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.

Insights into the regulation of protein abundance from proteomic and transcriptomic analyses

Abstract

Recent advances in next-generation DNA sequencing and proteomics provide an unprecedented ability to survey mRNA and protein abundances. Such proteome-wide surveys are illuminating the extent to which different aspects of gene expression help to regulate cellular protein abundances. Current data demonstrate a substantial role for regulatory processes occurring after mRNA is made — that is, post-transcriptional, translational and protein degradation regulation — in controlling steady-state protein abundances. Intriguing observations are also emerging in relation to cells following perturbation, single-cell studies and the apparent evolutionary conservation of protein and mRNA abundances. Here, we summarize current understanding of the major factors regulating protein expression.

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: Modes of translation and protein-degradation regulation.
Figure 2: Relationships between mRNA and protein abundances, as observed in large-scale proteome- and transcriptome-profiling experiments.

References

  1. de Sousa Abreu, R., Penalva, L. O., Marcotte, E. & Vogel, C. Global signatures of protein and mRNA expression levels. Mol. Biosyst. 5, 1512–1526 (2009).

    PubMed  Google Scholar 

  2. Hu, Q. et al. The Orbitrap: a new mass spectrometer. J. Mass Spectrom. 40, 430–443 (2005).

    Article  CAS  Google Scholar 

  3. Mallick, P. & Kuster, B. Proteomics: a pragmatic perspective. Nature Biotech. 28, 695–709 (2010).

    Article  CAS  Google Scholar 

  4. Lu, P., Vogel, C., Wang, R., Yao, X. & Marcotte, E. M. Absolute protein expression profiling estimates the relative contributions of transcriptional and translational regulation. Nature Biotech. 25, 117–124 (2007).

    Article  CAS  Google Scholar 

  5. Schwanhäusser, B. et al. Global quantification of mammalian gene expression control. Nature 473, 337–342 (2011).

    Article  Google Scholar 

  6. Malmstrom, J. et al. Proteome-wide cellular protein concentrations of the human pathogen Leptospira interrogans. Nature 460, 762–765 (2009).

    Article  Google Scholar 

  7. Gerber, S. A., Rush, J., Stemman, O., Kirschner, M. W. & Gygi, S. P. Absolute quantification of proteins and phosphoproteins from cell lysates by tandem MS. Proc. Natl Acad. Sci. USA 100, 6940–6945 (2003).

    Article  CAS  Google Scholar 

  8. Ong, S. E. et al. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol. Cell. Proteomics 1, 376–386 (2002).

    Article  CAS  Google Scholar 

  9. Ross, P. L. et al. Multiplexed protein quantitation in Saccharomyces cerevisiae using amine-reactive isobaric tagging reagents. Mol. Cell. Proteomics 3, 1154–1169 (2004).

    Article  CAS  Google Scholar 

  10. Bantscheff, M., Schirle, M., Sweetman, G., Rick, J. & Kuster, B. Quantitative mass spectrometry in proteomics: a critical review. Anal. Bioanal. Chem. 389, 1017–1031 (2007).

    Article  CAS  Google Scholar 

  11. Martin, J. A. & Wang, Z. Next-generation transcriptome assembly. Nature Rev. Genet. 12, 671–682 (2011).

    Article  CAS  Google Scholar 

  12. Ingolia, N. T., Ghaemmaghami, S., Newman, J. R. & Weissman, J. S. Genome-wide analysis in vivo of translation with nucleotide resolution using ribosome profiling. Science 324, 218–223 (2009).

    Article  CAS  Google Scholar 

  13. Taniguchi, Y. et al. Quantifying E. coli proteome and transcriptome with single-molecule sensitivity in single cells. Science 329, 533–538 (2010).

    Article  CAS  Google Scholar 

  14. Newman, J. R. et al. Single-cell proteomic analysis of S. cerevisiae reveals the architecture of biological noise. Nature 441, 840–846 (2006).

    Article  CAS  Google Scholar 

  15. Ghaemmaghami, S. et al. Global analysis of protein expression in yeast. Nature 425, 737–741 (2003).

    Article  CAS  Google Scholar 

  16. Belle, A., Tanay, A., Bitincka, L., Shamir, R. & O'Shea, E. K. Quantification of protein half-lives in the budding yeast proteome. Proc. Natl Acad. Sci. USA 103, 13004–13009 (2006).

    Article  CAS  Google Scholar 

  17. Eden, E. et al. Proteome half-life dynamics in living human cells. Science 331, 764–768 (2011).

    Article  CAS  Google Scholar 

  18. Yen, H. C., Xu, Q., Chou, D. M., Zhao, Z. & Elledge, S. J. Global protein stability profiling in mammalian cells. Science 322, 918–923 (2008).

