Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Exome sequencing as a tool for Mendelian disease gene discovery

Key Points

  • The development of methods that couple targeted capture and massively parallel DNA sequencing —termed exomesequencing — has made it possible to determine cost-effectively nearly all of the coding variation in an individual human genome.

  • Exome sequencing is a powerful and cost-effective new tool for dissecting the genetic basis of Mendelian diseases or traits that have proven intractable to conventional gene-discovery strategies.

  • Most Mendelian disorders that have been solved to date by exome sequencing have relied on comparison of variants found in a small number of unrelated or closely related affected individuals to identify shared novel or rare alleles of the same gene. An alternative to this discrete-filtering approach is to apply tests of association.

  • Exome sequencing of parent–child trios is a highly effective approach for identifying de novo coding mutations, as multiple de novo events occurring within a specific gene (or within a gene family or pathway) is an extremely unlikely event.

  • Solving the remaining several thousand Mendelian disorders by exome or whole-genome sequencing is possible and should be an imperative for the human and medical genetics community.

  • The widespread, useful, convenient and cost-effective use of exome sequencing and eventually whole-genome sequencing for clinical diagnosis and screening will necessitate overcoming a number of major challenges that currently limit its broad applicability.

Abstract

Exome sequencing — the targeted sequencing of the subset of the human genome that is protein coding — is a powerful and cost-effective new tool for dissecting the genetic basis of diseases and traits that have proved to be intractable to conventional gene-discovery strategies. Over the past 2 years, experimental and analytical approaches relating to exome sequencing have established a rich framework for discovering the genes underlying unsolved Mendelian disorders. Additionally, exome sequencing is being adapted to explore the extent to which rare alleles explain the heritability of complex diseases and health-related traits. These advances also set the stage for applying exome and whole-genome sequencing to facilitate clinical diagnosis and personalized disease-risk profiling.

This is a preview of subscription content, access via your institution

Access options

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

Figure 1: Exome coverage estimated as the percentage of bases called per gene.
Figure 2: Strategies for finding disease-causing rare variants using exome sequencing.
Figure 3: Estimated probability of identifying a single causal gene for a monogenic disorder under a discrete filtering framework.

Similar content being viewed by others

References

  1. McKusick, V. A. Mendelian Inheritance in Man and its online version, OMIM. Am. J. Hum. Genet. 80, 588–604 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Kaiser, J. Human genetics. Affordable 'exomes' fill gaps in a catalogue of rare diseases. Science 330, 903 (2010).

    CAS  PubMed  Google Scholar 

  3. Antonarakis, S. E. & Beckmann, J. S. Mendelian disorders deserve more attention. Nature Rev. Genet. 7, 277–282 (2006).

    CAS  PubMed  Google Scholar 

  4. Schork, N. J., Murray, S. S., Frazer, K. A. & Topol, E. J. Common vs. rare allele hypotheses for complex diseases. Curr. Opin. Genet. Dev. 19, 212–219 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Manolio, T. A. et al. Finding the missing heritability of complex diseases. Nature 461, 747–753 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. McClellan, J. & King, M. C. Genetic heterogeneity in human disease. Cell 141, 210–217 (2010).

    Article  CAS  PubMed  Google Scholar 

  7. Metzker, M. L. Sequencing technologies — the next generation. Nature Rev. Genet. 11, 31–46 (2010).

    CAS  PubMed  Google Scholar 

  8. Mamanova, L. et al. Target-enrichment strategies for next-generation sequencing. Nature Methods 7, 111–118 (2010).

    CAS  PubMed  Google Scholar 

  9. Biesecker, L. G. Exome sequencing makes medical genomics a reality. Nature Genet. 42, 13–14 (2010).

    CAS  PubMed  Google Scholar 

  10. Ng, S. B. et al. Targeted capture and massively parallel sequencing of 12 human exomes. Nature 461, 272–276 (2009). This was the first study to show the feasibility of using exome sequencing to identify disease-causing variants.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Ng, S. B. et al. Exome sequencing identifies the cause of a Mendelian disorder. Nature Genet. 42, 30–35 (2010). This was the first study to use exome sequencing to discover the genetic basis of a monogenic disorder.

