Abstract
It could be argued that the greatest transformative aspect of the Human Genome Project has been not the sequencing of the genome itself, but the resultant development of new technologies. A host of new approaches has fundamentally changed the way we approach problems in basic and translational research. Now, a new generation of high-throughput sequencing technologies promises to again transform the scientific enterprise, potentially supplanting array-based technologies and opening up many new possibilities. By allowing DNA/RNA to be assayed more rapidly than previously possible, these next-generation platforms promise a deeper understanding of genome regulation and biology. Significantly enhancing sequencing throughput will allow us to follow the evolution of viral and bacterial resistance in real time, to uncover the huge diversity of novel genes that are currently inaccessible, to understand nucleic acid therapeutics, to better integrate biological information for a complete picture of health and disease at a personalized level and to move to advances that we cannot yet imagine.
This is a preview of subscription content, access via your institution
Relevant articles
Open Access articles citing this article.
-
Using mechanistic models for the clinical interpretation of complex genomic variation
Scientific Reports Open Access 12 December 2019
-
Exploring the druggable space around the Fanconi anemia pathway using machine learning and mechanistic models
BMC Bioinformatics Open Access 02 July 2019
-
Performance assessment of variant calling pipelines using human whole exome sequencing and simulated data
BMC Bioinformatics Open Access 17 June 2019
Access options
Subscribe to this journal
Receive 12 print issues and online access
$209.00 per year
only $17.42 per issue
Rent or buy this article
Prices vary by article type
from$1.95
to$39.95
Prices may be subject to local taxes which are calculated during checkout



References
Bentley, D.R. Whole-genome re-sequencing. Curr. Opin. Genet. Dev. 16, 545–552 (2006).
Church, G.M. Genomes for all. Sci. Am. 294, 46–54 (2006).
Zwolak, M. & DiVentra, M. Physical approaches to DNA sequencing and detection. Rev. Mod. Phys. 80, 141–165 (2008).
Mardis, E.R. The impact of next-generation sequencing technology on genetics. Trends Genet. 24, 133–141 (2008).
Harris, T.D. et al. Single-Molecule DNA Sequencing of a Viral Genome. Science 320, 106–109 (2008).
International Human Genome Sequencing Consortium. Initial sequencing and analysis of the human genome. Nature 409, 860–921 (2001).
Drosophila 12 Genomes Consortium. Evolution of genes and genomes on the Drosophila phylogeny. Nature 450, 203–218 (2007).
Hillier, L.W. et al. Whole-genome sequencing and variant discovery in C. elegans . Nat. Methods 5, 183–188 (2008).
Wheeler, D.A. et al. The complete genome of an individual by massively parallel DNA sequencing. Nature 452, 872–876 (2008).
Millar, C.D., Huynen, L., Subramanian, S., Mohandesan, E. & Lamber, D.M. New developments in ancient genomics. Trends Ecol. Evol. 23, 386–393 (2008).
Ellegren, H. & Sheldon, B.C. Genetic basis of fitness differences in natural populations. Nature 452, 169–175 (2008).
Kruglyak, L. The road to genome-wide association studies. Nat. Rev. Genet. 9, 314–318 (2008).
Sachidanandam, R. et al. A map of human genome sequence variation containing 1.42 million single nucleotide polymorphisms. Nature 409, 928–933 (2001).
International HapMap Consortium. A haplotype map of the human genome. Nature 437, 1299–1320 (2005).
International HapMap Consortium. A second generation human haplotype map of over 3.1 million SNPs. Nature 449, 851–861 (2007).
Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447, 661–678 (2007).
Broadbent, H.M. et al. Susceptibility to coronary artery disease and diabetes is encoded by distinct, tightly linked SNPs in the ANRIL locus on chromosome 9p. Hum. Mol. Genet. 17, 806–814 (2008).
Ke, X., Taylor, M.S. & Cardon, L.R. Singleton SNPs in the human genome and implications for genome-wide association studies. Eur. J. Hum. Genet. 16, 506–515 (2008).