    Article  CAS  Google Scholar 

  19. Maier, T., Guell, M. & Serrano, L. Correlation of mRNA and protein in complex biological samples. FEBS Lett. 583, 3966–3973 (2009).

    Article  CAS  Google Scholar 

  20. de Godoy, L. M. et al. Comprehensive mass-spectrometry-based proteome quantification of haploid versus diploid yeast. Nature 455, 1251–1254 (2008).

    Article  CAS  Google Scholar 

  21. Lundberg, E. et al. Defining the transcriptome and proteome in three functionally different human cell lines. Mol. Syst. Biol. 6, 450 (2010).

    Article  Google Scholar 

  22. Sharova, L. V. et al. Database for mRNA half-life of 19 977 genes obtained by DNA microarray analysis of pluripotent and differentiating mouse embryonic stem cells. DNA Res. 16, 45–58 (2009).

    Article  CAS  Google Scholar 

  23. Selinger, D. W., Saxena, R. M., Cheung, K. J., Church, G. M. & Rosenow, C. Global RNA half-life analysis in Escherichia coli reveals positional patterns of transcript degradation. Genome Res. 13, 216–223 (2003).

    Article  CAS  Google Scholar 

  24. Vogel, C. et al. Sequence signatures and mRNA concentration can explain two-thirds of protein abundance variation in a human cell line. Mol. Syst. Biol. 6, 400 (2010).

    Article  Google Scholar 

  25. Lee, M. V. et al. A dynamic model of proteome changes reveals new roles for transcript alteration in yeast. Mol. Syst. Biol. 7, 514 (2011).

    Article  Google Scholar 

  26. Fournier, M. L. et al. Delayed correlation of mRNA and protein expression in rapamycin-treated cells and a role for Ggc1 in cellular sensitivity to rapamycin. Mol. Cell. Proteomics 9, 271–284 (2009).

    Article  Google Scholar 

  27. Vogel, C., Silva, G. M. & Marcotte, E. M. Protein expression regulation under oxidative stress. Mol. Cell. Proteomics 20 Sep 2011 (doi:10.1074/mcp.M111.009217).

    Article  Google Scholar 

  28. Maier, T. et al. Quantification of mRNA and protein and integration with protein turnover in a bacterium. Mol. Syst. Biol. 7, 511 (2011).

    Article  Google Scholar 

  29. Jayapal, K. P. et al. Uncovering genes with divergent mRNA-protein dynamics in Streptomyces coelicolor. PLoS ONE 3, e2097 (2008).

    Article  Google Scholar 

  30. Li, G. W. & Xie, X. S. Central dogma at the single-molecule level in living cells. Nature 475, 308–315 (2011).

    Article  CAS  Google Scholar 

  31. Fraser, H. B., Hirsh, A. E., Giaever, G., Kumm, J. & Eisen, M. B. Noise minimization in eukaryotic gene expression. PLoS Biol. 2, e137 (2004).

    Article  Google Scholar 

  32. Gandhi, S. J., Zenklusen, D., Lionnet, T. & Singer, R. H. Transcription of functionally related constitutive genes is not coordinated. Nature Struct. Mol. Biol. 18, 27–34 (2011).

    Article  CAS  Google Scholar 

  33. Pilpel, Y. Noise in biological systems: pros, cons, and mechanisms of control. Methods Mol. Biol. 759, 407–425 (2011).

    Article  CAS  Google Scholar 

  34. Plotkin, J. B. Transcriptional regulation is only half the story. Mol. Syst. Biol. 6, 406 (2010).

    Article  Google Scholar 

  35. Coghlan, A. & Wolfe, K. H. Relationship of codon bias to mRNA concentration and protein length in Saccharomyces cerevisiae. Yeast 16, 1131–1145 (2000).

    Article  CAS  Google Scholar 

  36. Ingolia, N. T., Lareau, L. F. & Weissman, J. S. Ribosome profiling of mouse embryonic stem cells reveals the complexity and dynamics of Mammalian proteomes. Cell 147, 789–802 (2011).

    Article  CAS  Google Scholar 

  37. Mayr, C. & Bartel, D. P. Widespread shortening of 3′UTRs by alternative cleavage and polyadenylation activates oncogenes in cancer cells. Cell 138, 673–684 (2009).