    CAS  PubMed  Google Scholar 

  12. Bilguvar, K. et al. Whole-exome sequencing identifies recessive WDR62 mutations in severe brain malformations. Nature 467, 207–210 (2010). This is an outstanding paper demonstrating the narrowing to a single candidate gene that is made possible by exome sequencing a single case in the context of a consanguineous pedigree and a recessive phenotype.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Stenson, P. D. et al. The Human Gene Mutation Database: providing a comprehensive central mutation database for molecular diagnostics and personalized genomics. Hum. Genomics 4, 69–72 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Kryukov, G. V., Pennacchio, L. A. & Sunyaev, S. R. Most rare missense alleles are deleterious in humans: implications for complex disease and association studies. Am. J. Hum. Genet. 80, 727–739 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Simpson, M. A. et al. Mutations in NOTCH2 cause Hajdu–Cheney syndrome, a disorder of severe and progressive bone loss. Nature Genet. 43, 303–305 (2011).

    CAS  PubMed  Google Scholar 

  16. Krawitz, P. M. et al. Identity-by-descent filtering of exome sequence data identifies PIGV mutations in hyperphosphatasia mental retardation syndrome. Nature Genet. 42, 827–829 (2010).

    CAS  PubMed  Google Scholar 

  17. Tsurusaki, Y. et al. Rapid detection of a mutation causing X-linked leucoencephalopathy by exome sequencing. J. Med. Genet. 48, 606–609 (2011).

    CAS  PubMed  Google Scholar 

  18. Liu, Y. et al. Confirmation by exome sequencing of the pathogenic role of NCSTN mutations in acne inversa (hidradenitis suppurativa). J. Invest. Dermatol. 131, 1570–1572 (2011).

    CAS  PubMed  Google Scholar 

  19. Yamaguchi, T. et al. Exome resequencing combined with linkage analysis identifies novel PTH1R variants in primary failure of tooth eruption in Japanese. J. Bone Miner. Res. 26, 1655–1661 (2011).

    CAS  PubMed  Google Scholar 

  20. Zuchner, S. et al. Whole-exome sequencing links a variant in DHDDS to retinitis pigmentosa. Am. J. Hum. Genet. 88, 201–206 (2011).

    PubMed  PubMed Central  Google Scholar 

  21. Otto, E. A. et al. Candidate exome capture identifies mutation of SDCCAG8 as the cause of a retinal–renal ciliopathy. Nature Genet. 42, 840–850 (2010).

    CAS  PubMed  Google Scholar 

  22. Haack, T. B. et al. Exome sequencing identifies ACAD9 mutations as a cause of complex I deficiency. Nature Genet. 42, 1131–1134 (2010).

    CAS  PubMed  Google Scholar 

  23. Ng, S. B. et al. Exome sequencing identifies MLL2 mutations as a cause of Kabuki syndrome. Nature Genet. 42, 790–793 (2010).

    CAS  PubMed  Google Scholar 

  24. Al Badr, W. et al. Exome capture and massively parallel sequencing identifies a novel HPSE2 mutation in a Saudi Arabian child with Ochoa (urofacial) syndrome. J. Pediatr. Urol. 28 Mar 2011 (doi:10.1016/j.jpurol.2011.02.034).

