请输入您要查询的百科知识:

 

词条 Single cell sequencing
释义

  1. Background

  2. Single cell genome (DNA) sequencing

      Method    Limitations    Applications  

  3. Single cell DNA methylome sequencing

  4. Single-cell RNA sequencing (scRNA-seq)

      Experimental procedures    Applications  

  5. Considerations

      Isolation of single cells    Number of cells to be analyzed    scRNA-Seq  

  6. See also

  7. References

Single cell sequencing examines the sequence information from individual cells with optimized next generation sequencing (NGS) technologies, providing a higher resolution of cellular differences and a better understanding of the function of an individual cell in the context of its microenvironment.[1] Sequencing the DNA of individual cells can give information about mutations carried by small populations of cells, for example in cancer, while sequencing the RNAs expressed by individual cells can give insight into the existence and behavior of different cell types, for example in development.[2]

Background

A typical human cell consists of about 2 x 3.3 billion base pairs of DNA and 600 million bases of mRNA. Usually a mix of millions of cells are used in sequencing the DNA or RNA using traditional methods like Sanger sequencing or Illumina sequencing. By using deep sequencing of DNA and RNA from a single cell, cellular functions can be investigated extensively.[1] Like typical NGS experiments, the protocols of single cell sequencing generally contain the following steps: isolation of a single cell, nucleic acid extraction and amplification, sequencing library preparation, sequencing and bioinformatic data analysis. It is more challenging to perform single cell sequencing in comparison with sequencing from cells in bulk. The minimal amount of starting materials from a single cell make degradation, sample loss and contamination exert pronounced effects on quality of sequencing data. In addition, due to the picogram level of the amount of nucleic acids used,[3] heavy amplification is often needed during sample preparation of single cell sequencing, resulting in the uneven coverage, noise and inaccurate quantification of sequencing data.

Recent technical improvements make single cell sequencing a promising tool for approaching a set of seemingly inaccessible problems. For example, heterogeneous samples, rare cell types, cell lineage relationships, mosaicism of somatic tissues, analyses of microbes that cannot be cultured, and disease evolution can all be elucidated through single cell sequencing.[4] Single cell sequencing was selected as the method of the year 2013 by Nature Publishing Group.[5]

Single cell genome (DNA) sequencing

Single cell DNA genome sequencing involves isolating a single cell, performing whole-genome-amplification (WGA), constructing sequencing libraries and then sequencing the DNA using a next-generation sequencer (ex. Illumina, Ion Torrent). A genome constructed in this fashion is commonly referred to as a single amplified genome or SAG. It can be used in microbiome studies, in order to obtain genomic data from uncultured microorganisms. In addition, it can be united with high throughput cell sorting of microorganisms and cancer. One popular method used for single cell genome sequencing is multiple displacement amplification and this enables research into various areas such as microbial genetics, ecology and infectious diseases. Furthermore, data obtained from microorganisms might establish processes for culturing in the future.[6] Some of the genome assembly tools that can be used in single cell genome sequencing include: SPAdes, IDBA-UD, Cortex and HyDA.[7]

Method

Multiple displacement amplification (MDA) is a widely used technique, enabling amplifying femtograms of DNA from bacterium to micrograms for the use of sequencing. Reagents required for MDA reactions include: random primers and DNA polymerase from bacteriophage phi29. In 30 degree isothermal reaction, DNA is amplified with included reagents. As the polymerases manufacture new strands, a strand displacement reaction takes place, synthesizing multiple copies from each template DNA. At the same time, the strands that were extended antecedently will be displaced. MDA products result in a length of about 12 kb and ranges up to around 100 kb, enabling its use in DNA sequencing.[6] In 2017, a major improvement to this technique, called WGA-X, was introduced by taking advantage of a thermostable mutant of the phi29 polymerase, leading to better genome recovery from individual cells, in particular those with high G+C content.[8] Other methods include MALBAC.[9]

Limitations

MDA of individual cell genomes results in highly uneven genome coverage, i.e. relative overrepresentation and underrepresentation of various regions of the template, leading to loss of some sequences. There are two components to this process: a) stochastic over- and under-amplification of random regions; and b) systematic bias against high %GC regions. The stochastic component may be addressed by pooling single-cell MDA reactions from the same cell type, by employing fluorescent in situ hybridization (FISH) and/or post-sequencing confirmation.[6] The bias of MDA against high %GC regions can be addressed by using thermostable polymerases, such as in the process called WGA-X.[8]

Single-nucleotide polymorphisms (SNPs), which are a big part of genetic variation in the human genome, and copy number variation (CNV), pose problems in single cell sequencing, as well as the limited amount of DNA extracted from a single cell. Due to scant amounts of DNA, accurate analysis of DNA poses problems even after amplification since coverage is low and susceptible to errors. With MDA, average genome coverage is less than 80% and SNPs that are not covered by sequencing reads will be opted out. In addition, MDA shows a high ratio of allele dropout, not detecting alleles from heterozygous samples. Various SNP algorithms are currently in use but none are specific to single cell sequencing. MDA with CNV also poses the problem of identifying false CNVs that conceal the real CNVs. To solve this, when patterns can be generated from false CNVs, algorithms can detect and eradicate this noise to produce true variants.[9]

