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:

T-REX on-demand redox targeting in live cells

This article has been updated

Abstract

This protocol describes targetable reactive electrophiles and oxidants (T-REX)—a live-cell-based tool designed to (i) interrogate the consequences of specific and time-resolved redox events, and (ii) screen for bona fide redox-sensor targets. A small-molecule toolset comprising photocaged precursors to specific reactive redox signals is constructed such that these inert precursors specifically and irreversibly tag any HaloTag-fused protein of interest (POI) in mammalian and Escherichia coli cells. Syntheses of the alkyne-functionalized endogenous reactive signal 4-hydroxynonenal (HNE(alkyne)) and the HaloTag-targetable photocaged precursor to HNE(alkyne) (also known as Ht-PreHNE or HtPHA) are described. Low-energy light prompts photo-uncaging (t1/2 <1–2 min) and target-specific modification. The targeted modification of the POI enables precisely timed and spatially controlled redox events with no off-target modification. Two independent pathways are described, along with a simple setup to functionally validate known targets or discover novel sensors. T-REX sidesteps mixed responses caused by uncontrolled whole-cell swamping with reactive signals. Modification and downstream response can be analyzed by in-gel fluorescence, proteomics, qRT-PCR, immunofluorescence, fluorescence resonance energy transfer (FRET)-based and dual-luciferase reporters, or flow cytometry assays. T-REX targeting takes 4 h from initial probe treatment. Analysis of targeted redox responses takes an additional 4–24 h, depending on the nature of the pathway and the type of readouts used.

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: Strategies for studying cellular redox responses.
Figure 2: On-target, on-demand redox signaling enabled by T-REX.
Figure 3: T-REX approach allows flexibility while enabling quantification of modification and response at numerous points.
Figure 4: Commercial HaloTag library allows discovery and validation of 'first responders' to a specific LDE using T-REX.
Figure 5: Assessment of N- versus C-terminal HaloTagging on T-REX functionality, exemplified by Keap1 LDE targeting.
Figure 6: T-REX targeting is equally efficient in human (HEK-293) cells and E. coli.
Figure 7: Flow-cytometry-based ARE-GFP reporter assay quantifying T-REX-mediated activation of antioxidant response in a subpopulation of live HEK-293 cells.
Figure 8: Immunofluorescence analysis of endogenous PIP3 phosphoinositide in fixed cells subsequent to PTEN-targeted redox modification enabled by T-REX in live cells.
Figure 9: FRET-based biosensor assay in live cells, reporting the levels of endogenous PIP3 subsequent to PTEN-targeted redox modification enabled by T-REX.
Figure 10

Similar content being viewed by others

Change history

  • 10 November 2016

    In the version of this article initially published online, there were three errors. (1) In the Reagent Setup, the instructions for preparing LB-ampicillin-chloramphenicol medium gave an incorrect concentration for ampicillin; it should read "100 μg/ml ampicillin." (2) The text for Step 11B(vi) incorrectly described the bromination procedure; it should read: "Brominate 0.35 g of the resulting alcohol 6 (1.4 mmol) in 20 ml of distilled DCM at 4 °C by adding 0.5 g of CBr4 (1.54 mmol) and 0.44 g of PPh3 (1.68 mmol). Stir the mixture for 15 min" (instead of "...by adding 0.5 g of CBr4 (1.54 mmol) and 0.44 g of PPh3 (1.68 mmol) to 20 ml of distilled DCM at 0 °C"). (3) In Figure 10, under the arrow between compounds 12 and 13, the label "(ii) Me2S" should be omitted. These errors have been corrected for all versions of the article.

References

  1. Jacob, C. & Winyard\, P.G. Redox Signaling and Regulation in Biology and Medicine (Wiley-VCH, 2009).

  2. Stadtman, E.R. Protein oxidation and aging. Science 257, 1220–1224 (1992).

    Article  CAS  PubMed  Google Scholar 

  3. Schopfer, F.J., Cipollina, C. & Freeman, B.A. Formation and signaling actions of electrophilic lipids. Chem. Rev. 111, 5997–6021 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Winterbourn, C.C. Reconciling the chemistry and biology of reactive oxygen species. Nat. Chem. Biol. 4, 278–286 (2008).