    PubMed  PubMed Central  Google Scholar 

  25. Bolze, A. et al. Whole-exome-sequencing-based discovery of human FADD deficiency. Am. J. Hum. Genet. 87, 873–881 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Caliskan, M. et al. Exome sequencing reveals a novel mutation for autosomal recessive non-syndromic mental retardation in the TECR gene on chromosome 19p13. Hum. Mol. Genet. 20, 1285–1289 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Glazov, E. A. et al. Whole-exome re-sequencing in a family quartet identifies POP1 mutations as the cause of a novel skeletal dysplasia. PLoS Genet. 7, e1002027 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Walsh, T. et al. Whole exome sequencing and homozygosity mapping identify mutation in the cell polarity protein GPSM2 as the cause of nonsyndromic hearing loss DFNB82. Am. J. Hum. Genet. 87, 90–94 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Johnston, J. J. et al. Massively parallel sequencing of exons on the X chromosome identifies RBM10 as the gene that causes a syndromic form of cleft palate. Am. J. Hum. Genet. 86, 743–748 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Norton, N. et al. Genome-wide studies of copy number variation and exome sequencing identify rare variants in BAG3 as a cause of dilated cardiomyopathy. Am. J. Hum. Genet. 88, 273–282 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Musunuru, K. et al. Exome sequencing, ANGPTL3 mutations, and familial combined hypolipidemia. N. Engl. J. Med. 363, 2220–2227 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Johnson, J. O. et al. Exome sequencing reveals VCP mutations as a cause of familial ALS. Neuron 68, 857–864 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Wang, J. L. et al. TGM6 identified as a novel causative gene of spinocerebellar ataxias using exome sequencing. Brain 133, 3510–3518 (2010).

    PubMed  Google Scholar 

  34. Gilissen, C. et al. Exome sequencing identifies WDR35 variants involved in Sensenbrenner syndrome. Am. J. Hum. Genet. 87, 418–423 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Lalonde, E. et al. Unexpected allelic heterogeneity and spectrum of mutations in Fowler syndrome revealed by next-generation exome sequencing. Hum. Mutat. 31, 918–923 (2010).

    CAS  PubMed  Google Scholar 

  36. Sirmaci, A. et al. MASP1 mutations in patients with facial, umbilical, coccygeal, and auditory findings of Carnevale, Malpuech, OSA, and Michels syndromes. Am. J. Hum. Genet. 87, 679–686 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Hoischen, A. et al. De novo mutations of SETBP1 cause Schinzel–Giedion syndrome. Nature Genet. 42, 483–485 (2010).

    CAS  PubMed  Google Scholar 

  38. Kalay, E. et al. CEP152 is a genome maintenance protein disrupted in Seckel syndrome. Nature Genet. 43, 23–26 (2011).

    CAS  PubMed  Google Scholar 

  39. Hubisz, M. J., Pollard, K. S. & Siepel, A. PHAST and RPHAST: phylogenetic analysis with space/time models. Brief. Bioinf. 12, 41–51 (2011).

    CAS  Google Scholar 

  40. Cooper, G. M. et al. Single-nucleotide evolutionary constraint scores highlight disease-causing mutations. Nature Methods 7, 250–251 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Kumar, P. H. & S. Ng, P. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nature Protoc. 4, 1073–1081 (2009).

    CAS  Google Scholar 

  42. Adzhubei, I. A. et al. A method and server for predicting damaging missense mutations. Nature Methods 7, 248–249 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Stone, E. A. & Sidow, A. Physicochemical constraint violation by missense substitutions mediates impairment of protein function and disease severity. Genome Res. 15, 978–986 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Nachman, M. W. & Crowell, S. L. Estimate of the mutation rate per nucleotide in humans. Genetics 156, 297–304 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Vissers, L. E. et al. A de novo paradigm for mental retardation. Nature Genet. 42, 1109–1112 (2010). This was the first study to use exome sequencing of parent–child trios of affected offspring and their unaffected parents to identify de novo variants and thus candidate genes for a complex trait characterized by substantial locus heterogeneity.

    CAS  PubMed  Google Scholar 

  46. Girard, S. L. et al. Increased exonic de novo mutation rate in individuals with schizophrenia. Nature Genet. 43, 860–863 (2011).