Applications

Microbiomes are among the main targets of single cell genomics due to the difficulty of culturing the majority of microorganisms in most environments. Single cell genomics is a powerful way to obtain microbial genome sequences without cultivation. This approach has been widely applied on marine, soil, subsurface, organismal, and other types of microbiomes in order to address a wide array of questions related to microbial ecology, evolution, public health and biotechnology potential.[10][11][12][13][14][15][16][17][18][19][8][20]

Cancer sequencing is also an emerging application of scDNAseq. Fresh or frozen tumors may be analyzed and categorized with respect to SCNAs, SNVs, and rearrangements quite well using whole genome DNAS approaches.[21] Cancer scDNAseq is particularly useful for examining the depth of complexity and compound mutations present in amplified therapeutic targets such as receptor tyrosine kinase genes (EGFR, PDGFRA etc.) where conventional population-level approaches of the bulk tumor are not able to resolve the co-occurrence patterns of these mutations within single cells of the tumor. Such overlap may provide redundancy of pathway activation and tumor cell resistance.

Single cell DNA methylome sequencing

Single cell DNA methylome sequencing quantifies DNA methylation. This is similar to single cell genome sequencing, but with the addition of a bisulfite treatment before sequencing. Forms include whole genome bisulfite sequencing,[22][23] and reduced representation bisulfite sequencing [24][25]

Single-cell RNA sequencing (scRNA-seq)

Standard methods such as microarrays and standard bulk RNA-seq analysis analyze the expression of RNAs from large populations of cells. In mixed cell populations, these measurements may obscure critical differences between individual cells within these populations.[33][26]

Single-cell RNA sequencing (scRNA-seq) provides the expression profiles of individual cells. Although it is not possible to obtain complete information on every RNA expressed by each cell, due to the small amount of material available, patterns of gene expression can be identified through gene clustering analyses. This can uncover the existence of rare cell types within a cell population that may never have been seen before. For example, rare specialized cells in the lung called pulmonary ionocytes that express the Cystic Fibrosis Transmembrane Conductance Regulator were identified in 2018 by two groups performing scRNA-Seq on lung airway epithelia.[27][28]

Experimental procedures

Current scRNA-seq protocols involve the following steps: isolation of single cell and RNA, reverse transcription (RT), amplification, library generation and sequencing. Early methods separated individual cells into separate wells; more recent methods encapsulate individual cells in droplets in a microfluidic device, where the reverse transcription reaction takes place, converting RNAs to cDNAs. Each droplet carries a DNA "barcode" that uniquely labels the cDNAs derived from a single cell. Once reverse transcription is complete, the cDNAs from many cells can be mixed together for sequencing; transcripts from a particular cell are identified by the unique barcode.[29][30]

Challenges for scRNA-Seq include preserving the initial relative abundance of mRNA in a cell and identifying rare transcripts.[31] The reverse transcription step is critical as the efficiency of the RT reaction determines how much of the cell’s RNA population will be eventually analyzed by the sequencer. The processivity of reverse transcriptases and the priming strategies used may affect full-length cDNA production and the generation of libraries biased toward 3’ or 5' end of genes.

In the amplification step, either PCR or in vitro transcription (IVT) is currently used to amplify cDNA. One of the advantages of PCR-based methods is the ability to generate full-length cDNA. However, different PCR efficiency on particular sequences (for instance, GC content and snapback structure) may also be exponentially amplified, producing libraries with uneven coverage. On the other hand, while libraries generated by IVT can avoid PCR-induced sequence bias, specific sequences may be transcribed inefficiently, thus causing sequence drop-out or generating incomplete sequences.[1][33]

Several scRNA-seq protocols have been published:

Tang et al.,[32]

STRT,[33]

SMART-seq,[34]

CEL-seq,[35]

RAGE-seq,[36]

, Quartz-seq.[37]

and C1-CAGE.[38] These protocols differ in terms of strategies for reverse transcription, cDNA synthesis and amplification, and the possibility to accommodate sequence-specific barcodes (i.e. UMIs) or the ability to process pooled samples.[39]

In 2017, two approaches were introduced to simultaneously measure single-cell mRNA and protein expression through oligonucleotide-labeled antibodies known as REAP-seq,[40] and CITE-seq.[41]

Applications

scRNA-Seq is becoming widely used across biological disciplines including Development, Neurology,[42] Oncology,[43][44][45] Autoimmune disease,[46] and Infectious disease.[47]

scRNA-Seq has provided considerable insight into the development of embryos and organisms, including the worm Caenorhabditis elegans,[48] and the regenerative planarian Schmidtea mediterranea.[49][50] The first vertebrate animals to be mapped in this way were Zebrafish[51][52] and Xenopus laevis.[53] In each case multiple stages of the embryo were studied, allowing the entire process of development to be mapped on a cell-by-cell basis.[2] Science recognized these advances as the 2018 Breakthrough of the Year.[54]