    Article  CAS  PubMed  Google Scholar 

  5. Murphy, M.P. et al. Unraveling the biological roles of reactive oxygen species. Cell Metab. 13, 361–366 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Holmstrom, K.M. & Finkel, T. Cellular mechanisms and physiological consequences of redox-dependent signalling. Nat. Rev. Mol. Cell Biol. 15, 411–421 (2014).

    Article  CAS  PubMed  Google Scholar 

  7. Jacobs, A.T. & Marnett, L.J. Systems analysis of protein modification and cellular responses induced by electrophile stress. Acc. Chem. Res. 43, 673–683 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Delmastro-Greenwood, M., Freeman, B.A. & Wendell, S.G. Redox-dependent anti-inflammatory signaling actions of unsaturated fatty acids. Annu. Rev. Physiol. 76, 79–105 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Crunkhorn, S. Deal watch: Abbott boosts investment in NRF2 activators for reducing oxidative stress. Nat. Rev. Drug Discov. 11, 96 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Dinkova-Kostova, A.T. & Kostov, R.V. Glucosinolates and isothiocyanates in health and disease. Trends Mol. Med. 18, 337–347 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. Gupta, S.C., Patchva, S. & Aggarwal, B.B. Therapeutic roles of curcumin: lessons learned from clinical trials. AAPS J. 15, 195–218 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Fernandez-Fernandez, B., Ortiz, A., Gomez-Guerrero, C. & Egido, J. Therapeutic approaches to diabetic nephropathy--beyond the RAS. Nat. Rev. Nephrol. 10, 325–346 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Bomprezzi, R. Dimethyl fumarate in the treatment of relapsing–remitting multiple sclerosis: an overview. Ther. Adv. Neurol. Disord. 8, 20–30 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hermann, C., Zeiher, A.M. & Dimmeler, S. Shear stress inhibits H2O2-induced apoptosis of human endothelial cells by modulation of the glutathione redox cycle and nitric oxide synthase. Arterioscler Thromb. Vasc. Biol. 17, 3588–3592 (1997).

    Article  CAS  PubMed  Google Scholar 

  15. De Keulenaer, G.W. et al. Oscillatory and steady laminar shear stress differentially affect human endothelial redox state: role of a superoxide-producing NADH oxidase. Circ. Res. 82, 1094–1101 (1998).

    Article  CAS  PubMed  Google Scholar 

  16. Cunningham, K.S. & Gotlieb, A.I. The role of shear stress in the pathogenesis of atherosclerosis. Lab. Invest. 85, 9–23 (2005).

    Article  CAS  PubMed  Google Scholar 

  17. Kim, S.K., Woodcroft, K.J., Oh, S.J., Abdelmegeed, M.A. & Novak, R.F. Role of mechanical and redox stress in activation of mitogen-activated protein kinases in primary cultured rat hepatocytes. Biochem. Pharmacol. 70, 1785–1795 (2005).

    Article  CAS  PubMed  Google Scholar 

  18. Lehoux, S. Redox signalling in vascular responses to shear and stretch. Cardiovasc Res. 71, 269–279 (2006).

    Article  CAS  PubMed  Google Scholar 

  19. Sundaresan, M., Yu, Z.X., Ferrans, V.J., Irani, K. & Finkel, T. Requirement for generation of H2O2 for platelet-derived growth factor signal transduction. Science 270, 296–299 (1995).