    CAS  PubMed  Google Scholar 

  47. O'Roak, B. J. et al. Exome sequencing in sporadic autism spectrum disorders identifies severe de novo mutations. Nature Genet. 43, 585–589 (2011).

    CAS  PubMed  Google Scholar 

  48. Blakemore, A. I. & Froguel, P. Investigation of Mendelian forms of obesity holds out the prospect of personalized medicine. Ann. N.Y. Acad. Sci. 1214, 180–189 (2010).

    CAS  PubMed  Google Scholar 

  49. Dietz, H. C. New therapeutic approaches to Mendelian disorders. N. Engl. J. Med. 363, 852–863 (2010).

    CAS  PubMed  Google Scholar 

  50. St. Hilaire, C. et al. NT5E mutations and arterial calcifications. N. Engl. J. Med. 364, 432–42 (2011).

    Google Scholar 

  51. Choi, M. et al. Genetic diagnosis by whole exome capture and massively parallel DNA sequencing. Proc. Natl Acad. Sci. USA 106, 19096–19101 (2009). This paper provides the first example of applying exome sequencing to make an unanticipated diagnosis in a clinical setting.

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Worthey, E. A. et al. Making a definitive diagnosis: successful clinical application of whole exome sequencing in a child with intractable inflammatory bowel disease. Genet. Med. 13, 255–262 (2011). This is an outstanding example of the clinical diagnosis of a rare disorder by exome sequencing leading to a subsequent, life-saving change in treatment.

    PubMed  Google Scholar 

  53. Bonnefond, A. et al. Molecular diagnosis of neonatal diabetes mellitus using next-generation sequencing of the whole exome. PLoS ONE 5, e13630 (2010).

    PubMed  PubMed Central  Google Scholar 

  54. Montenegro, G. et al. Exome sequencing allows for rapid gene identification in a Charcot–Marie–Tooth family. Ann. Neurol. 69, 464–470 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Chiu, R. W. et al. Noninvasive prenatal diagnosis of fetal chromosomal aneuploidy by massively parallel genomic sequencing of DNA in maternal plasma. Proc. Natl Acad. Sci. USA 105, 20458–20463 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Chiu, R. W. & Lo, Y. M. Non-invasive prenatal diagnosis by fetal nucleic acid analysis in maternal plasma: the coming of age. Semin. Fetal Neonatal Med. 16, 88–93 (2011).

    PubMed  Google Scholar 

  57. Bell, C. J. et al. Carrier testing for severe childhood recessive diseases by next-generation sequencing. Sci. Transl. Med. 3, 65ra4 (2011). This work reports on efforts to implement pre-conception carrier screening for over 400 recessive disorders by hybrid capture and next-generation sequencing.

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Ashley, E. A. et al. Clinical assessment incorporating a personal genome. Lancet 375, 1525–1535 (2010). This paper illustrates both the promise and challenges we face in the clinical interpretation of exome or genome sequences of individual patients.

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Kingsmore, S. F. & Saunders, C. J. Deep sequencing of patient genomes for disease diagnosis: when will it become routine? Sci. Transl. Med. 3, 87ps23 (2011).

    PubMed  PubMed Central  Google Scholar 

  60. Scriver, C. R. The PAH gene, phenylketonuria, and a paradigm shift. Hum. Mutat. 28, 831–845 (2007).

    CAS  PubMed  Google Scholar 

  61. Ormond, K. E. et al. Challenges in the clinical application of whole-genome sequencing. Lancet 375, 1749–1751 (2010).

    PubMed  Google Scholar 

  62. Tong, M. Y., Cassa, C. A. & Kohane, I. S. Automated validation of genetic variants from large databases: ensuring that variant references refer to the same genomic locations. Bioinformatics 27, 891–893 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Kohonen-Corish, M. R. et al. How to catch all those mutations—the report of the third Human Variome Project Meeting, UNESCO Paris, May 2010. Hum. Mutat. 31, 1374–1381 (2010).