Cohen, J.C. et al. Multiple rare alleles contribute to low plasma levels of HDL cholesterol. Science 305, 869–872 (2004).
McClellan, J.M., Susser, E. & King, M.-C. Schizophrenia: a common disease caused by multiple rare alleles. Br. J. Psychol. 190, 194–199 (2007).
Speicher, M.R. & Carter, N.P. The new cytogenetics: blurring the boundaries with molecular biology. Nat. Rev. Genet. 6, 782–792 (2005).
Iafrate, A.J. et al. Detection of large-scale variation in the human genome. Nat. Genet. 36, 949–951 (2004).
Freeman, J.L. et al. Copy number variation: new insights in genome diversity. Genome Res. 16, 949–961 (2006).
Komura, D. et al. Genome-wide detection of human copy number variations using high-density DNA oligonucleotide arrays. Genome Res. 16, 1575–1584 (2006).
Redon, R. et al. Global variation in copy number in the human genome. Nature 444, 444–454 (2006).
Levy, S. et al. The diploid genome sequence of an individual human. PLoS Biol. 5, e254 (2007).
Feuk, L., Marshall, C.R., Wintle, R.F., & Scherer, S.W. Structural variants: changing the landscape of chromosomes and design of disease studies. Hum. Mol. Genet. 15 Spec No 1, R57–R66 (2006).
McCarroll, S.A. & Althshuler, D.M. Copy-number variation and association studies of human disease. Nat. Genet. 39, S37–S42 (2008).
Sjoblom, T. et al. The consensus coding sequences of human breast and colorectal cancers. Science 314, 268–274 (2006).
Wood, L.D. et al. The genomic landscapes of human breast and colorectal cancers. Science 318, 1108–1113 (2007).
Chittenden, T. et al. Functional Classification Analysis of Somatically Mutated Genes in Human Breast and Colorectal Cancers. Genomics 91, 508–511 (2008).
Tomlins, S.A. et al. Distinct classes of chromosomal rearrangements create oncogenic ETS gene fusions in prostate cancer. Nature 448, 595–599 (2007).
Nardi, V. et al. Quantitative monitoring by polymerase colony assay of known mutations resistant to ABL kinase inhibitors. Oncogene 27, 775–782 (2008).
Hoffmann, C. et al. DNA bar coding and pyrosequencing to identify rare HIV drug resistance mutations. Nucleic Acids Res. 35, e91 (2007).
Wang, C., Mitsuya, Y., Gharizadeh, B., Ronaghi, M. & Shafer, R.W. Characterization of mutation spectra with ultra-deep pyrosequencing: application to HIV-1 drug resistance. Genome Res. 17, 1195–1201 (2007).
Loman, N.J. & Pallen, M.J. XDR-TB genome sequencing: a glimpse of the microbiology of the future. Future Microbiol. 3, 111–113 (2008).
Holt, K.E. et al. High-throughput sequencing provides insights into genome variation and evolution in Salmonella Typhi. Nat. Genet. 40, 987–993 (2008).
Feng, H., Shuda, M., Chang, Y. & Moore, P.S. Clonal integration of a polyomavirus in human Merkel cell carcinoma. Science 319, 1096–1100 (2007).
Palacios, G. et al. A new arenavirus in a cluster of fatal transplant-associated diseases. N. Engl. J. Med. 358, 991–998 (2008).
Cox-Foster, D.L. et al. A metagenomic survey of microbes in honey bee colony collapse disorder. Science 318, 283–287 (2007).
Woyke, T. et al. Symbiosis insights through metagenomic analysis of a microbial consortium. Nature 443, 950–955 (2006).
Marcy, Y. et al. Dissecting biological “dark matter” with single-cell genetic analysis of rare and uncultivated TM7 microbes from the human mouth. Proc. Natl. Acad. Sci. USA 104, 11889–11894 (2007).