Considerations

Isolation of single cells

There are several ways to isolate individual cells prior to whole genome amplification and sequencing. Fluorescence-activated cell sorting (FACS) is a widely used approach. Individual cells can also be collected by micromanipulation, for example by serial dilution or by using a patch pipette or nanotube to harvest a single cell.[55][56] The advantages of micromanipulation are ease and low cost, but they are laborious and susceptible to misidentification of cell types under microscope. Laser-capture microdissection (LCM) can also be used for collecting single cells. Although LCM preserves the knowledge of the spatial location of a sampled cell within a tissue, it is hard to capture a whole single cell without also collecting the materials from neighboring cells.[57][58][59]

High-throughput methods for single cell isolation also include microfluidics. Both FACS and microfluidics are accurate, automatic and capable of isolating unbiased samples. However, both methods require detaching cells from their microenvironments first, thereby causing perturbation to the transcriptional profiles in RNA expression analysis.[60][61]

Number of cells to be analyzed

scRNA-Seq

Generally speaking, for a typical bulk cell RNA-sequencing (RNA-seq) experiment, ten million reads are generated and a gene with higher than the threshold of 50 reads per kb per million reads (RPKM) is considered expressed. For a gene that is 1kb long, this corresponds to 500 reads and a minimum coefficient of variation (CV) of 4% under the assumption of the Poisson distribution. For a typical mammalian cell containing 200,000 mRNA, sequencing data from at least 50 single cells need to be pooled in order to achieve this minimum CV value. However, due to the efficiency of reverse transcription and other noise introduced in the experiments, more cells are required for accurate expression analyses and cell type identification.[57]

See also

  • Single-cell analysis
  • Single-cell transcriptomics
  • Single cell epigenomics
  • DNA sequencing