    Article  CAS  PubMed  Google Scholar 

  20. Bae, Y.S., Oh, H., Rhee, S.G. & Yoo, Y.D. Regulation of reactive oxygen species generation in cell signaling. Mol. Cells 32, 491–509 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Schieber, M. & Chandel, N.S. ROS function in redox signaling and oxidative stress. Curr. Biol. 24, R453–462 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Brewer, T.F., Garcia, F.J., Onak, C.S., Carroll, K.S. & Chang, C.J. Chemical approaches to discovery and study of sources and targets of hydrogen peroxide redox signaling through NADPH oxidase proteins. Annu. Rev. Biochem. 84, 765–790 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Brownlee, M. Biochemistry and molecular cell biology of diabetic complications. Nature 414, 813–820 (2001).

    Article  CAS  PubMed  Google Scholar 

  24. Green, K., Brand, M.D. & Murphy, M.P. Prevention of mitochondrial oxidative damage as a therapeutic strategy in diabetes. Diabetes 53 (Suppl. 1), S110–S118 (2004).

    Article  CAS  PubMed  Google Scholar 

  25. Nishikawa, T. & Araki, E. Impact of mitochondrial ROS production in the pathogenesis of diabetes mellitus and its complications. Antioxid. Redox Signal. 9, 343–353 (2007).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Codreanu, S.G. et al. Alkylation damage by lipid electrophiles targets functional protein systems. Mol. Cell. Proteomics 13, 849–859 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. 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  CAS  PubMed  Google Scholar 

  29. Niphakis, M.J. et al. A global map of lipid-binding proteins and their ligandability in cells. Cell 161, 1668–1680 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. 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 

  31. Kim, H.Y., Tallman, K.A., Liebler, D.C. & Porter, N.A. An azido-biotin reagent for use in the isolation of protein adducts of lipid-derived electrophiles by streptavidin catch and photorelease. Mol. Cell Proteomics 8, 2080–2089 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Furdui, C.M. & Poole, L.B. Chemical approaches to detect and analyze protein sulfenic acids. Mass Spectrom. Rev. 33, 126–146 (2014).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  34. Codreanu, S.G., Zhang, B., Sobecki, S.M., Billheimer, D.D. & Liebler, D.C. Global analysis of protein damage by the lipid electrophile 4-hydroxy-2-nonenal. Mol. Cell Proteomics 8, 670–680 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Rossi, A. et al. Anti-inflammatory cyclopentenone prostaglandins are direct inhibitors of IkappaB kinase. Nature 403, 103–108 (2000).

    Article  CAS  PubMed  Google Scholar 

  36. Straus, D.S. et al. 15-deoxy-delta 12,14-prostaglandin J2 inhibits multiple steps in the NF-kappa B signaling pathway. Proc. Natl. Acad. Sci. USA 97, 4844–4849 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ji, C., Kozak, K.R. & Marnett, L.J. IkappaB kinase, a molecular target for inhibition by 4-hydroxy-2-nonenal. J. Biol. Chem. 276, 18223–18228 (2001).

    Article  CAS  PubMed  Google Scholar 

  38. 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 

  39. Diab, A. et al. Peroxisome proliferator-activated receptor-gamma agonist 15-deoxy-Delta(12,14)-prostaglandin J(2) ameliorates experimental autoimmune encephalomyelitis. J. Immunol. 168, 2508–2515 (2002).

