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.

  • Protocol
  • Published:

A quantitative thiol reactivity profiling platform to analyze redox and electrophile reactive cysteine proteomes

Abstract

Cysteine is unique among all protein-coding amino acids, owing to its intrinsically high nucleophilicity. The cysteinyl thiol group can be covalently modified by a broad range of redox mechanisms or by various electrophiles derived from exogenous or endogenous sources. Measuring the response of protein cysteines to redox perturbation or electrophiles is critical for understanding the underlying mechanisms involved. Activity-based protein profiling based on thiol-reactive probes has been the method of choice for such analyses. We therefore adapted this approach and developed a new chemoproteomic platform, termed ‘QTRP’ (quantitative thiol reactivity profiling), that relies on the ability of a commercially available thiol-reactive probe IPM (2-iodo-N-(prop-2-yn-1-yl)acetamide) to covalently label, enrich and quantify the reactive cysteinome in cells and tissues. Here, we provide a detailed and updated workflow of QTRP that includes procedures for (i) labeling of the reactive cysteinome from cell or tissue samples (e.g., control versus treatment) with IPM, (ii) processing the protein samples into tryptic peptides and tagging the probe-modified peptides with isotopically labeled azido-biotin reagents containing a photo-cleavable linker via click chemistry reaction, (iii) capturing biotin-conjugated peptides with streptavidin beads, (iv) identifying and quantifying the photo-released peptides by mass spectrometry (MS)-based shotgun proteomics and (v) interpreting MS data by a streamlined informatic pipeline using a proteomics software, pFind 3, and an automatic post-processing algorithm. We also exemplified here how to use QTRP for mining H2O2-sensitive cysteines and for determining the intrinsic reactivity of cysteines in a complex proteome. We anticipate that this protocol should find broad applications in redox biology, chemical biology and the pharmaceutical industry. The protocol for sample preparation takes 3 d, whereas MS measurements and data analyses require 75 min and <30 min, respectively, per sample.

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

Fig. 1: Methodologies for indirect profiling of redox and/or electrophile reactive cysteines.
Fig. 2: Overview of QTRP.
Fig. 3: Display of LC-MS/MS data from a representative .raw file.
Fig. 4: Representative MS1-based quantification results of typical QTRP experiments.

Similar content being viewed by others

Data availability

The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE71 partner repository with the dataset identifier PXD016048. Software availability: post-processing algorithm (https://github.com/morpheusliu/Post-processing-program-for-pFind3-results).

References

  1. Paulsen, C. E. & Carroll, K. S. Cysteine-mediated redox signaling: chemistry, biology, and tools for discovery. Chem. Rev. 113, 4633–4679 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Chouchani, E. T. et al. Mitochondrial ROS regulate thermogenic energy expenditure and sulfenylation of UCP1. Nature 532, 112–116 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. O’Neill, J. S. & Reddy, A. B. Circadian clocks in human red blood cells. Nature 469, 498–503 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Edgar, R. S. et al. Peroxiredoxins are conserved markers of circadian rhythms. Nature 485, 459–464 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Yoo, S. K., Starnes, T. W., Deng, Q. & Huttenlocher, A. Lyn is a redox sensor that mediates leukocyte wound attraction in vivo. Nature 480, 109–112 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ruppersberg, J. P. et al. Regulation of fast inactivation of cloned mammalian IK(A) channels by cysteine oxidation. Nature 352, 711–714 (1991).

    Article  CAS  PubMed  Google Scholar 

  7. Guo, Z., Kozlov, S., Lavin, M. F., Person, M. D. & Paull, T. T. ATM activation by oxidative stress. Science 330, 517–521 (2010).

    Article  CAS  PubMed  Google Scholar 

  8. Rudolph, T. K. & Freeman, B. A. Transduction of redox signaling by electrophile-protein reactions. Sci. Signal. 2, re7 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Qin, W. et al. S-glycosylation-based cysteine profiling reveals regulation of glycolysis by itaconate. Nat. Chem. Biol. 15, 983–991 (2019).

