Fact-checked by Grok 2 weeks ago

Multiple displacement amplification

Multiple displacement amplification (MDA) is an isothermal DNA amplification technique that enables the whole-genome amplification of minute DNA samples, typically starting from as little as one to ten genomic copies, using random hexamer primers and the highly processive bacteriophage φ29 DNA polymerase to generate long, high-fidelity DNA products exceeding 10 in length. Developed in as an advancement from rolling circle amplification methods, MDA operates at a constant temperature of 30°C without the need for thermal cycling, relying on strand displacement synthesis where the polymerase displaces downstream strands to allow continuous priming and extension, resulting in a branched DNA network with minimal sequence bias. This process yields 20–30 μg of amplified DNA from low-input material, such as crude cell lysates or blood samples, making it suitable for applications where DNA is scarce. Key advantages of include its uniform genomic representation, with amplification bias typically less than threefold across loci—far superior to PCR-based methods that can exhibit biases of 10² to 10⁶-fold—and a low error rate due to the φ29 polymerase's 3'–5' activity, which is over 1,000 times more accurate than . The technique produces DNA fragments averaging over 10 kb and up to 100 kb, which supports downstream analyses requiring intact, long molecules, such as (RFLP), Southern blotting, and whole-genome sequencing. In single-cell applications, can generate approximately 40 μg of DNA, facilitating high-coverage sequencing comparable to unamplified samples. MDA has become widely adopted in genomics for amplifying low-abundance DNA from sources like forensic samples, unculturable microorganisms, and clinical specimens, enabling applications in SNP genotyping, chromosome painting, diagnostics, and pathogen detection. More recently, it has proven effective for whole-genome sequencing with long-read technologies like Oxford Nanopore, achieving high-quality assemblies from inputs as low as 0.025 pg—such as in Cryptosporidium oocysts or bacterial genomes—while supporting chromosomal-level resolutions with over 70x coverage depth, though it may introduce chimeric concatemers that require bioinformatic correction. Despite these benefits, limitations include reduced efficiency with highly degraded DNA and potential coverage gaps at ultra-low inputs, often necessitating replicates or tools like CADECT for concatemer removal.

Introduction

Definition and principles

Multiple displacement amplification (MDA) is an isothermal whole-genome amplification technique that enables the generation of microgram quantities of DNA from nanogram or picogram starting inputs, facilitating downstream genetic analyses from limited biological samples. This method relies on a strand-displacing DNA polymerase to synthesize long DNA strands continuously, producing high-molecular-weight products exceeding 10 kb in length without the need for thermal denaturation steps. MDA is particularly suited for amplifying low-abundance DNA, such as from single cells, where traditional methods may fail due to insufficient material. The core principles of MDA center on random priming, continuous strand displacement synthesis, and exponential amplification under constant temperature conditions. Random primers bind to denatured DNA templates, initiating polymerase activity that extends new strands while displacing the non-template strand, thereby exposing additional binding sites for more primers and enabling ongoing amplification. This process creates hyperbranched DNA structures that serve as new templates, driving exponential growth without discrete cycles, typically at 30°C for several hours. A key prerequisite is the function of strand-displacing DNA polymerases, which possess high processivity and fidelity, allowing them to synthesize DNA over tens of kilobases while displacing downstream strands to maintain reaction progression. Priming mechanisms involve short, sequence-independent oligonucleotides that anneal across the genome, ensuring broad coverage in an unbiased manner at an introductory level. In comparison to (PCR), MDA serves as a non-thermal alternative optimized for low-input samples, avoiding the biases introduced by repeated heating and cooling that can lead to incomplete representation in PCR-based whole-genome amplification. While PCR relies on cyclic denaturation, annealing, and extension for exponential amplification, MDA achieves similar growth through continuous displacement. This adaptation allows MDA to produce yields of 20–30 μg from as little as one to ten copies, with uniformity across loci typically under threefold .