    Article  CAS  PubMed  Google Scholar 

  40. Uchida, K. Redox-derived damage-associated molecular patterns: ligand function of lipid peroxidation adducts. Redox Biol. 1, 94–96 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Cubillos-Ruiz, J.R. et al. ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis. Cell 161, 1527–1538 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Doyle, K. & Fitzpatrick, F.A. Redox signaling, alkylation (carbonylation) of conserved cysteines inactivates class I histone deacetylases 1, 2, and 3 and antagonizes their transcriptional repressor function. J. Biol. Chem. 285, 17417–17424 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Fritz, K.S. et al. 4-Hydroxynonenal inhibits SIRT3 via thiol-specific modification. Chem. Res. Toxicol. 24, 651–662 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Galligan, J.J. et al. Stable histone adduction by 4-oxo-2-nonenal: a potential link between oxidative stress and epigenetics. J. Am. Chem. Soc. 136, 11864–11866 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kondo, M. et al. 15-Deoxy-Delta(12,14)-prostaglandin J(2): the endogenous electrophile that induces neuronal apoptosis. Proc. Natl. Acad. Sci. USA 99, 7367–7372 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Jacobs, A.T. & Marnett, L.J. HSF1-mediated BAG3 expression attenuates apoptosis in 4-hydroxynonenal-treated colon cancer cells via stabilization of anti-apoptotic Bcl-2 proteins. J. Biol. Chem. 284, 9176–9183 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Chipuk, J.E. et al. Sphingolipid metabolism cooperates with BAK and BAX to promote the mitochondrial pathway of apoptosis. Cell 148, 988–1000 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Droge, W. Free radicals in the physiological control of cell function. Physiol. Rev. 82, 47–95 (2002).

    Article  CAS  PubMed  Google Scholar 

  49. Foyer, C., Faragher, R. & Thornalley, P. (eds.) Redox Metabolism and Longevity Relationships in Animals and Plants. (Taylor & Francis, 2009).

  50. Stadtman, E.R. & Berlett, B.S. Reactive oxygen-mediated protein oxidation in aging and disease. Chem. Res. Toxicol. 10, 485–494 (1997).

    Article  CAS  PubMed  Google Scholar 

  51. Dalleau, S., Baradat, M., Gueraud, F. & Huc, L. Cell death and diseases related to oxidative stress: 4-hydroxynonenal (HNE) in the balance. Cell Death Differ. 20, 1615–1630 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Levine, R.L. Carbonyl modified proteins in cellular regulation, aging, and disease. Free Radic. Biol. Med. 32, 790–796 (2002).

    Article  CAS  PubMed  Google Scholar 

  53. Dalle-Donne, I., Rossi, R., Colombo, R., Giustarini, D. & Milzani, A. Biomarkers of oxidative damage in human disease. Clin. Chem. 52, 601–623 (2006).

    Article  CAS  PubMed  Google Scholar 

  54. 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 

  55. Fang, X. et al. Temporally controlled targeting of 4-hydroxynonenal to specific proteins in living cells. J. Am. Chem. Soc. 135, 14496–14499 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Lin, H.Y., Haegele, J.A., Disare, M.T., Lin, Q. & Aye, Y. A generalizable platform for interrogating target- and signal-specific consequences of electrophilic modifications in redox-dependent cell signaling. J. Am. Chem. Soc. 137, 6232–6244 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Parvez, S. et al. Substoichiometric hydroxynonenylation of a single protein recapitulates whole-cell-stimulated antioxidant response. J. Am. Chem. Soc. 137, 10–13 (2015).

    Article  CAS  PubMed  Google Scholar 

  58. Janssen, D.B. Evolving haloalkane dehalogenases. Curr. Opin. Chem. Biol. 8, 150–159 (2004).

    Article  CAS  PubMed  Google Scholar 

  59. Los, G.V. & Wood, K. A novel technology for cell imaging and protein analysis. Methods Mol. Biol. 356, 195–208 (2007).

    CAS  PubMed  Google Scholar 

  60. Los, G.V. et al. HaloTag: a novel protein labeling technology for cell imaging and protein analysis. ACS Chem. Biol. 3, 373–382 (2008).

    Article  CAS  PubMed  Google Scholar 

  61. Ohana, R.F. et al. HaloTag7: a genetically engineered tag that enhances bacterial expression of soluble proteins and improves protein purification. Protein Expr. Purif. 68, 110–120 (2009).

    Article  CAS  PubMed  Google Scholar 

  62. Long, M.J.C., Poganik, J.R. & Aye, Y. On-demand targeting: investigating biology with proximity-directed chemistry. J. Am. Chem. Soc. 138, 3610–3622 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Rogakou, E.P., Pilch, D.R., Orr, A.H., Ivanova, V.S. & Bonner, W.M. DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J. Biol. Chem. 273, 5858–5868 (1998).

