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.

  • Review Article
  • Published:

Comparative transcriptomics in human and mouse

Key Points

  • The mouse is the most widely used model organism to study human disease, but often mouse biology cannot be extrapolated to humans. A deep comparison of mouse and human physiology at the molecular level is essential for understanding under which circumstances the mouse can be a suitable model of human biology and for creating better mouse models. Advances in next-generation sequencing technologies fostered genome-wide annotation of functional DNA elements, enabling extensive comparison of the human and mouse genomes.

  • At the transcriptional level, human and mouse gene expression profiles are conserved overall, although the degree of conservation varies depending on the tissues and the genes that are compared. Therefore, the question of whether the human and mouse transcriptomes cluster preferentially by tissue or organ or by species does not have an answer overall, and it depends specifically on the genes being considered.

  • Conservation of expression is not a direct consequence of conservation in regulatory sequences, including promoters and enhancers. Although gene regulatory networks are preserved overall between human and mouse, transcription binding sites are often not conserved.

  • Inter-individual genetic variation can affect human gene expression, but such variation cannot be modelled in inbred strains of laboratory mice because their genetic variation is small compared to the human population. An expansion of the current studies on the relationship between genetic variation and gene expression in outbred mice might provide helpful insights to understand the same relationship in humans.

  • Emerging technologies — such single-cell genomics and single-cell spatial transcriptomics — and time series experiments will improve the annotation of human and mouse genomes, refine the current definitions of homologous cell types and homologous (molecular) phenotypes, and ultimately help scientists to identify which mouse models are the most appropriate to address a given biological question.

Abstract

Cross-species comparisons of genomes, transcriptomes and gene regulation are now feasible at unprecedented resolution and throughput, enabling the comparison of human and mouse biology at the molecular level. Insights have been gained into the degree of conservation between human and mouse at the level of not only gene expression but also epigenetics and inter-individual variation. However, a number of limitations exist, including incomplete transcriptome characterization and difficulties in identifying orthologous phenotypes and cell types, which are beginning to be addressed by emerging technologies. Ultimately, these comparisons will help to identify the conditions under which the mouse is a suitable model of human physiology and disease, and optimize the use of animal models.

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

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Homology of human and mouse genes and genomic elements.
Figure 2: Simplified clustering of human and mouse tissue samples and variance decomposition of gene expression.
Figure 3: Cellular composition of human and mouse pancreatic islets.
Figure 4: Multidimensional complexity of omics-layer integration across species.

Similar content being viewed by others

References

  1. Chinwalla, A. T. et al. Initial sequencing and comparative analysis of the mouse genome. Nature 420, 520–562 (2002). This article comprehensively characterizes the initial sequence of the mouse genome and is still a valuable reference for comparative genomics.

    Article  CAS  PubMed  Google Scholar 

  2. Adams, D. J. & van der Weyden, L. Contemporary approaches for modifying the mouse genome. Physiol. Genomics 34, 225–238 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Bedell, M. A., Jenkins, N. A. & Copeland, N. G. Mouse models of human disease. Part I: techniques and resources for genetic analysis in mice. Genes Dev. 11, 1–10 (1997).

    Article  CAS  PubMed  Google Scholar 

  4. Singh, P., Schimenti, J. C. & Bolcun-Filas, E. A mouse geneticist's practical guide to CRISPR applications. Genetics 199, 1–15 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Bult, C. J. et al. Mouse genome database 2016. Nucleic Acids Res. 44, D840–D847 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Qin, W. et al. Generating mouse models using CRISPR-Cas9-mediated genome editing. Curr. Protoc. Mouse Biol. 6, 39–66 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  7. European Commission. Report from the commission to the council and the European parliament: seventh report on the statistics on the number of animals used for experimental and other scientific purposes in the member states of the European Union. Eur-lex http://eur-lex.europa.eu/legal-content/EN/TXT/PDF/?uri=CELEX:52013DC0859&from=EN (2013).

  8. Home Office. Annual statistics of scientific procedures on living animals Great Britain 2014. Gov.uk https://www.gov.uk/government/uploads/system/uploads/attachment_data/file/469508/spanimals14.pdf (2014).

  9. Hay, M., Thomas, D. W., Craighead, J. L., Economides, C. & Rosenthal, J. Clinical development success rates for investigational drugs. Nat. Biotechnol. 32, 40–51 (2014). A comprehensive survey of clinical success rates across the drug industry.

    Article  CAS  PubMed  Google Scholar 

  10. Mak, I., Evaniew, N. & Ghert, M. Lost in translation: animal models and clinical trials in cancer treatment. Am. J. Transl Res. 6, 114–118 (2014).

    PubMed  PubMed Central  Google Scholar 

  11. Morgan, R. A. Human tumor xenografts: the good, the bad, and the ugly. Mol. Ther. 20, 882 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lonsdale, J. et al. The genotype-tissue expression (GTEx) project. Nat. Genet. 45, 580–585 (2013).

    Article  CAS  Google Scholar 

  13. Kundaje, A. et al. Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Abbott, A. Europe to map the human epigenome. Nature 477, 518 (2011).