Historical development

Multiple displacement amplification (MDA) was developed in 2002 by Frank B. Dean and colleagues primarily at Molecular Staging, Inc., in collaboration with Biotech (later ), as a method for achieving uniform whole-genome amplification from limited DNA quantities. Published in the Proceedings of the , the technique leveraged the strand-displacing activity of φ29 DNA polymerase to isothermally amplify genomic DNA with high fidelity and minimal bias, enabling comprehensive coverage of the from nanogram-scale inputs. The invention addressed key challenges in early 2000s , where traditional PCR-based methods often introduced amplification biases and incomplete coverage when starting from small samples, such as those from single cells or clinical biopsies. This built on 1990s research characterizing the exceptional processivity and strand displacement capabilities of φ29 DNA polymerase, originally isolated from phage φ29 in the 1970s by and Luis Blanco, whose properties were refined for amplification applications through studies on rolling-circle replication. Following its publication, was rapidly commercialized, with kits like REPLI-g from introduced in 2003 to facilitate routine laboratory use. Molecular Staging was acquired by in 2004, further advancing commercialization. By 2005, the method gained traction in single-cell applications, as demonstrated in studies amplifying genomes from individual bacterial cells and , expanding its utility beyond bulk samples. MDA's evolution shifted its primary focus from human genomics to microbial and environmental applications, driven by advances in sequencing technologies. Post-2020, integrations with long-read platforms like have enabled high-quality assemblies from ultra-low-input samples, such as those from uncultured microbes or single environmental cells, enhancing its role in ecological and biodiversity studies.

Mechanism

Key components

Multiple displacement amplification (MDA) relies on a specialized set of reagents and materials to enable isothermal, strand-displacing DNA synthesis. The core enzyme driving the reaction is Phi29 DNA polymerase, a replicative polymerase derived from the bacteriophage phi29 that infects Bacillus subtilis. This enzyme exhibits exceptional processivity, capable of synthesizing DNA strands exceeding 70 kb in length without dissociating from the template, which is essential for comprehensive genome coverage. Additionally, Phi29 possesses 3'→5' exonuclease activity that contributes to its high fidelity, with an error rate of approximately 1 in 10⁶–10⁷ bases, and inherent strand displacement capability that allows continuous amplification without thermal cycling. Initiation of amplification occurs through random hexamer primers, which are synthetic oligonucleotides consisting of six nucleotides with randomized sequences to ensure broad annealing across the template DNA. These primers typically anneal at temperatures between 30–40°C, facilitating unbiased priming sites throughout the genome and promoting multiple initiation points for exponential amplification. In standard protocols, phosphorothioate modifications may be incorporated at the 3' ends to enhance stability and resistance to degradation, though unmodified versions are also effective. The reaction provides the optimal chemical environment for enzymatic activity and includes triphosphates (dNTPs) at concentrations of approximately 1 mM each (dATP, dCTP, dGTP, dTTP) to serve as building blocks for new DNA strands. Magnesium ions (Mg²⁺), typically at 10–20 mM as MgCl₂, act as cofactors essential for and strand displacement. (BSA) is included at 0.1–0.2 mg/mL to stabilize the , reduce non-specific interactions, and mitigate potential inhibition from contaminants. The is maintained at a of 7.5–8.0, often using 40–50 mM Tris-HCl, to support the 's optimal activity under isothermal conditions. For effective MDA, the input DNA template must be of sufficient quality and quantity, with standard reactions requiring 1–10 ng of high-molecular-weight genomic DNA to achieve reliable amplification yields. Advanced applications, such as single-cell or low-input , can utilize as little as a single DNA molecule (approximately 6 pg for a equivalent), though this demands careful optimization to minimize biases.