References

1. ^{{cite journal | vauthors = Eberwine J, Sul JY, Bartfai T, Kim J | title = The promise of single-cell sequencing | journal = Nature Methods | volume = 11 | issue = 1 | pages = 25–7 | date = January 2014 | pmid = 24524134 | doi = 10.1038/nmeth.2769 }}
2. ^{{cite journal | vauthors = Pennisi E | title = Chronicling embryos, cell by cell, gene by gene | journal = Science | volume = 360 | issue = 6387 | pages = 367 | date = April 2018 | pmid = 29700246 | doi = 10.1126/science.360.6387.367 }}
3. ^{{cite journal | vauthors = Shintaku H, Nishikii H, Marshall LA, Kotera H, Santiago JG | title = On-chip separation and analysis of RNA and DNA from single cells | journal = Analytical Chemistry | volume = 86 | issue = 4 | pages = 1953–7 | date = February 2014 | pmid = 24499009 | doi = 10.1021/ac4040218 }}
4. ^{{cite journal | vauthors = Nawy T | title = Single-cell sequencing | journal = Nature Methods | volume = 11 | issue = 1 | pages = 18 | date = January 2014 | pmid = 24524131 | doi = 10.1038/nmeth.2771 }}
5. ^{{cite journal|title=Method of the Year 2013|journal=Nat Methods|year=2014|volume=11|issue=1|pages=1|doi=10.1038/nmeth.2801|pmid=24524124}}
6. ^"{{cite journal | vauthors = Lasken RS | title = Single-cell genomic sequencing using Multiple Displacement Amplification | journal = Current Opinion in Microbiology | volume = 10 | issue = 5 | pages = 510–6 | date = October 2007 | pmid = 17923430 | doi = 10.1016/j.mib.2007.08.005 }}"
7. ^{{cite journal | vauthors = Taghavi Z, Movahedi NS, Draghici S, Chitsaz H | title = Distilled single-cell genome sequencing and de novo assembly for sparse microbial communities | journal = Bioinformatics | volume = 29 | issue = 19 | pages = 2395–401 | date = October 2013 | pmid = 23918251 | pmc = 3777112 | doi = 10.1093/bioinformatics/btt420 | arxiv = 1305.0062 }}
8. ^{{cite journal | vauthors = Stepanauskas R, Fergusson EA, Brown J, Poulton NJ, Tupper B, Labonté JM, Becraft ED, Brown JM, Pachiadaki MG, Povilaitis T, Thompson BP, Mascena CJ, Bellows WK, Lubys A | title = Improved genome recovery and integrated cell-size analyses of individual uncultured microbial cells and viral particles | journal = Nature Communications | volume = 8 | issue = 1 | pages = 84 | date = July 2017 | pmid = 28729688 | pmc = 5519541 | doi = 10.1038/s41467-017-00128-z | bibcode = 2017NatCo...8...84S }}
9. ^"{{cite journal | vauthors = Ning L, Liu G, Li G, Hou Y, Tong Y, He J | title = Current challenges in the bioinformatics of single cell genomics | journal = Frontiers in Oncology | volume = 4 | issue = 7 | pages = 7 | year = 2014 | pmid = 24478987 | pmc = 3902584 | doi = 10.3389/fonc.2014.00007 }}"
10. ^{{cite journal | vauthors = Blainey PC, Quake SR | title = Dissecting genomic diversity, one cell at a time | journal = Nature Methods | volume = 11 | issue = 1 | pages = 19–21 | date = January 2014 | pmid = 24524132 | pmc = 3947563 | doi = 10.1038/nmeth.2783 | hdl = 1721.1/106574 }}
11. ^{{cite journal | vauthors = Zhang K, Martiny AC, Reppas NB, Barry KW, Malek J, Chisholm SW, Church GM | title = Sequencing genomes from single cells by polymerase cloning | journal = Nature Biotechnology | volume = 24 | issue = 6 | pages = 680–6 | date = June 2006 | pmid = 16732271 | doi = 10.1038/nbt1214 }}
12. ^{{cite journal | vauthors = Stepanauskas R, Sieracki ME | title = Matching phylogeny and metabolism in the uncultured marine bacteria, one cell at a time | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 104 | issue = 21 | pages = 9052–7 | date = May 2007 | pmid = 17502618 | pmc = 1885626 | doi = 10.1073/pnas.0700496104 | bibcode = 2007PNAS..104.9052S }}
13. ^{{cite journal | vauthors = Yoon HS, Price DC, Stepanauskas R, Rajah VD, Sieracki ME, Wilson WH, Yang EC, Duffy S, Bhattacharya D | title = Single-cell genomics reveals organismal interactions in uncultivated marine protists | journal = Science | volume = 332 | issue = 6030 | pages = 714–7 | date = May 2011 | pmid = 21551060 | doi = 10.1126/science.1203163 | bibcode = 2011Sci...332..714Y }}
14. ^{{cite journal | vauthors = Swan BK, Martinez-Garcia M, Preston CM, Sczyrba A, Woyke T, Lamy D, Reinthaler T, Poulton NJ, Masland ED, Gomez ML, Sieracki ME, DeLong EF, Herndl GJ, Stepanauskas R | title = Potential for chemolithoautotrophy among ubiquitous bacteria lineages in the dark ocean | journal = Science | volume = 333 | issue = 6047 | pages = 1296–300 | date = September 2011 | pmid = 21885783 | doi = 10.1126/science.1203690 | bibcode = 2011Sci...333.1296S }}
15. ^{{cite journal | vauthors = Woyke T, Xie G, Copeland A, González JM, Han C, Kiss H, Saw JH, Senin P, Yang C, Chatterji S, Cheng JF, Eisen JA, Sieracki ME, Stepanauskas R | title = Assembling the marine metagenome, one cell at a time | journal = PLOS One | volume = 4 | issue = 4 | pages = e5299 | date = 2009-04-23 | pmid = 19390573 | pmc = 2668756 | doi = 10.1371/journal.pone.0005299 | bibcode = 2009PLoSO...4.5299W }}
