DNA sequencing

Source: Wikipedia, the free encyclopedia.

DNA sequencing is the process of determining the

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.[1][2]

Knowledge of DNA sequences has become indispensable for basic biological research, DNA Genographic Projects and in numerous applied fields such as medical diagnosis, biotechnology, forensic biology, virology and biological systematics. Comparing healthy and mutated DNA sequences can diagnose different diseases including various cancers,[3] characterize antibody repertoire,[4] and can be used to guide patient treatment.[5] Having a quick way to sequence DNA allows for faster and more individualized medical care to be administered, and for more organisms to be identified and cataloged.[4]

The rapid speed of sequencing attained with modern DNA sequencing technology has been instrumental in the sequencing of complete DNA sequences, or

genomes, of numerous types and species of life, including the human genome and other complete DNA sequences of many animal, plant, and microbial species.

An example of the results of automated chain-termination DNA sequencing.

The first DNA sequences were obtained in the early 1970s by academic researchers using laborious methods based on two-dimensional chromatography. Following the development of fluorescence-based sequencing methods with a DNA sequencer,[6] DNA sequencing has become easier and orders of magnitude faster.[7][8]

Applications

DNA sequencing may be used to determine the sequence of individual

forensics, and anthropology
.

Molecular biology

Sequencing is used in molecular biology to study genomes and the proteins they encode. Information obtained using sequencing allows researchers to identify changes in genes and noncoding DNA (including regulatory sequences), associations with diseases and phenotypes, and identify potential drug targets.

Evolutionary biology

Since DNA is an informative macromolecule in terms of transmission from one generation to another, DNA sequencing is used in evolutionary biology to study how different organisms are related and how they evolved. In February 2021, scientists reported, for the first time, the sequencing of DNA from animal remains, a mammoth in this instance, over a million years old, the oldest DNA sequenced to date.[9][10]

Metagenomics

The field of metagenomics involves identification of organisms present in a body of water, sewage, dirt, debris filtered from the air, or swab samples from organisms. Knowing which organisms are present in a particular environment is critical to research in ecology, epidemiology, microbiology, and other fields. Sequencing enables researchers to determine which types of microbes may be present in a microbiome, for example.

Virology

As most viruses are too small to be seen by a light microscope, sequencing is one of the main tools in virology to identify and study the virus.[11] Viral genomes can be based in DNA or RNA. RNA viruses are more time-sensitive for genome sequencing, as they degrade faster in clinical samples.[12] Traditional Sanger sequencing and next-generation sequencing are used to sequence viruses in basic and clinical research, as well as for the diagnosis of emerging viral infections, molecular epidemiology of viral pathogens, and drug-resistance testing. There are more than 2.3 million unique viral sequences in GenBank.[11] Recently, NGS has surpassed traditional Sanger as the most popular approach for generating viral genomes.[11]

During the 1990 avian influenza outbreak, viral sequencing determined that the influenza sub-type originated through reassortment between quail and poultry. This led to legislation in Hong Kong that prohibited selling live quail and poultry together at market. Viral sequencing can also be used to estimate when a viral outbreak began by using a molecular clock technique.[12]

Medicine

Medical technicians may sequence genes (or, theoretically, full genomes) from patients to determine if there is risk of genetic diseases. This is a form of genetic testing, though some genetic tests may not involve DNA sequencing.

As of 2013 DNA sequencing was increasingly used to diagnose and treat rare diseases. As more and more genes are identified that cause rare genetic diseases, molecular diagnoses for patients become more mainstream. DNA sequencing allows clinicians to identify genetic diseases, improve disease management, provide reproductive counseling, and more effective therapies.[13] Gene sequencing panels are used to identify multiple potential genetic causes of a suspected disorder.[14]

Also, DNA sequencing may be useful for determining a specific bacteria, to allow for more precise antibiotics treatments, hereby reducing the risk of creating antimicrobial resistance in bacteria populations.[15][16][17][18][19][20]

Forensic investigation

DNA sequencing may be used along with DNA profiling methods for forensic identification[21] and paternity testing. DNA testing has evolved tremendously in the last few decades to ultimately link a DNA print to what is under investigation. The DNA patterns in fingerprint, saliva, hair follicles, etc. uniquely separate each living organism from another. Testing DNA is a technique which can detect specific genomes in a DNA strand to produce a unique and individualized pattern.

The four canonical bases

The canonical structure of DNA has four bases:

5 methyl cytosine) common in humans, may or may not be detected.[25]

In almost all organisms, DNA is synthesized in vivo using only the 4 canonical bases; modification that occurs post replication creates other bases like 5 methyl C. However, some bacteriophage can incorporate a non standard base directly.[26]

In addition to modifications, DNA is under constant assault by environmental agents such as UV and Oxygen radicals. At the present time, the presence of such damaged bases is not detected by most DNA sequencing methods, although PacBio has published on this https://www.pacb.com/publications/direct-detection-and-sequencing-of-damaged-dna-bases/

History

Discovery of DNA structure and function

Deoxyribonucleic acid (DNA) was first discovered and isolated by Friedrich Miescher in 1869, but it remained under-studied for many decades because proteins, rather than DNA, were thought to hold the genetic blueprint to life. This situation changed after 1944 as a result of some experiments by Oswald Avery, Colin MacLeod, and Maclyn McCarty demonstrating that purified DNA could change one strain of bacteria into another. This was the first time that DNA was shown capable of transforming the properties of cells.

In 1953,

double-helix model of DNA, based on crystallized X-ray structures being studied by Rosalind Franklin. According to the model, DNA is composed of two strands of nucleotides coiled around each other, linked together by hydrogen bonds and running in opposite directions. Each strand is composed of four complementary nucleotides – adenine (A), cytosine (C), guanine (G) and thymine (T) – with an A on one strand always paired with T on the other, and C always paired with G. They proposed that such a structure allowed each strand to be used to reconstruct the other, an idea central to the passing on of hereditary information between generations.[27]

Frederick Sanger, a pioneer of sequencing. Sanger is one of the few scientists who was awarded two Nobel prizes, one for the sequencing of proteins, and the other for the sequencing of DNA.