    Article  CAS  PubMed  Google Scholar 

  64. Morgan, M.J. & Liu, Z.G. Crosstalk of reactive oxygen species and NF-kappaB signaling. Cell Res. 21, 103–115 (2011).

    Article  CAS  PubMed  Google Scholar 

  65. Tjalkens, R.B., Cook, L.W. & Petersen, D.R. Formation and export of the glutathione conjugate of 4-hydroxy-2, 3-E-nonenal (4-HNE) in hepatoma cells. Arch. Biochem. Biophys. 361, 113–119 (1999).

    Article  CAS  PubMed  Google Scholar 

  66. Volkel, W. et al. Glutathione conjugates of 4-hydroxy-2(E)-nonenal as biomarkers of hepatic oxidative stress-induced lipid peroxidation in rats. Free Radic. Biol. Med. 38, 1526–1536 (2005).

    Article  CAS  PubMed  Google Scholar 

  67. Banerjee, R. Redox Biochemistry (Wiley, 2007).

  68. Cao, Z., Hardej, D., Trombetta, L.D. & Li, Y. The role of chemically induced glutathione and glutathione S-transferase in protecting against 4-hydroxy-2-nonenal-mediated cytotoxicity in vascular smooth muscle cells. Cardiovasc. Toxicol. 3, 165–177 (2003).

    Article  CAS  PubMed  Google Scholar 

  69. Kolb, H.C., Finn, M.G. & Sharpless, K.B. Click chemistry: diverse chemical function from a few good reactions. Angew. Chem. Int. Ed. Engl. 40, 2004–2021 (2001).

    Article  CAS  PubMed  Google Scholar 

  70. Aye, Y., Li, M., Long, M.J.C. & Weiss, R.S. Ribonucleotide reductase and cancer: biological mechanisms and targeted therapies. Oncogene 34, 2011–2021 (2015).

    Article  CAS  PubMed  Google Scholar 

  71. Hayes, J.D. & Dinkova-Kostova, A.T. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem. Sci. 39, 199–218 (2014).

    Article  CAS  PubMed  Google Scholar 

  72. Kobayashi, A. et al. Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Mol. Cell Biol. 24, 7130–7139 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Krief, S. et al. Identification and characterization of cvHsp: a novel human small stress protein selectively expressed in cardiovascular and insulin-sensitive tissues. J. Biol. Chem. 274, 36592–36600 (1999).

    Article  CAS  PubMed  Google Scholar 

  74. Rosenfeld, G.E., Mercer, E.J., Mason, C.E. & Evans, T. Small heat shock proteins Hspb7 and Hspb12 regulate early steps of cardiac morphogenesis. Dev. Biol. 381, 389–400 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Marvin, M. et al. Developmental expression patterns of the zebrafish small heat shock proteins. Dev. Dyn. 237, 454–463 (2008).

    Article  CAS  PubMed  Google Scholar 

  76. Ma, Q. Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol. 53, 401–426 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Sporn, M.B. & Liby, K.T. NRF2 and cancer: the good, the bad and the importance of context. Nat. Rev. Cancer 12, 564–571 (2012).

    Article  CAS  PubMed  Google Scholar 

  78. Garber, K. Biochemistry: a radical treatment. Nature 489, S4–S6 (2012).

    Article  CAS  PubMed  Google Scholar 

  79. Uchida, K. & Stadtman, E.R. Modification of histidine residues in proteins by reaction with 4-hydroxynonenal. Proc. Natl. Acad. Sci. USA 89, 4544–4548 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Uchida, K. & Stadtman, E.R. Covalent attachment of 4-hydroxynonenal to glyceraldehyde-3-phosphate dehydrogenase. A possible involvement of intra- and intermolecular cross-linking reaction. J. Biol. Chem. 268, 6388–6393 (1993).

