Massive parallel sequencing

Last updated

Massive parallel sequencing or massively parallel sequencing is any of several high-throughput approaches to DNA sequencing using the concept of massively parallel processing; it is also called next-generation sequencing (NGS) or second-generation sequencing. Some of these technologies emerged between 1993 and 1998 [1] [2] [3] [4] [5] and have been commercially available since 2005. These technologies use miniaturized and parallelized platforms for sequencing of 1 million to 43 billion short reads (50 to 400 bases each) per instrument run.

Contents

Many NGS platforms differ in engineering configurations and sequencing chemistry. They share the technical paradigm of massive parallel sequencing via spatially separated, clonally amplified DNA templates or single DNA molecules in a flow cell. This design is very different from that of Sanger sequencing—also known as capillary sequencing or first-generation sequencing—which is based on electrophoretic separation of chain-termination products produced in individual sequencing reactions. [6] This methodology allows sequencing to be completed on a larger scale. [7]

NGS platforms

DNA sequencing with commercially available NGS platforms is generally conducted with the following steps. First, DNA sequencing libraries are generated by clonal amplification by PCR in vitro. Second, the DNA is sequenced by synthesis, such that the DNA sequence is determined by the addition of nucleotides to the complementary strand rather than through chain-termination chemistry. Third, the spatially segregated, amplified DNA templates are sequenced simultaneously in a massively parallel fashion without the requirement for a physical separation step. These steps are followed in most NGS platforms, but each utilizes a different strategy. [8]

NGS parallelization of the sequencing reactions generates hundreds of megabases to gigabases of nucleotide sequence reads in a single instrument run. This has enabled a drastic increase in available sequence data and fundamentally changed genome sequencing approaches in the biomedical sciences. [9] Newly emerging NGS technologies and instruments have further contributed to a significant decrease in the cost of sequencing nearing the mark of $1000 per genome sequencing. [10] [11]

As of 2014, massively parallel sequencing platforms are commercially available and their features are summarized in the table. As the pace of NGS technologies is advancing rapidly, technical specifications and pricing are in flux.

An Illumina HiSeq 2000 sequencing machine HiSeq 2000.JPG
An Illumina HiSeq 2000 sequencing machine
NGS platforms
PlatformTemplate preparationChemistryMax read length (bases)Run times (days)Max Gb per Run
Roche 454Clonal-emPCRPyrosequencing400‡0.420.40-0.60
GS FLX TitaniumClonal-emPCRPyrosequencing400‡0.420.035
Illumina MiSeqClonal Bridge AmplificationReversible Dye Terminator2x3000.17-2.715
Illumina HiSeqClonal Bridge AmplificationReversible Dye Terminator2x1500.3-11 [12] 1000 [13]
Illumina Genome Analyzer IIXClonal Bridge AmplificationReversible Dye Terminator [14] [15] 2x1502-1495
Life Technologies SOLiD4Clonal-emPCROligonucleotide 8-mer Chained Ligation [16] 20-454-735-50
Life Technologies Ion Proton [17] Clonal-emPCRNative dNTPs, proton detection2000.5100
Complete GenomicsGridded DNA-nanoballsOligonucleotide 9-mer Unchained Ligation [18] [19] [20] 7x10113000
Helicos Biosciences HeliscopeSingle MoleculeReversible Dye Terminator35‡825
Pacific Biosciences SMRTSingle MoleculePhospholinked Fluorescent Nucleotides10,000 (N50); 30,000+ (max) [21] 0.080.5 [22]


Run times and gigabase (Gb) output per run for single-end sequencing are noted. Run times and outputs approximately double when performing paired-end sequencing. ‡Average read lengths for the Roche 454 and Helicos Biosciences platforms. [23]

Template preparation methods for NGS

Two methods are used in preparing templates for NGS reactions: amplified templates originating from single DNA molecules, and single DNA molecule templates. For imaging systems which cannot detect single fluorescence events, amplification of DNA templates is required. The three most common amplification methods are emulsion PCR (emPCR), rolling circle and solid-phase amplification. The final distribution of templates can be spatially random or on a grid.