Pernthaler, A. et al. Diverse syntrophic partnerships from deep-sea methane vents revealed by direct cell capture and metagenomics. Proc. Natl. Acad. Sci. USA 105, 7052–7057 (2008).
Dinsdale, E.A. et al. Functional metagenomic profiling of nine biomes. Nature 452, 629–632 (2008).
Bundy, J.G. et al. 'Systems toxicology' approach identifies coordinated metabolic responses to copper in a terrestrial non-model invertebrate, the earthworm Lumbricus rubellus . BMC Biol. 6, 25 (2008).
van Straalen, N.M. & Roelofs, D. Genomics technology for assessing soil pollution. J. Biol. 7, 19 (2008).
Turnbaugh, P.J. et al. The human microbiome project. Nature 449, 804–810 (2007).
Rauch, T.A. et al. High-resolution mapping of DNA hypermethylation and hypomethylation in lung cancer. Proc. Natl. Acad. Sci. USA 105, 252–257 (2007).
Yasui, D.H. et al. Integrated epigenomic analyses of neuronal MeCP2 reveal a role for long-range interaction with active genes. Proc. Natl. Acad. Sci. USA 104, 19416–19421 (2007).
Beck, S. & Rakyan, V.K. The methylome: approaches for global DNA methylation profiling. Trends Genet. 24, 231–237 (2008).
Gitan, R.S., Shi, H., Chen, C.M., Yan, P.S. & Huang, T.H. Methylation-specific oligonucleotide microarray: a new potential for high-throughput methylation analysis. Genome Res. 12, 158–164 (2002).
Hu, M. et al. Distinct epigenetic changes in the stromal cells of breast cancers. Nat. Genet. 37, 899–905 (2005).
Agrelo, R. et al. Epigenetic inactivation of the premature aging Werner syndrome gene in human cancer. Proc. Natl. Acad. Sci. USA 103, 8822–8827 (2006).
Brena, R.M., Huang, T.H. & Plass, C. Quantitative assessment of DNA methylation: potential applications for disease diagnosis, classification, and prognosis in clinical settings. J. Mol. Med. 84, 365–377 (2006).
Fraga, M.F. et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc. Natl. Acad. Sci. USA 102, 10604–10609 (2005).
Jones, P.A. & Martienssen, R. A blueprint for a Human Epigenome Project: the AACR Human Epigenome Workshop. Cancer Res. 65, 11241–11246 (2005).
Garber, K. Momentum building for human epigenome project. J. Natl. Cancer Inst. 98, 84–86 (2006).
Esteller, M. The necessity of a human epigenome project. Carcinogenesis 27, 1121–1125 (2006).
Eckhardt, F. et al. DNA methylation profiling of human chromosomes 6, 20 and 22. Nat. Genet. 38, 1378–1385 (2006).
Zhang, X. et al. Genome-wide high-resolution mapping and functional analysis of DNA methylation in Arabidopsis . Cell 126, 1189–1201 (2006).
Mavrich, T.N. et al. Nucleosome organization in the Drosophila genome. Nature 453, 358–362 (2008).
Valouev, A. et al. A high-resolution, nucleosome position map of C. elegans reveals a lack of universal sequence-dictated positioning. Genome Res. 18, 1051–1063 (2008).
Kim, T.H. & Ren, B. Genome-wide analysis of protein-DNA interactions. Annu. Rev. Genomics Hum. Genet. 7, 81–102 (2006).
Massie, C.E. & Mills, I.G. ChIPping away at gene regulation. EMBO Rep. 9, 337–343 (2008).
Mendenhall, E.M. & Bernstein, B.E. Chromatin state maps: new technologies, new insights. Curr. Opin. Genet. Dev. 18, 109–115 (2008).
Robertson, G. et al. Genome-wide profiles of STAT1 DNA association using chromatin immunoprecipitation and massively parallel sequencing. Nat. Methods 4, 651–657 (2007).
Euskirchen, G.M. et al. Mapping of transcription factor binding regions in mammalian cells by ChIP: comparison of array- and sequencing-based technologies. Genome Res. 17, 898–909 (2007).