16. ^{{cite journal | vauthors = Swan BK, Tupper B, Sczyrba A, Lauro FM, Martinez-Garcia M, González JM, Luo H, Wright JJ, Landry ZC, Hanson NW, Thompson BP, Poulton NJ, Schwientek P, Acinas SG, Giovannoni SJ, Moran MA, Hallam SJ, Cavicchioli R, Woyke T, Stepanauskas R | title = Prevalent genome streamlining and latitudinal divergence of planktonic bacteria in the surface ocean | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 110 | issue = 28 | pages = 11463–8 | date = July 2013 | pmid = 23801761 | pmc = 3710821 | doi = 10.1073/pnas.1304246110 | bibcode = 2013PNAS..11011463S }}
17. ^{{cite journal | vauthors = Rinke C, Schwientek P, Sczyrba A, Ivanova NN, Anderson IJ, Cheng JF, Darling A, Malfatti S, Swan BK, Gies EA, Dodsworth JA, Hedlund BP, Tsiamis G, Sievert SM, Liu WT, Eisen JA, Hallam SJ, Kyrpides NC, Stepanauskas R, Rubin EM, Hugenholtz P, Woyke T | title = Insights into the phylogeny and coding potential of microbial dark matter | journal = Nature | volume = 499 | issue = 7459 | pages = 431–7 | date = July 2013 | pmid = 23851394 | doi = 10.1038/nature12352 | bibcode = 2013Natur.499..431R }}
18. ^{{cite journal | vauthors = Kashtan N, Roggensack SE, Rodrigue S, Thompson JW, Biller SJ, Coe A, Ding H, Marttinen P, Malmstrom RR, Stocker R, Follows MJ, Stepanauskas R, Chisholm SW | title = Single-cell genomics reveals hundreds of coexisting subpopulations in wild Prochlorococcus | journal = Science | volume = 344 | issue = 6182 | pages = 416–20 | date = April 2014 | pmid = 24763590 | doi = 10.1126/science.1248575 | bibcode = 2014Sci...344..416K }}
19. ^{{cite journal | vauthors = Wilson WH, Gilg IC, Moniruzzaman M, Field EK, Koren S, LeCleir GR, Martínez Martínez J, Poulton NJ, Swan BK, Stepanauskas R, Wilhelm SW | title = Genomic exploration of individual giant ocean viruses | journal = The ISME Journal | volume = 11 | issue = 8 | pages = 1736–1745 | date = August 2017 | pmid = 28498373 | pmc = 5520044 | doi = 10.1038/ismej.2017.61 }}
20. ^{{cite journal | vauthors = Pachiadaki MG, Sintes E, Bergauer K, Brown JM, Record NR, Swan BK, Mathyer ME, Hallam SJ, Lopez-Garcia P, Takaki Y, Nunoura T, Woyke T, Herndl GJ, Stepanauskas R | title = Major role of nitrite-oxidizing bacteria in dark ocean carbon fixation | journal = Science | volume = 358 | issue = 6366 | pages = 1046–1051 | date = November 2017 | pmid = 29170234 | doi = 10.1126/science.aan8260 | bibcode = 2017Sci...358.1046P }}
21. ^{{cite journal | vauthors = Francis JM, Zhang CZ, Maire CL, Jung J, Manzo VE, Adalsteinsson VA, Homer H, Haidar S, Blumenstiel B, Pedamallu CS, Ligon AH, Love JC, Meyerson M, Ligon KL | title = EGFR variant heterogeneity in glioblastoma resolved through single-nucleus sequencing | journal = Cancer Discovery | volume = 4 | issue = 8 | pages = 956–71 | date = August 2014 | pmid = 24893890 | pmc = 4125473 | doi = 10.1158/2159-8290.CD-13-0879 }}
22. ^{{cite journal | vauthors = Farlik M, Sheffield NC, Nuzzo A, Datlinger P, Schönegger A, Klughammer J, Bock C | title = Single-cell DNA methylome sequencing and bioinformatic inference of epigenomic cell-state dynamics | journal = Cell Reports | volume = 10 | issue = 8 | pages = 1386–97 | date = March 2015 | pmid = 25732828 | pmc = 4542311 | doi = 10.1016/j.celrep.2015.02.001 }}
23. ^{{cite journal | vauthors = Smallwood SA, Lee HJ, Angermueller C, Krueger F, Saadeh H, Peat J, Andrews SR, Stegle O, Reik W, Kelsey G | title = Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity | journal = Nature Methods | volume = 11 | issue = 8 | pages = 817–820 | date = August 2014 | pmid = 25042786 | pmc = 4117646 | doi = 10.1038/nmeth.3035 }}
24. ^{{cite journal | vauthors = Guo H, Zhu P, Wu X, Li X, Wen L, Tang F | title = Single-cell methylome landscapes of mouse embryonic stem cells and early embryos analyzed using reduced representation bisulfite sequencing | journal = Genome Research | volume = 23 | issue = 12 | pages = 2126–35 | date = December 2013 | pmid = 24179143 | pmc = 3847781 | doi = 10.1101/gr.161679.113 }}
25. ^{{cite journal | vauthors = Guo H, Zhu P, Guo F, Li X, Wu X, Fan X, Wen L, Tang F | title = Profiling DNA methylome landscapes of mammalian cells with single-cell reduced-representation bisulfite sequencing | journal = Nature Protocols | volume = 10 | issue = 5 | pages = 645–59 | date = May 2015 | pmid = 25837417 | doi = 10.1038/nprot.2015.039 }}
26. ^{{cite journal | vauthors = Kolodziejczyk AA, Kim JK, Svensson V, Marioni JC, Teichmann SA | title = The technology and biology of single-cell RNA sequencing | journal = Molecular Cell | volume = 58 | issue = 4 | pages = 610–20 | date = May 2015 | pmid = 26000846 | doi = 10.1016/j.molcel.2015.04.005 }}