Emulsion PCR

In emulsion PCR methods, a DNA library is first generated through random fragmentation of genomic DNA. Single-stranded DNA fragments (templates) are attached to the surface of beads with adaptors or linkers, and one bead is attached to a single DNA fragment from the DNA library. The surface of the beads contains oligonucleotide probes with sequences that are complementary to the adaptors binding the DNA fragments. The beads are then compartmentalized into water-oil emulsion droplets. In the aqueous water-oil emulsion, each of the droplets capturing one bead is a PCR microreactor that produces amplified copies of the single DNA template. [24] [25] [26]

Gridded rolling circle nanoballs

Amplification of a population of single DNA molecules by rolling circle amplification in solution is followed by capture on a grid of spots sized to be smaller than the DNAs to be immobilized. [27] [28] [29] [30]

DNA colony generation (Bridge amplification)

Forward and reverse primers are covalently attached at high-density to the slide in a flow cell. The ratio of the primers to the template on the support defines the surface density of the amplified clusters. The flow cell is exposed to reagents for polymerase-based extension, and priming occurs as the free/distal end of a ligated fragment "bridges" to a complementary oligo on the surface. Repeated denaturation and extension results in localized amplification of DNA fragments in millions of separate locations across the flow cell surface. Solid-phase amplification produces 100–200 million spatially separated template clusters, providing free ends to which a universal sequencing primer is then hybridized to initiate the sequencing reaction. [24] [25] This technology was filed for a patent in 1997 from Glaxo-Welcome's Geneva Biomedical Research Institute (GBRI), by Pascal Mayer, Eric Kawashima, and Laurent Farinelli, [4] [5] and was publicly presented for the first time in 1998. [31] In 1994 Chris Adams and Steve Kron filed a patent on a similar, but non-clonal, surface amplification method, named “bridge amplification” [3] adapted for clonal amplification in 1997 by Church and Mitra. [27] [28]

Single-molecule templates

Protocols requiring DNA amplification are often cumbersome to implement and may introduce sequencing errors. The preparation of single-molecule templates is more straightforward and does not require PCR, which can introduce errors in the amplified templates. AT-rich and GC-rich target sequences often show amplification bias, which results in their underrepresentation in genome alignments and assemblies. Single molecule templates are usually immobilized on solid supports using one of at least three different approaches. In the first approach, spatially distributed individual primer molecules are covalently attached to the solid support. The template, which is prepared by randomly fragmenting the starting material into small sizes (for example,~200–250 bp) and adding common adapters to the fragment ends, is then hybridized to the immobilized primer. In the second approach, spatially distributed single-molecule templates are covalently attached to the solid support by priming and extending single-stranded, single-molecule templates from immobilized primers. A common primer is then hybridized to the template. In either approach, DNA polymerase can bind to the immobilized primed template configuration to initiate the NGS reaction. Both of the above approaches are used by Helicos BioSciences. In a third approach, spatially distributed single polymerase molecules are attached to the solid support, to which a primed template molecule is bound. This approach is used by Pacific Biosciences. Larger DNA molecules (up to tens of thousands of base pairs) can be used with this technique and, unlike the first two approaches, the third approach can be used with real-time methods, resulting in potentially longer read lengths.

Sequencing approaches

Sequencing by synthesis

The objective for sequential sequencing by synthesis (SBS) is to determine the sequencing of a DNA sample by detecting the incorporation of a nucleotide by a DNA polymerase. An engineered polymerase is used to synthesize a copy of a single strand of DNA and the incorporation of each nucleotide is monitored. The principle of sequencing by synthesis was first described in 1993 [1] with improvements published some years later. [32] The key parts are highly similar for all embodiments of SBS and include (1) amplification of DNA to enhance the subsequent signal and to attach the DNA to be sequenced to a solid support,  (2) generation of single stranded DNA on the solid support, (3) incorporation of nucleotides using an engineered polymerase and (4) detection of the incorporation of nucleotide. Then steps 3-4 are repeated and the sequence is assembled from the signals obtained in step 4. This principle of sequencing-by-synthesis has been used for almost all massive parallel sequencing instruments, including 454, PacBio, IonTorrent, Illumina and MGI.

