• advantage 2 polymerase mix user manual

    advantage 2 polymerase mix user manual

    Download Link: ➡ advantage 2 polymerase mix user manual



    File Name: advantage 2 polymerase mix user manual.pdf
    Size: 1303 KB
    Type: PDF, ePub, eBook
    Category: Book
    Uploaded: 8 May 2019, 16:30 PM
    Rating: 4.6/5 from 841 votes.


    Last checked: 8 Minutes ago!









    ⬆ LINK ⬆




















    In order to read or download advantage 2 polymerase mix user manual ebook, you need to create a FREE account.

    eBook includes PDF, ePub and Kindle version



    ✔ Register a free 1 month Trial Account.
    ✔ Download as many books as you like (Personal use)
    ✔ Cancel the membership at any time if not satisfied.
    ✔ Join Over 80000 Happy Readers


    Book Descriptions:

    advantage 2 polymerase mix user manual

    Simply reconstitute the mix by adding PCR-grade water along with primers and template. They may not be used for any other purpose, including, but not limited to, use in humans, therapeutic or diagnostic use, or commercial use of any kind. Our products may not be transferred to third parties, resold, modified for resale, or used to manufacture commercial products or to provide a service to third parties without our prior written approval. The premix contains all neceeary reagents for a high yield and high-fidelity PCR. Please refer to User Manual for the composition. EcoDry Premix are packaged as individual 8-well tube strips with optically clear sealing caps.They may not be used for any other purpose, including, but not limited to, use in humans, therapeutic or diagnostic use, or commercial use of any kind. Our products may not be transferred to third parties, resold, modified for resale, or used to manufacture commercial products or to provide a service to third parties without our prior written approval. Please see the Kit Components List to determine kit components. Certificates of Analysis and Kit Components Lists are located under the Documents tab. Use this guide to prevent common PCR problems. All Rights Reserved. All trademarks are the property of Takara Bio Inc. Certain trademarks may not be registered in all jurisdictions. Additional product, intellectual property, and restricted use information is available at takarabio.com. Through our Takara, Clontech, and Cellartis brands, our mission is to develop high-quality innovative tools and services to accelerate discovery. We have a variety of best-in-class products that can help your AD research move forward. This product utilizes our novel Capturem technology in a spin column format with membrane-immobilized trypsin.

    • advantage 2 polymerase mix user manual, advantage 2 polymerase mix user manual, advantage 2 polymerase mix user manual pdf, advantage 2 polymerase mix user manual download, advantage 2 polymerase mix user manual free, advantage 2 polymerase mix user manual 2017.

    Read our cookie policy.With an automatic hot-start feature, our Advantage 2 Polymerase Mix can readily amplify a wide range of DNA templates, including long templates up to 18 kb, and complex genomic DNA up to 6kb. The Advantage 2 Polymerase Mix includes two optimized buffers.With an automatic hot-start feature, our Advantage 2 Polymerase Mix can readily amplify a wide range of DNA templates, including long templates up to 18 kb, and complex genomic DNA up to 6 kb. The Advantage 2 Polymerase Mix includes two optimized buffers.Advantage 2 Polymerase Mix is highly recommended for applications downstream of our SMART(er) method for cDNA synthesis. The Mix is ideal for amplification of long templates up to 18 kb and complex genomic DNA up to 6 kb. Two different optimized buffers are supplied with the polymerase mix.They may not be used for any other purpose, including, but not limited to, use in humans, therapeutic or diagnostic use, or commercial use of any kind. Our products may not be transferred to third parties, resold, modified for resale, or used to manufacture commercial products or to provide a service to third parties without our prior written approval. The Mix is ideal for amplification of long templates up to 18 kb and complex genomic DNA up to 6 kb. Two different optimized buffers are supplied with the polymerase mix.They may not be used for any other purpose, including, but not limited to, use in humans, therapeutic or diagnostic use, or commercial use of any kind. Our products may not be transferred to third parties, resold, modified for resale, or used to manufacture commercial products or to provide a service to third parties without our prior written approval. The premix contains all neceeary reagents for a high-yield and high-fidelity PCR. Please refer to User Manual for the composition. EcoDry Premix are packaged as individual 8-well tube strips with optically clear sealing caps.