27. ^{{cite journal | vauthors = Montoro DT, Haber AL, Biton M, Vinarsky V, Lin B, Birket SE, Yuan F, Chen S, Leung HM, Villoria J, Rogel N, Burgin G, Tsankov AM, Waghray A, Slyper M, Waldman J, Nguyen L, Dionne D, Rozenblatt-Rosen O, Tata PR, Mou H, Shivaraju M, Bihler H, Mense M, Tearney GJ, Rowe SM, Engelhardt JF, Regev A, Rajagopal J | title = A revised airway epithelial hierarchy includes CFTR-expressing ionocytes | journal = Nature | volume = 560 | issue = 7718 | pages = 319–324 | date = August 2018 | pmid = 30069044 | pmc = 6295155 | doi = 10.1038/s41586-018-0393-7 }}
28. ^{{cite journal | vauthors = Plasschaert LW, Žilionis R, Choo-Wing R, Savova V, Knehr J, Roma G, Klein AM, Jaffe AB | title = A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte | journal = Nature | volume = 560 | issue = 7718 | pages = 377–381 | date = August 2018 | pmid = 30069046 | pmc = 6108322 | doi = 10.1038/s41586-018-0394-6 }}
29. ^{{cite journal | vauthors = Klein AM, Mazutis L, Akartuna I, Tallapragada N, Veres A, Li V, Peshkin L, Weitz DA, Kirschner MW | title = Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells | journal = Cell | volume = 161 | issue = 5 | pages = 1187–1201 | date = May 2015 | pmid = 26000487 | pmc = 4441768 | doi = 10.1016/j.cell.2015.04.044 }}
30. ^{{cite journal | vauthors = Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, Tirosh I, Bialas AR, Kamitaki N, Martersteck EM, Trombetta JJ, Weitz DA, Sanes JR, Shalek AK, Regev A, McCarroll SA | title = Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets | journal = Cell | volume = 161 | issue = 5 | pages = 1202–1214 | date = May 2015 | pmid = 26000488 | pmc = 4481139 | doi = 10.1016/j.cell.2015.05.002 }}
31. ^"{{cite journal | vauthors = Hebenstreit D | title = Methods, Challenges and Potentials of Single Cell RNA-seq | journal = Biology | volume = 1 | issue = 3 | pages = 658–67 | date = November 2012 | pmid = 24832513 | pmc = 4009822 | doi = 10.3390/biology1030658 }}"
32. ^{{cite journal | vauthors = Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, Xu N, Wang X, Bodeau J, Tuch BB, Siddiqui A, Lao K, Surani MA | title = mRNA-Seq whole-transcriptome analysis of a single cell | journal = Nature Methods | volume = 6 | issue = 5 | pages = 377–82 | date = May 2009 | pmid = 19349980 | doi = 10.1038/NMETH.1315 }}
33. ^{{cite journal | vauthors = Islam S, Kjällquist U, Moliner A, Zajac P, Fan JB, Lönnerberg P, Linnarsson S | title = Characterization of the single-cell transcriptional landscape by highly multiplex RNA-seq | journal = Genome Research | volume = 21 | issue = 7 | pages = 1160–7 | date = July 2011 | pmid = 21543516 | pmc = 3129258 | doi = 10.1101/gr.110882.110 }}
34. ^{{cite journal | vauthors = Ramsköld D, Luo S, Wang YC, Li R, Deng Q, Faridani OR, Daniels GA, Khrebtukova I, Loring JF, Laurent LC, Schroth GP, Sandberg R | title = Full-length mRNA-Seq from single-cell levels of RNA and individual circulating tumor cells | journal = Nature Biotechnology | volume = 30 | issue = 8 | pages = 777–82 | date = August 2012 | pmid = 22820318 | pmc = 3467340 | doi = 10.1038/nbt.2282 }}
35. ^{{cite journal | vauthors = Hashimshony T, Wagner F, Sher N, Yanai I | title = CEL-Seq: single-cell RNA-Seq by multiplexed linear amplification | journal = Cell Reports | volume = 2 | issue = 3 | pages = 666–73 | date = September 2012 | pmid = 22939981 | doi = 10.1016/j.celrep.2012.08.003 }}
36. ^{{cite journal| vauthors = Singh M, Al-Eryani G, Carswell S, Ferguson JM, Blackburn J, Barton K, Roden D, Luciani F, Phan T, Junankar S, Jackson K, Goodnow CC, Smith MA, Swarbrick A | title=High-throughput targeted long-read single cell sequencing reveals the clonal and transcriptional landscape of lymphocytes |journal=bioRxiv|year=2018 |doi=10.1101/424945 }}
37. ^{{cite journal | vauthors = Sasagawa Y, Nikaido I, Hayashi T, Danno H, Uno KD, Imai T, Ueda HR | title = Quartz-Seq: a highly reproducible and sensitive single-cell RNA sequencing method, reveals non-genetic gene-expression heterogeneity | journal = Genome Biology | volume = 14 | issue = 4 | pages = R31 | date = April 2013 | pmid = 23594475 | pmc = 4054835 | doi = 10.1186/gb-2013-14-4-r31 }}
38. ^{{Cite journal|last=Shin|first=Jay W.|last2=Plessy|first2=Charles|last3=Carninci|first3=Piero|last4=Arner|first4=Erik|last5=Hon|first5=Chung-Chau|last6=Lassmann|first6=Timo|last7=Kasukawa|first7=Takeya|last8=Suzuki|first8=Harukazu|last9=West|first9=Jay|date=2019-01-21|title=C1 CAGE detects transcription start sites and enhancer activity at single-cell resolution|journal=Nature Communications|volume=10|issue=1|pages=360|doi=10.1038/s41467-018-08126-5|pmid=30664627|pmc=6341120|issn=2041-1723}}