Pyrosequencing

The principle of Pyrosequencing was first described in 1993 [1] by combining a solid support with an engineered DNA polymerase lacking 3´to 5´exonuclease activity (proof-reading) and luminescence real-time detection using the firefly luciferase. All the key concepts of sequencing by synthesis were introduced, including (1) amplification of DNA to enhance the subsequent signal and attach the DNA to be sequenced (template) to a solid support, (2) generation of single stranded DNA on the solid support (3) incorporation of nucleotides using an engineered polymerase and (4) detection of the incorporated nucleotide by light detection in real-time. In a follow-up article, [2] the concept was further developed and in 1998, an article [32] was published in which the authors showed that non-incorporated nucleotides could be removed with a fourth enzyme (apyrase) allowing sequencing by synthesis to be performed without the need for washing away non-incorporated nucleotides.

Sequencing by reversible terminator chemistry

This approach uses reversible terminator-bound dNTPs in a cyclic method that comprises nucleotide incorporation, fluorescence imaging and cleavage. A fluorescently-labeled terminator is imaged as each dNTP is added and then cleaved to allow incorporation of the next base. These nucleotides are chemically blocked such that each incorporation is a unique event. An imaging step follows each base incorporation step, then the blocked group is chemically removed to prepare each strand for the next incorporation by DNA polymerase. This series of steps continues for a specific number of cycles, as determined by user-defined instrument settings. The 3' blocking groups were originally conceived as either enzymatic [33] or chemical reversal [14] [15] The chemical method has been the basis for the Solexa and Illumina machines. Sequencing by reversible terminator chemistry can be a four-colour cycle such as used by Illumina/Solexa, or a one-colour cycle such as used by Helicos BioSciences. Helicos BioSciences used “virtual Terminators”, which are unblocked terminators with a second nucleoside analogue that acts as an inhibitor. These terminators have the appropriate modifications for terminating or inhibiting groups so that DNA synthesis is terminated after a single base addition. [25] [34] [35]

Sequencing-by-ligation mediated by ligase enzymes

In this approach, the sequence extension reaction is not carried out by polymerases but rather by DNA ligase and either one-base-encoded probes or two-base-encoded probes. In its simplest form, a fluorescently labelled probe hybridizes to its complementary sequence adjacent to the primed template. DNA ligase is then added to join the dye-labelled probe to the primer. Non-ligated probes are washed away, followed by fluorescence imaging to determine the identity of the ligated probe. The cycle can be repeated either by using cleavable probes to remove the fluorescent dye and regenerate a 5′-PO4 group for subsequent ligation cycles (chained ligation [16] [36] ) or by removing and hybridizing a new primer to the template (unchained ligation [18] [19] ).

Phospholinked Fluorescent Nucleotides or Real-time sequencing

Pacific Biosciences is currently leading this method. The method of real-time sequencing involves imaging the continuous incorporation of dye-labelled nucleotides during DNA synthesis: single DNA polymerase molecules are attached to the bottom surface of individual zero-mode waveguide detectors (Zmw detectors) that can obtain sequence information while phospholinked nucleotides are being incorporated into the growing primer strand. Pacific Biosciences uses a unique DNA polymerase which better incorporates phospholinked nucleotides and enables the resequencing of closed circular templates. While single-read accuracy is 87%, consensus accuracy has been demonstrated at 99.999% with multi-kilobase read lengths. [37] [38] In 2015, Pacific Biosciences released a new sequencing instrument called the Sequel System, which increases capacity approximately 6.5-fold. [39] [40]

See also

Related Research Articles

<span class="mw-page-title-main">Polymerase chain reaction</span> Laboratory technique to multiply a DNA sample for study

The polymerase chain reaction (PCR) is a method widely used to make millions to billions of copies of a specific DNA sample rapidly, allowing scientists to amplify a very small sample of DNA sufficiently to enable detailed study. PCR was invented in 1983 by American biochemist Kary Mullis at Cetus Corporation. Mullis and biochemist Michael Smith, who had developed other essential ways of manipulating DNA, were jointly awarded the Nobel Prize in Chemistry in 1993.

In genetics and biochemistry, sequencing means to determine the primary structure of an unbranched biopolymer. Sequencing results in a symbolic linear depiction known as a sequence which succinctly summarizes much of the atomic-level structure of the sequenced molecule.

<span class="mw-page-title-main">DNA sequencer</span> A scientific instrument used to automate the DNA sequencing process

A DNA sequencer is a scientific instrument used to automate the DNA sequencing process. Given a sample of DNA, a DNA sequencer is used to determine the order of the four bases: G (guanine), C (cytosine), A (adenine) and T (thymine). This is then reported as a text string, called a read. Some DNA sequencers can be also considered optical instruments as they analyze light signals originating from fluorochromes attached to nucleotides.

Pyrosequencing is a method of DNA sequencing based on the "sequencing by synthesis" principle, in which the sequencing is performed by detecting the nucleotide incorporated by a DNA polymerase. Pyrosequencing relies on light detection based on a chain reaction when pyrophosphate is released. Hence, the name pyrosequencing.

<span class="mw-page-title-main">DNA sequencing</span> Process of determining the nucleic acid sequence

DNA sequencing is the process of determining the nucleic acid sequence – the order of nucleotides in DNA. It includes any method or technology that is used to determine the order of the four bases: adenine, guanine, cytosine, and thymine. The advent of rapid DNA sequencing methods has greatly accelerated biological and medical research and discovery.

<span class="mw-page-title-main">Sanger sequencing</span> Method of DNA sequencing developed in 1977

Sanger sequencing is a method of DNA sequencing that involves electrophoresis and is based on the random incorporation of chain-terminating dideoxynucleotides by DNA polymerase during in vitro DNA replication. After first being developed by Frederick Sanger and colleagues in 1977, it became the most widely used sequencing method for approximately 40 years. It was first commercialized by Applied Biosystems in 1986. More recently, higher volume Sanger sequencing has been replaced by next generation sequencing methods, especially for large-scale, automated genome analyses. However, the Sanger method remains in wide use for smaller-scale projects and for validation of deep sequencing results. It still has the advantage over short-read sequencing technologies in that it can produce DNA sequence reads of > 500 nucleotides and maintains a very low error rate with accuracies around 99.99%. Sanger sequencing is still actively being used in efforts for public health initiatives such as sequencing the spike protein from SARS-CoV-2 as well as for the surveillance of norovirus outbreaks through the Center for Disease Control and Prevention's (CDC) CaliciNet surveillance network.

454 Life Sciences was a biotechnology company based in Branford, Connecticut that specialized in high-throughput DNA sequencing. It was acquired by Roche in 2007 and shut down by Roche in 2013 when its technology became noncompetitive, although production continued until mid-2016.

SNP genotyping is the measurement of genetic variations of single nucleotide polymorphisms (SNPs) between members of a species. It is a form of genotyping, which is the measurement of more general genetic variation. SNPs are one of the most common types of genetic variation. An SNP is a single base pair mutation at a specific locus, usually consisting of two alleles. SNPs are found to be involved in the etiology of many human diseases and are becoming of particular interest in pharmacogenetics. Because SNPs are conserved during evolution, they have been proposed as markers for use in quantitative trait loci (QTL) analysis and in association studies in place of microsatellites. The use of SNPs is being extended in the HapMap project, which aims to provide the minimal set of SNPs needed to genotype the human genome. SNPs can also provide a genetic fingerprint for use in identity testing. The increase of interest in SNPs has been reflected by the furious development of a diverse range of SNP genotyping methods.

<span class="mw-page-title-main">Bisulfite sequencing</span> Lab procedure detecting 5-methylcytosines in DNA

Bisulfitesequencing (also known as bisulphite sequencing) is the use of bisulfite treatment of DNA before routine sequencing to determine the pattern of methylation. DNA methylation was the first discovered epigenetic mark, and remains the most studied. In animals it predominantly involves the addition of a methyl group to the carbon-5 position of cytosine residues of the dinucleotide CpG, and is implicated in repression of transcriptional activity.