Amplification process

The amplification process of multiple displacement amplification (MDA) initiates with the annealing of random hexamer primers to the template DNA in an isothermal reaction at 30°C, without requiring initial heat denaturation due to the high primer concentration that facilitates invasion of double-stranded regions. The highly processive Phi29 DNA polymerase binds to the 3' ends of these annealed primers and begins DNA synthesis, displacing any downstream strands through its inherent strand displacement activity, which generates single-stranded templates for further priming. This displacement enables continuous, branching amplification as newly displaced strands are immediately available for additional hexamer annealing and extension, leading to exponential propagation of DNA copies without the need for thermal cycling or denaturation steps. The reaction typically proceeds in volumes of 10–100 μL, such as 50 μL for standard setups, and is incubated at 30°C for 8–18 hours to allow completion, yielding microgram quantities of high-molecular-weight, hyperbranched products from minimal input DNA. Upon completion, the reaction is terminated by heating to 65°C for 3–10 minutes to inactivate the . Cleanup follows via purification methods, such as column-based kits or magnetic bead selection, to remove unincorporated primers, , and enzymes; in certain protocols, treatments like are applied to cleave single-stranded regions in branched structures, facilitating .

Product characteristics

The MDA-amplified DNA typically yields 20–30 μg of product from a 100-μl reaction volume starting with 100 fg to 10 ng of input DNA, representing over 1,000-fold amplification for small inputs. Purity is assessed via , with an A260/A280 ratio of approximately 1.8 indicating high-quality, protein-free genomic DNA. Amplicon lengths are characteristically high molecular weight, often exceeding 100 kb, due to the formation of long, concatenated networks from the strand-displacing activity of φ29 . These extended structures are resolved and confirmed using techniques such as , which reveals minimal shearing under standard conditions. Genome coverage uniformity in MDA products reaches 80–95% for bacterial genomes, as measured by sequencing depth or quantitative across loci, though it can vary more in eukaryotic samples due to inherent dynamics. Quality metrics include low fragmentation, with less than 1% short fragments (<5 kb) observed in gel analyses, and an error rate of approximately 10^{-6} to 10^{-7} base substitutions per nucleotide owing to the high fidelity of φ29 polymerase. Chimeric junctions occur at a low frequency of about 1 per 20–25 kb, primarily as inverted sequences, detectable via mapping of amplified sequences to reference genomes.

Advantages

Fidelity and yield

Multiple displacement amplification (MDA) achieves high fidelity through the use of Φ29 DNA polymerase, which possesses 3'–5' exonuclease proofreading activity that corrects errors during synthesis, resulting in an error rate of approximately 10^{-6} to 10^{-7} errors per base—over 100 times lower than the ~10^{-4} errors per base typical of Taq polymerase in PCR-based methods. This superior accuracy has been validated in mutation assays comparing amplified products to unamplified references, showing minimal introduction of spurious variants even at high amplification levels. In terms of yield, MDA demonstrates efficient scaling from linear to exponential amplification based on input DNA quantity, routinely achieving greater than 10^6-fold amplification from single-cell inputs (equivalent to ~6 pg human genomic DNA), enabling sufficient product for downstream applications like next-generation sequencing (NGS). Amplification efficiency supports yields of 20–60 μg from low-input reactions under optimized conditions. MDA provides genomic coverage with relatively low representation bias compared to earlier PCR-based alternatives such as degenerate oligonucleotide-primed PCR (DOP-PCR), though conventional MDA exhibits coefficient of variation (CV) values often exceeding 100% across chromosomal regions due to amplification bias. Improved variants, such as emulsion-based MDA, achieve lower CV (e.g., ~0.36). For instance, MDA attains up to ~98% genome recovery in single-cell sequencing under high-coverage conditions, outperforming DOP-PCR's ~20–40% coverage.

Practical benefits

Multiple displacement amplification (MDA) operates isothermally at a constant temperature of 30°C, relying on the strand-displacing activity of φ29 DNA polymerase, which eliminates the need for thermal cycling equipment such as thermocyclers. This simplifies laboratory workflows and enables deployment in resource-limited or field settings, where portable incubators suffice for amplification. MDA reactions typically complete within 16–18 hours, producing yields of 20–30 μg DNA from standard 100-μL volumes, and can be scaled from microliter reactions to multi-well plate formats for high-throughput processing. Compatibility with automation systems, such as robotic liquid handlers and 384-well plates, further enhances scalability, allowing parallel amplification of numerous samples with minimal manual intervention. The method demonstrates versatility across a wide range of input DNA quantities, from as low as 1 pg (equivalent to sub-single-cell levels) up to micrograms, making it particularly suitable for amplifying precious or limited samples such as those from clinical biopsies or ancient DNA extracts. MDA offers cost-effectiveness through commercial reagent kits, which is lower than the cumulative expense of multiple rounds of traditional PCR, while also reducing hands-on time due to its straightforward, single-step protocol.