    CAS  PubMed  Google Scholar 

  81. Nadkarni, D.V. & Sayre, L.M. Structural definition of early lysine and histidine adduction chemistry of 4-hydroxynonenal. Chem. Res. Toxicol. 8, 284–291 (1995).

    Article  CAS  PubMed  Google Scholar 

  82. Papa, A. et al. Cancer-associated PTEN mutants act in a dominant-negative manner to suppress PTEN protein function. Cell 157, 595–610 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Covey, T.M., Edes, K., Coombs, G.S., Virshup, D.M. & Fitzpatrick, F.A. Alkylation of the tumor suppressor PTEN activates Akt and β-catenin signaling: a mechanism linking inflammation and oxidative stress with cancer. PLoS One 5, e13545 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Shearn, C.T. et al. Increased carbonylation of the lipid phosphatase PTEN contributes to Akt2 activation in a murine model of early alcohol-induced steatosis. Free Radic. Biol. Med. 65, 680–692 (2013).

    Article  CAS  PubMed  Google Scholar 

  85. Shearn, C.T. et al. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) inhibition by 4-hydroxynonenal leads to increased Akt activation in hepatocytes. Mol. Pharmacol. 79, 941–952 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Leslie, N.R. & Foti, M. Non-genomic loss of PTEN function in cancer: not in my genes. Trends Pharmacol. Sci. 32, 131–140 (2011).

    Article  CAS  PubMed  Google Scholar 

  87. Trotman, L.C. et al. Pten dose dictates cancer progression in the prostate. PLoS Biol. 1, E59 (2003).

    Article  PubMed  PubMed Central  Google Scholar 

  88. Covey, T.M., Edes, K. & Fitzpatrick, F.A. Akt activation by arachidonic acid metabolism occurs via oxidation and inactivation of PTEN tumor suppressor. Oncogene 26, 5784–5792 (2007).

    Article  CAS  PubMed  Google Scholar 

  89. Ananthanarayanan, B., Ni, Q. & Zhang, J. Signal propagation from membrane messengers to nuclear effectors revealed by reporters of phosphoinositide dynamics and Akt activity. Proc. Natl. Acad. Sci. USA 102, 15081–15086 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Antal, C.E. & Newton, A.C. Spatiotemporal dynamics of phosphorylation in lipid second messenger signaling. Mol. Cell Proteomics 12, 3498–3508 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Doudna, J.A. & Charpentier, E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346, 1258096 (2014).

    Article  CAS  PubMed  Google Scholar 

  92. Fellmann, C.L. & Scott, W. Stable RNA interference rules for silencing. Nat. Cell Biol. 16, 10–18 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Assou, S. et al. Dynamic changes in gene expression during human early embryo development: from fundamental aspects to clinical applications. Hum. Reprod. Update 17, 272–290 (2011).

    Article  CAS  PubMed  Google Scholar 

  94. García-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 

  95. Lo Conte, M., Lin, J., Wilson, M.A. & Carroll, K.S. A chemical approach for the detection of protein sulfinylation. ACS Chem. Biol. 10, 1825–1830 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. 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 

  97. Gautier, A. et al. An engineered protein tag for multiprotein labeling in living cells. Chem. Biol. 15, 128–136 (2008).

    Article  CAS  PubMed  Google Scholar 

  98. Grune, T., Siems, W.G., Zollner, H. & Esterbauer, H. Metabolism of 4-hydroxynonenal, a cytotoxic lipid peroxidation product, in Ehrlich mouse ascites cells at different proliferation stages. Cancer Res. 54, 5231–5235 (1994).

    CAS  PubMed  Google Scholar 

  99. Srivastava, S. et al. Metabolism of the lipid peroxidation product, 4-hydroxy-trans-2-nonenal, in isolated perfused rat heart. J. Biol. Chem. 273, 10893–10900 (1998).

    Article  CAS  PubMed  Google Scholar 

  100. Blankespoor, R.L. et al. Photochemistry of 1-alkoxy- and 1-(benzyloxy)-9,10-anthraquinones in methanol: a facile process for the preparation of aldehydes and ketones. J. Org. Chem. 60, 6852–6859 (1995).