39. ^{{cite journal | vauthors = Dal Molin A, Di Camillo B | title = How to design a single-cell RNA-sequencing experiment: pitfalls, challenges and perspectives | journal = Briefings in Bioinformatics | volume = | issue = | pages = bby007 | date = January 2018 | pmid = 29394315 | doi = 10.1093/bib/bby007 }}
40. ^{{Cite journal|last=Klappenbach|first=Joel A.|last2=Sadekova|first2=Svetlana|last3=McClanahan|first3=Terrill K.|last4=Moore|first4=Renee|last5=Douglas C. Wilson|last6=Li|first6=Lixia|last7=Wong|first7=Jerelyn|last8=Kumar|first8=Namit|last9=Zhang|first9=Kelvin Xi|date=October 2017|title=Multiplexed quantification of proteins and transcripts in single cells|journal=Nature Biotechnology|volume=35|issue=10|pages=936–939|doi=10.1038/nbt.3973|pmid=28854175|issn=1546-1696}}
41. ^{{Cite journal|last=Smibert|first=Peter|last2=Satija|first2=Rahul|last3=Swerdlow|first3=Harold|last4=Pratip K. Chattopadhyay|last5=Houck-Loomis|first5=Brian|last6=Stephenson|first6=William|last7=Hafemeister|first7=Christoph|last8=Stoeckius|first8=Marlon|date=September 2017|title=Simultaneous epitope and transcriptome measurement in single cells|journal=Nature Methods|volume=14|issue=9|pages=865–868|doi=10.1038/nmeth.4380|issn=1548-7105|pmc=5669064|pmid=28759029}}
42. ^{{cite journal | vauthors = Raj B, Wagner DE, McKenna A, Pandey S, Klein AM, Shendure J, Gagnon JA, Schier AF | title = Simultaneous single-cell profiling of lineages and cell types in the vertebrate brain | journal = Nature Biotechnology | volume = 36 | issue = 5 | pages = 442–450 | date = June 2018 | pmid = 29608178 | pmc = 5938111 | doi = 10.1038/nbt.4103 }}
43. ^{{cite journal | vauthors = Olmos D, Arkenau HT, Ang JE, Ledaki I, Attard G, Carden CP, Reid AH, A'Hern R, Fong PC, Oomen NB, Molife R, Dearnaley D, Parker C, Terstappen LW, de Bono JS | title = Circulating tumour cell (CTC) counts as intermediate end points in castration-resistant prostate cancer (CRPC): a single-centre experience | journal = Annals of Oncology | volume = 20 | issue = 1 | pages = 27–33 | date = January 2009 | pmid = 18695026 | doi = 10.1093/annonc/mdn544 }}
44. ^{{Cite journal|last=Sims|first=Peter A.|last2=Yuan|first2=Jinzhou|last3=Levitin|first3=Hanna Mendes|date=2018-04-01|title=Single-Cell Transcriptomic Analysis of Tumor Heterogeneity|url=https://www.cell.com/trends/cancer/abstract/S2405-8033(18)30038-4|journal=Trends in Cancer|language=English|volume=4|issue=4|pages=264–268|doi=10.1016/j.trecan.2018.02.003|issn=2405-8033|pmc=5993208|pmid=29606308}}
45. ^{{Cite journal|last=Regev|first=Aviv|last2=Izar|first2=Benjamin|last3=Yoon|first3=Charles H.|last4=Garraway|first4=Levi A.|last5=Rozenblatt-Rosen|first5=Orit|last6=Rotem|first6=Asaf|last7=Johnson|first7=Bruce E.|last8=Schadendorf|first8=Dirk|last9=Allen|first9=Eliezer M. Van|date=2018-11-01|title=A Cancer Cell Program Promotes T Cell Exclusion and Resistance to Checkpoint Blockade|url=https://www.cell.com/cell/abstract/S0092-8674(18)31178-4|journal=Cell|language=English|volume=175|issue=4|pages=984–997.e24|doi=10.1016/j.cell.2018.09.006|issn=0092-8674|pmid=30388455}}
46. ^{{Cite journal|last=Satija|first=Rahul|last2=Swerdlow|first2=Harold P.|last3=Darnell|first3=Robert B.|last4=Orange|first4=Dana E.|last5=Bykerk|first5=Vivian P.|last6=Ivashkiv|first6=Lionel B.|last7=Goodman|first7=Susan M.|last8=Rashidfarrokhi|first8=Ali|last9=Bracken|first9=Bernadette|date=2018-02-23|title=Single-cell RNA-seq of rheumatoid arthritis synovial tissue using low-cost microfluidic instrumentation|journal=Nature Communications|volume=9|issue=1|pages=791|doi=10.1038/s41467-017-02659-x|issn=2041-1723|pmc=5824814|pmid=29476078}}
47. ^{{cite journal|vauthors=Avraham R, Haseley N, Brown D, Penaranda C, Jijon HB, Trombetta JJ, Satija R, Shalek AK, Xavier RJ, Regev A, Hung DT|date=September 2015|title=Pathogen Cell-to-Cell Variability Drives Heterogeneity in Host Immune Responses|journal=Cell|volume=162|issue=6|pages=1309–21|doi=10.1016/j.cell.2015.08.027|pmc=4578813|pmid=26343579}}
48. ^{{cite journal | vauthors = Cao J, Packer JS, Ramani V, Cusanovich DA, Huynh C, Daza R, Qiu X, Lee C, Furlan SN, Steemers FJ, Adey A, Waterston RH, Trapnell C, Shendure J | title = Comprehensive single-cell transcriptional profiling of a multicellular organism | journal = Science | volume = 357 | issue = 6352 | pages = 661–667 | date = August 2017 | pmid = 28818938 | pmc = 5894354 | doi = 10.1126/science.aam8940 }}
49. ^{{cite journal | vauthors = Plass M, Solana J, Wolf FA, Ayoub S, Misios A, Glažar P, Obermayer B, Theis FJ, Kocks C, Rajewsky N | title = Cell type atlas and lineage tree of a whole complex animal by single-cell transcriptomics | journal = Science | volume = 360 | issue = 6391 | pages = eaaq1723 | date = May 2018 | pmid = 29674432 | doi = 10.1126/science.aaq1723 }}