Limitations

Amplification biases

Multiple displacement amplification (MDA) introduces several biases that result in uneven genome coverage, primarily due to the stochastic nature of the isothermal amplification process using phi29 DNA polymerase and random hexamer primers. These biases manifest as allelic dropout (ADO) and preferential amplification of certain genomic regions, leading to under- or over-representation of sequences and complicating downstream analyses such as variant calling. Such inconsistencies arise from inefficiencies in primer annealing and strand displacement, particularly in low-input samples like single cells, where template availability is limited. Allelic dropout (ADO) refers to the failure to amplify one allele at heterozygous loci, often resulting in apparent homozygosity or allele loss during SNP calling. In single-cell applications, ADO rates typically range from 60% to 90%, with commercial MDA kits like and exhibiting rates exceeding 60%, attributed to stochastic priming failures and uneven reagent distribution around target sequences. This bias is quantified by measuring heterozygosity loss, where only about 10-30% of heterozygous retain both alleles post-amplification, with higher rates near genomic regions of low complexity like centromeres and telomeres. Preferential amplification further exacerbates coverage nonuniformity by favoring certain genomic features, such as GC-rich regions, which can show slight over-representation, typically less than threefold, compared to AT-rich areas. This occurs due to thermodynamic differences in primer annealing, where higher GC content facilitates more stable hybridization and thus disproportionate amplification. Open chromatin regions may also be preferentially amplified owing to better accessibility, though this is less pronounced in MDA than in PCR-based methods. Quantification of these biases commonly involves assessing coverage variance through techniques like array comparative genomic hybridization (CGH) or whole-genome sequencing, where metrics such as the coefficient of variation (CV) in read depth or heterozygote concordance rates (e.g., 31% ± 21% for heterozygotes) highlight nonuniformity. The Gini coefficient, a measure of amplification uniformity, ideally falls below 0.3 for reliable coverage, but MDA products often exceed this threshold, indicating significant bias. Mitigation strategies focus on protocol optimizations rather than complete elimination of biases, including the use of refined buffer compositions and primer mixtures to enhance annealing efficiency across diverse sequences. Pre-amplification screening with assays like TaqMan qPCR can identify high-quality samples (e.g., >60% concordance predicting >80% SNP call rates), while commercial variants like REPLI-g reduce compared to standard through improved reaction conditions. As of 2025, commercial kits continue to show rates exceeding 60%, inferior to non-MDA methods like Ampli1 (~16% ). Advanced approaches, such as barcoded or droplet-based implementations, further improve uniformity by compartmentalizing reactions and minimizing effects.