    Article  CAS  Google Scholar 

  101. Wood, K.V. Luciferase Assay Method. US patent 5,283,179 (1994).

  102. Sherf, B.A., Wood, K.V. & Schenborn, E.T. Quenching Reagents and Assays for Enzyme-mediated Luminescence. US patent 5,744,320 (1998).

  103. Grune, T. & Davies, K.J.A. The proteasomal system and HNE-modified proteins. Mol. Aspects Med. 24, 195–204 (2003).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the laboratories of T. Evans (Weill Cornell Medicine, New York) and J. Zhang (University of California, San Diego) for plasmids encoding zebrafish hspb7 and the lnPAkt (PIP3-reporter) construct, respectively. We acknowledge all of the Aye Laboratory members who have contributed to T-REX redox targeting protocols, particularly J. Li, X. Fang and Y. Fu, as well as Q. Lin of the State University of New York at Albany for assistance with the LC–MS/MS analysis of modifications on the protein pulled down from cells. Funding was provided by the NIH Director's New Innovator award (1DP2GM114850), the National Science Foundation (NSF) CAREER award (CHE-1351400), the Beckman Young Investigator award and the Sloan Research Fellowship (to Y.A.) and the Burroughs Wellcome Funds CTRG (to principal investigator (PI) Y.A. and host T. Evans). S.P. is a Howard Hughes Medical Institute international predoctoral fellow (59108350). J.A.H. acknowledges the CBI training grant (T32GM008500, PI – H. Lin). V.N.P. thanks the Douglas family for an undergraduate research fellowship. D.K.L. thanks the Cornell University P3 scholars program. Imaging and flow cytometry data were acquired at the Cornell University Biotechnology Resource Center (NIH 1S10RR025502) and the Cornell University cytometry core (supported in part by the Empire State Stem Cell Fund), respectively. We acknowledge the NSF (NSF MRI: CHE-1531632 to Y.A. (PI)) for NMR instrumentation support at Cornell University.

Author information

Authors and Affiliations

Authors

Contributions

H.-Y.L. and Y.Z. were joint second authors of this work. S.P., M.J.C.L., H.-Y.L., Y.Z., J.A.H., V.N.P., D.K.L. and Y.A. developed protocols. S.P. and V.N.P. contributed to the data associated with T-REX in E. coli cells. M.J.C.L., Y.Z., J.A.H. and D.K.L. obtained the data associated with T-REX in cultured human cells. H.-Y.L. collected LC-MS/MS data. H.-Y.L. and Y.Z. contributed to chemical synthesis. S.P., M.J.C.L. and H.-Y.L. wrote the protocols. Y.A. wrote the manuscript with proofreading/editing contributions from S.P., M.J.C.L., Y.Z. and J.A.H.

Corresponding author

Correspondence to Yimon Aye.

Ethics declarations

Competing interests

Licensing application on this work is being pursued.

Integrated supplementary information

Supplementary Figure 1 Executing T-REX in mammalian cells

HEK-293 cells cultured (a) in 2x 55 cm2 adherent cell culture plates, and (b and c) in a 48-well multi-well adherent cell culture plate. No cover was placed on the plates during photo-uncaging. See Main Text for detailed experimental conditions and equipment specifications. Also see Supplementary Videos 1, 2

Supplementary Figure 2 Evaluation of time-dependent redox signal release in cells in T-REX method and validation that HaloTag does not react with HNE.