    Article  CAS  PubMed  Google Scholar 

  15. The FANTOM Consortium and the RIKEN PMI and CLST (DGT). A promoter-level mammalian expression atlas. Nature 507, 462–470 (2014).

  16. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

  17. Yue, F. et al. A comparative encyclopedia of DNA elements in the mouse genome. Nature 515, 355–364 (2014). This paper from the Mouse ENCODE Consortium presents an extensive catalogue of mouse DNA elements identified through hundreds of next-generation sequencing assays.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Lander, E. S. et al. Initial sequencing and analysis of the human genome. Nature 409, 860–921 (2001).

    Article  CAS  PubMed  Google Scholar 

  19. Venter, J. C. et al. The sequence of the human genome. Science 291, 1304–1351 (2001).

    Article  CAS  PubMed  Google Scholar 

  20. Harrow, J. et al. GENCODE: the reference human genome annotation for The ENCODE Project. Genome Res. 22, 1760–1774 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Mudge, J. M. & Harrow, J. Creating reference gene annotation for the mouse C57BL6/J genome assembly. Mamm. Genome 26, 366–378 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Pervouchine, D. et al. Enhanced transcriptome maps from multiple mouse tissues reveal evolutionary constraint in gene expression for thousands of genes. Nat. Commun. 6, 5903 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Herrero, J. et al. Ensembl comparative genomics resources. Database (Oxford) 2016, bav096 (2016).

  24. Wang, Y., Gao, L., Conrad, C. G. & Andreadis, A. Saitohin, which is nested within the tau gene, interacts with tau and Abl and its human-specific allele influences Abl phosphorylation. J. Cell. Biochem. 112, 3482–3488 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Shi, X., Sun, M., Liu, H., Yao, Y. & Song, Y. Long non-coding RNAs: a new frontier in the study of human diseases. Cancer Lett. 339, 159–166 (2013).

    Article  CAS  PubMed  Google Scholar 

  26. Wapinski, O. & Chang, H. Y. Long noncoding RNAs and human disease. Trends Cell Biol. 21, 354–361 (2011).

    Article  CAS  PubMed  Google Scholar 

  27. Esteller, M. Non-coding RNAs in human disease. Nat. Rev. Genet. 12, 861–874 (2011).

    Article  CAS  PubMed  Google Scholar 

  28. Derrien, T. et al. The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression. Genome Res. 22, 1775–1789 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Cabili, M. N. et al. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev. 25, 1915–1927 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ulitsky, I. Evolution to the rescue: using comparative genomics to understand long non-coding RNAs. Nat. Rev. Genet. 17, 601–614 (2016). This Review summarizes current strategies for identifying lncRNAs and their function through comparative analysis across different species.

    Article  CAS  PubMed  Google Scholar 

  31. Nawrocki, E. P. et al. Rfam 12.0: updates to the RNA families database. Nucleic Acids Res. 43, D130–D137 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Pignatelli, M. et al. ncRNA orthologies in the vertebrate lineage. Database (Oxford) 2016, bav127 (2016).

  33. Hezroni, H. et al. Principles of long noncoding RNA evolution derived from direct comparison of transcriptomes in 17 species. Cell Rep. 11, 1110–1122 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Necsulea, A. et al. The evolution of lncRNA repertoires and expression patterns in tetrapods. Nature 505, 635–640 (2014).

    CAS  PubMed  Google Scholar 

  35. Washietl, S., Kellis, M. & Garber, M. Evolutionary dynamics and tissue specificity of human long noncoding RNAs in six mammals. Genome Res. 24, 616–628 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Chen, J. et al. Evolutionary analysis across mammals reveals distinct classes of long non-coding RNAs. Genome Biol. 17, 19 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Engström, P. G. et al. Complex loci in human and mouse genomes. PLoS Genet. 2, e47 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Faghihi, M. A. & Wahlestedt, C. Regulatory roles of natural antisense transcripts. Nat. Rev. Mol. Cell Biol. 10, 637–643 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Roux, J., Gonzalez-Porta, M. & Robinson-Rechavi, M. Comparative analysis of human and mouse expression data illuminates tissue-specific evolutionary patterns of miRNAs. Nucleic Acids Res. 40, 5890–5900 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Meunier, J. et al. Birth and expression evolution of mammalian microRNA genes. Genome Res. 23, 34–45 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kutter, C. et al. Pol III binding in six mammals shows conservation among amino acid isotypes despite divergence among tRNA genes. Nat. Genet. 43, 948–955 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Zhang, B. et al. Changes in snoRNA and snRNA abundance in the human, chimpanzee, macaque, and mouse brain. Genome Biol. Evol. 8, 840–850 (2016).

    PubMed  PubMed Central  Google Scholar 

  43. Matera, A. G., Terns, R. M. & Terns, M. P. Non-coding RNAs: lessons from the small nuclear and small nucleolar RNAs. Nat. Rev. Mol. Cell Biol. 8, 209–220 (2007).