Johnson, D.S., Mortazavi, A., Myers, R.M. & Wold, B. Genome-wide mapping of in vivo protein-DNA interactions. Science 316, 1497–1502 (2007).
Barski, A. et al. High-resolution profiling of histone methylations in the human genome. Cell 129, 823–837 (2007).
Roh, T.Y., Wei, G., Farrell, C.M. & Zhao, K. Genome-wide prediction of conserved and nonconserved enhancers by histone acetylation patterns. Genome Res. 17, 74–81 (2007).
Minsky, N. et al. Monoubiquitinated H2B is associated with the transcribed region of highly expressed genes in human cells. Nat. Cell Biol. 10, 483–488 (2008).
Mikkelsen, T.S. et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature 448, 553–560 (2007).
Meissner, A. et al. Genome-scale DNA methylation maps of pluirpotent and differentiated cells. Nature 454, 766–770 (2008).
Boyer, L.A. et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell 122, 947–956 (2005).
Lee, T.I. et al. Control of developmental regulators by Polycomb in human embryonic stem cells. Cell 125, 301–313 (2006).
Gilchrist, M. et al. Systems biology approaches identify ATF3 as a negative regulator of Toll-like receptor 4. Nature 441, 173–178 (2006).
Petersen, D. et al. Three microarray platforms: an analysis of their concordance in profiling gene expression. BMC Genomics 6, 63 (2005).
Encode Project Consortium. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature 447, 799–816 (2007).
Kapranov, P. et al. Large-scale transcriptional activity in chromosomes 21 and 22. Science 296, 916–919 (2002).
Cheng, J. et al. Transcriptional maps of 10 human chromosomes at 5-nucleotide resolution. Science 308, 1149–1154 (2005).
Kapranov, P. et al. Examples of the complex architecture of the human transcriptome revealed by RACE and high-density tiling arrays. Genome Res. 15, 987–997 (2005).
Palatnik, J.F. et al. Control of leaf morphogenesis by microRNAs. Nature 425, 257–263 (2003).
Lu, J. et al. MicroRNA expression profiles classify human cancers. Nature 435, 834–838 (2005).
Abbott, A.L. et al. The let-7 MicroRNA family members mir-48, mir-84, and mir-241 function together to regulate developmental timing in Caenorhabditis elegans . Dev. Cell 9, 403–414 (2005).
Calin, G.A. & Croce, C.M. MicroRNA signatures in human cancers. Nat. Rev. Cancer 6, 857–866 (2006).
Calin, G.A. & Croce, C.M. MicroRNA-cancer connection: the beginning of a new tale. Cancer Res. 66, 7390–7394 (2006).
Huang, Q. et al. The microRNAs miR-373 and miR-520c promote tumour invasion and metastasis. Nat. Cell Biol. 10, 202–210 (2008).
Morin, R.D. et al. Application of massively parallel sequencing to microRNA profiling and discovery in human embryonic stem cells. Genome Res. 18, 610–621 (2008).
Yu, W. et al. Epigenetic silencing of tumour suppressor gene p15 by its antisense RNA. Nature 451, 202–206 (2008).
Lein, E.S. et al. Genome-wide atlas of gene expression in the adult mouse brain. Nature 445, 168–176 (2007).
Liang, W.S. et al. Gene expression profiles in anatomically and functionally distinct regions of the normal aged human brain. Physiol. Genomics 28, 311–322 (2006).
Myers, A.J. et al. A survey of genetic human cortical gene expression. Nat. Genet. 39, 1494–1499 (2007).
Fehlbaum, P., Guihal, C., Bracco, L. & Cochet, O. A microarray configuration to quantify expression levels and relative abundance of splice variants. Nucleic Acids Res. 33, e47 (2005).
Kwan, T. et al. Genome-wide analysis of transcript isoform variation in humans. Nat. Genet. 40, 225–231 (2008).