<span class="mw-page-title-main">2 base encoding</span>

2 Base Encoding, also called SOLiD, is a next-generation sequencing technology developed by Applied Biosystems and has been commercially available since 2008. These technologies generate hundreds of thousands of small sequence reads at one time. Well-known examples of such DNA sequencing methods include 454 pyrosequencing, the Solexa system and the SOLiD system. These methods have reduced the cost from $0.01/base in 2004 to nearly $0.0001/base in 2006 and increased the sequencing capacity from 1,000,000 bases/machine/day in 2004 to more than 100,000,000 bases/machine/day in 2006.

Single-molecule real-time (SMRT) sequencing is a parallelized single molecule DNA sequencing method. Single-molecule real-time sequencing utilizes a zero-mode waveguide (ZMW). A single DNA polymerase enzyme is affixed at the bottom of a ZMW with a single molecule of DNA as a template. The ZMW is a structure that creates an illuminated observation volume that is small enough to observe only a single nucleotide of DNA being incorporated by DNA polymerase. Each of the four DNA bases is attached to one of four different fluorescent dyes. When a nucleotide is incorporated by the DNA polymerase, the fluorescent tag is cleaved off and diffuses out of the observation area of the ZMW where its fluorescence is no longer observable. A detector detects the fluorescent signal of the nucleotide incorporation, and the base call is made according to the corresponding fluorescence of the dye.

<span class="mw-page-title-main">Helicos Biosciences</span> Former life science company

Helicos BioSciences Corporation was a publicly traded life science company headquartered in Cambridge, Massachusetts focused on genetic analysis technologies for the research, drug discovery and diagnostic markets. The firm's Helicos Genetic Analysis Platform was the first DNA-sequencing instrument to operate by imaging individual DNA molecules. In May 2010, the company announced a 50% layoff and a re-focusing on molecular diagnostics. After long financial troubles, in November 2010, Helicos was delisted from NASDAQ.

Optical mapping is a technique for constructing ordered, genome-wide, high-resolution restriction maps from single, stained molecules of DNA, called "optical maps". By mapping the location of restriction enzyme sites along the unknown DNA of an organism, the spectrum of resulting DNA fragments collectively serves as a unique "fingerprint" or "barcode" for that sequence. Originally developed by Dr. David C. Schwartz and his lab at NYU in the 1990s this method has since been integral to the assembly process of many large-scale sequencing projects for both microbial and eukaryotic genomes. Later technologies use DNA melting, DNA competitive binding or enzymatic labelling in order to create the optical mappings.

<span class="mw-page-title-main">DNA nanoball sequencing</span>

DNA nanoball sequencing is a high throughput sequencing technology that is used to determine the entire genomic sequence of an organism. The method uses rolling circle replication to amplify small fragments of genomic DNA into DNA nanoballs. Fluorescent nucleotides bind to complementary nucleotides and are then polymerized to anchor sequences bound to known sequences on the DNA template. The base order is determined via the fluorescence of the bound nucleotides This DNA sequencing method allows large numbers of DNA nanoballs to be sequenced per run at lower reagent costs compared to other next generation sequencing platforms. However, a limitation of this method is that it generates only short sequences of DNA, which presents challenges to mapping its reads to a reference genome. After purchasing Complete Genomics, the Beijing Genomics Institute (BGI) refined DNA nanoball sequencing to sequence nucleotide samples on their own platform.

<span class="mw-page-title-main">Illumina dye sequencing</span> DNA sequencing method

Illumina dye sequencing is a technique used to determine the series of base pairs in DNA, also known as DNA sequencing. The reversible terminated chemistry concept was invented by Bruno Canard and Simon Sarfati at the Pasteur Institute in Paris. It was developed by Shankar Balasubramanian and David Klenerman of Cambridge University, who subsequently founded Solexa, a company later acquired by Illumina. This sequencing method is based on reversible dye-terminators that enable the identification of single nucleotides as they are washed over DNA strands. It can also be used for whole-genome and region sequencing, transcriptome analysis, metagenomics, small RNA discovery, methylation profiling, and genome-wide protein-nucleic acid interaction analysis.

In DNA sequencing, a read is an inferred sequence of base pairs corresponding to all or part of a single DNA fragment. A typical sequencing experiment involves fragmentation of the genome into millions of molecules, which are size-selected and ligated to adapters. The set of fragments is referred to as a sequencing library, which is sequenced to produce a set of reads.

Multiple Annealing and Looping Based Amplification Cycles (MALBAC) is a quasilinear whole genome amplification method. Unlike conventional DNA amplification methods that are non-linear or exponential, MALBAC utilizes special primers that allow amplicons to have complementary ends and therefore to loop, preventing DNA from being copied exponentially. This results in amplification of only the original genomic DNA and therefore reduces amplification bias. MALBAC is “used to create overlapped shotgun amplicons covering most of the genome”. For next generation sequencing, MALBAC is followed by regular PCR which is used to further amplify amplicons.

Magnetic sequencing is a single-molecule sequencing method in development. A DNA hairpin, containing the sequence of interest, is bound between a magnetic bead and a glass surface. A magnetic field is applied to stretch the hairpin open into single strands, and the hairpin refolds after decreasing of the magnetic field. The hairpin length can be determined by direct imaging of the diffraction rings of the magnetic beads using a simple microscope. The DNA sequences are determined by measuring the changes in the hairpin length following successful hybridization of complementary nucleotides.

<span class="mw-page-title-main">Duplex sequencing</span>

Duplex sequencing is a library preparation and analysis method for next-generation sequencing (NGS) platforms that employs random tagging of double-stranded DNA to detect mutations with higher accuracy and lower error rates.

Reverse complement polymerase chain reaction (RC-PCR) is a modification of the polymerase chain reaction (PCR). It is primarily used to generate amplicon libraries for DNA sequencing by next generation sequencing (NGS). The technique permits both the amplification and the ability to append sequences or functional domains of choice independently to either end of the generated amplicons in a single closed tube reaction. RC-PCR was invented in 2013 by Daniel Ward and Christopher Mattocks at Salisbury NHS Foundation Trust, UK.