The foundation for sequencing proteins was first laid by the work of Frederick Sanger who by 1955 had completed the sequence of all the amino acids in insulin, a small protein secreted by the pancreas. This provided the first conclusive evidence that proteins were chemical entities with a specific molecular pattern rather than a random mixture of material suspended in fluid. Sanger's success in sequencing insulin spurred on x-ray crystallographers, including Watson and Crick, who by now were trying to understand how DNA directed the formation of proteins within a cell. Soon after attending a series of lectures given by Frederick Sanger in October 1954, Crick began developing a theory which argued that the arrangement of nucleotides in DNA determined the sequence of amino acids in proteins, which in turn helped determine the function of a protein. He published this theory in 1958.[28]

RNA sequencing

University of Ghent (Ghent, Belgium), in 1972[29] and 1976.[30] Traditional RNA sequencing methods require the creation of a cDNA molecule which must be sequenced.[31]

Early DNA sequencing methods

The first method for determining DNA sequences involved a location-specific primer extension strategy established by Ray Wu at Cornell University in 1970.[32] DNA polymerase catalysis and specific nucleotide labeling, both of which figure prominently in current sequencing schemes, were used to sequence the cohesive ends of lambda phage DNA.[33][34][35] Between 1970 and 1973, Wu, R Padmanabhan and colleagues demonstrated that this method can be employed to determine any DNA sequence using synthetic location-specific primers.[36][37][8] Frederick Sanger then adopted this primer-extension strategy to develop more rapid DNA sequencing methods at the MRC Centre, Cambridge, UK and published a method for "DNA sequencing with chain-terminating inhibitors" in 1977.[38] Walter Gilbert and Allan Maxam at Harvard also developed sequencing methods, including one for "DNA sequencing by chemical degradation".[39][40] In 1973, Gilbert and Maxam reported the sequence of 24 basepairs using a method known as wandering-spot analysis.[41] Advancements in sequencing were aided by the concurrent development of recombinant DNA technology, allowing DNA samples to be isolated from sources other than viruses.

Sequencing of full genomes

bacteriophage φX174. Each coloured block represents a gene.

The first full DNA genome to be sequenced was that of

Epstein-Barr virus in 1984, finding it contained 172,282 nucleotides. Completion of the sequence marked a significant turning point in DNA sequencing because it was achieved with no prior genetic profile knowledge of the virus.[43][8]

A non-radioactive method for transferring the DNA molecules of sequencing reaction mixtures onto an immobilizing matrix during

The Institute for Genomic Research (TIGR) published the first complete genome of a free-living organism, the bacterium Haemophilus influenzae. The circular chromosome contains 1,830,137 bases and its publication in the journal Science[50]
marked the first published use of whole-genome shotgun sequencing, eliminating the need for initial mapping efforts.

By 2001, shotgun sequencing methods had been used to produce a draft sequence of the human genome.[51][52]

High-throughput sequencing (HTS) methods

History of sequencing technology [53]

Several new methods for DNA sequencing were developed in the mid to late 1990s and were implemented in commercial

DNA sequencers by 2000. Together these were called the "next-generation" or "second-generation" sequencing (NGS) methods, in order to distinguish them from the earlier methods, including Sanger sequencing
. In contrast to the first generation of sequencing, NGS technology is typically characterized by being highly scalable, allowing the entire genome to be sequenced at once. Usually, this is accomplished by fragmenting the genome into small pieces, randomly sampling for a fragment, and sequencing it using one of a variety of technologies, such as those described below. An entire genome is possible because multiple fragments are sequenced at once (giving it the name "massively parallel" sequencing) in an automated process.

NGS technology has tremendously empowered researchers to look for insights into health, anthropologists to investigate human origins, and is catalyzing the "Personalized Medicine" movement. However, it has also opened the door to more room for error. There are many software tools to carry out the computational analysis of NGS data, often compiled at online platforms such as CSI NGS Portal, each with its own algorithm. Even the parameters within one software package can change the outcome of the analysis. In addition, the large quantities of data produced by DNA sequencing have also required development of new methods and programs for sequence analysis. Several efforts to develop standards in the NGS field have been attempted to address these challenges, most of which have been small-scale efforts arising from individual labs. Most recently, a large, organized, FDA-funded effort has culminated in the BioCompute standard.

On 26 October 1990,

Roger Tsien, Pepi Ross, Margaret Fahnestock and Allan J Johnston filed a patent describing stepwise ("base-by-base") sequencing with removable 3' blockers on DNA arrays (blots and single DNA molecules).[54]
In 1996, Pål Nyrén and his student Mostafa Ronaghi at the Royal Institute of Technology in Stockholm published their method of pyrosequencing.[55]

On 1 April 1997,

Illumina
's Hi-Seq genome sequencers.

In 1998, Phil Green and Brent Ewing of the University of Washington described their phred quality score for sequencer data analysis,[57] a landmark analysis technique that gained widespread adoption, and which is still the most common metric for assessing the accuracy of a sequencing platform.[58]

Lynx Therapeutics published and marketed

DNA sequencers were sold to independent laboratories.[59]

Basic methods

Maxam-Gilbert sequencing

Allan Maxam and Walter Gilbert published a DNA sequencing method in 1977 based on chemical modification of DNA and subsequent cleavage at specific bases.[39] Also known as chemical sequencing, this method allowed purified samples of double-stranded DNA to be used without further cloning. This method's use of radioactive labeling and its technical complexity discouraged extensive use after refinements in the Sanger methods had been made.

Maxam-Gilbert sequencing requires radioactive labeling at one 5' end of the DNA and purification of the DNA fragment to be sequenced. Chemical treatment then generates breaks at a small proportion of one or two of the four nucleotide bases in each of four reactions (G, A+G, C, C+T). The concentration of the modifying chemicals is controlled to introduce on average one modification per DNA molecule. Thus a series of labeled fragments is generated, from the radiolabeled end to the first "cut" site in each molecule. The fragments in the four reactions are electrophoresed side by side in denaturing acrylamide gels for size separation. To visualize the fragments, the gel is exposed to X-ray film for autoradiography, yielding a series of dark bands each corresponding to a radiolabeled DNA fragment, from which the sequence may be inferred.[39]

This method is mostly obsolete as of 2023.[60]

Chain-termination methods

The chain-termination method developed by Frederick Sanger and coworkers in 1977 soon became the method of choice, owing to its relative ease and reliability.[38][61] When invented, the chain-terminator method used fewer toxic chemicals and lower amounts of radioactivity than the Maxam and Gilbert method. Because of its comparative ease, the Sanger method was soon automated and was the method used in the first generation of DNA sequencers.

Sanger sequencing is the method which prevailed from the 1980s until the mid-2000s. Over that period, great advances were made in the technique, such as fluorescent labelling, capillary electrophoresis, and general automation. These developments allowed much more efficient sequencing, leading to lower costs. The Sanger method, in mass production form, is the technology which produced the first human genome in 2001, ushering in the age of genomics. However, later in the decade, radically different approaches reached the market, bringing the cost per genome down from $100 million in 2001 to $10,000 in 2011.[62]

Sequencing by synthesis

The objective for sequential sequencing by synthesis (SBS) is to determine the sequencing of a

IonTorrent, Illumina and MGI
.