    Article  CAS  Google Scholar 

  38. Sandberg, R., Neilson, J. R., Sarma, A., Sharp, P. A. & Burge, C. B. Proliferating cells express mRNAs with shortened 3′ untranslated regions and fewer microRNA target sites. Science 320, 1643–1647 (2008).

    Article  CAS  Google Scholar 

  39. Schrimpf, S. P. et al. Comparative functional analysis of the Caenorhabditis elegans and Drosophila melanogaster proteomes. PLoS Biol. 7, e48 (2009).

    Article  Google Scholar 

  40. Laurent, J. et al. Protein abundances are more conserved than mRNA abundances across diverse taxa. Proteomics 10, 4209–4212 (2010).

    Article  CAS  Google Scholar 

  41. Weiss, M., Schrimpf, S., Hengartner, M. O., Lercher, M. J. & von Mering, C. Shotgun proteomics data from multiple organisms reveals remarkable quantitative conservation of the eukaryotic core proteome. Proteomics 10, 1297–1306 (2010).

    Article  CAS  Google Scholar 

  42. Ramakrishnan, S. R. et al. Integrating shotgun proteomics and mRNA expression data to improve protein identification. Bioinformatics 25, 1397–1403 (2009).

    Article  CAS  Google Scholar 

  43. Golding, I., Paulsson, J., Zawilski, S. M. & Cox, E. C. Real-time kinetics of gene activity in individual bacteria. Cell 123, 1025–1036 (2005).

    Article  CAS  Google Scholar 

  44. Raj, A., Peskin, C. S., Tranchina, D., Vargas, D. Y. & Tyagi, S. Stochastic mRNA synthesis in mammalian cells. PLoS Biol. 4, e309 (2006).

    Article  Google Scholar 

  45. Mukherji, S. et al. MicroRNAs can generate thresholds in target gene expression. Nature Genet. 43, 854–859 (2011).

    Article  CAS  Google Scholar 

  46. Baek, D. et al. The impact of microRNAs on protein output. Nature 455, 64–71 (2008).

    Article  CAS  Google Scholar 

  47. Selbach, M. et al. Widespread changes in protein synthesis induced by microRNAs. Nature 455, 58–63 (2008).

    Article  CAS  Google Scholar 

  48. Jacobs, J. M. et al. Utilizing human blood plasma for proteomic biomarker discovery. J. Proteome Res. 4, 1073–1085 (2005).

    Article  CAS  Google Scholar 

  49. Hebenstreit, D. et al. RNA sequencing reveals two major classes of gene expression levels in metazoan cells. Mol. Syst. Biol. 7, 497 (2011).

    Article  Google Scholar 

  50. Dahan, O., Gingold, H. & Pilpel, Y. Regulatory mechanisms and networks couple the different phases of gene expression. Trends Genet. 27, 316–322 (2011).

    Article  CAS  Google Scholar 

  51. Auld, K. L. & Silver, P. A. Transcriptional regulation by the proteasome as a mechanism for cellular protein homeostasis. Cell Cycle 5, 1503–1505 (2006).

    Article  CAS  Google Scholar 

  52. Kodadek, T. No splicing, no dicing: non-proteolytic roles of the ubiquitin-proteasome system in transcription. J. Biol. Chem. 285, 2221–2226 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors acknowledge support from the US National Institutes of Health and the Welch Foundation (F1515, to E.M.M.). We also thank T. Lionnet for helpful discussions.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Christine Vogel or Edward M. Marcotte.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

FURTHER INFORMATION

Christine Vogel's homepage

Edward M. Marcotte's homepage

Glossary

High-resolution tandem mass spectrometry

The use of two consecutive mass spectrometry steps to measure mass-to-charge ratios for peptides and their fragment ions, respectively. Modern technology enables a mass accuracy of <0.01 Da.

Nanoflow chromatography

In the context of peptides, this method separates a peptide mixture by differences in biophysical properties. It operates at flow rates of nanolitres per minute to increase separation efficiency and decrease sample volumes.

PEST sequences

Protein sequence motif enriched for proline (P), glutamate (E), serine (S) and threonine (T) that serves as a protein degradation signal.

Ribosome footprinting

Identification of ribosomal binding sites on mRNA through ribosome stalling and next-generation sequencing of the bound RNA fragments.

Stable isotopic labelling with amino acids in cell culture

(SILAC). A widely used technique for estimating relative protein concentrations by mass spectrometry.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Vogel, C., Marcotte, E. Insights into the regulation of protein abundance from proteomic and transcriptomic analyses. Nat Rev Genet 13, 227–232 (2012). https://doi.org/10.1038/nrg3185

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg3185

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