    Article  CAS  PubMed  Google Scholar 

  10. Kulkarni, R. A. et al. A chemoproteomic portrait of the oncometabolite fumarate. Nat. Chem. Biol. 15, 391–400 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Gersch, M., Kreuzer, J. & Sieber, S. A. Electrophilic natural products and their biological targets. Nat. Prod. Rep. 29, 659–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  12. Singh, J., Petter, R. C., Baillie, T. A. & Whitty, A. The resurgence of covalent drugs. Nat. Rev. Drug Discov. 10, 307–317 (2011).

    Article  CAS  PubMed  Google Scholar 

  13. Yang, J., Carroll, K. S. & Liebler, D. C. The expanding landscape of the thiol redox proteome. Mol. Cell. Proteom. 15, 1–11 (2016).

    Article  Google Scholar 

  14. Lee, S. R., Kwon, K. S., Kim, S. R. & Rhee, S. G. Reversible inactivation of protein-tyrosine phosphatase 1B in A431 cells stimulated with epidermal growth factor. J. Biol. Chem. 273, 15366–15372 (1998).

    Article  CAS  PubMed  Google Scholar 

  15. Shiio, Y. & Aebersold, R. Quantitative proteome analysis using isotope-coded affinity tags and mass spectrometry. Nat. Protoc. 1, 139–145 (2006).

    Article  CAS  PubMed  Google Scholar 

  16. Thompson, A. et al. Tandem mass tags: a novel quantification strategy for comparative analysis of complex protein mixtures by MS/MS. Anal. Chem. 75, 1895–1904 (2003).

    Article  CAS  PubMed  Google Scholar 

  17. Vajrychova, M. et al. Quantification of cellular protein and redox imbalance using SILAC-iodoTMT methodology. Redox Biol. 24, 101227 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Sethuraman, M., McComb, M. E., Heibeck, T., Costello, C. E. & Cohen, R. A. Isotope-coded affinity tag approach to identify and quantify oxidant-sensitive protein thiols. Mol. Cell. Proteom. 3, 273–278 (2004).

    Article  CAS  Google Scholar 

  19. Fu, C. et al. Quantitative analysis of redox-sensitive proteome with DIGE and ICAT. J. Proteome Res. 7, 3789–3802 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Leichert, L. I. et al. Quantifying changes in the thiol redox proteome upon oxidative stress in vivo. Proc. Natl Acad. Sci. USA 105, 8197–8202 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Topf, U. et al. Quantitative proteomics identifies redox switches for global translation modulation by mitochondrially produced reactive oxygen species. Nat. Commun. 9, 324 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Weerapana, E. et al. Quantitative reactivity profiling predicts functional cysteines in proteomes. Nature 468, 790–795 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Weerapana, E., Speers, A. E. & Cravatt, B. F. Tandem orthogonal proteolysis-activity-based protein profiling (TOP-ABPP)—a general method for mapping sites of probe modification in proteomes. Nat. Protoc. 2, 1414–1425 (2007).

    Article  CAS  PubMed  Google Scholar 

  24. Backus, K. M. et al. Proteome-wide covalent ligand discovery in native biological systems. Nature 534, 570–574 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bar-Peled, L. et al. Chemical proteomics identifies druggable vulnerabilities in a genetically defined cancer. Cell 171, 696–709.e23 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. van Esbroeck, A. C. M. et al. Activity-based protein profiling reveals off-target proteins of the FAAH inhibitor BIA 10-2474. Science 356, 1084–1087 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Wang, C., Weerapana, E., Blewett, M. M. & Cravatt, B. F. A chemoproteomic platform to quantitatively map targets of lipid-derived electrophiles. Nat. Methods 11, 79–85 (2014).