(a) Measurements of HNE release efficiency in cells. HEK293T cells expressing HaloTag alone treated under standard T-REX conditions with Ht-PreHNE were exposed to UV light (0.3 mW/cm2, 365 nm) for the indicated time periods at which point the cells were harvested, lysed, and subjected to Click coupling and in-gel fluorescence analysis followed by western blot. Error bars designate SD (N=3). (b) Controls to show that HaloTag does not react with HNE. Purified recombinant HaloTag was treated with either the photocaged precursor Ht-PreHNE (Figure 2, inset, and Figure 10) (2 equiv., lane a, positive control), or directly with reactive electrophile HNE (Figure 10) (0, 2, 4, 8, 16 equiv., lane b, c, d, e, f, respectively). After 20-min incubation, the samples were analyzed by in-gel fluorescence. M, molecular weight ladder.

Supplementary Figure 3 UV light exposure employed in T-REX is non-invasive: representative data for γ-H2AX1 and NF-κB2, markers for DNA damage and inflammatory signaling, respectively.

(a) HEK293T cells were exposed to UV light (0.3 mW/cm2, 340 nm) for the indicated time periods. Mitomycin C (10 μg/ml for 24 h)3 and aphidicolin (10 μg/ml for 36 h)4 serve as positive controls. After 12 hours post the end of UV illumination, cells were fixed and analyzed by standard immunofluorescence imaging method using γ-H2AX antibody (Millipore 05-636 at 1:1000 dilution). Data show mean +/- S.D. N > 50 cells. (b) HEK293T cells stably expressing NRE-inducible firefly luciferase5 were transfected with the respective plasmids encoding indicated transgene (empty vector, HaloTag alone, or Halo-Keap1 and Renilla luciferase) under constitutive CMV promoters. 24 hours post transfection, half of the plates were exposed to UV light (0.3 mW/cm2, 365 nm) over 20 min. Phorbol 12-myristate 13-acetate (PMA) (10 ng/mL, 18 h) was used as a positive control for NRE activation5. NRE activation was measured after 18 hrs. Error bars designate S.D. (N = 8 biological replicates).

Supplementary Figure 4 T-REX screen of Halo ORF clones for the discovery of novel electrophile-sensitive targets and pulldown validation of expressed proteins exemplified by zebrafish HSPB7.

(a) T-REX-enabled gel-based screen for bona fide HNE-sensitive targets using Halo-ORFeome library (Promega). Individual wells in a 48-well plate contained live HEK-293 cells ectopically expressing a unique HaloTagged gene of interest. The cells were subjected to T-REX-HNE(alkyne) targeting on demand. Post cell lysis, all samples were treated with TEV protease and subsequently subjected to Click coupling reaction with Cy5 azide. Probing with Halo antibody allowed evaluation of expression level (and/or solubility under the lysis conditions used). The “hit” bands on Cy5-fluorescent gel were judged against Halo protein level revealed by western blot. For example, RRM1, PRKCD, p53R2, and Keap1 (positive control) had roughly similar expression levels. Only RRM1 and Keap1 were HNE-sensitive although all four targets have been previously identified to be potentially redox/HNE-sensitive6–10. See Main Text for discussion. , a non-specific band. Also see Figure 4 and procedural details in Main Text. (b) Zebrafish HSPB7 expression and protein ID of the band shown in Figure 4a was validated by enrichment from HEK-293 cells ectopically expressing Halo-HSPB7 with the use of HaloTag PEG-Biotin ligand (Promega G8592) and streptavidin sepharose beads (GE Healthcare, cat. no. 17-5113-01), and subsequent on-bead TEV-protease cleavage followed by gel electrophoresis analysis. Theoretical MW of HSPB7 ~ 18 kDa. L, MW ladder.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–4 and Supplementary Tables 1 and 2 (PDF 2343 kb)

Setting up T-REX in cultured mammalian cells:

Caption text: video shows a proper setup for the light illumination of cultured mammalian cells in a 48-well plate. (MP4 35071 kb)

Setting up T-REX in E. coli

Caption text: video shows a proper setup for the light illumination step. (MP4 3685 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Parvez, S., Long, M., Lin, HY. et al. T-REX on-demand redox targeting in live cells. Nat Protoc 11, 2328–2356 (2016). https://doi.org/10.1038/nprot.2016.114

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2016.114

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

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