Large-scale sequencing and de novo sequencing

Genomic DNA is fragmented into random pieces and cloned as a bacterial library. DNA from individual bacterial clones is sequenced and the sequence is assembled by using overlapping DNA regions.

Large-scale sequencing often aims at sequencing very long DNA pieces, such as whole

DNA vector and amplified in a bacterial host such as Escherichia coli. Short DNA fragments purified from individual bacterial colonies are individually sequenced and assembled electronically into one long, contiguous sequence. Studies have shown that adding a size selection step to collect DNA fragments of uniform size can improve sequencing efficiency and accuracy of the genome assembly. In these studies, automated sizing has proven to be more reproducible and precise than manual gel sizing.[65][66][67]

The term "de novo sequencing" specifically refers to methods used to determine the sequence of DNA with no previously known sequence. De novo translates from Latin as "from the beginning". Gaps in the assembled sequence may be filled by

sequence repeats
often causing gaps in genome assembly.

Most sequencing approaches use an in vitro cloning step to amplify individual DNA molecules, because their molecular detection methods are not sensitive enough for single molecule sequencing. Emulsion PCR

Life Technologies).[69][70][71] Emulsion PCR is also used in the GemCode and Chromium platforms developed by 10x Genomics.[72]

Shotgun sequencing

Shotgun sequencing is a sequencing method designed for analysis of DNA sequences longer than 1000 base pairs, up to and including entire chromosomes. This method requires the target DNA to be broken into random fragments. After sequencing individual fragments using the chain termination method, the sequences can be reassembled on the basis of their overlapping regions.[73]

High-throughput methods

Multiple, fragmented sequence reads must be assembled together on the basis of their overlapping areas.

High-throughput sequencing, which includes next-generation "short-read" and third-generation "long-read" sequencing methods,

ChIP-sequencing), and epigenome characterization.[74]

The high demand for low-cost sequencing has driven the development of high-throughput sequencing technologies that parallelize the sequencing process, producing thousands or millions of sequences concurrently.[75][76][77] High-throughput sequencing technologies are intended to lower the cost of DNA sequencing beyond what is possible with standard dye-terminator methods.[78] In ultra-high-throughput sequencing as many as 500,000 sequencing-by-synthesis operations may be run in parallel.[79][80][81] Such technologies led to the ability to sequence an entire human genome in as little as one day.[82] As of 2019, corporate leaders in the development of high-throughput sequencing products included Illumina, Qiagen and ThermoFisher Scientific.[82]

Comparison of high-throughput sequencing methods[83][84]
Method Read length Accuracy (single read not consensus) Reads per run Time per run Cost per 1 billion bases (in US$) Advantages Disadvantages
Single-molecule real-time sequencing (Pacific Biosciences) 30,000 bp (
N50
);

maximum read length >100,000 bases[85][86][87]

87% raw-read accuracy[88] 4,000,000 per Sequel 2 SMRT cell, 100–200 gigabases[85][89][90] 30 minutes to 20 hours[85][91] $7.2-$43.3 Fast. Detects 4mC, 5mC, 6mA.[92] Moderate throughput. Equipment can be very expensive.
Ion semiconductor (Ion Torrent sequencing) up to 600 bp[93] 99.6%[94] up to 80 million 2 hours $66.8-$950 Less expensive equipment. Fast. Homopolymer errors.
Pyrosequencing (454) 700 bp 99.9% 1 million 24 hours $10,000 Long read size. Fast. Runs are expensive. Homopolymer errors.
Sequencing by synthesis (Illumina) MiniSeq, NextSeq: 75–300 bp;

MiSeq: 50–600 bp;

HiSeq 2500: 50–500 bp;

HiSeq 3/4000: 50–300 bp;

HiSeq X: 300 bp

99.9% (Phred30) MiniSeq/MiSeq: 1–25 Million;

NextSeq: 130-00 Million;

HiSeq 2500: 300 million – 2 billion;

HiSeq 3/4000 2.5 billion;

HiSeq X: 3 billion

1 to 11 days, depending upon sequencer and specified read length[95] $5 to $150 Potential for high sequence yield, depending upon sequencer model and desired application. Equipment can be very expensive. Requires high concentrations of DNA.
Combinatorial probe anchor synthesis (cPAS- BGI/MGI) BGISEQ-50: 35-50bp;

MGISEQ 200: 50-200bp;

BGISEQ-500, MGISEQ-2000: 50-300bp[96]

99.9% (Phred30) BGISEQ-50: 160M;

MGISEQ 200: 300M;

BGISEQ-500: 1300M per flow cell;

MGISEQ-2000: 375M FCS flow cell, 1500M FCL flow cell per flow cell.

1 to 9 days depending on instrument, read length and number of flow cells run at a time. $5– $120
Sequencing by ligation (SOLiD sequencing) 50+35 or 50+50 bp 99.9% 1.2 to 1.4 billion 1 to 2 weeks $60–130 Low cost per base. Slower than other methods. Has issues sequencing palindromic sequences.[97]
Nanopore Sequencing Dependent on library preparation, not the device, so user chooses read length (up to 2,272,580 bp reported[98]). ~92–97% single read dependent on read length selected by user data streamed in real time. Choose 1 min to 48 hrs $7–100 Longest individual reads. Accessible user community. Portable (Palm sized). Lower throughput than other machines, Single read accuracy in 90s.
GenapSys Sequencing Around 150 bp single-end 99.9% (Phred30) 1 to 16 million Around 24 hours $667 Low-cost of instrument ($10,000)
Chain termination (Sanger sequencing) 400 to 900 bp 99.9% N/A 20 minutes to 3 hours $2,400,000 Useful for many applications. More expensive and impractical for larger sequencing projects. This method also requires the time-consuming step of plasmid cloning or PCR.

Long-read sequencing methods

Single molecule real time (SMRT) sequencing

SMRT sequencing is based on the sequencing by synthesis approach. The DNA is synthesized in zero-mode wave-guides (ZMWs) – small well-like containers with the capturing tools located at the bottom of the well. The sequencing is performed with use of unmodified polymerase (attached to the ZMW bottom) and fluorescently labelled nucleotides flowing freely in the solution. The wells are constructed in a way that only the fluorescence occurring by the bottom of the well is detected. The fluorescent label is detached from the nucleotide upon its incorporation into the DNA strand, leaving an unmodified DNA strand. According to Pacific Biosciences (PacBio), the SMRT technology developer, this methodology allows detection of nucleotide modifications (such as cytosine methylation). This happens through the observation of polymerase kinetics. This approach allows reads of 20,000 nucleotides or more, with average read lengths of 5 kilobases.[89][99] In 2015, Pacific Biosciences announced the launch of a new sequencing instrument called the Sequel System, with 1 million ZMWs compared to 150,000 ZMWs in the PacBio RS II instrument.[100][101] SMRT sequencing is referred to as "third-generation" or "long-read" sequencing.