References

  1. 1 2 3 Nyren, P.; Pettersson, B.; Uhlen, M. (January 1993). "Solid Phase DNA Minisequencing by an Enzymatic Luminometric Inorganic Pyrophosphate Detection Assay". Analytical Biochemistry. 208 (1): 171–175. doi:10.1006/abio.1993.1024.
  2. 1 2 Ronaghi M, Karamohamed S, Pettersson B, Uhlén M, Nyrén P (November 1996). "Real-time DNA sequencing using detection of pyrophosphate release". Analytical Biochemistry. 242 (1): 84–89. doi:10.1006/abio.1996.0432. PMID   8923969.
  3. 1 2 US 5641658,Adams CP, Kron SJ,"Method for performing amplification of nucleic acid with two primers bound to a single solid support",published 1997-06-24, assigned to Mosaic Technologies Inc.and Whitehead Institute for Biomedical Research
  4. 1 2 EP 0972081,Farinelli L, Kawashima E, Mayer P ),"Method of nucleic acid amplification",published 2007-06-13, assigned to Solexa Ltd.
  5. 1 2 EP 0975802,Kawashima E, Farinellit L, Mayer P,"Method of nucleic acid sequencing",published 2004-06-23
  6. Voelkerding KV, Dames SA, Durtschi JD (April 2009). "Next-generation sequencing: from basic research to diagnostics". Clinical Chemistry. 55 (4): 641–658. doi: 10.1373/clinchem.2008.112789 . PMID   19246620.
  7. Ballard D, Winkler-Galicki J, Wesoły J (July 2020). "Massive parallel sequencing in forensics: advantages, issues, technicalities, and prospects". International Journal of Legal Medicine. 134 (4): 1291–1303. doi:10.1007/s00414-020-02294-0. PMC   7295846 . PMID   32451905.
  8. Anderson MW, Schrijver I (May 2010). "Next generation DNA sequencing and the future of genomic medicine". Genes. 1 (1): 38–69. doi: 10.3390/genes1010038 . PMC   3960862 . PMID   24710010.
  9. Tucker T, Marra M, Friedman JM (August 2009). "Massively parallel sequencing: the next big thing in genetic medicine". American Journal of Human Genetics. 85 (2): 142–154. doi:10.1016/j.ajhg.2009.06.022. PMC   2725244 . PMID   19679224.
  10. von Bubnoff A (March 2008). "Next-generation sequencing: the race is on". Cell. 132 (5): 721–723. doi: 10.1016/j.cell.2008.02.028 . PMID   18329356. S2CID   8413828.
  11. "2008 Release: NHGRI Seeks DNA Sequencing Technologies Fit for Routine Laboratory and Medical Use". Genome.gov. Retrieved 2012-08-05.
  12. "Specifications for HiSeq 2500". Archived from the original on 2014-12-06. Retrieved 2014-11-06.
  13. "HiSeq v4 is here… and it delivers | Edinburgh Genomics". Archived from the original on 2014-11-06. Retrieved 2014-11-06.
  14. 1 2 USpatent 7790869,Ju J, Li Z, Edwards JR, Itagaki Y,"Massive parallel method for decoding DNA and RNA",published 2010-09-07, assigned to The Trustees of Columbia University in the City of New York
  15. 1 2 Bentley DR, Balasubramanian S, Swerdlow HP, Smith GP, Milton J, Brown CG, et al. (November 2008). "Accurate whole human genome sequencing using reversible terminator chemistry". Nature. 456 (7218): 53–59. Bibcode:2008Natur.456...53B. doi:10.1038/nature07517. PMC   2581791 . PMID   18987734.
  16. 1 2 McKernan KJ, Peckham HE, Costa GL, McLaughlin SF, Fu Y, Tsung EF, et al. (September 2009). "Sequence and structural variation in a human genome uncovered by short-read, massively parallel ligation sequencing using two-base encoding". Genome Research. 19 (9): 1527–1541. doi:10.1101/gr.091868.109. PMC   2752135 . PMID   19546169.
  17. "Ion Torrent". Archived from the original on 30 December 2013. Retrieved 1 Jan 2014.
  18. 1 2 Drmanac R, Sparks AB, Callow MJ, Halpern AL, Burns NL, Kermani BG, et al. (January 2010). "Human genome sequencing using unchained base reads on self-assembling DNA nanoarrays". Science. 327 (5961): 78–81. Bibcode:2010Sci...327...78D. doi: 10.1126/science.1181498 . PMID   19892942. S2CID   17309571.
  19. 1 2 Shendure J, Porreca GJ, Reppas NB, Lin X, McCutcheon JP, Rosenbaum AM, et al. (September 2005). "Accurate multiplex polony sequencing of an evolved bacterial genome". Science. 309 (5741): 1728–1732. Bibcode:2005Sci...309.1728S. doi:10.1126/science.1117389. PMID   16081699. S2CID   11405973.
  20. Peters BA, Kermani BG, Sparks AB, Alferov O, Hong P, Alexeev A, et al. (July 2012). "Accurate whole-genome sequencing and haplotyping from 10 to 20 human cells". Nature. 487 (7406): 190–195. Bibcode:2012Natur.487..190P. doi:10.1038/nature11236. PMC   3397394 . PMID   22785314.