    Article  CAS  PubMed  Google Scholar 

  44. Huang, Y. et al. Molecular functions of small regulatory noncoding RNA. Biochemistry (Mosc.) 78, 221–230 (2013).

    Article  CAS  Google Scholar 

  45. Li, Y. & Kowdley, K. V. MicroRNAs in common human diseases. Genomics Proteomics Bioinformatics 10, 246–253 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Lin, S. & Gregory, R. I. MicroRNA biogenesis pathways in cancer. Nat. Rev. Cancer 15, 321–333 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Park, C. Y., Choi, Y. & McManus, M. T. Analysis of microRNA knockouts in mice. Hum. Mol. Genet. 19, R169–R175 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Kozomara, A. & Griffiths-Jones, S. miRBase: integrating microRNA annotation and deep-sequencing data. Nucleic Acids Res. 39, D152–D157 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Landgraf, P. et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 129, 1401–1414 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Goodenbour, J. M. & Pan, T. Diversity of tRNA genes in eukaryotes. Nucleic Acids Res. 34, 6137–6146 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chan, P. P. & Lowe, T. M. GtRNAdb: a database of transfer RNA genes detected in genomic sequence. Nucleic Acids Res. 37, D93–D97 (2009).

    Article  CAS  PubMed  Google Scholar 

  52. Zheng-Bradley, X., Rung, J., Parkinson, H. & Brazma, A. Large scale comparison of global gene expression patterns in human and mouse. Genome Biol. 11, R124 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Jolliffe, I. Principal Component Analysis (Wiley Online Library, 2002).

    Google Scholar 

  54. Cox, M. A. & Cox, T. F. in Handbook of Data Visualization (eds Chen, C., Härdle, W. & Unwin, A.) 315–347 (Springer, 2008).

    Book  Google Scholar 

  55. van der Maaten, L. & Hinton, G. Visualizing data using t-SNE. J. Mach. Learn. Res. 9, 2579–2605 (2008).

    Google Scholar 

  56. McCall, M. N., Uppal, K., Jaffee, H. A., Zilliox, M. J. & Irizarry, R. A. The gene expression barcode: leveraging public data repositories to begin cataloging the human and murine transcriptomes. Nucleic Acids Res. 39, D1011–D1015 (2011).

    Article  CAS  PubMed  Google Scholar 

  57. Liao, B.-Y. & Zhang, J. Evolutionary conservation of expression profiles between human and mouse orthologous genes. Mol. Biol. Evol. 23, 530–540 (2006). This was the first paper to highlight the importance of normalization in comparative transcriptomics studies.

    Article  CAS  PubMed  Google Scholar 

  58. Yanai, I., Graur, D. & Ophir, R. Incongruent expression profiles between human and mouse orthologous genes suggest widespread neutral evolution of transcription control. OMICS 8, 15–24 (2004).

    Article  CAS  PubMed  Google Scholar 

  59. Pishesha, N. et al. Transcriptional divergence and conservation of human and mouse erythropoiesis. Proc. Natl Acad. Sci. USA 111, 4103–4108 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Schroder, K. et al. Conservation and divergence in Toll-like receptor 4-regulated gene expression in primary human versus mouse macrophages. Proc. Natl Acad. Sci. USA 109, E944–E953 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Jubb, A. W., Young, R. S., Hume, D. A. & Bickmore, W. A. Enhancer turnover is associated with a divergent transcriptional response to glucocorticoid in mouse and human macrophages. J. Immunol. 196, 813–822 (2016).

    Article  CAS  PubMed  Google Scholar 

  62. Seok, J. et al. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc. Natl Acad. Sci. USA 110, 3507–3512 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Takao, K. & Miyakawa, T. Genomic responses in mouse models greatly mimic human inflammatory diseases. Proc. Natl Acad. Sci. USA 112, 1167–1172 (2015).

    Article  CAS  PubMed  Google Scholar 

  64. Warren, H. S. et al. Mice are not men. Proc. Natl Acad. Sci. USA 112, E345 (2015).

    Article  CAS  PubMed  Google Scholar 

  65. Shay, T., Lederer, J. A. & Benoist, C. Genomic responses to inflammation in mouse models mimic humans: we concur, apples to oranges comparisons won't do. Proc. Natl Acad. Sci. USA 112, E346 (2015).

    Article  CAS  PubMed  Google Scholar 

  66. Romero, I. G., Ruvinsky, I. & Gilad, Y. Comparative studies of gene expression and the evolution of gene regulation. Nat. Rev. Genet. 13, 505–516 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Necsulea, A. & Kaessmann, H. Evolutionary dynamics of coding and non-coding transcriptomes. Nat. Rev. Genet. 15, 734–748 (2014). This is a Review on comparative transcriptomics studies in vertebrates.

    Article  CAS  PubMed  Google Scholar 

  68. 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).