Nanopore DNA sequencing

The DNA passing through the nanopore changes its ion current. This change is dependent on the shape, size and length of the DNA sequence. Each type of the nucleotide blocks the ion flow through the pore for a different period of time. The method does not require modified nucleotides and is performed in real time. Nanopore sequencing is referred to as "third-generation" or "long-read" sequencing, along with SMRT sequencing.

Early industrial research into this method was based on a technique called 'exonuclease sequencing', where the readout of electrical signals occurred as nucleotides passed by alpha(α)-hemolysin pores covalently bound with cyclodextrin.[102] However the subsequent commercial method, 'strand sequencing', sequenced DNA bases in an intact strand.

Two main areas of nanopore sequencing in development are solid state nanopore sequencing, and protein based nanopore sequencing. Protein nanopore sequencing utilizes membrane protein complexes such as α-hemolysin, MspA (Mycobacterium smegmatis Porin A) or CssG, which show great promise given their ability to distinguish between individual and groups of nucleotides.[103] In contrast, solid-state nanopore sequencing utilizes synthetic materials such as silicon nitride and aluminum oxide and it is preferred for its superior mechanical ability and thermal and chemical stability.[104] The fabrication method is essential for this type of sequencing given that the nanopore array can contain hundreds of pores with diameters smaller than eight nanometers.[103]

The concept originated from the idea that single stranded DNA or RNA molecules can be electrophoretically driven in a strict linear sequence through a biological pore that can be less than eight nanometers, and can be detected given that the molecules release an ionic current while moving through the pore. The pore contains a detection region capable of recognizing different bases, with each base generating various time specific signals corresponding to the sequence of bases as they cross the pore which are then evaluated.[104] Precise control over the DNA transport through the pore is crucial for success. Various enzymes such as exonucleases and polymerases have been used to moderate this process by positioning them near the pore's entrance.[105]

Short-read sequencing methods

Massively parallel signature sequencing (MPSS)

The first of the high-throughput sequencing technologies,

cDNA for measurements of gene expression levels.[59]

Polony sequencing

The

Life Technologies, now part of Thermo Fisher Scientific
.

454 pyrosequencing

A parallelized version of

picoliter-volume wells each containing a single bead and sequencing enzymes. Pyrosequencing uses luciferase to generate light for detection of the individual nucleotides added to the nascent DNA, and the combined data are used to generate sequence reads.[69] This technology provides intermediate read length and price per base compared to Sanger sequencing on one end and Solexa and SOLiD on the other.[78]

Illumina (Solexa) sequencing

Illumina, was founded by Shankar Balasubramanian and David Klenerman in 1998, and developed a sequencing method based on reversible dye-terminators technology, and engineered polymerases.[107] The reversible terminated chemistry concept was invented by Bruno Canard and Simon Sarfati at the Pasteur Institute in Paris.[108][109] It was developed internally at Solexa by those named on the relevant patents. In 2004, Solexa acquired the company Manteia Predictive Medicine in order to gain a massively parallel sequencing technology invented in 1997 by Pascal Mayer and Laurent Farinelli.[56]
It is based on "DNA clusters" or "DNA colonies", which involves the clonal amplification of DNA on a surface. The cluster technology was co-acquired with Lynx Therapeutics of California. Solexa Ltd. later merged with Lynx to form Solexa Inc.

An Illumina HiSeq 2500 sequencer
Illumina NovaSeq 6000 flow cell

In this method, DNA molecules and primers are first attached on a slide or flow cell and amplified with

fluorescently labeled
nucleotides. Then the dye, along with the terminal 3' blocker, is chemically removed from the DNA, allowing for the next cycle to begin. Unlike pyrosequencing, the DNA chains are extended one nucleotide at a time and image acquisition can be performed at a delayed moment, allowing for very large arrays of DNA colonies to be captured by sequential images taken from a single camera.

An Illumina MiSeq sequencer

Decoupling the enzymatic reaction and the image capture allows for optimal throughput and theoretically unlimited sequencing capacity. With an optimal configuration, the ultimately reachable instrument throughput is thus dictated solely by the analog-to-digital conversion rate of the camera, multiplied by the number of cameras and divided by the number of pixels per DNA colony required for visualizing them optimally (approximately 10 pixels/colony). In 2012, with cameras operating at more than 10 MHz A/D conversion rates and available optics, fluidics and enzymatics, throughput can be multiples of 1 million nucleotides/second, corresponding roughly to 1 human genome equivalent at 1x coverage per hour per instrument, and 1 human genome re-sequenced (at approx. 30x) per day per instrument (equipped with a single camera).[110]

Combinatorial probe anchor synthesis (cPAS)

This method is an upgraded modification to combinatorial probe anchor ligation technology (cPAL) described by

BGI in 2013.[112] The two companies have refined the technology to allow for longer read lengths, reaction time reductions and faster time to results. In addition, data are now generated as contiguous full-length reads in the standard FASTQ file format and can be used as-is in most short-read-based bioinformatics analysis pipelines.[113][citation needed
]

The two technologies that form the basis for this high-throughput sequencing technology are DNA nanoballs (DNB) and patterned arrays for nanoball attachment to a solid surface.[111] DNA nanoballs are simply formed by denaturing double stranded, adapter ligated libraries and ligating the forward strand only to a splint oligonucleotide to form a ssDNA circle. Faithful copies of the circles containing the DNA insert are produced utilizing Rolling Circle Amplification that generates approximately 300–500 copies. The long strand of ssDNA folds upon itself to produce a three-dimensional nanoball structure that is approximately 220 nm in diameter. Making DNBs replaces the need to generate PCR copies of the library on the flow cell and as such can remove large proportions of duplicate reads, adapter-adapter ligations and PCR induced errors.[113][citation needed]

A BGI MGISEQ-2000RS sequencer

The patterned array of positively charged spots is fabricated through photolithography and etching techniques followed by chemical modification to generate a sequencing flow cell. Each spot on the flow cell is approximately 250 nm in diameter, are separated by 700 nm (centre to centre) and allows easy attachment of a single negatively charged DNB to the flow cell and thus reducing under or over-clustering on the flow cell.[111][citation needed]

Sequencing is then performed by addition of an oligonucleotide probe that attaches in combination to specific sites within the DNB. The probe acts as an anchor that then allows one of four single reversibly inactivated, labelled nucleotides to bind after flowing across the flow cell. Unbound nucleotides are washed away before laser excitation of the attached labels then emit fluorescence and signal is captured by cameras that is converted to a digital output for base calling. The attached base has its terminator and label chemically cleaved at completion of the cycle. The cycle is repeated with another flow of free, labelled nucleotides across the flow cell to allow the next nucleotide to bind and have its signal captured. This process is completed a number of times (usually 50 to 300 times) to determine the sequence of the inserted piece of DNA at a rate of approximately 40 million nucleotides per second as of 2018.[citation needed]

SOLiD sequencing

Library preparation for the SOLiD platform
Two-base encoding scheme. In two-base encoding, each unique pair of bases on the 3' end of the probe is assigned one out of four possible colors. For example, "AA" is assigned to blue, "AC" is assigned to green, and so on for all 16 unique pairs. During sequencing, each base in the template is sequenced twice, and the resulting data are decoded according to this scheme.