Artifact formation

Multiple displacement amplification (MDA) generates non-biological artifacts, primarily chimeras, which are artificial DNA junctions linking non-contiguous genomic regions. These arise from incomplete strand displacement, where displaced 3'-ends fail to fully separate and re-anneal erroneously to nearby 5'-strands, or from primer switching, in which random hexamer primers bind to unintended templates during synthesis. Additional mechanisms include polymerase jumping, where the highly processive Phi29 DNA polymerase dissociates and re-initiates on a different strand, and branch migration, facilitating mispriming within hyperbranched DNA structures formed by ongoing strand displacement. Chimera rates typically range from 1-10% of sequencing reads in next-generation sequencing data, though they can reach up to 76% in long-read platforms like PacBio due to the detection of larger concatemeric structures. Primer-primer interactions in MDA contribute to artifact formation through self-annealing of random hexamers, particularly in AT-rich regions due to lower DNA stability, leading to non-specific amplification products. These interactions promote mispriming events, generating extraneous sequences that mimic genomic fragments but originate from primer dimers or aberrant annealing rather than template DNA. Such artifacts are more prevalent in low-input samples, exacerbating uneven coverage in AT-biased genomes. Other artifacts include concatemers, which manifest as branched DNA networks resulting from repeated strand displacement and re-annealing without resolution, creating hyperbranched, high-molecular-weight products that appear as fused repeats in sequencing assemblies. Low-level mutations, occurring at rates of approximately 1 in 10^6-10^7 bases due to Phi29's , can also arise from environmental factors such as suboptimal reaction temperatures, high primer concentrations, or contaminants inducing or during amplification. Detection of these artifacts relies on bioinformatics tools tailored to MDA products, such as ChimeraMiner, which identifies chimeric junctions by analyzing read alignments and secondary structures, improving structural variant detection by up to 84%. Long-read sequencing validation, using platforms like PacBio or Oxford Nanopore, distinguishes true chimeras from biological variants by resolving branch points in concatemers. Recent 2023 studies in metagenomics have quantified chimera rates post-MDA, reporting reductions to 6-10% with optimized protocols, emphasizing the need for integrated correction pipelines to enhance assembly accuracy in low-biomass environmental samples.

Applications

Single-cell genomics

Multiple displacement amplification (MDA) has been adapted for single-cell genomics through protocols that enable efficient DNA amplification from minimal input, typically involving cell lysis in small volumes followed by direct amplification without intermediate purification steps. In standard workflows, single cells are lysed in volumes ranging from 0.1 to 5 µL using buffers containing detergents like Tween-20 or Triton X-100, often combined with proteinase K to degrade cellular proteins and release genomic DNA. This lysed material is then directly added to MDA reaction mixes containing phi29 DNA polymerase, random hexamer primers, and deoxynucleotide triphosphates, allowing isothermal amplification at 30°C for 8-16 hours. These adaptations minimize loss of material from low-input samples and reduce contamination risks, with commercial kits like REPLI-g Single Cell (QIAGEN) optimizing the process for 1-50 cells per reaction. Yields from these protocols typically range from tens to hundreds of nanograms of amplified DNA per single cell, representing a 10,000- to 100,000-fold increase over the native ~6 pg diploid human genome, with commercial kits achieving up to 40 µg—sufficient for downstream whole-genome sequencing (WGS) or other analyses. MDA enables comprehensive WGS from individual cells, achieving genome coverage of over 80% at sequencing depths of 30× or higher, which is critical for detecting copy number variations (CNVs) and single-nucleotide variants (SNVs). This high coverage facilitates the study of cellular heterogeneity, such as in cancer where MDA-amplified single-cell genomes have revealed subclonal mutations and aneuploidy in tumor cells, or in preimplantation embryos to assess mosaic aneuploidies. For instance, droplet-based MDA variants have demonstrated 70-93% coverage at 20-60× depth in human cancer cell lines, enabling accurate mapping of variants to the reference genome with low allelic dropout rates. MDA's role in large-scale single-cell genomic projects is exemplified by its use in the Human Cell Atlas (HCA) initiative since 2016, where it supports WGS to map genomic variations across diverse human cell types and tissues. Additionally, MDA integrates with single-cell RNA sequencing (scRNA-seq) workflows for multi-omics profiling, such as in transcriptogenomics approaches where genomic DNA is amplified post-transcriptional capture to link and . Recent advances post-2020 include combinations with platforms, where MDA is applied to genomic fractions in multi-ome kits for simultaneous assessment of , CNVs, and transcriptomes in heterogeneous populations like tumor samples, enhancing resolution in studies of disease progression.