    Article  CAS  PubMed  Google Scholar 

  69. Stamatoyannopoulos, J. A. et al. An encyclopedia of mouse DNA elements (Mouse ENCODE). Genome Biol. 13, 418 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Lin, S. et al. Comparison of the transcriptional landscapes between human and mouse tissues. Proc. Natl Acad. Sci. USA 111, 17224–17229 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Sudmant, P. H., Alexis, M. S. & Burge, C. B. Meta-analysis of RNA-seq expression data across species, tissues and studies. Genome Biol. 16, 287 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Su, A. I. et al. Large-scale analysis of the human and mouse transcriptomes. Proc. Natl Acad. Sci. USA 99, 4465–4470 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Chan, E. T. et al. Conservation of core gene expression in vertebrate tissues. J. Biol. 8, 33 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Gilad, Y. & Mizrahi-Man, O. A reanalysis of mouse ENCODE comparative gene expression data. F1000Res. 4, 121 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Brawand, D. et al. The evolution of gene expression levels in mammalian organs. Nature 478, 343–348 (2011).

    Article  CAS  PubMed  Google Scholar 

  76. Barbosa-Morais, N. L. et al. The evolutionary landscape of alternative splicing in vertebrate species. Science 338, 1587–1593 (2012).

    Article  CAS  PubMed  Google Scholar 

  77. Merkin, J., Russell, C., Chen, P. & Burge, C. B. Evolutionary dynamics of gene and isoform regulation in mammalian tissues. Science 338, 1593–1599 (2012). References 76 and 77 are the first systematic studies on alternative splicing evolution across vertebrates by RNA-seq.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Breschi, A. et al. Gene-specific patterns of expression variation across organs and species. Genome Biol. 17, 151 (2016). This study investigated the pattern of gene expression variation across tissues and species individually for each gene along the set of vertebrate orthologues.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Bortvin, A. et al. Incomplete reactivation of Oct4-related genes in mouse embryos cloned from somatic nuclei. Development 130, 1673–1680 (2003).

    Article  CAS  PubMed  Google Scholar 

  80. Hardison, R. C. A guide to translation of research results from model organisms to human. Genome Biol. 17, 161 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Soumillon, M. et al. Cellular source and mechanisms of high transcriptome complexity in the mammalian testis. Cell Rep. 3, 2179–2190 (2013).

    Article  CAS  PubMed  Google Scholar 

  82. Liao, B.-Y. & Zhang, J. Low rates of expression profile divergence in highly ex-pressed genes and tissue-specific genes during mammalian evolution. Mol. Biol. Evol. 23, 1119–1128 (2006).

    Article  CAS  PubMed  Google Scholar 

  83. Wang, Y. & Rekaya, R. A comprehensive analysis of gene expression evolution between humans and mice. Evol. Bioinform. Online 5, 81–90 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Koonin, E. V. & Wolf, Y. I. Constraints and plasticity in genome and molecular-phenome evolution. Nat. Rev. Genet. 11, 487–498 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Vakhrusheva, O. A., Bazykin, G. A. & Kondrashov, A. S. Genome-level analysis of selective constraint without apparent sequence conservation. Genome Biol. Evol. 5, 532–541 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Carvunis, A.-R. et al. Evidence for a common evolutionary rate in metazoan transcriptional networks. eLife 4, e11615 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  87. Weirauch, M. T. & Hughes, T. R. Conserved expression without conserved regulatory sequence: the more things change, the more they stay the same. Trends Genet. 26, 66–74 (2010).

    Article  CAS  PubMed  Google Scholar 

  88. Pai, A. A. & Gilad, Y. Comparative studies of gene regulatory mechanisms. Curr. Opin. Genet. Dev. 29, 68–74 (2014).

    Article  CAS  PubMed  Google Scholar 

  89. Odom, D. T. et al. Tissue-specific transcriptional regulation has diverged significantly between human and mouse. Nat. Genet. 39, 730–732 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Johnson, R. et al. Evolution of the vertebrate gene regulatory network controlled by the transcriptional repressor REST. Mol. Biol. Evol. 26, 1491–1507 (2009).

    Article  CAS  PubMed  Google Scholar 

  91. Schmidt, D. et al. Five-vertebrate ChIP-seq reveals the evolutionary dynamics of transcription factor binding. Science 328, 1036–1040 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Kunarso, G. et al. Transposable elements have rewired the core regulatory network of human embryonic stem cells. Nat. Genet. 42, 631–634 (2010).

    Article  CAS  PubMed  Google Scholar 

  93. Ballester, B. et al. Multi-species, multi-transcription factor binding highlights con-served control of tissue-specific biological pathways. eLife 3, e02626 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Ernst, J. & Kellis, M. ChromHMM: automating chromatin-state discovery and char-acterization. Nat. Methods 9, 215–216 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Cheng, Y. et al. Principles of regulatory information conservation between mouse and human. Nature 515, 371–375 (2014). A comparative analysis of the binding sites of 32 transcription factors through ChIP–seq in human and mouse cell lines.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Denas, O. et al. Genome-wide comparative analysis reveals human–mouse regulatory landscape and evolution. BMC Genomics 16, 87 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  97. Vierstra, J. et al. Mouse regulatory DNA landscapes reveal global principles of cis-regulatory evolution. Science 346, 1007–1012 (2014). An analysis of open chromatin regions in 45 mouse cell and tissue types by DNase-seq, with a comparison to humans.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Bourque, G. et al. Evolution of the mammalian transcription factor binding repertoire via transposable elements. Genome Res. 18, 1752–1762 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Villar, D. et al. Enhancer evolution across 20 mammalian species. Cell 160, 554–566 (2015). This study compares the liver enhancer landscape of 20 mammals, as inferred from ChIP–seq of H3K27ac and H3K4me3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Stergachis, A. B. et al. Conservation of trans-acting circuitry during mammalian regulatory evolution. Nature 515, 365–370 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Young, R. S. Lineage-specific genomics: frequent birth and death in the human genome. Bioessays 38, 654–663 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Visel, A. et al. Ultraconservation identifies a small subset of extremely constrained developmental enhancers. Nat. Genet. 40, 158–160 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Pennacchio, L. A. et al. In vivo enhancer analysis of human conserved non-coding sequences. Nature 444, 499–502 (2006).