Life Technologies brand) SOLiD technology employs sequencing by ligation. Here, a pool of all possible oligonucleotides of a fixed length are labeled according to the sequenced position. Oligonucleotides are annealed and ligated; the preferential ligation by DNA ligase for matching sequences results in a signal informative of the nucleotide at that position. Each base in the template is sequenced twice, and the resulting data are decoded according to the 2 base encoding scheme used in this method. Before sequencing, the DNA is amplified by emulsion PCR. The resulting beads, each containing single copies of the same DNA molecule, are deposited on a glass slide.[114] The result is sequences of quantities and lengths comparable to Illumina sequencing.[78] This sequencing by ligation method has been reported to have some issue sequencing palindromic sequences.[97]

Ion Torrent semiconductor sequencing

Ion Torrent Systems Inc. (now owned by

homopolymer repeats are present in the template sequence, multiple nucleotides will be incorporated in a single cycle. This leads to a corresponding number of released hydrogens and a proportionally higher electronic signal.[115]

Sequencing of the TAGGCT template with IonTorrent, PacBioRS and GridION

DNA nanoball sequencing

genomic sequence of an organism. The company Complete Genomics uses this technology to sequence samples submitted by independent researchers. The method uses rolling circle replication to amplify small fragments of genomic DNA into DNA nanoballs. Unchained sequencing by ligation is then used to determine the nucleotide sequence.[116] This method of DNA sequencing allows large numbers of DNA nanoballs to be sequenced per run and at low reagent costs compared to other high-throughput sequencing platforms.[117] However, only short sequences of DNA are determined from each DNA nanoball which makes mapping the short reads to a reference genome difficult.[116]

Heliscope single molecule sequencing

Heliscope sequencing is a method of single-molecule sequencing developed by Helicos Biosciences. It uses DNA fragments with added poly-A tail adapters which are attached to the flow cell surface. The next steps involve extension-based sequencing with cyclic washes of the flow cell with fluorescently labeled nucleotides (one nucleotide type at a time, as with the Sanger method). The reads are performed by the Heliscope sequencer.[118][119] The reads are short, averaging 35 bp.[120] What made this technology especially novel was that it was the first of its class to sequence non-amplified DNA, thus preventing any read errors associated with amplification steps.[121] In 2009 a human genome was sequenced using the Heliscope, however in 2012 the company went bankrupt.[122]

Microfluidic Systems

There are two main microfluidic systems that are used to sequence DNA; droplet based microfluidics and digital microfluidics. Microfluidic devices solve many of the current limitations of current sequencing arrays.

Abate et al. studied the use of droplet-based microfluidic devices for DNA sequencing.[4] These devices have the ability to form and process picoliter sized droplets at the rate of thousands per second. The devices were created from polydimethylsiloxane (PDMS) and used Forster resonance energy transfer, FRET assays to read the sequences of DNA encompassed in the droplets. Each position on the array tested for a specific 15 base sequence.[4]

Fair et al. used digital microfluidic devices to study DNA pyrosequencing.[123] Significant advantages include the portability of the device, reagent volume, speed of analysis, mass manufacturing abilities, and high throughput. This study provided a proof of concept showing that digital devices can be used for pyrosequencing; the study included using synthesis, which involves the extension of the enzymes and addition of labeled nucleotides.[123]

Boles et al. also studied pyrosequencing on digital microfluidic devices.[124] They used an electro-wetting device to create, mix, and split droplets. The sequencing uses a three-enzyme protocol and DNA templates anchored with magnetic beads. The device was tested using two protocols and resulted in 100% accuracy based on raw pyrogram levels. The advantages of these digital microfluidic devices include size, cost, and achievable levels of functional integration.[124]

DNA sequencing research, using microfluidics, also has the ability to be applied to the sequencing of RNA, using similar droplet microfluidic techniques, such as the method, inDrops.[125] This shows that many of these DNA sequencing techniques will be able to be applied further and be used to understand more about genomes and transcriptomes.

Methods in development

DNA sequencing methods currently under development include reading the sequence as a DNA strand transits through

atomic force microscopy or transmission electron microscopy that are used to identify the positions of individual nucleotides within long DNA fragments (>5,000 bp) by nucleotide labeling with heavier elements (e.g., halogens) for visual detection and recording.[128][129]
Third generation technologies aim to increase throughput and decrease the time to result and cost by eliminating the need for excessive reagents and harnessing the processivity of DNA polymerase.[130]

Tunnelling currents DNA sequencing

Another approach uses measurements of the electrical tunnelling currents across single-strand DNA as it moves through a channel. Depending on its electronic structure, each base affects the tunnelling current differently,[131] allowing differentiation between different bases.[132]

The use of tunnelling currents has the potential to sequence orders of magnitude faster than ionic current methods and the sequencing of several DNA oligomers and micro-RNA has already been achieved.[133]

Sequencing by hybridization

Sequencing by hybridization is a non-enzymatic method that uses a DNA microarray. A single pool of DNA whose sequence is to be determined is fluorescently labeled and hybridized to an array containing known sequences. Strong hybridization signals from a given spot on the array identifies its sequence in the DNA being sequenced.[134]

This method of sequencing utilizes binding characteristics of a library of short single stranded DNA molecules (oligonucleotides), also called DNA probes, to reconstruct a target DNA sequence. Non-specific hybrids are removed by washing and the target DNA is eluted.[135] Hybrids are re-arranged such that the DNA sequence can be reconstructed. The benefit of this sequencing type is its ability to capture a large number of targets with a homogenous coverage.[136] A large number of chemicals and starting DNA is usually required. However, with the advent of solution-based hybridization, much less equipment and chemicals are necessary.[135]