Metagenomics and environmental DNA

Multiple displacement amplification (MDA) is particularly suited for of low-biomass environmental samples, where DNA yields are often below 1 ng, such as in , , borehole fluids, and ancient sediments. By employing phi29 for isothermal amplification, MDA enables the generation of sufficient material for downstream sequencing from picogram quantities, thereby facilitating the study of microbial communities in nutrient-poor or dilute environments like and ocean . This approach minimizes the need for extensive sample concentration, which can introduce artifacts, and helps amplify microbial signals in (eDNA) matrices that may contain trace host or extraneous DNA, effectively reducing relative contamination through targeted enrichment. In terms of outcomes, MDA has demonstrated high recovery rates, achieving over 90% genome coverage for low-GC microbial genomes in mock communities simulating environmental mixtures, particularly when using bulk or emulsion-based protocols. These methods have been integral to recovering diverse microbial taxa from complex samples, supporting functional profiling comparable to unamplified high-input metagenomes. For instance, in marine eDNA studies, MDA amplified picogram levels of 13C-labeled DNA from methanol-incubated seawater, yielding fosmid libraries that revealed novel methylotroph pathways with minimal amplification bias. Notable applications include the characterization of viral metagenomes from ocean samples in the 2010s, where MDA preferentially amplified single-stranded DNA viruses like Microviridae from concentrates, producing libraries with 85-96% ssDNA content and higher assembly rates (66-73%) compared to linker-amplified methods, though with biases toward circular genomes. More recently, in 2024 studies integrating MDA with sequencing, high-quality draft genomes of uncultured cable were assembled from low-input environmental isolates, closing nearly complete genomes and highlighting MDA's role in resolving uncultured microbial diversity in samples. To address amplification biases inherent to , such as skews that underrepresent high-GC (>45%) taxa, strategies like MDA— which compartmentalizes reactions to promote even displacement—or primase-based variants of have been developed, improving uniformity and reducing chimeric artifacts in community-level metagenomes. These corrections enhance the reliability of for ecological surveys, though chimeras from strand displacement remain a noted limitation in diverse mixtures.

Clinical and forensic analysis

Multiple displacement amplification (MDA) has emerged as a valuable tool in clinical settings for amplifying low-abundance DNA from liquid biopsies, enabling next-generation sequencing (NGS) analysis of circulating cell-free DNA (cfDNA) to detect rare mutations associated with cancer and other diseases. In liquid biopsies, total cell-free DNA (cfDNA) is typically present in low quantities, around 0.5–10 ng per 100 µL of plasma, of which circulating tumor DNA (ctDNA) represents a small fraction (often <1 ng per 100 µL), making amplification necessary for analysis. MDA facilitates whole-genome amplification to yields exceeding 1 µg from such inputs, supporting comprehensive genomic profiling for precision oncology applications. For instance, template-dependent MDA variants have been optimized for RNA-based liquid biopsies, enhancing sensitivity for transcriptomic analysis in circulating tumor cells (CTCs) while minimizing biases in low-input samples. This approach has been integrated into workflows for serial monitoring of tumor evolution, as demonstrated in studies amplifying cfDNA for variant detection in non-small cell lung cancer patients. In epigenetics research with clinical relevance, is employed to amplify immunoprecipitated () DNA from limited cell numbers, enabling -seq to map modifications and patterns in cancer biopsies from the 2000s onward. Low-input protocols using have achieved sufficient library complexity for sequencing, revealing epigenetic alterations in tumor suppressor genes, such as aberrant enrichment in samples, which informs therapeutic targeting. These applications underscore 's role in bridging epigenomic profiling with clinical diagnostics, particularly for heterogeneous tumors where traditional methods fail due to insufficient material. Forensic applications of MDA center on amplifying trace or degraded DNA for short tandem repeat (STR) profiling in human identification, such as from touch DNA, bone fragments, or aged evidence, where inputs are often below 1 ng. Validation studies from the mid-2000s demonstrated MDA's efficacy, achieving over 95% success rates in generating interpretable STR profiles from sub-nanogram quantities, outperforming direct PCR in challenging samples by providing uniform amplification across loci. Commercial kits like REPLI-g, based on φ29 polymerase-driven MDA, have been widely adopted for forensic casework, reducing allelic imbalance and enabling downstream multiplexing for database matching. Recent advancements in MDA protocols, including those evaluated in 2022 reviews, have further minimized allele dropout to below 10% in low-template scenarios, enhancing reliability for legal proceedings. Regulatory aspects of MDA in clinical and forensic contexts emphasize rigorous validation to meet diagnostic and evidentiary standards. While no standalone FDA-approved MDA kits exist for routine diagnostics as of 2023, MDA-integrated workflows have undergone validation for diagnostic use in targeted applications, such as tuberculosis detection via MDA-PCR assays, ensuring compliance with (CLIA) guidelines. In forensics, MDA methods align with Scientific Working Group on DNA Analysis Methods (SWGDAM) recommendations, with studies confirming reproducibility and low error rates in accredited laboratories for court-admissible profiles. These validations prioritize to avoid adventitious artifacts, supporting MDA's integration into certified pipelines for human-related sample analysis.