  21. Inc, Pacific Biosciences of California (October 3, 2013). "Pacific Biosciences Introduces New Chemistry With Longer Read Lengths to Detect Novel Features in DNA Sequence and Advance Genome Studies of Large Organisms". GlobeNewswire News Room.{{cite web}}: |last= has generic name (help)
  22. Nederbragt L (2013-07-05). "De novo bacterial genome assembly: a solved problem?".
  23. Voelkerding KV, Dames S, Durtschi JD (September 2010). "Next generation sequencing for clinical diagnostics-principles and application to targeted resequencing for hypertrophic cardiomyopathy: a paper from the 2009 William Beaumont Hospital Symposium on Molecular Pathology". The Journal of Molecular Diagnostics. 12 (5): 539–551. doi:10.2353/jmoldx.2010.100043. PMC   2928417 . PMID   20805560.
  24. 1 2 Chee-Seng K, Yun LE, Yudi P, Kee-Seng C (April 2010). "Next Generation Sequencing Technologies and Their Applications.". Encyclopedia of Life Sciences (ELS). Chichester: John Wiley & Sons, Ltd.
  25. 1 2 3 Metzker ML (January 2010). "Sequencing technologies - the next generation". Nature Reviews. Genetics. 11 (1): 31–46. doi:10.1038/nrg2626. PMID   19997069. S2CID   205484500.
  26. Dressman D, Yan H, Traverso G, Kinzler KW, Vogelstein B (July 2003). "Transforming single DNA molecules into fluorescent magnetic particles for detection and enumeration of genetic variations". Proceedings of the National Academy of Sciences of the United States of America. 100 (15): 8817–8822. Bibcode:2003PNAS..100.8817D. doi: 10.1073/pnas.1133470100 . PMC   166396 . PMID   12857956.
  27. 1 2 USpatent 6485944,Church GM, Mitra R,"Replica amplification of nucleic acid arrays",published 2002-11-26, assigned to President and Fellows of Harvard College
  28. 1 2 Mitra RD, Church GM (December 1999). "In situ localized amplification and contact replication of many individual DNA molecules". Nucleic Acids Research. 27 (24): 34e–34. doi:10.1093/nar/27.24.e34. PMC   148757 . PMID   10572186.
  29. US 9624538,Church GM, Porreca GJ, Shendure J, Rosenbaum AM,"Nanogrid rolling circle DNA sequencing",published 2017-04-18, assigned to President and Fellows of Harvard College
  30. USpatent 8445194,Drmanac R, Callow MJ, Drmanac S, Hauser BK, Yeung G,"Single molecule arrays for genetic and chemical analysis",published 2013-05-21, assigned to Callida Genomics Inc.
  31. Mayer P, Matton G, Adessi C, Turcatti G, Mermod JJ, Kawashima E (October 7–10, 1998). A very large scale, high throughput and low cost DNA sequencing method based on a new 2-dimensional DNA auto-patterning process. Fifth International Automation in Mapping and DNA Sequencing Conference. St. Louis, MO, USA. DNA colony massively parallel sequencing ams98 presentation
  32. 1 2 Ronaghi, Mostafa; Uhlén, Mathias; Nyrén, Pål (1998-07-17). "A Sequencing Method Based on Real-Time Pyrophosphate". Science. 281 (5375): 363–365. doi:10.1126/science.281.5375.363. ISSN   0036-8075. PMID   9705713. S2CID   26331871.
  33. US 6833246,Balasubramanian S,"Polynucleotide sequencing",published 2004-12-21, assigned to Solexa Ltd.
  34. "Assay Technology". Illumina. Archived from the original on 2012-08-26. Retrieved 2012-08-05.
  35. "True Single Molecule Sequencing (tSMS™): Helicos BioSciences". Helicosbio.com. Archived from the original on 2012-03-11. Retrieved 2012-08-05.
  36. "Fundamentals of 2 Base Encoding and Color Space". Appliedbiosystems.cnpg.com. Retrieved 2012-08-05.
  37. Chin CS, Alexander DH, Marks P, Klammer AA, Drake J, Heiner C, et al. (June 2013). "Nonhybrid, finished microbial genome assemblies from long-read SMRT sequencing data". Nature Methods. 10 (6): 563–569. doi:10.1038/nmeth.2474. PMID   23644548. S2CID   205421576.
  38. Monica Heger (March 5, 2013). "PacBio Users Report Progress in Long Reads for Plant Genome Assembly, Tricky Regions of Human Genome".
  39. "PacBio Launches Higher-Throughput, Lower-Cost Single-Molecule Sequencing System". October 2015.
  40. "PacBio Announces Sequel Sequencing System - Bio-IT World". www.bio-itworld.com. Archived from the original on 2015-10-02.