    PubMed  PubMed Central  Google Scholar 

  64. Roach, J. C. et al. Analysis of genetic inheritance in a family quartet by whole-genome sequencing. Science 328, 636–639 (2010). This was the first study to report the sequencing of the entire genome for each member of a family with a Mendelian disorder.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Beskow, L. M. & Burke, W. Offering individual genetic research results: context matters. Sci. Transl. Med. 2, 38cm20 (2010).

    PubMed  PubMed Central  Google Scholar 

  66. Richards, C. S. et al. ACMG recommendations for standards for interpretation and reporting of sequence variations: revisions 2007. Genet. Med. 10, 294–300 (2008).

    CAS  PubMed  Google Scholar 

  67. Fabsitz, R. R. et al. Ethical and practical guidelines for reporting genetic research results to study participants: updated guidelines from a National Heart, Lung, and Blood Institute working group. Circ. Cardiovasc. Genet. 3, 574–580 (2011).

    Google Scholar 

  68. Caulfield, T. et al. Research ethics recommendations for whole-genome research: consensus statement. PLoS Biol. 6, e73 (2008).

    PubMed  PubMed Central  Google Scholar 

  69. Wolf, S. M. et al. Managing incidental findings in human subjects research: analysis and recommendations. J. Law Med. Ethics 36, 219–248 (2008).

    PubMed  PubMed Central  Google Scholar 

  70. Ravitsky, V. & Wilfond, B. S. Disclosing individual genetic results to research participants. Am. J. Bioeth. 6, 8–17 (2006).

    PubMed  Google Scholar 

  71. Green, E. D. & Guyer, M. S. Charting a course for genomic medicine from base pairs to bedside. Nature 470, 204–213 (2011).

    CAS  PubMed  Google Scholar 

  72. Dahl, F. et al. Multigene amplification and massively parallel sequencing for cancer mutation discovery. Proc. Natl Acad. Sci. USA 104, 9387–9392 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Fredriksson, S. et al. Multiplex amplification of all coding sequences within 10 cancer genes by Gene-Collector. Nucleic Acids Res. 35, e47 (2007).

    PubMed  PubMed Central  Google Scholar 

  74. Gnirke, A. et al. Solution hybrid selection with ultra-long oligonucleotides for massively parallel targeted sequencing. Nature Biotech. 27, 182–189 (2009).

    CAS  Google Scholar 

  75. Okou, D. T. et al. Microarray-based genomic selection for high-throughput resequencing. Nature Methods 4, 907–909 (2007).

    CAS  PubMed  Google Scholar 

  76. Porreca, G. J. et al. Multiplex amplification of large sets of human exons. Nature Methods 4, 931–936 (2007).

    CAS  PubMed  Google Scholar 

  77. Albert, T. J. et al. Direct selection of human genomic loci by microarray hybridization. Nature Methods 4, 903–905 (2007).

    CAS  PubMed  Google Scholar 

  78. Turner, E. H., Lee, C., Ng, S. B., Nickerson, D. A. & Shendure, J. Massively parallel exon capture and library-free resequencing across 16 genomes. Nature Methods 6, 315–316 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Turner, E. H., Ng, S. B., Nickerson, D. A. & Shendure, J. Methods for genomic partitioning. Annu. Rev. Genomics Hum. Genet. 10, 263–284 (2009).

    CAS  PubMed  Google Scholar 

  80. Nielsen, R., Paul, J. S., Albrechtsen, A. & Song, Y. S. Genotype and SNP calling from next-generation sequencing data. Nature Rev. Genet. 12, 443–451 (2011).

    CAS  PubMed  Google Scholar 

  81. Alkan, C., Coe, B. P. & Eichler, E. E. Genome structural variation discovery and genotyping. Nature Rev. Genet. 12, 363–376 (2011).