    Article  CAS  PubMed  Google Scholar 

  104. Visel, A., Minovitsky, S., Dubchak, I. & Pennacchio, L. A. VISTA Enhancer Browser — a database of tissue-specific human enhancers. Nucleic Acids Res. 35, D88–D92 (2007).

    Article  CAS  PubMed  Google Scholar 

  105. Welter, D. et al. The NHGRI GWAS Catalog, a curated resource of SNP-trait associations. Nucleic Acids Res. 42, D1001–D1006 (2014).

    Article  CAS  PubMed  Google Scholar 

  106. 1000 Genomes Project Consortium et al. A global reference for human genetic variation. Nature 526, 68–74 (2015). A milestone paper on human genetic variation from the 1000 Genomes Project Consortium.

  107. Levy, S. et al. The diploid genome sequence of an individual human. PLoS Biol. 5, e254 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Lappalainen, T. et al. Transcriptome and genome sequencing uncovers functional variation in humans. Nature 501, 506–511 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. GTEx Consortium et al. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648–660 (2015).

  110. Yang, H. et al. Subspecific origin and haplotype diversity in the laboratory mouse. Nat. Genet. 43, 648–655 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Beck, J. A. et al. Genealogies of mouse inbred strains. Nat. Genet. 24, 23–25 (2000).

    Article  CAS  PubMed  Google Scholar 

  112. Wade, C. M. et al. The mosaic structure of variation in the laboratory mouse genome. Nature 420, 574–578 (2002). This paper explains how the most common laboratory mouse strains were created from wild mice.

    Article  CAS  PubMed  Google Scholar 

  113. Yang, H., Bell, T. A., Churchill, G. A. & de Villena, F. P.-M. On the subspecific origin of the laboratory mouse. Nat. Genet. 39, 1100–1107 (2007).

    Article  CAS  PubMed  Google Scholar 

  114. Keane, T. M. et al. Mouse genomic variation and its effect on phenotypes and gene regulation. Nature 477, 289–294 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Matsuo, N. et al. Behavioral profiles of three C57BL/6 substrains. Front. Behav. Neurosci. 4, 29 (2010).

    PubMed  PubMed Central  Google Scholar 

  116. Kiselycznyk, C. & Holmes, A. All (C57BL/6) mice are not created equal. Front. Neurosci. 5, 10 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  117. Doran, A. G. et al. Deep genome sequencing and variation analysis of 13 inbred mouse strains defines candidate phenotypic alleles, private variation, and homozygous truncating mutations. Genome Biol. 17, 167 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Turk, R. et al. Gene expression variation between mouse inbred strains. BMC Genomics 5, 57 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Holgersen, K. et al. High-resolution gene expression profiling using RNA sequencing in patients with inflammatory bowel disease and in mouse models of colitis. J. Crohns Colitis 9, 492–506 (2015).

    Article  PubMed  Google Scholar 

  120. Mostafavi, S. et al. Variation and genetic control of gene expression in primary immunocytes across inbred mouse strains. J. Immunol. 193, 4485–4496 (2014).

    Article  CAS  PubMed  Google Scholar 

  121. Pritchard, C. C., Hsu, L., Delrow, J. & Nelson, P. S. Project normal: defining normal variance in mouse gene expression. Proc. Natl Acad. Sci. USA 98, 13266–13271 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Bogue, M. A., Churchill, G. A. & Chesler, E. J. Collaborative cross and diversity outbred data resources in the Mouse Phenome Database. Mamm. Genome 26, 511–520 (2015). This paper illustrates the current status of the Mouse Phenome Database as an established resource for studying mouse genetic variation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Chick, J. M. et al. Defining the consequences of genetic variation on a proteome-wide scale. Nature 534, 500–505 (2016). This paper describes the relationship between eQTLs and protein quantitative trait loci in outbred mice.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Deng, Q., Ramskold, D., Reinius, B. & Sandberg, R. Single-cell RNA-seq reveals dynamic, random monoallelic gene expression in mammalian cells. Science 343, 193–196 (2014).

    Article  CAS  PubMed  Google Scholar 

  125. Vickaryous, M. K. & Hall, B. K. Human cell type diversity, evolution, development, and classification with special reference to cells derived from the neural crest. Biol. Rev. 81, 425–455 (2006).