    Article  PubMed  Google Scholar 

  28. Deng, X. et al. Proteome-wide quantification and characterization of oxidation-sensitive cysteines in pathogenic bacteria. Cell Host Microbe 13, 358–370 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Medina-Cleghorn, D. et al. Mapping proteome-wide targets of environmental chemicals using reactivity-based chemoproteomic platforms. Chem. Biol. 22, 1394–1405 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Huang, Z. et al. Global portrait of protein targets of metabolites of the neurotoxic compound BIA 10-2474. ACS Chem. Biol. 14, 192–197 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Maurais, A. J. & Weerapana, E. Reactive-cysteine profiling for drug discovery. Curr. Opin. Chem. Biol. 50, 29–36 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Senkane, K. et al. The proteome-wide potential for reversible covalency at cysteine. Angew. Chem. Int. Ed. Engl. 58, 11385–11389 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Fu, L. et al. Systematic and quantitative assessment of hydrogen peroxide reactivity with cysteines across human proteomes. Mol. Cell. Proteom. 16, 1815–1828 (2017).

    Article  CAS  Google Scholar 

  34. Guo, J. et al. Resin-assisted enrichment of thiols as a general strategy for proteomic profiling of cysteine-based reversible modifications. Nat. Protoc. 9, 64–75 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Garcia-Santamarina, S. et al. Monitoring in vivo reversible cysteine oxidation in proteins using ICAT and mass spectrometry. Nat. Protoc. 9, 1131–1145 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Xiao, H. et al. A quantitative tissue-specific landscape of protein redox regulation during aging. Cell 180, 968–983.e24 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Paulsen, C. E. et al. Peroxide-dependent sulfenylation of the EGFR catalytic site enhances kinase activity. Nat. Chem. Biol. 8, 57–64 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Gupta, V., Yang, J., Liebler, D. C. & Carroll, K. S. Diverse redoxome reactivity profiles of carbon nucleophiles. J. Am. Chem. Soc. 139, 5588–5595 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Akter, S. et al. Chemical proteomics reveals new targets of cysteine sulfinic acid reductase. Nat. Chem. Biol. 14, 995–1004 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Yang, J., Gupta, V., Carroll, K. S. & Liebler, D. C. Site-specific mapping and quantification of protein S-sulphenylation in cells. Nat. Commun. 5, 4776 (2014).

    Article  CAS  PubMed  Google Scholar 

  41. Yang, J. et al. Global, in situ, site-specific analysis of protein S-sulfenylation. Nat. Protoc. 10, 1022–1037 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Huang, J. et al. Mining for protein S-sulfenylation in Arabidopsis uncovers redox-sensitive sites. Proc. Natl Acad. Sci. USA 116, 21256–21261 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Fu, L., Liu, K., Ferreira, R. B., Carroll, K. S. & Yang, J. Proteome-wide analysis of cysteine S-sulfenylation using a benzothiazine-based probe. Curr. Protoc. Protein Sci. 95, e76 (2019).

    Article  PubMed  Google Scholar 

  44. Fu, L. et al. Direct proteomic mapping of cysteine persulfidation. Antioxid. Redox Signal. Available at https://doi.org/10.1089/ars.2019.7777 (2019).

  45. Petrova, B. et al. Dynamic redox balance directs the oocyte-to-embryo transition via developmentally controlled reactive cysteine changes. Proc. Natl Acad. Sci. USA 115, E7978–E7986 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Tian, C. et al. Multiplexed thiol reactivity profiling for target discovery of electrophilic natural products. Cell Chem. Biol. 24, 1416–1427.e15 (2017).

    Article  CAS  PubMed  Google Scholar 

  47. Wang, W. et al. An Arabidopsis secondary metabolite directly targets expression of the bacterial type III secretion system to inhibit bacterial virulence. Cell Host Microbe 27, 601–613 (2020).

    Article  CAS  PubMed  Google Scholar 

  48. Sun, R. et al. A chemoproteomic platform to assess bioactivation potential of drugs. Chem. Res. Toxicol. 30, 1797–1803 (2017).