References

  1. [1]
    Comprehensive human genome amplification using multiple ... - NIH
    We describe a method, termed multiple displacement amplification (MDA), which provides a highly uniform representation across the genome.
  2. [2]
    Multiple displacement amplification WGA - QIAGEN
    Multiple displacement amplification (MDA) involves the binding of random hexamers to denatured DNA followed by strand displacement synthesis at a constant ...
  3. [3]
    Evaluating the Benefits and Limits of Multiple Displacement ...
    Feb 28, 2025 · Multiple displacement amplification (MDA) outperforms conventional PCR in long fragment and whole-genome amplification, making it attractive ...
  4. [4]
    Comprehensive human genome amplification using multiple ... - PNAS
    We describe a method, termed multiple displacement amplification (MDA), which provides a highly uniform representation across the genome.Materials And Methods · Dna Preparation From Blood... · ResultsMissing: paper | Show results with:paper
  5. [5]
    Recent advances and application in whole‐genome multiple ...
    Dec 1, 2020 · Multiple displacement amplification (MDA) is the most widely used WGA technique. Results. Compared with amplification methods based on PCR and ...Phi29 Dna Polymerase... · Improved Mda-Based... · Mda ApplicationsMissing: seminal | Show results with:seminal
  6. [6]
  7. [7]
    My life with bacteriophage phi29 - PubMed
    Dec 28, 2012 · Very relevant was the discovery of the ϕ29 DNA polymerase, with its properties of extremely high processivity and strand displacement capacity, ...
  8. [8]
    Genomic DNA Amplification from a Single Bacterium - ASM Journals
    Jun 1, 2005 · Genomic DNA was amplified about 5 billion-fold from single, flow-sorted bacterial cells by the multiple displacement amplification (MDA) reaction.
  9. [9]
    the impact of multiple displacement amplification on microbial ecology
    Feb 7, 2008 · A method called multiple displacement amplification (MDA) has been used to circumvent these limitations by amplifying DNA from microbial communities in low- ...
  10. [10]
    Mechanism of chimera formation during the Multiple Displacement ...
    Apr 12, 2007 · The reaction proceeds through a strand displacement mechanism with the phi29 DNA polymerase extending 3'-termini while concurrently displacing ...
  11. [11]
    Assessing the utility of whole-genome amplified serum DNA for ...
    Dec 18, 2009 · Multiple displacement amplification (MDA), in particular, is an ... DNA quantity and A260/A280 ratios were measured using Nanodrop-ND ...
  12. [12]
    Chimera: The spoiler in multiple displacement amplification - PMC
    Long-read sequencing technologies, such as those provided by Pacific Biosciences and Oxford Nanopore Technology, also require high molecular weight ... 100 kb [54] ...
  13. [13]
    A Quantitative Comparison of Single-Cell Whole Genome ...
    Here, we compare three state-of-the-art methods on both bulk and single-cell samples of E. coli DNA: Multiple Displacement Amplification (MDA), Multiple ...
  14. [14]
    Single duplex DNA sequencing with CODEC detects mutations with ...
    Apr 27, 2023 · ... 10−7) was similar to that of duplex sequencing (4.3 × 10−7; Fig. 2a). Because error rates are affected by multiple factors other than a ...
  15. [15]
    Taq Polymerase - an overview | ScienceDirect Topics
    The Taq polymerase, for example, is a low-fidelity enzyme, belonging to the A family, with an error rate of approximately 10−4 bp−1. In addition, this enzyme ...
  16. [16]
    High-resolution mapping of DNA polymerase fidelity using ... - NIH
    Apr 30, 2018 · For Phi29, the estimated error rate fell 2.7-fold from the mean of the two reported literature error rates.Missing: 4.3 × | Show results with:4.3 ×
  17. [17]
    Genomic DNA Amplification from a Single Bacterium - PMC - NIH