    Please try again or contact Customer Service. Traditional methods of cloning a DNA sequence into a vector and replicating it in a living cell often require days or weeks of work, but amplification of DNA sequences by PCR requires only hours. While most biochemical analyses, including nucleic acid detection with radioisotopes, require the input of significant amounts of biological material, the PCR process requires very little. Thus, PCR can achieve more sensitive detection and higher levels of amplification of specific sequences in less time than previously used methods. These features make the technique extremely useful, not only in basic research, but also in commercial uses, including genetic identity testing, forensics, industrial quality control and in vitro diagnostics. Basic PCR is commonplace in many molecular biology labs where it is used to amplify DNA fragments and detect DNA or RNA sequences within a cell or environment. However, PCR has evolved far beyond simple amplification and detection, and many extensions of the original PCR method have been described. This chapter provides an overview of different types of PCR methods, applications and optimization. Once assembled, the reaction is placed in a thermal cycler, an instrument that subjects the reaction to a series of different temperatures for set amounts of time. This series of temperature and time adjustments is referred to as one cycle of amplification. Each PCR cycle theoretically doubles the amount of targeted sequence (amplicon) in the reaction. Ten cycles theoretically multiply the amplicon by a factor of about one thousand; 20 cycles, by a factor of more than a million in a matter of hours. In the denaturation process, the two intertwined strands of DNA separate from one another, producing the necessary single-stranded DNA template for replication by the thermostable DNA polymerase.

    Capturem Trypsin Columns may be used to completely digest protein samples in less than a minute with digestion efficiencies (protein coverage) comparable to or better than those obtained using in-solution trypsin digestion. Careful planning, dedicated researchers, and the right tools. At Takara Bio, we thoughtfully develop best-in-class products to tackle your most challenging research problems, and have an expert team of technical support professionals to help you along the way, all at superior value. As a member of the Takara Bio Group, TBUSA is part of a company that holds a leadership position in the global market and is committed to improving the human condition through biotechnology. Our mission is to develop high-quality innovative tools and services to accelerate discovery. The Mix is ideal for amplification of long templates up to 18 kb and complex genomic DNA up to 6 kb. Two different optimized buffers are supplied with the polymerase mix.With an automatic hot-start feature, our Advantage 2 Polymerase Mix can readily amplify a wide range of DNA templates, including long templates up to 18 kb, and complex genomic DNA up to 6kb. The Advantage 2 Polymerase Mix includes two optimized buffers.By continuing to view our website we will assume that you are happy with it. Ok. Your commerce experience may be limited. Please update your browser to Internet Explorer 11 or above. When you select your country, you agree that we can place these functional cookies on your device. After that, you will need to contact Customer Service to unlock your account. Please try again or contact Customer Service. Please request another reset link. Please try again or contact Customer Service. A verified email address is required to access the full functionality of your Promega.com account. Please try again or contact Customer Service. Please try again or contact Customer Service. Please try again or contact Customer Service. Please check your network settings and try again.

    This newly synthesized region then acts as a template for primer extension and synthesis of undesired amplification products.The formation of nonspecific products and primer-dimer can compete for reagent availability with amplification of the desired product. Thus, hot-start PCR can improve the yield of specific PCR products. This omission prevents the polymerase from extending primers until the critical component is added at the higher temperature where primer annealing is more stringent. However, this method is tedious and increases the risk of contamination. A second, less labor-intensive approach involves the reversible inactivation or physical separation of one or more critical components in the reaction. Finally, the DNA polymerase can be maintained in an inactive state through chemical modification (Moretti, T. et al 1998). Activity is restored during initial denaturation, allowing hot-start PCR. While basic PCR works well when smaller fragments are amplified, amplification efficiency (and therefore the yield of amplified fragments) decreases significantly as the amplicon size increases over 5kb. This decrease in yield can be attributed to the accumulation of truncated products, which are not suitable substrates for additional cycles of amplification. These products appear as smeared, as opposed to discrete, bands on a gel. They devised an approach using a mixture of two thermostable polymerases to synthesize longer PCR products. Presumably, when the nonproofreading DNA polymerase (e.g., Taq DNA polymerase) misincorporates a dNTP, subsequent extension of the newly synthesized DNA either proceeds very slowly or stops completely. The proofreading polymerase (e.g., Pfu DNA polymerase or Tli DNA polymerase) serves to remove the misincorporated nucleotide, allowing the DNA polymerases to continue extension of the new strand. In addition, template quality is crucial.