    Article  CAS  PubMed  Google Scholar 

  49. Pei, J.-F. et al. Diurnal oscillations of endogenous H2O2 sustained by p66Shc regulate circadian clocks. Nat. Cell Biol. 21, 1553–1564 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Martell, J. et al. Global cysteine-reactivity profiling during impaired insulin/IGF-1 signaling in C. elegans identifies uncharacterized mediators of longevity. Cell Chem. Biol. 23, 955–966 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chen, X. et al. Discovery of heteroaromatic sulfones as a new class of biologically compatible thiol-selective reagents. ACS Chem. Biol. 12, 2201–2208 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Reisz, J. A., Bechtold, E., King, S. B., Poole, L. B. & Furdui, C. M. Thiol-blocking electrophiles interfere with labeling and detection of protein sulfenic acids. FEBS J. 280, 6150–6161 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Cuevasanta, E. et al. Reaction of hydrogen sulfide with disulfide and sulfenic acid to form the strongly nucleophilic persulfide. J. Biol. Chem. 290, 26866–26880 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Yang, J. Sweetly profiling the cysteinome. Nat. Chem. Biol. 15, 935–936 (2019).

    Article  CAS  PubMed  Google Scholar 

  55. Yang, J., Tallman, K. A., Porter, N. A. & Liebler, D. C. Quantitative chemoproteomics for site-specific analysis of protein alkylation by 4-hydroxy-2-nonenal in cells. Anal. Chem. 87, 2535–2541 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Zhang, B. et al. Proteogenomic characterization of human colon and rectal cancer. Nature 513, 382–387 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Chambers, M. C. et al. A cross-platform toolkit for mass spectrometry and proteomics. Nat. Biotechnol. 30, 918–920 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Kim, S. & Pevzner, P. A. MS-GF+ makes progress towards a universal database search tool for proteomics. Nat. Commun. 5, 5277 (2014).

    Article  CAS  PubMed  Google Scholar 

  59. Ma, Z. Q. et al. IDPicker 2.0: improved protein assembly with high discrimination peptide identification filtering. J. Proteome Res. 8, 3872–3881 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Pino, L. K. et al. The Skyline ecosystem: informatics for quantitative mass spectrometry proteomics. Mass Spectrom. Rev. 39, 229–244 (2017).

    Article  PubMed  Google Scholar 

  61. Chi, H. et al. Comprehensive identification of peptides in tandem mass spectra using an efficient open search engine. Nat. Biotechnol. 36, 1059–1061 (2018).

    Article  CAS  Google Scholar 

  62. Liu, C. et al. pQuant improves quantitation by keeping out interfering signals and evaluating the accuracy of calculated ratios. Anal. Chem. 86, 5286–5294 (2014).

    Article  CAS  PubMed  Google Scholar 

  63. Tyanova, S., Temu, T. & Cox, J. The MaxQuant computational platform for mass spectrometry-based shotgun proteomics. Nat. Protoc. 11, 2301–2319 (2016).

    Article  CAS  PubMed  Google Scholar 

  64. Tran, N. H. et al. Deep learning enables de novo peptide sequencing from data-independent-acquisition mass spectrometry. Nat. Methods 16, 63–66 (2019).

    Article  PubMed  Google Scholar 

  65. Luo, Q., Tao, Y., Sheng, W., Lu, J. & Wang, H. Dinitroimidazoles as bifunctional bioconjugation reagents for protein functionalization and peptide macrocyclization. Nat. Commun. 10, 142 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Yu, J., Yang, X., Sun, Y. & Yin, Z. Highly reactive and tracelessly cleavable cysteine-specific modification of proteins via 4-substituted cyclopentenone. Angew. Chem. Int. Ed. Engl. 57, 11598–11602 (2018).

    Article  CAS  PubMed  Google Scholar 

  67. Abegg, D. et al. Proteome-wide profiling of targets of cysteine reactive small molecules by using ethynyl benziodoxolone reagents. Angew. Chem. Int. Ed. Engl. 54, 10852–10857 (2015).

    Article  CAS  PubMed  Google Scholar 

  68. Uttamapinant, C., Sanchez, M. I., Liu, D. S., Yao, J. Z. & Ting, A. Y. Site-specific protein labeling using PRIME and chelation-assisted click chemistry. Nat. Protoc. 8, 1620–1634 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Rappsilber, J., Mann, M. & Ishihama, Y. Protocol for micro-purification, enrichment, pre-fractionation and storage of peptides for proteomics using StageTips. Nat. Protoc. 2, 1896–1906 (2007).