    CAS  PubMed  Google Scholar 

  82. Adey, A. et al. Rapid, low-input, low-bias construction of shotgun fragment libraries by high-density in vitro transposition. Genome Biol. 11, R119 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Cirulli, E. T. & Goldstein, D. B. Uncovering the roles of rare variants in common disease through whole-genome sequencing. Nature Rev. Genet. 11, 415–425 (2010).

    CAS  PubMed  Google Scholar 

  84. Lanktree, M. B., Hegele, R. A., Schork, N. J. & Spence, J. D. Extremes of unexplained variation as a phenotype: an efficient approach for genome-wide association studies of cardiovascular disease. Circ. Cardiovasc. Genet. 3, 215–221 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Cohen, J. C. et al. Multiple rare alleles contribute to low plasma levels of HDL cholesterol. Science 305, 869–872 (2004). This was an important study that demonstrated the effectiveness of sequencing candidate genes at the extremes of a phenotype to find rare alleles influencing risk for a complex trait.

    CAS  PubMed  Google Scholar 

  86. Li, B. & Leal, S. M. Methods for detecting associations with rare variants for common diseases: application to analysis of sequence data. Am. J. Hum. Genet. 83, 311–321 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Morris, A. P. & Zeggini, E. An evaluation of statistical approaches to rare variant analysis in genetic association studies. Genet. Epidemiol. 34, 188–193 (2010).

    PubMed  Google Scholar 

  88. Price, A. L. et al. Pooled association tests for rare variants in exon-resequencing studies. Am. J. Hum. Genet. 86, 832–838 (2010).

    PubMed  PubMed Central  Google Scholar 

  89. Madsen, B. E. & Browning, S. R. A groupwise association test for rare mutations using a weighted sum statistic. PLoS Genet. 5, e1000384 (2009).

    PubMed  PubMed Central  Google Scholar 

  90. Bansal, V., Libiger, O., Torkamani, A. & Schork, N. J. Statistical analysis strategies for association studies involving rare variants. Nature Rev. Genet. 11, 773–785 (2010).

    CAS  PubMed  Google Scholar 

  91. DePristo, M. A. et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nature Genet. 43, 491–498 (2011).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the US National Institutes of Health (NIH)/National Heart, Lung, and Blood Institute (NHLBI) Exome Sequencing Project (Lung Grand Opportunity (GO) Sequencing Project (HL-102923 to M.J.B.), the US Women's Health Initiative (WHI) GO Sequencing Project (HL-102924), the Heart GO Sequencing Project (HL-103010), the Broad GO Sequencing Project (HL-102925) and the Seattle GO Sequencing Project (HL-102926 to D.A.N. and J.S.) for early data release that proved useful for demonstrating filtering strategies. Our work was supported in part by grants from the NIH/NHLBI (5R01HL094976 to D.A.N. and J.S.), the NIH/National Human Genome Research Institute (5R21HG004749 to J.S., 1RC2HG005608 to M.J.B., D.A.N. and J.S., and 5RO1HG004316 to H.K.T.), NIH/National Institute of Environmental Health Sciences (HHSN273200800010C to D.N.), the Life Sciences Discovery Fund (2065508 and 0905001), the Washington Research Foundation and the NIH/National Institute of Child Health and Human Development (1R01HD048895 to M.J.B.). S.B.N. is supported by the Agency for Science, Technology and Research, Singapore. A.W.B. is supported by a training fellowship from the NIH/National Human Genome Research Institute (T32HG00035).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Michael J. Bamshad or Jay Shendure.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary information S1 (table)

Diseases identified to date via exome sequencing (PDF 253 kb)

Related links

Related links

DATABASES

1000 Genomes Project

dbSNP

EntrezGene

GeneReviews

Human Gene Mutation Database (HGMD)

miRBase

OMIM

arterial calcifications

Bartter syndrome

congenital chloride-losing diarrhoea

Charcot–Marie–Tooth syndrome

cystic fibrosis

Schinzel–Giedion syndrome

sickle cell anaemia

X-linked lymphoproliferative syndrome type 2

RefSeq

FURTHER INFORMATION

Burrows–Wheeler alignment tool

Consensus coding sequence (CCDS) project

Exome Variant Server

Generic exome analysis plan

Genetic Evolutionary Rate Profiling (GERP)