    Finally, the synthesis of new DNA begins as the reaction temperature is raised to the optimum for the DNA polymerase.If the temperature during the annealing and extension steps are similar, these two steps can be combined into a single step in which both primer annealing and extension take place.Yet numerous instances exist in which amplification of RNA would be preferred. To apply PCR to the study of RNA, the RNA sample must first be converted to cDNA to provide the necessary DNA template for the thermostable polymerase (Figure 1). This process is called reverse transcription (RT), hence the name RT-PCR. Reverse transcriptases have been adapted for use in a variety of in vitro applications including real-time and endpoint RT-PCR, labeled-cDNA probe generation and cDNA library construction. The ideal reverse transcriptase is robust (highly active under a variety of conditions) and converts all primed RNA within a sample to cDNA, regardless of its abundance, length or secondary structure. Genetic engineering and development of proprietary RT-enhancing buffers have led to the commercial availability of new enzymes that offer superior performance over these naturally occurring RTs. Some thermostable DNA polymerases (e.g., Tth DNA polymerase) possess a reverse transcriptase activity, which can be activated by using manganese instead of magnesium as a cofactor (Myers and Gelfand, 1991). After this initial reverse transcription step to produce the cDNA template, basic PCR is carried out to amplify the target sequence. The efficiency of the first-strand synthesis reaction, which can be related to the RNA quality, also will significantly affect amplification results. At room temperature, PCR primers can anneal to template sequences that are not perfectly complementary. Since thermostable DNA polymerases have activity at these low temperatures (although in most cases the activity is less than 25%) the polymerase can extend misannealed primers.

    Because fluorescence of these dyes increases dramatically in the presence of double-stranded DNA, DNA synthesis can be monitored as an increase in fluorescent signal. However, preliminary work often must be done to ensure that the PCR conditions yield only specific product. In subsequent reactions, specific amplification can verified by a melt curve analysis. The product length and sequence affect melting temperature (Tm), so the melt curve is used to characterize amplicon homogeneity. Nonspecific amplification can be identified by broad peaks in the melt curve or peaks with unexpected Tm values. By distinguishing specific and nonspecific amplification products, the melt curve adds a quality control aspect during routine use.These probes also can be used to detect single nucleotide polymorphisms (Lee et al. 1993; Bernard et al. 1998). There are several general categories of real-time PCR probes, including hydrolysis, hairpin and simple hybridization probes. These probes contain a complementary sequence that allows the probe to anneal to the accumulating PCR product, but probes can differ in the number and location of the fluorescent reporters. During the annealing step, the probe hybridizes to the PCR product generated in previous amplification cycles. With this approach, preliminary experiments must be performed prior to the quantitation experiments to show that the signal generated is proportional to the amount of the desired PCR product and that nonspecific amplification does not occur. The hairpin probe is designed so that the probe binds preferentially to the target DNA rather than retains the hairpin structure. As the reaction progresses, increasing amounts of the probe anneal to the accumulating PCR product, and as a result, the fluor and quencher become physically separated. The fluor is no longer quenched, and the level of fluorescence increases.