    Article  CAS  PubMed  Google Scholar 

  70. Chick, J. M. et al. A mass-tolerant database search identifies a large proportion of unassigned spectra in shotgun proteomics as modified peptides. Nat. Biotechnol. 33, 743–749 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Perez-Riverol, Y. et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 47, D442–D450 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Albertolle of The Scripps Research Institute and A. Foti of the Max Planck Institute for Infection Biology Cellular Microbiology for insightful comments. We thank K. S. Carroll of The Scripps Research Institute for kindly providing the S-sulfenic acid specific probe BTD. The work was supported by grants from the National Key R&D Program of China (2016YFA0501303 to J.Y.), the Natural Science Foundation of China (21922702, 81973279 and 31770885 to J.Y. and 31800036 to L.F.) and the State Key Laboratory of Proteomics (SKLP-K201703 and SKLP-K201804 to J.Y.). We also thank B. Fu and B. Zhong from the MS facility of the National Center for Protein Sciences • Beijing for their help and technical support.

Author information

Authors and Affiliations

Authors

Contributions

L.F. and J.Y. developed the QTRP protocol and designed the studies. L.F., Z.L. C.T., Jixiang He and Jingyang He performed the experiments and analyzed the data. K.L. developed the post-processing algorithm. F.H., P.X. and J.Y. supervised the project. J.Y. wrote the manuscript.

Corresponding author

Correspondence to Jing Yang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Protocols thanks Wei-Jun Qian, Johannes Herrmann and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

Key references using this protocol

Fu, L. et al. Mol. Cell. Proteomics 16, 1815–1828 (2017): https://www.mcponline.org/content/16/10/1815.long

Sun, R. et al. Chem. Res. Toxicol. 30, 1797–1803 (2017): https://pubs.acs.org/doi/10.1021/acs.chemrestox.7b00183

Petrova, B. et al. Proc. Natl Acad. Sci. USA 115, E7978–E7986 (2018): http://www.pnas.org/content/115/34/E7978

Pei, J.-F. et al. Nat. Cell Biol. 21, 1553–1564 (2019): https://www.nature.com/articles/s41556-019-0420-4

Wang, W. et al. Cell Host Microbe 27, 601–613 (2020): https://www.sciencedirect.com/science/article/abs/pii/S1931312820301682

Supplementary information

Supplementary Information

Supplementary Figs. 1–13.

Reporting Summary

Supplementary Data 1

Proteome-wide mapping of oxidation-sensitive cysteines in HEK293T cells by QTRP. HEK293T cell lysates stimulated with or without H2O2 (1 mM, 15 min, 37 °C) and labeled with 100 mM IPM. The IPM-labeled proteome was then processed into tryptic peptides. The resulting probe-labeled peptides were conjugated with both L and H azido-UV-cleavable-biotin (Az-UV-biotin) reagents (1:1) via CuAAC. The L and H ‘Click’ reaction mixtures were cleaned with or without SCX. The biotinylated peptides were captured with streptavidin and photoreleased for LC-MS/MS–based identification and quantification. High RH/L values are indicative of cysteines with less free thiol being available after H2O2 treatment, suggesting potential redox-sensitive targets. A schematic workflow is shown in Fig. 2a.

Supplementary Data 2

Quantitative profiling of the intrinsic reactivity of proteomic cysteines. HEK293T proteomes were labeled with low (10 µM) and high (100 µM) levels of IPM. The probe-labeled proteins were processed into tryptic peptides. The resulting probe-labeled peptides were conjugated with both L and H azido-UV-cleavable-biotin (Az-UV-biotin) reagents (1:1) via CuAAC. The light and heavy ‘Click’ reaction mixtures were cleaned with or without SCX. The biotinylated peptides were captured with streptavidin and photoreleased for LC-MS/MS–based identification and quantification. Those with a calculated ratio of H to L (100 µM versus 10 µM, R10:1) <2.0 were defined as hyperreactive. A schematic workflow is shown in Fig. 2b.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fu, L., Li, Z., Liu, K. et al. A quantitative thiol reactivity profiling platform to analyze redox and electrophile reactive cysteine proteomes. Nat Protoc 15, 2891–2919 (2020). https://doi.org/10.1038/s41596-020-0352-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-020-0352-2

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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