Sequencing with mass spectrometry

Mass spectrometry may be used to determine DNA sequences. Matrix-assisted laser desorption ionization time-of-flight mass spectrometry, or MALDI-TOF MS, has specifically been investigated as an alternative method to gel electrophoresis for visualizing DNA fragments. With this method, DNA fragments generated by chain-termination sequencing reactions are compared by mass rather than by size. The mass of each nucleotide is different from the others and this difference is detectable by mass spectrometry. Single-nucleotide mutations in a fragment can be more easily detected with MS than by gel electrophoresis alone. MALDI-TOF MS can more easily detect differences between RNA fragments, so researchers may indirectly sequence DNA with MS-based methods by converting it to RNA first.[137]

The higher resolution of DNA fragments permitted by MS-based methods is of special interest to researchers in forensic science, as they may wish to find

single-nucleotide polymorphisms in human DNA samples to identify individuals. These samples may be highly degraded so forensic researchers often prefer mitochondrial DNA for its higher stability and applications for lineage studies. MS-based sequencing methods have been used to compare the sequences of human mitochondrial DNA from samples in a Federal Bureau of Investigation database[138] and from bones found in mass graves of World War I soldiers.[139]

Early chain-termination and TOF MS methods demonstrated read lengths of up to 100 base pairs.[140] Researchers have been unable to exceed this average read size; like chain-termination sequencing alone, MS-based DNA sequencing may not be suitable for large de novo sequencing projects. Even so, a recent study did use the short sequence reads and mass spectroscopy to compare single-nucleotide polymorphisms in pathogenic Streptococcus strains.[141]

Microfluidic Sanger sequencing

In microfluidic Sanger sequencing the entire thermocycling amplification of DNA fragments as well as their separation by electrophoresis is done on a single glass wafer (approximately 10 cm in diameter) thus reducing the reagent usage as well as cost.[142] In some instances researchers have shown that they can increase the throughput of conventional sequencing through the use of microchips.[143] Research will still need to be done in order to make this use of technology effective.

Microscopy-based techniques

This approach directly visualizes the sequence of DNA molecules using electron microscopy. The first identification of DNA base pairs within intact DNA molecules by enzymatically incorporating modified bases, which contain atoms of increased atomic number, direct visualization and identification of individually labeled bases within a synthetic 3,272 base-pair DNA molecule and a 7,249 base-pair viral genome has been demonstrated.[144]

RNAP sequencing

This method is based on use of RNA polymerase (RNAP), which is attached to a polystyrene bead. One end of DNA to be sequenced is attached to another bead, with both beads being placed in optical traps. RNAP motion during transcription brings the beads in closer and their relative distance changes, which can then be recorded at a single nucleotide resolution. The sequence is deduced based on the four readouts with lowered concentrations of each of the four nucleotide types, similarly to the Sanger method.[145] A comparison is made between regions and sequence information is deduced by comparing the known sequence regions to the unknown sequence regions.[146]

In vitro virus high-throughput sequencing

A method has been developed to analyze full sets of protein interactions using a combination of 454 pyrosequencing and an in vitro virus mRNA display method. Specifically, this method covalently links proteins of interest to the mRNAs encoding them, then detects the mRNA pieces using reverse transcription PCRs. The mRNA may then be amplified and sequenced. The combined method was titled IVV-HiTSeq and can be performed under cell-free conditions, though its results may not be representative of in vivo conditions.[147]

Market share

While there are many different ways to sequence DNA, only a few dominate the market. In 2022, Illumina had about 80% of the market; the rest of the market is taken by only a few players (PacBio, Oxford, 454, MGI)[148]

Sample preparation

The success of any DNA sequencing protocol relies upon the DNA or RNA sample extraction and preparation from the biological material of interest.

  • A successful DNA extraction will yield a DNA sample with long, non-degraded strands.
  • A successful RNA extraction will yield a RNA sample that should be converted to complementary DNA (cDNA) using reverse transcriptase—a DNA polymerase that synthesizes a complementary DNA based on existing strands of RNA in a PCR-like manner.[149] Complementary DNA can then be processed the same way as genomic DNA.

After DNA or RNA extraction, samples may require further preparation depending on the sequencing method. For Sanger sequencing, either cloning procedures or PCR are required prior to sequencing. In the case of next-generation sequencing methods, library preparation is required before processing.[150] Assessing the quality and quantity of nucleic acids both after extraction and after library preparation identifies degraded, fragmented, and low-purity samples and yields high-quality sequencing data.[151]

The high-throughput nature of current DNA/RNA sequencing technologies has posed a challenge for sample preparation method to scale-up. Several liquid handling instruments are being used for the preparation of higher numbers of samples with a lower total hands-on time:

company Liquid handlers / Automation lower_mark_USD upper_mark_USD landing_url
Opentrons OpenTrons OT-2 $6,500 $20,000 https://www.opentrons.com/
Gilson Gilson Pipetmax $20,000 $40,000 https://gb.gilson.com/GBSV/system-pipetmax.html
Neotec Neotec EzMate $25,000 $45,000 http://neotec.co.il/pipetting-device/
Formulatrix Formulatrix Mantis $40,000 $60,000 https://formulatrix.com/liquid-handling-systems/mantis-liquid-handler/
Hudson Robotics Hudson Robotics SOLO $40,000 $50,000 https://hudsonrobotics.com/products/applications/automated-solutions-next-generation-sequencing-ngs/
Hamilton Hamilton Microlab NIMBUS $40,000 $80,000 https://www.hamiltoncompany.com/automated-liquid-handling/platforms/microlab-nimbus#specifications
TTP Labtech TTP Labtech Mosquito HV Genomics $45,000 $80,000 https://www.sptlabtech.com/products/liquid-handling/mosquito-hv-genomics/
Beckman Coulter Biomek 4000 $50,000 $65,000 https://www.mybeckman.uk/liquid-handlers/biomek-4000/b22640
Hamilton Hamilton Genomic STARlet $50,000 $100,000 https://www.hamiltoncompany.com/automated-liquid-handling/assay-ready-workstations/genomic-starlet
Eppendorf Eppendorf epMotion 5075t $95,000 $110,000 https://www.eppendorf.com/epmotion/
Beckman Coulter Beckman Coulter Biomek i5 $100,000 $150,000 https://www.beckman.com/liquid-handlers/biomek-i5
Hamilton Hamilton NGS STAR $100,000 $200,000 http://www.hamiltonrobotics.com/
PerkinElmer PerkinElmer Sciclone G3 NGS and NGSx Workstation $150,000 $220,000 https://www.perkinelmer.com/uk/product/sciclone-g3-ngs-workstation-cls145321
Agilent Agilent Bravo NGS $170,000 $290,000 https://www.agilent.com/en/products/automated-liquid-handling/automated-liquid-handling-applications/bravo-ngs
Beckman Coulter Beckman Coulter Biomek i7 $200,000 $250,000 https://www.beckman.com/liquid-handlers/biomek-i7
Labcyte Echo 525 Beckman Coulter Labcyte Echo 525 $260,000 $300,000 https://www.labcyte.com/products/liquid-handling/echo-525-liquid-handler
Tecan Tecan NGS $270,000 $350,000 https://lifesciences.tecan.com/ngs-sample-preparation

Development initiatives

Total cost of sequencing a human genome over time as calculated by the NHGRI.