    Article  PubMed  Google Scholar 

  126. Abdulreda, M., Caicedo, A. & Berggren, P. A natural body window to study human pancreatic islet cell function and survival. CellR4 1, 111–122 (2013).

    CAS  Google Scholar 

  127. Mabbott, N. A., Baillie, J. K., Brown, H., Freeman, T. C. & Hume, D. A. An expression atlas of human primary cells: inference of gene function from coexpression networks. BMC Genomics 14, 632 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Hume, D. A., Summers, K. M., Raza, S., Baillie, J. K. & Freeman, T. C. Functional clustering and lineage markers: insights into cellular differentiation and gene function from large-scale microarray studies of purified primary cell populations. Genomics 95, 328–338 (2010).

    Article  CAS  PubMed  Google Scholar 

  129. Lee, Y.-s., Krishnan, A., Zhu, Q. & Troyanskaya, O. G. Ontology-aware classification of tissue and cell-type signals in gene expression profiles across platforms and technologies. Bioinformatics 29, 3036–3044 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Li, J. et al. Single-cell transcriptomes reveal characteristic features of human pancreatic islet cell types. EMBO Rep. 17, 178–187 (2016).

    Article  CAS  PubMed  Google Scholar 

  131. Saliba, A.-E., Westermann, A. J., Gorski, S. A. & Vogel, J. Single-cell RNA-seq: advances and future challenges. Nucleic Acids Res. 42, 8845–8860 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Macosko, E. Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Kolodziejczyk, A. A., Kim, J. K., Svensson, V., Marioni, J. C. & Teichmann, S. A. The technology and biology of single-cell RNA sequencing. Mol. Cell 58, 610–620 (2015).

    Article  CAS  PubMed  Google Scholar 

  134. Trapnell, C. Defining cell types and states with single-cell genomics. Genome Res. 25, 1491–1498 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Zeisel, A. et al. Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq. Science 347, 1138–1142 (2015).

    Article  CAS  PubMed  Google Scholar 

  136. Treutlein, B. et al. Reconstructing lineage hierarchies of the distal lung epithelium using single-cell RNA-seq. Nature 509, 371–375 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Grün, D. et al. Single-cell messenger RNA sequencing reveals rare intestinal cell types. Nature 525, 251–255 (2015).

    Article  CAS  PubMed  Google Scholar 

  138. Darmanis, S. et al. A survey of human brain transcriptome diversity at the single cell level. Proc. Natl Acad. Sci. USA 112, 7285–7290 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Jaitin, D. A. et al. Massively parallel single-cell RNA-seq for marker-free decomposition of tissues into cell types. Science 343, 776–779 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Paul, F. et al. Transcriptional heterogeneity and lineage commitment in myeloid progenitors. Cell 163, 1663–1677 (2015).

    Article  CAS  PubMed  Google Scholar 

  141. Scialdone, A. et al. Resolving early mesoderm diversification through single-cell expression profiling. Nature 535, 289–293 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Xue, Z. et al. Genetic programs in human and mouse early embryos revealed by single-cell RNA sequencing. Nature 500, 593–597 (2013). A dynamic transcriptional study of human and mouse early embryo development at single-cell resolution.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Blekhman, R., Marioni, J. C., Zumbo, P., Stephens, M. & Gilad, Y. Sex-specific and lineage-specific alternative splicing in primates. Genome Res. 20, 180–189 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Mele, M. et al. The human transcriptome across tissues and individuals. Science 348, 660–665 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Zhang, R., Lahens, N. F., Ballance, H. I., Hughes, M. E. & Hogenesch, J. B. A circadian gene expression atlas in mammals: implications for biology and medicine. Proc. Natl Acad. Sci. USA 111, 16219–16224 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Romero, I. G., Pai, A. A., Tung, J. & Gilad, Y. RNA-seq: impact of RNA degradation on transcript quantification. BMC Biol. 12, 42 (2014).

    Article  CAS  Google Scholar 

  147. Crosetto, N., Bienko, M. & van Oudenaarden, A. Spatially resolved transcriptomics and beyond. Nat. Rev. Genet. 16, 57–66 (2015).

    Article  CAS  PubMed  Google Scholar 

  148. Chen, K. H., Boettiger, A. N., Moffitt, J. R., Wang, S. & Zhuang, X. Spatially re-solved, highly multiplexed RNA profiling in single cells. Science 348, aaa6090 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Gharib, S. A. et al. Of mice and men: comparative proteomics of bronchoalveolar fluid. Eur. Respir. J. 35, 1388–1395 (2010).

    Article  CAS  PubMed  Google Scholar 

  151. Maher, B. ENCODE: the human encyclopaedia. Nature 489, 46–48 (2012).

    Article  PubMed  Google Scholar 

  152. Von Meyenn, F. et al. Comparative principles of DNA methylation reprogramming during human and mouse in vitro primordial germ cell specification. Dev. Cell 39, 104–115 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Nakamura, T. et al. A developmental coordinate of pluripotency among mice, monkeys and humans. Nature 537, 57–62 (2016).