Genome Analysis Toolkit (GATK)

Human Variome Project

Multivariate Analysis of Protein Polymorphism (MAPP)

Nature Reviews Genetics series on Applications of Next-Generation Sequencing

Nature Reviews Genetics series on Translational Genetics

Nature Reviews Genetics series on Study Designs

NHLBI Grand Opportunity Exome Sequencing Project

Phylogenetic analysis with space/time models (PHAST)

phastphyloP

Polymorphism Phenotyping v2 (Polyphen2)

Seqanswers

SIFT

Glossary

Mendelian disorders

Phenotypes caused by a mutation (or mutations) in a single gene and inherited in a dominant, recessive or X-linked pattern.

Penetrance

The proportion of individuals with a specific phenotype among carriers of a particular genotype.

Locus heterogeneity

The appearance of phenotypically similar characteristics resulting from mutations at different genetic loci. Differences in effect size or in replication between studies and samples are often ascribed to different loci leading to the same disease.

Genome-wide association studies

(GWASs). Studies that search for a population association between a phenotype and a particular allele by screening loci (most commonly by genotyping SNPs) across the entire genome.

Complex traits

Traits that are influenced by the environment and/or through a combination of variants in at least several genes, each of which has a small effect.

Heritability

The proportion of the total phenotypic variation in a given characteristic that can be attributed to additive genetic effects.

Next-generation DNA sequencing

Highly parallelized DNA-sequencing technologies that produce many hundreds of thousands or millions of short reads (25–500bp) for a low cost and in a short time.

Exome

The subset of a genome that is protein coding. In addition to the exome, commercially available capture probes target non-coding exons, sequences flanking exons and microRNAs.

Homozygosity mapping

Narrowing down the location of a gene underlying a trait by searching for regions of the genome in which both chromosomal segments are inherited identically-by-descent.

Sample indexing

Sequencing more than one sample in a single sequencing lane.

RefSeq

An open-access, annotated and curated collection of publicly available nucleotide sequences (DNA and RNA) and their protein translations.

Ultra-conserved elements

Subsequences of the genome that appear to be under extremely high levels of sequence constraint based on phylogenetic comparisons.

Purifying selection

Selection against a functionally deleterious allele.

Parametric tests

Statistical significance tests for which P values are based on models or assumed formulae for the distribution of the test statistic.

Permutation test

A statistical test in which the data are randomized many times to determine the statistical significance of the experimental outcome.

Multiplex families

Families in which two or more individuals are affected by the same disorder.

Identity-by-descent

Alleles on different chromosomes that are identical because they are inherited from a shared common ancestor.

Identity-by-state

Alleles on different chromosomes that are identical but do not share a common ancestor with respect to a pedigree or population of interest.

Haplotype

A combination of alleles on a single chromosome.

Processed pseudogenes

Copies of the coding sequences of genes that lack promoters and introns, contain poly(A) tails and are flanked by target-site duplications.

Posterior probability

The probability of an event after combining prior knowledge of the event with the likelihood of that event given by observed data.

Bootstrap

A type of statistical analysis that is generally used for measuring the reliability of a sample estimate. It proceeds by the repeated sampling, with replacement, of the original data set. In the application described here bootstrapping is used to assess the probability of identifying the causal variant for a genetic condition in a population.

Incidental findings

Findings that are not explicitly related to the original research hypotheses (that is, primary findings).>

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bamshad, M., Ng, S., Bigham, A. et al. Exome sequencing as a tool for Mendelian disease gene discovery. Nat Rev Genet 12, 745–755 (2011). https://doi.org/10.1038/nrg3031

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg3031

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research