Blencowe, B.J. Alternative splicing: new insights from global analyses. Cell 126, 37–47 (2006).
Velculescu, V.E., Zhang, L., Vogelstein, B. & Kinzler, K.W. Serial analysis of gene expression. Science 270, 484–487 (1995).
Sun, M. et al. SAGE is far more sensitive than EST for detecting low-abundance transcripts. BMC Genomics 5, 1–4 (2004).
Hirst, M. et al. LongSAGE profiling of nine human embryonic stem cell lines. Genome Biol. 8, R113 (2007).
Mortazavi, A., Williams, B.A., McCue, K., Schaeffer, L. & Wold, B. Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat. Methods 5, 621–628 (2008).
Cloonan, N. et al. Stem cell transcriptome profiling via massive-scale mRNA sequencing. Nat. Methods 5, 613–619 (2008).
Shin, H. et al. Transcriptome analysis for Caenorhabditis elegans based on novel expressed sequence tags. BMC Biol. 6, 30 (2008).
Morin, R. et al. Profiling the HeLa S3 transcriptome using randomly primed cDNA and massively parallel short-read sequencing. Biotechniques 45, 81–94 (2008).
Nagalakshmi, U. et al. The transcriptional landscape of the yeast genome defined by RNA sequencing. Science 320, 1344–1349 (2008).
Wilhelm, B.T. et al. Dynamic repertoire of a eukaryotic transcriptome surveyed at single-nucleotide resolution. Nature 453, 1239–1243 (2008).
Rockman, M.V. & Kruglyak, L. Genetics of global gene expression. Nat. Rev. Genet. 7, 862–872 (2006).
Schadt, E.E. et al. Genetics of gene expression surveyed in maize, mouse and man. Nature 422, 297–302 (2003).
Chen, Y. et al. Variations in DNA elucidate molecular networks that cause disease. Nature 452, 429–435 (2008).
De Gobbi, M. et al. A regulatory SNP causes a human genetic disease by creating a new transcriptional promoter. Science 312, 1215–1217 (2006).
Lister, R. et al. Highly integrated single-base resolution maps of the epigenome in Arabidopsis . Cell 133, 523–536 (2008).
Chou, C.C., Chen, C.H., Lee, T.T. & Peck, K. Optimization of probe length and the number of probes per gene for optimal microarray analysis of gene expression. Nucleic Acids Res. 32, e99 (2004).
Shendure, J. The beginning of the end for microarrays? Nat. Genet. 5, 585–586 (2008).
Pop, M. & Salzburg, S.L. Bioinformatics challenges of new sequencing technology. Trends Genet. 24, 142–149 (2008).
Acknowledgements
We thank Jennifer MacArthur (Helicos BioSciences) for assistance and critical reading of the manuscript and Stephen Quake (Stanford University) for critical reading and comments.
Author information
Authors and Affiliations
Corresponding author
Ethics declarations
Competing interests
A.K. and J.F.T. are employees and J.Q. is on the scientific advisory board of Helicos BioSciences Corp.
Rights and permissions
About this article
Cite this article
Kahvejian, A., Quackenbush, J. & Thompson, J. What would you do if you could sequence everything?. Nat Biotechnol 26, 1125–1133 (2008). https://doi.org/10.1038/nbt1494
Issue Date:
DOI: https://doi.org/10.1038/nbt1494
This article is cited by
-
Species concepts of Dothideomycetes: classification, phylogenetic inconsistencies and taxonomic standardization
Fungal Diversity (2021)
-
Exploring the druggable space around the Fanconi anemia pathway using machine learning and mechanistic models
BMC Bioinformatics (2019)
-
Performance assessment of variant calling pipelines using human whole exome sequencing and simulated data
BMC Bioinformatics (2019)
-
Using mechanistic models for the clinical interpretation of complex genomic variation
Scientific Reports (2019)
-
SMARTcleaner: identify and clean off-target signals in SMART ChIP-seq analysis
BMC Bioinformatics (2018)