50. ^{{cite journal | vauthors = Fincher CT, Wurtzel O, de Hoog T, Kravarik KM, Reddien PW | title = Schmidtea mediterranea | journal = Science | volume = 360 | issue = 6391 | pages = eaaq1736 | date = May 2018 | pmid = 29674431 | doi = 10.1126/science.aaq1736 }}
51. ^{{cite journal | vauthors = Wagner DE, Weinreb C, Collins ZM, Briggs JA, Megason SG, Klein AM | title = Single-cell mapping of gene expression landscapes and lineage in the zebrafish embryo | journal = Science | volume = 360 | issue = 6392 | pages = 981–987 | date = June 2018 | pmid = 29700229 | pmc = 6083445 | doi = 10.1126/science.aar4362 }}
52. ^{{cite journal | vauthors = Farrell JA, Wang Y, Riesenfeld SJ, Shekhar K, Regev A, Schier AF | title = Single-cell reconstruction of developmental trajectories during zebrafish embryogenesis | journal = Science | volume = 360 | issue = 6392 | pages = eaar3131 | date = June 2018 | pmid = 29700225 | pmc = 6247916 | doi = 10.1126/science.aar3131 }}
53. ^{{cite journal | vauthors = Briggs JA, Weinreb C, Wagner DE, Megason S, Peshkin L, Kirschner MW, Klein AM | title = The dynamics of gene expression in vertebrate embryogenesis at single-cell resolution | journal = Science | volume = 360 | issue = 6392 | pages = eaar5780 | date = June 2018 | pmid = 29700227 | pmc = 6038144 | doi = 10.1126/science.aar5780 }}
54. ^{{cite web |url= https://vis.sciencemag.org/breakthrough2018/finalists/ |title=Science's 2018 Breakthrough of the Year: tracking development cell by cell|last=You|first=Jia | name-list-format = vanc | work = Science Magazine | publisher = American Association for the Advancement of Science }}
55. ^{{cite journal | vauthors = Zong C, Lu S, Chapman AR, Xie XS | title = Genome-wide detection of single-nucleotide and copy-number variations of a single human cell | journal = Science | volume = 338 | issue = 6114 | pages = 1622–6 | date = December 2012 | pmid = 23258894 | pmc = 3600412 | doi = 10.1126/science.1229164 | bibcode = 2012Sci...338.1622Z }}
56. ^{{cite journal | vauthors = Kurimoto K, Yabuta Y, Ohinata Y, Saitou M | title = Global single-cell cDNA amplification to provide a template for representative high-density oligonucleotide microarray analysis | journal = Nature Protocols | volume = 2 | issue = 3 | pages = 739–52 | year = 2007 | pmid = 17406636 | doi = 10.1038/nprot.2007.79 }}
57. ^"{{cite journal | vauthors = Shapiro E, Biezuner T, Linnarsson S | title = Single-cell sequencing-based technologies will revolutionize whole-organism science | journal = Nature Reviews. Genetics | volume = 14 | issue = 9 | pages = 618–30 | date = September 2013 | pmid = 23897237 | doi = 10.1038/nrg3542 }}"
58. ^{{cite journal | vauthors = Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, Kamiyama M, Hruban RH, Eshleman JR, Nowak MA, Velculescu VE, Kinzler KW, Vogelstein B, Iacobuzio-Donahue CA | title = Distant metastasis occurs late during the genetic evolution of pancreatic cancer | journal = Nature | volume = 467 | issue = 7319 | pages = 1114–7 | date = October 2010 | pmid = 20981102 | pmc = 3148940 | doi = 10.1038/nature09515 | bibcode = 2010Natur.467.1114Y }}
59. ^{{cite journal | vauthors = Frumkin D, Wasserstrom A, Itzkovitz S, Harmelin A, Rechavi G, Shapiro E | title = Amplification of multiple genomic loci from single cells isolated by laser micro-dissection of tissues | journal = BMC Biotechnology | volume = 8 | issue = 17 | pages = 17 | date = February 2008 | pmid = 18284708 | pmc = 2266725 | doi = 10.1186/1472-6750-8-17 }}
60. ^{{cite journal | vauthors = Dalerba P, Kalisky T, Sahoo D, Rajendran PS, Rothenberg ME, Leyrat AA, Sim S, Okamoto J, Johnston DM, Qian D, Zabala M, Bueno J, Neff NF, Wang J, Shelton AA, Visser B, Hisamori S, Shimono Y, van de Wetering M, Clevers H, Clarke MF, Quake SR | title = Single-cell dissection of transcriptional heterogeneity in human colon tumors | journal = Nature Biotechnology | volume = 29 | issue = 12 | pages = 1120–7 | date = November 2011 | pmid = 22081019 | pmc = 3237928 | doi = 10.1038/nbt.2038 }}
61. ^{{cite journal | vauthors = White AK, VanInsberghe M, Petriv OI, Hamidi M, Sikorski D, Marra MA, Piret J, Aparicio S, Hansen CL | title = High-throughput microfluidic single-cell RT-qPCR | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 108 | issue = 34 | pages = 13999–4004 | date = August 2011 | pmid = 21808033 | pmc = 3161570 | doi = 10.1073/pnas.1019446108 | bibcode = 2011PNAS..10813999W }}
{{Portal bar|Molecular and cellular biology}}{{Breakthrough of the Year}}

3 : DNA sequencing|Molecular biology techniques|Biotechnology

随便看

 

开放百科全书收录14589846条英语、德语、日语等多语种百科知识,基本涵盖了大多数领域的百科知识,是一部内容自由、开放的电子版国际百科全书。

 

Copyright © 2023 OENC.NET All Rights Reserved
京ICP备2021023879号 更新时间:2024/9/20 18:56:06