    Article  CAS  PubMed  Google Scholar 

  154. Rosenthal, N. & Brown, S. The mouse ascending: perspectives for human-disease models. Nat. Cell Biol. 9, 993–999 (2007).

    Article  CAS  PubMed  Google Scholar 

  155. Onos, K. D., Rizzo, S. J. S., Howell, G. R. & Sasner, M. Toward more predictive genetic mouse models of Alzheimer's disease. Brain Res. Bull. 122, 1–11 (2016).

    Article  CAS  PubMed  Google Scholar 

  156. Blesa, J., Phani, S., Jackson-Lewis, V. & Przedborski, S. Classic and new animal models of Parkinson's disease. J. Biomed. Biotechnol. 2012, 845618 (2012).

  157. Ribeiro, F. M., Camargos, E. R., de Souza, L. C. & Teixeira, A. L. Animal models of neurodegenerative diseases. Rev. Bras. Psiquiatr. 35 (Suppl. 2), S82–S91 (2013).

    Article  PubMed  Google Scholar 

  158. Antonarakis, S. E. Down syndrome and the complexity of genome dosage imbalance. Nat. Rev. Genet. 18, 147–163 (2016).

    Article  CAS  PubMed  Google Scholar 

  159. Rueda, N., Florez, J. & Martinez-Cue, C. Mouse models of Down syndrome as a tool to unravel the causes of mental disabilities. Neural Plast. 2012, 584071 (2012).

  160. Steimer, T. Animal models of anxiety disorders in rats and mice: some conceptual issues. Dialogues Clin. Neurosci. 13, 495–506 (2011).

    PubMed  PubMed Central  Google Scholar 

  161. Schweinfurth, N. & Lang, U. E. Behavioral testing of mice concerning anxiety and depression. Z. Psychol. 223, 151–156 (2015).

    Google Scholar 

  162. Lynch, W. J., Nicholson, K. L., Dance, M. E., Morgan, R. W. & Foley, P. L. Animal models of substance abuse and addiction: implications for science, animal welfare, and society. Comp. Med. 60, 177–188 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Ellacott, K. L., Morton, G. J., Woods, S. C., Tso, P. & Schwartz, M. W. Assessment of feeding behavior in laboratory mice. Cell Metab. 12, 10–17 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Mestas, J. & Hughes, C. C. Of mice and not men: differences between mouse and human immunology. J. Immunol. 172, 2731–2738 (2004).

    Article  CAS  PubMed  Google Scholar 

  165. Karpel, M. E., Boutwell, C. L. & Allen, T. M. BLT humanized mice as a small animal model of HIV infection. Curr. Opin. Virol. 13, 75–80 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Chayama, K. et al. Animal model for study of human hepatitis viruses. J. Gastroenterol. Hepatol. 26, 13–18 (2011).

    Article  PubMed  Google Scholar 

  167. Silverman, J. L., Yang, M., Lord, C. & Crawley, J. N. Behavioural phenotyping assays for mouse models of autism. Nat. Rev. Neurosci. 11, 490–502 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Vanhooren, V. & Libert, C. The mouse as a model organism in aging research: usefulness, pitfalls and possibilities. Ageing Res. Rev. 12, 8–21 (2013).

    Article  PubMed  Google Scholar 

  169. Bult, C. J. et al. Mouse Tumor Biology (MTB): a database of mouse models for human cancer. Nucleic Acids Res. 43, D818–D824 (2015).

    Article  CAS  PubMed  Google Scholar 

  170. Justice, M. J. & Dhillon, P. Using the mouse to model human disease: increasing validity and reproducibility. Dis. Model. Mech. 9, 101–103 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Rangarajan, A. & Weinberg, R. A. Comparative biology of mouse versus human cells: modelling human cancer in mice. Nat. Rev. Cancer 3, 952–959 (2003).

    Article  CAS  PubMed  Google Scholar 

  172. Egan, M. E. How useful are cystic fibrosis mouse models? Drug Discov. Today Dis. Models 6, 35–41 (2009).

    Article  CAS  Google Scholar 

  173. Fisher, J. T., Zhang, Y. & Engelhardt, J. F. Comparative biology of cystic fibrosis animal models. Methods Mol. Biol. 742, 311–334 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. McGreevy, J. W., Hakim, C. H., McIntosh, M. A. & Duan, D. Animal models of Duchenne muscular dystrophy: from basic mechanisms to gene therapy. Dis. Model. Mech. 8, 195–213 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Modrek, B. & Lee, C. J. Alternative splicing in the human, mouse and rat genomes is associated with an increased frequency of exon creation and/or loss. Nat. Genet. 34, 177–180 (2003).