    Even a 0.1% efficiency corresponds to a 106- to 107-fold DNA amplification from the single cell and will enable many kinds of specific primer/probe applications ...
  18. [18]
    Robust high-performance nanoliter-volume single-cell ... - PNAS
    Robust high-performance nanoliter-volume single-cell multiple displacement amplification on planar substrates ... 100 kb, and 10 kb (mean of 100 reads per ...
  19. [19]
    Uniform and accurate single-cell sequencing based on emulsion ...
    We demonstrate the proof-of-principle of eWGA with multiple displacement amplification (MDA), a popular WGA method. This easy-to-operate approach enables ...Sign Up For Pnas Alerts · Results And Discussion · Emda Sequencing Library...Missing: invention | Show results with:invention
  20. [20]
    Comparison of variations detection between whole-genome ...
    Aug 6, 2015 · We found that MDA-2 amplified data had the highest mean genome coverage (8.84 %), even higher than that of MALBAC (8.06 %). MDA and MALBAC ...Missing: CV | Show results with:CV
  21. [21]
    Comparison of Multiple Displacement Amplification (MDA) and ...
    Dec 8, 2014 · Compared with PCR-based methods, MDA reduces amplification bias by three to four orders of magnitude and generates much longer amplicons with ...Missing: input usable
  22. [22]
    Recent advances and application of whole genome amplification in ...
    In this paper, we reviewed multiple WGA methods along with their amplification principles, advantages, and disadvantages. ... Comparison of multiple displacement ...
  23. [23]
    A Simplified Improvement to Multiple Displacement Amplification for ...
    Feb 21, 2023 · We present a volume reduction approach that significantly reduces costs while improving genome coverage and uniformity of DNA amplification products in ...
  24. [24]
  25. [25]
  26. [26]
  27. [27]
    Mechanism of chimera formation during the Multiple Displacement ...
    Apr 12, 2007 · Results: Here, we characterize the major types of chimeras formed by carrying out an MDA whole genome amplification from a single E. coli cell ...
  28. [28]
    [PDF] REPLI-g® Single Cell Handbook - QIAGEN
    The REPLI-g Single Cell Kit allows uniform amplification of whole genomic DNA from small samples, enabling a greater variety and number of analyses to be.
  29. [29]
    Optimized Workflow for Whole Genome and Transcriptome Next ...
    May 11, 2023 · The protocol describes the QIAseq single cell procedure for WGA from single cells, or very small quantities of purified genomic DNA using MDA.
  30. [30]
    Massively parallel whole genome amplification for single-cell ...
    Jul 12, 2017 · We introduce single droplet multiple displacement amplification (sd-MDA), a method that enables massively parallel amplification of single cell genomes.
  31. [31]
    Single-Cell Sequencing for Precise Cancer Research
    The MDA method is most popularly applied in WGA due to its high fidelity and simplicity. Several updated MDA kits are suitable for single-cell WGA (scWGA), such ...
  32. [32]
    HSCGD: a comprehensive database of single-cell whole-genome ...
    Oct 29, 2024 · ... cells (5.31%). The database supports various single-cell whole-genome amplification methods, including multiple displacement amplification (MDA) ...
  33. [33]
    Methods and applications for single-cell and spatial multi-omics - PMC
    Mar 2, 2023 · Single-cell transcriptogenomics applies a similar principle as G&T-seq and is compatible with targeted DNA exome sequencing following MDA. The ...
  34. [34]
    Advances in single-cell omics and multiomics for high-resolution ...
    Mar 5, 2024 · This review provides an overview of the recent advances in single-cell and multimodal omics for high-resolution molecular profiling.
  35. [35]
  36. [36]
  37. [37]