In October 2006, the

full genome sequencing technologies, called the Archon X Prize, intending to award $10 million to "the first Team that can build a device and use it to sequence 100 human genomes within 10 days or less, with an accuracy of no more than one error in every 100,000 bases sequenced, with sequences accurately covering at least 98% of the genome, and at a recurring cost of no more than $10,000 (US) per genome."[152]

Each year the National Human Genome Research Institute, or NHGRI, promotes grants for new research and developments in genomics. 2010 grants and 2011 candidates include continuing work in microfluidic, polony and base-heavy sequencing methodologies.[153]

Computational challenges

The sequencing technologies described here produce raw data that needs to be assembled into longer sequences such as complete genomes (

repetitive sequences that often prevent complete genome assemblies because they occur in many places of the genome. As a consequence, many sequences may not be assigned to particular chromosomes. The production of raw sequence data is only the beginning of its detailed bioinformatical analysis.[154] Yet new methods for sequencing and correcting sequencing errors were developed.[155]

Read trimming

Sometimes, the raw reads produced by the sequencer are correct and precise only in a fraction of their length. Using the entire read may introduce artifacts in the downstream analyses like genome assembly, SNP calling, or gene expression estimation. Two classes of trimming programs have been introduced, based on the window-based or the running-sum classes of algorithms.[156] This is a partial list of the trimming algorithms currently available, specifying the algorithm class they belong to:

Read Trimming Algorithms
Name of algorithm Type of algorithm Link
Cutadapt[157] Running sum Cutadapt
ConDeTri[158] Window based ConDeTri
ERNE-FILTER[159] Running sum ERNE-FILTER
FASTX quality trimmer Window based FASTX quality trimmer
PRINSEQ[160] Window based PRINSEQ
Trimmomatic[161] Window based Trimmomatic
SolexaQA[162] Window based SolexaQA
SolexaQA-BWA Running sum SolexaQA-BWA
Sickle Window based Sickle

Ethical issues

Human genetics have been included within the field of

cell line). However, individuals have a right to informed consent regarding removal and use of cells. Regarding the data produced through DNA sequencing, Moore gives the individual no rights to the information derived from their DNA.[164]

As DNA sequencing becomes more widespread, the storage, security and sharing of genomic data has also become more important.[164][165] For instance, one concern is that insurers may use an individual's genomic data to modify their quote, depending on the perceived future health of the individual based on their DNA.[165][166] In May 2008, the Genetic Information Nondiscrimination Act (GINA) was signed in the United States, prohibiting discrimination on the basis of genetic information with respect to health insurance and employment.[167][168] In 2012, the US Presidential Commission for the Study of Bioethical Issues reported that existing privacy legislation for DNA sequencing data such as GINA and the Health Insurance Portability and Accountability Act were insufficient, noting that whole-genome sequencing data was particularly sensitive, as it could be used to identify not only the individual from which the data was created, but also their relatives.[169][170]

In most of the United States, DNA that is "abandoned", such as that found on a licked stamp or envelope, coffee cup, cigarette, chewing gum, household trash, or hair that has fallen on a public sidewalk, may legally be collected and sequenced by anyone, including the police, private investigators, political opponents, or people involved in paternity disputes. As of 2013, eleven states have laws that can be interpreted to prohibit "DNA theft".[171]

Ethical issues have also been raised by the increasing use of genetic variation screening, both in newborns, and in adults by companies such as 23andMe.[172][173] It has been asserted that screening for genetic variations can be harmful, increasing anxiety in individuals who have been found to have an increased risk of disease.[174] For example, in one case noted in Time, doctors screening an ill baby for genetic variants chose not to inform the parents of an unrelated variant linked to dementia due to the harm it would cause to the parents.[175] However, a 2011 study in The New England Journal of Medicine has shown that individuals undergoing disease risk profiling did not show increased levels of anxiety.[174] Also, the development of Next Generation sequencing technologies such as Nanopore based sequencing has also raised further ethical concerns.[176]