    Article  CAS  PubMed  Google Scholar 

  176. Abril, J. F., Castelo, R. & Guigo, R. Comparison of splice sites in mammals and chicken. Genome Res. 15, 111–119 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Sorek, R. & Ast, G. Intronic sequences flanking alternatively spliced exons are conserved between human and mouse. Genome Res. 13, 1631–1637 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Zambelli, F., Pavesi, G., Gissi, C., Horner, D. S. & Pesole, G. Assessment of orthologous splicing isoforms in human and mouse orthologous genes. BMC Genomics 11, 534 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Tilgner, H. et al. Comprehensive transcriptome analysis using synthetic long-read sequencing reveals molecular co-association of distant splicing events. Nat. Biotechnol. 33, 736–742 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Sharon, D., Tilgner, H., Grubert, F. & Snyder, M. A single-molecule long-read survey of the human transcriptome. Nat. Biotechnol. 31, 1009–1014 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors acknowledge G. Schiavo and D. Cirillo for insightful discussion. The authors acknowledge support of the Spanish Ministry of Economy and Competitiveness, 'Centro de Excelencia Severo Ochoa 2013–2017' of the Centres de Recerca de Catalunya Programme/Generalitat de Catalunya and the Programa de Ayudas Formación de Personal investigador (FPI) del Ministerio de Economia y Competitividad (BES-2012-055848).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Roderic Guigó.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

PowerPoint slides

Glossary

Synteny

Preserved genomic order and orientation of genes or other elements between species.

Xenograft models of cancer

Created when cancerous tissue from a person's primary tumour is implanted directly into an immunodeficient mouse.

Chromatin domains

Functionally distinct chromosomal regions that confer structural organization to eukaryotic genomes and represent regulatory units for gene expression and chromosome behaviour.

Cap analysis of gene expression

(CAGE). In CAGE, short ( 20-nucleotide) sequence tags originating from the 5′ end of full-length mRNAs are sequenced to identify transcription events on a genome-wide scale.

GENCODE annotation

The GENCODE project produces high-quality reference gene annotation and experimental validation for human and mouse genomes.

Long non-coding RNAs

(lncRNAs). Non-protein coding transcripts that are longer than 200 nucleotides. This somewhat arbitrary limit distinguishes lncRNAs from small regulatory RNAs.

Orthologous

Homologous genes in different species that have evolved from a common ancestral gene by speciation.

MicroRNAs

(miRNAs). Derived from primary transcripts with features similar to mRNAs that are enzymatically processed to their mature length of 21–24 nucleotides by Drosha and Dicer enzymes.

Transfer RNAs

(tRNAs). Adaptor RNA molecules (76–90 nucleotides) that serve as the physical link between the mRNA and the amino acid sequence of proteins by carrying an amino acid to the ribosome, as directed by the codon in an mRNA.

Small nuclear RNAs and small nucleolar RNAs

(snRNAs and snoRNAs). Classes of short non-coding RNAs (100–200 nucleotides) that have important regulatory roles in nuclear ribonucleoprotein complexes.

Homologues

A pair of genes that descended from a common ancestral gene.

Hierarchical clustering

A statistical method in which objects (for example, gene expression profiles for different individuals or tissue samples) are grouped into a hierarchy, which is visualized in a dendrogram. Objects close to each other in the hierarchy, as measured by tracing the branch heights, are also close by some measure of distance — for example, between gene expression profiles. Individuals or samples with similar expression profiles will be close together in terms of branch lengths.

Euclidean distance

The Euclidean distance between points p and q is the length of the line segment connecting them in a multidimensional space. In gene expression analysis, p and q are usually vectors of expression values in two samples or conditions.

Dimensionality reduction techniques

These reduce multidimensional data to a minimal number of dimensions for visualization by identifying those dimensions that capture the most important information underlying the data structure.

Principal component analysis

(PCA). Orthogonal linear transformation that transforms the original data to a new coordinate system, such that the greatest variance of the projected data comes to lie on the first coordinate (called the first principal component), the second greatest variance on the second coordinate, and so on.

Multidimensional scaling

(MDS). A technique used to display the information contained in a distance matrix. It aims to place each object in N-dimensional space such that the between-object distances are preserved as well as possible.

t-Distributed stochastic neighbour embedding

(t-SNE). A nonlinear dimensionality reduction technique that is based on the probability distribution over pairs of high-dimensional objects that are embedded into a space of two or three dimensions. Similar objects are modelled by nearby points, and dissimilar objects are modelled by distant points.

Normalization

Methods used to adjust measurements so that they can be appropriately compared among samples. For example, in microarray analysis, methods such as quantile normalization manipulate common characteristics of the data.

DNA exaptation

The shift in the function of a DNA sequence during evolution.

Expression quantitative trait loci

(eQTLs). Genomic loci that contribute to variation in the expression levels of mRNAs.

Allele-specific expression

Expression variation between the two haplotypes of a diploid individual, as distinguished by heterozygous sites.

Ischaemic time

In the case of organ donors, the time elapsed between the death of a donor and the organ extraction.

Pseudogenes

Segments of DNA that originate from functional genes, but have lost at least some of the ability of the parent gene in terms of expression or coding potential.

Precision medicine

An emerging approach for disease treatment and prevention that takes into account individual variability in genes, environment and lifestyle for each person.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Breschi, A., Gingeras, T. & Guigó, R. Comparative transcriptomics in human and mouse. Nat Rev Genet 18, 425–440 (2017). https://doi.org/10.1038/nrg.2017.19

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg.2017.19

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing