu polymerase 3mM PPi and 0.2u Sac PPase
7 + 8 9°N exo- polymerase OmM PPi and no PPase
9 + 10 9°N exo- polymerase 3mM PPi and no PPase
11 + 12 9°N exo- polymerase 3mM PPi and 0.2u Sac PPase
Bottom Row
1 + 2 VENT polymerase OmM PPi and no PPase 3 + 4 VENT polymerase 3mM PPi and no PPase 5 + 6 VENT polymerase 3mM PPi and 0.2u Sac PPase
Fig 8a Angiotensin assay without PPi and without Sac PPase (with and without Betaine)
Lanes
1 + 2 Taq polymerase without betaine
3 + 4 Tag polymerase with betaine
5 + 6 Accurase polymerase without betaine
7 + 8 Accurase polymerase with betaine
9 + 10 Tbr polymerase without betaine
11+ 12 Tbr polymerase with betaine
13 + 14 Tth polymerase without betaine
15 + 16 Tth polymerase with betaine
Fig 8b Angiotensin assay with PPi and Sac PPase (with and without Betaine) Control Lanes 1-4 (Top Row) and 12-16 (Bottom Row)
Lanes
Top Row
1 + 2 Taq polymerase without betaine but plus 3mM PPi
No Sac PPase 3 + 4 Tag polymerase with betaine but plus 3mM PPi -
No Sac PPase
All below with 3mM PPi and 0.2u Sac PPase
5 + 6 Taq polymerase without betaine
7 + 8 Tag polymerase with betaine
9 + 10 Accurase polymerase without betaine
11 + 12 Accurase polymerase with betaine
13 + 14 TJbr polymerase without betaine
15 + 16 TJbr polymerase with betaine
Bottom Row
All below with 3mM PPi and 0.2u Sac PPase
1 + 2 Tth polymerase without betaine
3 + 4 Tth polymerase with betaine
5 + 6 TspNH polymerase without betaine
7 + 8 TspNH polymerase with betaine
9 + 10 Pfu polymerase without betaine
11 + 12 Pfu polymerase with betaine
13 + 14 Tag polymerase control without betaine and no PPi or PPase 15 + 16 Tag polymerase control with betaine and no PPi or
PPase
All DNA polymerases tested were inhibited by PPi and that inhibition could be overcome with Sac PPase. Comparative Example 8
Comparison of method of invention with conventional "Hotstart" methodologies
We have some initial results (Figure 9 and 10) that show that a chemically modified Tag polymerase (modified as described in US Patent No 5,677,152) does generate some false PCR products in the absence of betaine but gives the correct product in the presence of betaine.
Figure 9 Angiotensin assay
Lanes
1 + 2 Tag polymerase without betaine
3 + 4 Tag polymerase with betaine
5 + 6 Chemically modified Tag without betaine
7 + 8 Chemically modified Tag with betaine
9 + 10 Method of the invention (3mM PPi and 2u Sac PPase) without betiane 11 + 12 Method of the invention (3mM PPi and 2u Sac' PPase) with betaine
It appears that under these circumstances, the chemically modified enzyme is inactive until it has a 10 min activation at 95°C. Without this preliminary incubation, negligible PCR product was generated. The apparent false priming and generation of wrong PCR products in the absence of betaine is difficult to explain however, since the chemically modified Tag is inactive at room temperature.
Figure 10 Angiotensin assay with Tag and anti-Tag antibody
Lanes
1 + 2 Anti-Tag antibody plus Tag polymerase without betaine 3 + 4 Anti-Tag antibody plus Tag polymerase with betaine
In an anti-Tag DNA polymerase antibody mediated HotStart, a substantial number of false products are generated in the absence of betaine (similar to a standard Taq polymerase PCR without betaine) and a minor false product is also generated along with the correct product in the presence of betaine.
The method of the invention appears to give a rapid PCR reaction which is more specific than both of these commercial HotStart methodologies .
Example 9 Isolation of inorganic pyrophosphatase from Aeropyrum pernix Aeropyrum pernix was obtained from the J.C.M. culture collection. The inorganic pyrophosphatase enzyme was cloned, expressed and purified.
Cloning and expression of inorganic pyrophosphatase from A. pernix
The genome sequence comprising the pyrophosphatase gene of Aeropyrum pernix is shown in Figure 11. The primers used were designed from the genome sequence of Aeropyrum pernix. These are shown below as 5' to 3' with the restriction sites shown in bold.
Upper primer, introducing the Nde I site: (SEQ ID NO 18)
TGCATGCATATGACAGGCTGTCTGAAAATTG
Lower primer, introducing the Hind III site: (SEQ ID NO 19)
TAAGTGTAAGCTTGACTGTGGGGGCGGTGAAAG
Aligning the putative sequence from the genome with other pyrophosphates genes suggested that a later ATG should be the start methionine and not the one shown in the databank (shown in italics in SEQ ID NO.l in Figure 11) and that the amino acid sequence of the enzyme is, in fact, as shown in SEQ ID NO 25. Primers were therefore designed corresponding to the later methionine (shown in bold in SEQ ID NO.l in Figure 11). A PCR was run using lOOng of the Aeropyrum pernix DNA in a lOOμl volume with 50pM of the above primers. 20 cycles were run with 55°C annealing and a 45 second extension time.
Initial hold of 3mins at 94°C
20 Cycles of 94°C, 10 sees, 55°C, 10 sees, 68°C, 45 sees.
Final hold of 72°C 7 minutes
PCR conditions.
50pM Upper Primer (5' .. TGCATGCATATGACAGGCTGTCTGAAAATTG..3' -SEQ
ID NO 18) 5OpM Lower Primer (5' .. TAAGTGTAAGCTTGACTGTGGGGGCGGTGAAAG..3' -
SEQ ID NO 19)
1.5mM MgCl2
1.25u Accurase DNA polymerase (Cat. No. AC001, GeneSys Ltd.)
75mM Tris, pH 8.8 2OmM Ammonium sulphate
0.1% (w/v) Tween20 lOOng Aeropyrum pernix genomic DNA
The PCR product was 686 base pairs long as shown in Figure 13. The PCR product was Prepanol™ (Cat. No. P001, GeneSys Ltd.) precipitated following the manufacturers recommended conditions and finally re-suspended in lOmM Tris, O.lmM EDTA.
The PCR product was digested with restriction enzymes Nde I and Hind III, phenol extracted, precipitated with ethanol and re- suspended in lOmM Tris, O.lmM EDTA.
pTTQ18NHK vector (shown in Figure 15) had also been digested with Nde I and Hind III, phenol extracted, ethanol precipitated and re-suspended in lOmM Tris, O.lmM EDTA.
lOOng cut PCR sequence was ligated with lμg of cut pTTQlδNHK vector (see Figure 16) in a total volume of lOμl, overnight at 16°C in lχ NEB ligation buffer using 200u of New England Biolabs T4 DNA ligase. The plasmid vector was pTTQ18NHK, a modified form of vector pTTQ18 (Stark MJ, Gene, 1987; 51(2- 3) : 255-67) containing a kanamycin antibiotic gene inserted at the unique Eco0109 I restriction enzyme site and a replacement polylinker (see Figure 14) inserted between the EcoR I site and Hind III site of the original vector.
20μl of water was added and the reaction heated to 70°C for
20mins. 1/10 volume of 3M sodium acetate, pH 5.2 and 2 volumes of ethanol added. It was mixed and stored at -20°C for 1 hour. After microfuging at 10,000g for lOmins, the supernatant was removed from the pelleted DNA and the DNA re-suspended in 5μl water.
0.5μl was electroporated into E. coli TOP10F' cells and fpllowing 1 hour recovery at 37°C, aliquots of the cells were plated on Kanamycin Luria Broth agar plates. The plates were incubated at 37°C overnight.
Colonies were gridded in duplicate on both a fresh Kanamycin Luria Broth agar plate and a Kanamycin Luria Broth agar plate prepared by addition of lμl of 20mg/ml XGAL and lμl of 0.5M IPTG per ml of agar gel (KIX' plate) .
Following overnight incubation at 37°C, white colonies on the KIX plate were screened by PCR with M13 forward and reverse primers for the presence of an insert corresponding to the Aeropyrum pernix PCR product.
9 colonies containing a 701bp product were grown up in 20ml LB plus lOOμg/ml Kanamycin to an OD600 of 1.0 then expression was induced by addition of IPTG to 0.5mM final. Cells were grown for a further 4hours and then the cells harvested and stored frozen at -20°c. Cells were lysed by addition of 0.5ml 50mM Tris-HCl, pH 7.9, 50mM dextrose, ImM EDTA and 0.5ml lOmM Tris-HCl, pH7.9, 50mM KCl, ImM EDTA, 0.5% v/v) Tween 20, 0.5% (v/v) Nonidet-P40 and incubation at 80°C for 15 minutes.
Following centrifugation at 10,000g for 10 minutes at room temperature, an aliquot from each lysed cells were analysed by SDS polyacrylamaide gel electrophoresis using a 12% gel. The gel was run then stained with Coomassie blue R250. All samples showed a band of approx 23kDa, which corresponds to the size of the putative PPase.
The same samples were then assayed for PPase activity at 75°C using the colorimetric assay of Jukka K. Heinonen, Reijo J. Lahti. (1981) Analytical Biochemistry, Vol.113, pp313-317.
All samples showed as positive, confirming that the expressed protein possessed thermophilic inorganic pyrophosphatase activity.
The first clone was subsequently used for larger scale production of the protein.
Purification of the Pyrophosphatase This clone was in 24 litres of LB. Once the ODe00 reached approximately 1.5, the culture was induced with 0.5mM IPTG and left to grow for a further 4 hours. The cells were then harvested and the cell pellet lysed. The expressed enzyme was purified by standard column chromatography on phenyl-sepharose CL4B (Amersham Pharmacia Biotech) , hydroxylapatite (Bio-rad Laboratories) and Hi-Performance Q Sepharose (Amersham Pharmacia Biotech) , finally being stored at -20°C in 20mM Tris- HCl, pH 8.0, lOOmM NaCl, 0.5% (v/v) Tween 20, 0.5% (v/v) Nonidet P40, O.lmM EDTA, ImM dithiothreitol and 50% glycerol . Example 10
PCR Assay using the A, pernix inorganic pyrophosphatase enzyme The method of the present invention was carried out using the A. pernix inorganic pyrophosphatase enzyme. The assay is based around the amplification of the human B-actin gene.
In this assay, a kit was used which was obtained from from Eurogentec S.A., Pare Scientifique du Sart-Tilman, rue Bois Saint-Jean 14, 4102 SERAING, Belgium (Cat. No. RT-QP73-05) . The standard Taq polymerase was substituted for the HotStart Taq polymerase provided with the kit.
PCR reaction mixture lx Reaction Buffer 200μM, dATP, dCTP, dGTP and 400μM dUTP
0.025u/μl unmodified Taq polymerase
0.002u/μl Aeropyrum pernix inorganic pyrophosphatase
0.3μM 5' Primer (5' GAC TCG TCA TAC TCC TGC TTG CT 3' - SEQ ID
NO 22) 0.3μM 3' Primer (5' CAT TGC CGA CAG GAT GCA GAA 3' - SEQ ID NO
23)
0 . 15μM Taqman probe ( FAM-ATCCACATCTGCTGGAAGGTGGACAGT-TAMRA -
SEQ ID NO 24 )
5mM MgCl2 2mM NaPPi
Passive Reference
1 in 4 dilutions of Human genomic DNA starting with 7.5ng (2500 copies)
Cycling conditions
Initial denaturation of 94°C 3 minutes
40 cycles of 94°C, 15 seconds and 60°C, 60 seconds
The results are shown in Figure 19. In conclusion we believe that using the method of the invention, by using pyrophosphate to inhibit a PCR and then removing that inhibition, for example at 80°C-95°C through the use of a thermostable PPase, behaves in the same manner as HotStart PCR but at a rapid rate with the additional benefit of increased specificity.
All references mentioned in the above specification are herein incorporated by reference. Other modifications of the present invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with the specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention, which are obvious to those skilled in the art, are intended to be within the scope of the following claims.

Claims (26)

Claims
1. A method for conducting a nucleic acid amplification reaction, said method comprising forming an amplification reaction mixture in the presence of sufficient of a pyrophosphate salt to prevent primer extension taking place, digesting said pyrophosphate salt with a pyrophosphatase enzyme (PPase) , and subjecting said reaction mixture to conditions such that an amplification reaction may proceed.
2. A method according to claim 1 wherein the amplification reaction is a polymerase chain reaction (PCR) and the reaction mixture contains reagents suitable for conducting such a reaction.
3. A method according to claim 1 or claim 2 wherein the reaction mixture contains a DNA polymerase which is selected from Thermus aquaticus polymerase ( Taq) , Thermus thermophilus polymerase { Tth) , Thermus species NH polymerase (TspNH), Thermus brockianus polymerase { Tbr) , Pyrococcus furiosus polymerase (Pfu) , 9°N7 exo-DNa polymerase, and Thermococcus literalis DNA polymerase.
4. A method according to any one of the preceding claims wherein the inorganic pyrophosphate is an alkali earth metal pyrophosphate.
5. A method according to claim 4 wherein the inorganic pyrophosphate is tetrasodium pyrophosphate of formula Na4P207.
6. A method according to any one of the preceding claims wherein the pyrophosphate is present in the reaction mixture at a concentration of at least 0.5mM.
7. A method according to claim 6 wherein the pyrophosphate is present at a concentration of from 1-1OmM.
8. A method according to any one of the preceding claims wherein the said digestion is effected using a thermostable PPase.
9. A method according to claim 8 wherein the thermostable PPase is Sulfolbus acidicaldarius inorganic pyrophosphatase, (Sac PPase) , thermococcus litoralis inorganic pyrophosphatase or Aeropyrum pernix inorganic pyrophosphatase.
10. A method according to claim 8 or claim 9 wherein the thermostable PPase is added to the reaction mixture on formation thereof.
11. A method according to claim 10 which includes a incubation step prior to the amplification reaction at elevated temperature in order to allow the PPase to digest inorganic pyrophosphate present.
12. A method according to any one of the preceding claims wherein the PPase is added to the reaction mixture at a concentration of at least 0.04u per 50μL PCR reaction mixture.
13. A method according to claim 12 wherein the PPase is added to the reaction mixture at a concentration of 0.08u per 50μL
PCR reaction mixture .
14. A method according to claim 12 or claim 13 wherein the PPase is added to the reaction mixture at a concentration of from 0.2-10u per 50μL PCR reaction mixture.
15. A kit for conducting an amplification reaction, said kit comprising a pyrophosphate salt, an pyrophosphatase enzyme, and optionally one or more reagents required for use in an amplification reaction.
16. A kit according to claim 15 which further comprises one or more primers necessary to carry out amplification of a particular target nucleic acid.
17. A kit according to claim 15 or claim 16 which further includes one or more fluorescently labelled reagents .
18. A kit according to claim 17 wherein the fluorescently labelled reagents are selected from one or more of an intercalating dye, a fluorescently labelled probe, a fluorescently labelled primer or a fluorescently labelled nucleotide.
19. The use of a pyrophosphate salt in a method for carrying out amplification reactions as claimed in any one of claims 1 to 14.
20. The use of a pyrophosphatase enzyme in a method for carrying out amplification reactions as claimed in any one of claims 1 to 14.
21. A pyrophosphatase enzyme isolated from Aeropyrum pernix.
22. A pyrophosphatase enzyme encoded by the polynucleotide sequence as shown in SEQ ID NO.26 or a variant or fragment thereof.
23. A pyrophosphatase enzyme comprising the amino acid sequence as shown in SEQ ID NO. 25 or a variant or fragment thereof.
24. An isolated polynucleotide which encodes an enzyme according to claim 22 or claim 23.
25. The use of a pyrophosphatase enzyme as claimed in any one of claims 21 to 23 in a method for carrying out amplification reactions as claimed in any one of claims 1 to 14.
26. A method for conducting an amplification reaction substantially as herein before described with reference to the Examples .
AU2002249444A 2001-04-30 2002-04-22 Amplification process Ceased AU2002249444B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
GB0110501.4 2001-04-30
GBGB0110501.4A GB0110501D0 (en) 2001-04-30 2001-04-30 Amplification process
PCT/GB2002/001861 WO2002088387A2 (en) 2001-04-30 2002-04-22 Amplification process

Publications (2)

Publication Number Publication Date
AU2002249444A1 true AU2002249444A1 (en) 2003-04-17
AU2002249444B2 AU2002249444B2 (en) 2007-07-12

Family

ID=9913697

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2002249444A Ceased AU2002249444B2 (en) 2001-04-30 2002-04-22 Amplification process

Country Status (14)

Country Link
US (2) US6951744B2 (en)
EP (1) EP1390532B1 (en)
JP (1) JP4177118B2 (en)
KR (2) KR20100005245A (en)
CN (1) CN1318604C (en)
AR (1) AR035238A1 (en)
AT (1) ATE502118T1 (en)
AU (1) AU2002249444B2 (en)
CA (1) CA2445864C (en)
DE (1) DE60239456D1 (en)
GB (2) GB0110501D0 (en)
NZ (1) NZ528919A (en)
TW (1) TWI323284B (en)
WO (1) WO2002088387A2 (en)

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0112868D0 (en) 2001-05-25 2001-07-18 Secr Defence Detection system
CN1500887A (en) * 2002-10-01 2004-06-02 松下电器产业株式会社 Method for detecting primer elongation reaction, method and apparatus for distinguishing kinds of basic groups
US20060051796A1 (en) * 2004-09-09 2006-03-09 Inga Boell Real time PCR with the addition of pyrophosphatase
EP1634965B1 (en) 2004-09-09 2010-01-20 Roche Diagnostics GmbH Real time PCR with the addition of pyrophosphatase
GB0502010D0 (en) * 2005-02-01 2005-03-09 Enigma Diagnostics Ltd Biochemical reagents and their uses
EP2032714B1 (en) 2006-06-01 2011-03-16 TriLink BioTechnologies Chemically modified oligonucleotide primers for nucleic acid amplification
KR101098764B1 (en) * 2007-10-29 2011-12-26 (주)바이오니아 Dried Composition for hot-start PCR with Long-Term Stability
WO2009151921A1 (en) 2008-05-27 2009-12-17 Trilink Biotechnologies Chemically modified nucleoside 5'-triphosphates for thermally initiated amplification of nucleic acid
CA2793970A1 (en) * 2010-04-30 2011-11-03 F. Hoffmann-La Roche Ag System and method for purification and use of inorganic pyrophosphatase from aquifex aeolicus
US20110312751A1 (en) * 2010-06-17 2011-12-22 Geneasys Pty Ltd Microfluidic device for detection of mitochondrial dna via fluorescence modulated by hybridization
KR101870311B1 (en) * 2012-03-09 2018-06-25 (주)바이오니아 Compositions for hot start reverse transcription reaction or hot start reverse transcription polymerase chain reaction
EP2861757B1 (en) 2012-06-14 2019-11-06 Life Technologies Corporation Novel compositions, methods and kits for polymerase chain reaction (pcr)
GB201301457D0 (en) 2013-01-28 2013-03-13 Fluorogenics Ltd Freeze-dried composition
JP6467829B2 (en) * 2014-09-03 2019-02-13 東洋紡株式会社 Improved RT-PCR reaction
CN104862288B (en) * 2015-05-08 2019-10-29 苏州大学 A kind of synthesis of New temperature responsiveness inorganic pyrophosphate enzyme conjugates and its application in enhancing polymerase chain reaction
EP3402880B1 (en) 2016-01-15 2024-01-03 Thermo Fisher Scientific Baltics UAB Thermophilic dna polymerase mutants
US11618891B2 (en) 2017-06-26 2023-04-04 Thermo Fisher Scientific Baltics Uab Thermophilic DNA polymerase mutants
JP7234598B2 (en) * 2018-01-26 2023-03-08 東ソー株式会社 Nucleic acid amplification reagent and nucleic acid amplification control method using the reagent
CN113481180A (en) * 2021-07-05 2021-10-08 吉林大学 Alkaline thermophilic inorganic pyrophosphatase and application thereof in enhancing polymerase chain reaction and UDP-galactose synthesis reaction