    One advantage of this technique is that hairpin probes are less likely to mismatch than hydrolysis probes (Tyagi et al. 1998). However, preliminary experiments must be performed to show that the signal is specific for the desired PCR product and that nonspecific amplification does not occur. In the first approach, the energy emitted by the fluor on one probe is absorbed by a fluor on the second probe, which hybridizes nearby. In the second approach, the emitted energy is absorbed by a second fluor that is incorporated into the PCR product as part of the primer. Both of these approaches result in increased fluorescence of the energy acceptor and decreased fluorescence of the energy donor. The use of hybridization probes can be simplified even further so that only one labeled probe is required. In this approach, quenching of the fluor by deoxyguanosine is used to bring about a change in fluorescence (Crockett and Wittwer, 2001; Kurata et al. 2001). The labeled probe anneals so that the fluor is in close proximity to G residues within the target sequence, and as probe annealing increases, fluorescence decreases due to deoxyguanosine quenching. With this approach, the location of probe is limited because the probe must hybridize so that the fluorescent dye is very near a G residue. The advantage of simple hybridization probes is their ability to be multiplexed more easily than hydrolysis and hairpin probes through the use of differently colored fluors and probes with different melting temperatures (reviewed in Wittwer et al. 2001). These probes also can be used to detect single nucleotide polymorphisms (Lee et al. 1993; Bernard et al. 1998). There are several general categories of real-time PCR probes, including hydrolysis, hairpin and simple hybridization probes. These probes contain a complementary sequence that allows the probe to anneal to the accumulating PCR product, but probes can differ in the number and location of the fluorescent reporters.

    Depurination of the template, which is promoted at elevated temperatures and lower pH, will result in more partial products and decreased overall yield. Additives, such as glycerol and dimethyl sulfoxide (DMSO), also help lower the strand-separation and primer-annealing temperatures, alleviating some of the depurination effects of high temperatures.This optimized enzyme mixture allows efficient amplification of up to 40kb from lambda DNA or 30kb from human genomic DNA. However, a wide variety of applications, such as determining viral load, measuring responses to therapeutic agents and characterizing gene expression, would be improved by quantitative determination of target abundance. Theoretically, this should be easy to achieve, given the exponential nature of PCR, because a linear relationship exists between the number of amplification cycles and the logarithm of the number of molecules. In practice, however, amplification efficiency is decreased because of contaminants (inhibitors), competitive reactions, substrate exhaustion, polymerase inactivation and target reannealing. As the number of cycles increases, the amplification efficiency decreases, eventually resulting in a plateau effect. This point must be determined empirically for different reactions because of the numerous factors that can affect amplification efficiency. Because the measurement is taken prior to the reaction plateau, quantitative PCR uses fewer amplification cycles than basic PCR. This can cause problems in detecting the final product because there is less product to detect. One such approach includes a second primer pair that is specific for a “housekeeping” gene (i.e., a gene that has constant expression levels among the samples compared) in the reaction (Gaudette and Crain, 1991; Murphy et al. 1990).

    Amplification of housekeeping genes verifies that the target nucleic acid and reaction components were of acceptable quality but does not account for differences in amplification efficiencies due to differences in product size or primer annealing efficiency between the internal standard and target being quantified. In competitive PCR, a known amount of a control template is added to the reaction. This template is amplified using the same primer pair as the experimental target molecule but yields a distinguishable product (e.g., different size, restriction digest pattern, etc.). The amounts of control and test product are compared after amplification. While these approaches control for the quality of the target nucleic acid, buffer components and primer annealing efficiencies, they have their own limitations (Siebert and Larrick, 1993; McCulloch et al. 1995), including the fact that many depend on final analysis by electrophoresis.Some of these analyses rely on blotting techniques, which introduce another variable due to nucleic acid transfer efficiencies, while other assays were developed to eliminate the need for gel electrophoresis yet provide the requisite specificity. Real-time PCR, which provides the ability to view the results of each amplification cycle, is a popular way of overcoming the need for analysis by electrophoresis. Specially designed instruments perform both thermal cycling to amplify the target and fluorescence detection to monitor PCR product accumulation. DNA-binding dyes are easy to use but do not differentiate between specific and nonspecific PCR products and are not conducive to multiplex reactions. Fluorescently labeled nucleic acid probes have the advantage that they react with only specific PCR products, but they can be expensive and difficult to design. Some qPCR technologies employ fluorescently labeled PCR primers instead of probes. The dye is simply added to the reaction, and fluorescence is measured at each PCR cycle.

    GoTaq qPCR Systems contain BRYT Green Dye, which provides maximum amplification efficiency and greater fluorescence than SYBR Green. Many of these suggestions also apply when using other DNA polymerases. Template DNA concentration, chelating agents present in the sample (e.g., EDTA or citrate), dNTP concentration and the presence of proteins all can affect the amount of free magnesium in the reaction. In the absence of adequate free magnesium, Taq DNA polymerase is inactive. Excess free magnesium reduces enzyme fidelity (Eckert and Kunkel, 1990) and may increase the level of nonspecific amplification (Williams, 1989; Ellsworth et al. 1993). For these reasons, researchers should empirically determine the optimal magnesium concentration for each target. The effect of magnesium concentration and the optimal concentration range can vary with the particular DNA polymerase. Before assembling the reactions, be sure to thaw the magnesium solution completely prior to use and vortex the magnesium solution for several seconds before pipetting. Magnesium chloride solutions can form concentration gradients as a result of multiple freeze-thaw cycles, and vortex mixing is required to obtain a uniform solution. These two steps, though seemingly simple, eliminate the cause of many failed experiments. The free magnesium changes of 0.6mM observed in their experiments dramatically affected amplification yields in an allele-specific manner. They postulated that magnesium chloride precipitates as a result of multiple freeze-thaw cycles. Amplifications were performed using various Mg concentrations to demonstrate the effect on the amplification of a 1.8kb target luciferase gene. The reaction products were analyzed by agarose gel electrophoresis followed by ethidium bromide staining.The buffer regulates the pH of the reaction, which affects DNA polymerase activity and fidelity.

    The buffer also contains a compound that increases the density of the sample so that it will sink into the well of the agarose gel, allowing reactions to be directly loaded onto an agarose gel without the need for loading dye. The yellow dye migrates at a rate faster than primers ( 2 at a concentration of 7.5mM for a final concentration of 1.5mM. In most cases, this is an excess of enzyme, and adding more enzyme will not significantly increase product yield.Accurate dispensing of small volumes of enzyme solutions in 50% glycerol is difficult, so we strongly recommend preparing a reaction master mix, which requires a larger volume of each reagent, to reduce pipetting errors. Also, avoid primers with intra- or intermolecular complementary sequences to minimize the production of primer-dimer. Intramolecular regions of secondary structure can interfere with primer annealing to the template and should be avoided. Primers can be designed to include sequences that are useful for downstream applications.Reagents commonly used to purify nucleic acids (salts, guanidine, proteases, organic solvents and SDS) are potent inactivators of DNA polymerases. In some cases, the inhibitor is not introduced into the reaction with the nucleic acid template. A good example of this is an inhibitory substance that can be released from polystyrene or polypropylene upon exposure to ultraviolet light (Pao et al. 1993; Linquist et al. 1998). Failure to amplify the control DNA usually indicates the presence of an inhibitor. Additional steps to clean up the DNA preparation, such as phenol:chloroform extraction or ethanol precipitation, may be necessary. For example, of a 4kb plasmid containing a 1kb target sequence, 25% of the input DNA is the target of interest. Thus, approximately 1,000,000-fold more human genomic DNA is required to maintain the same number of target copies per reaction.

    Common mistakes include using too much plasmid DNA, too much PCR product or too little genomic DNA as the template. Reactions with too little DNA template will have low yields, while reactions with too much DNA template can be plagued by nonspecific amplification. When reamplifying a PCR product, the concentration of the specific PCR product is often not known. We recommend diluting the previous amplification reaction 1:10 to 1:10,000 before reamplification. The lengths and temperatures for the other steps of a PCR cycle do not usually vary significantly. However in some cases, the denaturation cycle can be shortened or a lower denaturation temperature used to reduce the number of depurination events, which can lead to mutations in the PCR products. Using an annealing temperature slightly higher than the primer Tm will increase annealing stringency and can minimize nonspecific primer annealing and decrease the amount of undesired products synthesized. Using an annealing temperature lower than the primer Tm can result in higher yields, as the primers anneal more efficiently at the lower temperature. In many cases, nonspecific amplification and primer-dimer formation can be reduced through optimization of annealing temperature, but if undesirable PCR products remain a problem, consider incorporating one of the many hot-start PCR methods. The Tm also can be calculated using the Biomath Calculators. Numerous formulas exist to determine the theoretical Tm of nucleic acids (Baldino, Jr. et al. 1989; Rychlik et al. 1990). The formula below can be used to estimate the melting temperature for oligonucleotides: The risk of undesirable PCR products appearing in the reaction increases as the cycle number increases, so we recommend performing only enough cycles to synthesize the desired amount of product.

    If nonspecific amplification products accumulate before sufficient amounts of PCR product can be synthesized, consider diluting the products of the first reaction and performing a second amplification with the same primers or primers that anneal to sequences within the desired PCR product (nested primers). There are many PCR enhancers, which can act through a number of different mechanisms. These reagents will not enhance all PCRs; the beneficial effects are often template- and primer-specific and will need to be determined empirically. Some of the more common enhancing agents are discussed below. GC-rich templates can be problematic due to inefficient separation of the two DNA strands or the tendency for the complementary, GC-rich primers to form intermolecular secondary structures, which will compete with primer annealing to the template. Betaine reduces the amount of energy required to separate DNA strands (Rees et al. 1993). DMSO and formamide are thought to aid amplification in a similar manner by interfering with hydrogen bond formation between two DNA strands (Geiduschek and Herskovits, 1961). Concentrations of DMSO greater than 10% and formamide greater than 5% can inhibit Taq DNA polymerase and presumably other DNA polymerases as well (Varadaraj and Skinner, 1994). These additives can increase DNA polymerase stability and reduce the loss of reagents through adsorption to tube walls. Ammonium ions can make an amplification reaction more tolerant of nonoptimal conditions.Use separate work areas and pipettors for pre- and post-amplification steps. Use positive displacement pipettes or aerosol-resistant tips to reduce cross-contamination during pipetting. Wear gloves, and change them often. PCR reagents can be treated with isopsoralen, a photo-activated, cross-linking reagent that intercalates into double-stranded DNA molecules and forms covalent, interstrand crosslinks, to prevent DNA denaturation and replication.

    These inter-strand crosslinks effectively render contaminating DNA unamplifiable. UNG treatment prevents replication of uracil-containing DNA by causing the DNA polymerase to stall at the resulting abasic sites. For UNG to be an effective safeguard against contamination, the products of previous amplifications must be synthesized in the presence of dUTP. This is easily accomplished by substituting dUTP for some or all of the dTTP in the reaction. Nonproofreading polymerases will readily incorporate dUTP into a PCR product, but proofreading polymerases incorporate dUTP much less efficiently (Slupphaug et al. 1993; Greagg et al. 1999; Lasken et al. 1996). Since dUTP incorporation has no noticeable effect on the intensity of ethidium bromide staining or electrophoretic mobility of the PCR product, reactions can be analyzed by standard agarose gel electrophoresis. While both methods are effective (Rys and Persing, 1993), UNG treatment has the advantage that both single-stranded and double-stranded DNA templates will be rendered unamplifiable (Longo et al. 1990). Many of the important parameters discussed there also apply to RT-PCR. For a discussion of reverse transcriptases commonly used for RT-PCR, see the Thermostable Polymerases and Reverse Transcriptases section (below). Procedures for creating and maintaining a ribonuclease-free (RNase-free) environment to minimize RNA degradation are described in Blumberg, 1987. For optimal results, the RNA template, whether a total RNA preparation, an mRNA population or a synthesized RNA transcript, should be DNA-free to avoid amplification of contaminating DNA. The most commonly used DNA polymerases for PCR have no reverse transcriptase activity under standard reaction conditions, and thus, amplification products will be generated only if the template contains trace amounts of DNA with similar sequences. The specific 540bp amplicon is indicated.For example, a primer that anneals specifically to the 3?


  • Commentaires

    Aucun commentaire pour le moment

    Suivre le flux RSS des commentaires


    Ajouter un commentaire

    Nom / Pseudo :

    E-mail (facultatif) :

    Site Web (facultatif) :

    Commentaire :