See also

Notes

References

  1. ^ "Introducing 'dark DNA' – the phenomenon that could change how we think about evolution". 24 August 2017.
  2. PMID 23986538
    .
  3. .
  4. ^ .
  5. .
  6. .
  7. ^ .
  8. ^ Hunt, Katie (17 February 2021). "World's oldest DNA sequenced from a mammoth that lived more than a million years ago". CNN. Retrieved 17 February 2021.
  9. PMID 33597786
    .
  10. ^ .
  11. ^ .
  12. .
  13. .
  14. .
  15. .
  16. ^ Mykrobe predictor –Antibiotic resistance prediction for S. aureus and M. tuberculosis from whole genome sequence data
  17. PMID 26686880
    .
  18. ^ "Michael Mosley vs the superbugs". Archived from the original on 24 November 2020. Retrieved 21 October 2019.
  19. ^ Mykrobe, Mykrobe-tools, 24 December 2022, retrieved 2 January 2023
  20. ^ Curtis C, Hereward J (29 August 2017). "From the crime scene to the courtroom: the journey of a DNA sample". The Conversation.
  21. PMID 11493010
    .
  22. .
  23. .
  24. .
  25. .
  26. .
  27. ^ Marks, L. "The path to DNA sequencing: The life and work of Frederick Sanger". What is Biotechnology?. Retrieved 27 June 2023.
  28. S2CID 4153893
    .
  29. .
  30. .
  31. ^ "Ray Wu Faculty Profile". Cornell University. Archived from the original on 4 March 2009.
  32. PMID 4526223
    .
  33. .
  34. .
  35. .
  36. .
  37. ^ .
  38. ^ .
  39. ^ Gilbert, W. DNA sequencing and gene structure. Nobel lecture, 8 December 1980.
  40. PMID 4587255
    .
  41. .
  42. ^ Marks, L. "The next frontier: Human viruses". What is Biotechnology?. Retrieved 27 June 2023.
  43. PMID 6396083
    .
  44. ^ United States Patent 4,631,122 (1986)
  45. PMID 7813418
    .
  46. .
  47. .
  48. .
  49. .
  50. .
  51. .
  52. .
  53. ^ "Espacenet – Bibliographic data". worldwide.espacenet.com.
  54. PMID 8923969
    .
  55. ^ a b Kawashima, Eric H.; Laurent Farinelli; Pascal Mayer (12 May 2005). "Patent: Method of nucleic acid amplification". Archived from the original on 22 February 2013. Retrieved 22 December 2012.
  56. PMID 9521922
    .
  57. ^ "Quality Scores for Next-Generation Sequencing" (PDF). Illumina. 31 October 2011. Retrieved 8 May 2018.
  58. ^
    S2CID 13884154
    .
  59. ^ "maxam gilbert sequencing". PubMed.
  60. PMID 1100841
    .
  61. ^ Wetterstrand, Kris. "DNA Sequencing Costs: Data from the NHGRI Genome Sequencing Program (GSP)". National Human Genome Research Institute. Retrieved 30 May 2013.
  62. PMID 8382019
    .
  63. .
  64. .
  65. .
  66. .
  67. .
  68. ^ .
  69. .
  70. ^ "Applied Biosystems – File Not Found (404 Error)". 16 May 2008. Archived from the original on 16 May 2008.
  71. S2CID 8295541
    .
  72. .
  73. .
  74. .
  75. .(subscription required)
  76. ^ .
  77. ]
  78. ^ .
  79. ^ a b c "New Software, Polymerase for Sequel System Boost Throughput and Affordability – PacBio". 7 March 2018.
  80. ^ "After a Year of Testing, Two Early PacBio Customers Expect More Routine Use of RS Sequencer in 2012". GenomeWeb. 10 January 2012.(registration required)
  81. ^ "Pacific Biosciences Introduces New Chemistry With Longer Read Lengths to Detect Novel Features in DNA Sequence and Advance Genome Studies of Large Organisms" (Press release). 2013.
  82. S2CID 205421576
    .
  83. ^ a b "De novo bacterial genome assembly: a solved problem?". 5 July 2013.
  84. PMID 21793740.Open access icon
  85. .(subscription required)
  86. .
  87. ^ "Ion 520 & Ion 530 ExT Kit-Chef – Thermo Fisher Scientific". thermofisher.com.
  88. ^ "Raw accuracy". Archived from the original on 30 March 2018. Retrieved 29 March 2018.
  89. PMID 19735299.Open access icon
  90. ^ "BGI and MGISEQ". en.mgitech.cn. Retrieved 5 July 2018.
  91. ^
    PMID 23281822
    .
  92. .
  93. ^ "PacBio Sales Start to Pick Up as Company Delivers on Product Enhancements". 12 February 2013.
  94. ^ "Bio-IT World". bio-itworld.com. Archived from the original on 29 July 2020. Retrieved 16 November 2015.
  95. ^ "PacBio Launches Higher-Throughput, Lower-Cost Single-Molecule Sequencing System". October 2015.
  96. PMID 19350039
    .
  97. ^ .
  98. ^ .
  99. .
  100. ^ .
  101. .
  102. ^ Canard B, Sarfati S (13 October 1994), Novel derivatives usable for the sequencing of nucleic acids, retrieved 9 March 2016
  103. PMID 7523248
    .
  104. .
  105. ^ .
  106. ^ brandonvd. "About Us – Complete Genomics". Complete Genomics. Retrieved 2 July 2018.
  107. ^
    PMID 28379488
    .
  108. .
  109. .
  110. ^ .
  111. .
  112. ^ "HeliScope Gene Sequencing / Genetic Analyzer System : Helicos BioSciences". 2 November 2009. Archived from the original on 2 November 2009.
  113. PMID 20890904
    .
  114. ^ "tSMS SeqLL Technical Explanation". SeqLL. Archived from the original on 8 August 2014. Retrieved 9 August 2015.
  115. PMID 26554401
    .
  116. .
  117. ^ .
  118. ^ .
  119. .
  120. ^ "The Harvard Nanopore Group". Mcb.harvard.edu. Archived from the original on 21 February 2002. Retrieved 15 November 2009.
  121. ^ "Nanopore Sequencing Could Slash DNA Analysis Costs".
  122. ^ US patent 20060029957, ZS Genetics, "Systems and methods of analyzing nucleic acid polymers and related components", issued 2005-07-14 
  123. PMID 19904762
    .
  124. .
  125. .
  126. .
  127. .
  128. .
  129. ^ .
  130. .
  131. .
  132. .
  133. .
  134. .
  135. .
  136. .
  137. .
  138. .
  139. .
  140. .
  141. .
  142. ^ "2022 Sequencing Market Share – Same as It Ever Was (For Now)". 25 June 2023.
  143. PMID 18222633
    .
  144. .
  145. ^ "Scalable Nucleic Acid Quality Assessments for Illumina Next-Generation Sequencing Library Prep" (PDF). Retrieved 27 December 2017.
  146. ^ "Archon Genomics XPRIZE". Archon Genomics XPRIZE. Archived from the original on 17 June 2013. Retrieved 9 August 2007.
  147. ^ "Grant Information". National Human Genome Research Institute (NHGRI).
  148. S2CID 26575621
    .
  149. S2CID 2737235. Archived from the original
    (PDF) on 31 May 2020. Retrieved 10 January 2014.
  150. .
  151. .
  152. .
  153. .
  154. .
  155. .
  156. .
  157. .
  158. ^ .
  159. ^ a b Henderson, Mark (9 September 2013). "Human genome sequencing: the real ethical dilemmas". The Guardian. Retrieved 20 May 2015.
  160. ^ Harmon, Amy (24 February 2008). "Insurance Fears Lead Many to Shun DNA Tests". The New York Times. Retrieved 20 May 2015.
  161. ^ Statement of Administration policy, Executive Office of the President, Office of Management and Budget, 27 April 2007
  162. ^ National Human Genome Research Institute (21 May 2008). "President Bush Signs the Genetic Information Nondiscrimination Act of 2008". Retrieved 17 February 2014.
  163. ^ Baker, Monya (11 October 2012). "US ethics panel reports on DNA sequencing and privacy". Nature News Blog.
  164. ^ "Privacy and Progress in Whole Genome Sequencing" (PDF). Presidential Commission for the Study of Bioethical Issues. Archived from the original (PDF) on 12 June 2015. Retrieved 20 May 2015.
  165. ^ Hartnett, Kevin (12 May 2013). "The DNA in your garbage: up for grabs". The Boston Globe. Retrieved 2 January 2023.
  166. PMID 22298675
    .
  167. ^ Hughes, Virginia (7 January 2013). "It's Time To Stop Obsessing About the Dangers of Genetic Information". Slate Magazine. Retrieved 22 May 2015.
  168. ^
    PMID 21226570
    .
  169. ^ Rochman, Bonnie (25 October 2012). "What Your Doctor Isn't Telling You About Your DNA". Time.com. Retrieved 22 May 2015.
  170. S2CID 257803254
    .

External links