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4868103A (en) 1986-02-19 1989-09-19 Enzo Biochem, Inc. Analyte detection by means of energy transfer
CH670709A5 (en) * 1986-04-24 1989-06-30 Univ Moskovsk
US5498523A (en) * 1988-07-12 1996-03-12 President And Fellows Of Harvard College DNA sequencing with pyrophosphatase
EP0467953A4 (en) * 1989-04-12 1992-06-17 The President And Fellows Of Harvard College Improved primer extension reactions
KR100236506B1 (en) 1990-11-29 2000-01-15 퍼킨-엘머시터스인스트루먼츠 Apparatus for polymerase chain reaction
FI923911A (en) * 1992-09-01 1994-03-02 Vsevolod Kiselev DNA molecules in vitro syntheses
US5565339A (en) 1992-10-08 1996-10-15 Hoffmann-La Roche Inc. Compositions and methods for inhibiting dimerization of primers during storage of polymerase chain reaction reagents
US5491063A (en) 1994-09-01 1996-02-13 Hoffmann-La Roche Inc. Methods for in-solution quenching of fluorescently labeled oligonucleotide probes
US5773258A (en) 1995-08-25 1998-06-30 Roche Molecular Systems, Inc. Nucleic acid amplification using a reversibly inactivated thermostable enzyme
US5665551A (en) * 1995-09-13 1997-09-09 Roche Molecular Systems, Inc. Purified nucleic acid encoding a thermostable pyrophosphatase
US6291164B1 (en) * 1996-11-22 2001-09-18 Invitrogen Corporation Methods for preventing inhibition of nucleic acid synthesis by pyrophosphate
GB9716052D0 (en) 1996-12-06 1997-10-01 Secr Defence Reaction vessels
GB9725197D0 (en) 1997-11-29 1998-01-28 Secr Defence Detection system
GB9725237D0 (en) 1997-11-29 1998-01-28 Secr Defence Amplification system
GB9803382D0 (en) 1998-02-19 1998-04-15 Secr Defence Detection system
US6183998B1 (en) 1998-05-29 2001-02-06 Qiagen Gmbh Max-Volmer-Strasse 4 Method for reversible modification of thermostable enzymes
GB9812768D0 (en) 1998-06-13 1998-08-12 Zeneca Ltd Methods
KR100292883B1 (en) * 1999-02-09 2001-06-15 박한오 Hot Start PCR Mixture Comprising Pyrophosphate and Pyrophosphatase
ATE441726T1 (en) 2000-02-23 2009-09-15 Hope City PYROPHOSPHOROLYSIS-ACTIVATED POLYMERIZATION (PAP): APPLICATION FOR ALLEL-SPECIFIC AMPLIFICATION AND SEQUENCE DETERMINATION OF NUCLEIC ACIDS
KR20050005626A (en) * 2003-07-07 2005-01-14 주식회사 포스코 improvement method of charging coal bulk density in a coke oven

Similar Documents

Publication Publication Date Title
US7449312B2 (en) Kit for conducting a polymerase chain reaction
AU2002249444A1 (en) Amplification process
US7122355B2 (en) Composition and method for hot start nucleic acid amplification
EP1866434B1 (en) Isothermal nucleic acid amplification
JP5286261B2 (en) Specialized oligonucleotides and their use in nucleic acid amplification and detection
KR101590175B1 (en) Detection of Target Nucleic Acid Sequences by Cyclic Exonucleolytic Reactions
EP1989324B1 (en) Pcr hot start by magnesium sequestration
JP2005503755A (en) Methods and compositions for making multiple copies of nucleic acid sequences and methods for detecting them
CN102770556A (en) Td probe and its uses
US20090181401A1 (en) Detection format for hot start real time polymerase chain reaction
US20220316001A1 (en) Methods and devices related to amplifying nucleic acid at a variety of temperatures
JP2004524853A5 (en)
US20050026166A1 (en) Concurrent enzymatic polynucleotide synthesis and detectable signal generation
WO2013093432A1 (en) Method for enriching and detection of variant target nucleic acids
CN110446791B (en) Polynucleotide adaptors and methods of use thereof
US10227642B2 (en) Detergent free polymerases
US9617588B2 (en) Enzyme mixture
EP1277841B1 (en) New composition and method for hot start nucleic acid amplification
EP1502961B1 (en) New detection format for hot start real time polymerase chain reaction
WO2002101041A1 (en) Method of amplifying nucleic acid and method of detecting nucleotide polymorphism with the use of nucleotide analog