Skip to main content

Advertisement

Log in

OregonFluor enables quantitative intracellular paired agent imaging to assess drug target availability in live cells and tissues

  • Article
  • Published:

From Nature Chemistry

View current issue Submit your manuscript

A Publisher Correction to this article was published on 28 April 2023

This article has been updated

Abstract

Non-destructive fluorophore diffusion across cell membranes to provide an unbiased fluorescence intensity readout is critical for quantitative imaging applications in live cells and tissues. Commercially available small-molecule fluorophores have been engineered for biological compatibility, imparting high water solubility by modifying rhodamine and cyanine dye scaffolds with multiple sulfonate groups. The resulting net negative charge, however, often renders these fluorophores cell-membrane-impermeant. Here we report the design and development of our biologically compatible, water-soluble and cell-membrane-permeable fluorophores, termed OregonFluor (ORFluor). By adapting previously established ratiometric imaging methodology using bio-affinity agents, it is now possible to use small-molecule ORFluor-labelled therapeutic inhibitors to quantitatively visualize their intracellular distribution and protein target-specific binding, providing a chemical toolkit for quantifying drug target availability in live cells and tissues.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: Design strategies to develop water-soluble and cell-membrane-permeant fluorophores for quantitative, live-cell and tissue imaging.
Fig. 2: ORFluors are a class of zwitterionic, water-soluble, cell-membrane-permeant fluorophores suitable for live-cell imaging.
Fig. 3: Erlotinib iPAI probe design.
Fig. 4: Superior live-cell imaging of drug distribution with ORFluor-labelled iPAI probes.
Fig. 5: Quantitative imaging of erlotinib iPAI probes in live cells.
Fig. 6: iPAI erlotinib imaging enables quantification of DTA in tissues.

Similar content being viewed by others

Data availability

All data associated with this study are presented in the article, extended data, source data, supplementary files or Supplementary Information. The data generated during the study are also available from the corresponding author upon reasonable request. Source data are provided with this paper.

Code availability

Custom­-written MATLAB code used to calculate drug target availability is available at https://doi.org/10.5281/zenodo.4004647.

Change history

References

  1. Proceedings of the World Molecular Imaging Congress 2021, October 5–8, 2021: Late-Breaking Abstracts. Mol. Imaging Biol. 23, 1739–2027 (2021).

  2. Lavis, L. D. Teaching old dyes new tricks: biological probes built from fluoresceins and rhodamines. Annu. Rev. Biochem. 86, 825–843 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. Nasu, Y., Shen, Y., Kramer, L. & Campbell, R. E. Structure- and mechanism-guided design of single fluorescent protein-based biosensors. Nat. Chem. Biol. 17, 509–518 (2021).

    Article  CAS  PubMed  Google Scholar 

  4. Wolfbeis, O. S. An overview of nanoparticles commonly used in fluorescent bioimaging. Chem. Soc. Rev. 44, 4743–4768 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Day, R. N. & Davidson, M. W. The fluorescent protein palette: tools for cellular imaging. Chem. Soc. Rev. 38, 2887–2921 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Hoffman, R. M. The multiple uses of fluorescent proteins to visualize cancer in vivo. Nat. Rev. Cancer 5, 796–806 (2005).

    Article  CAS  PubMed  Google Scholar 

  7. Rodriguez, E. A. et al. The growing and glowing toolbox of fluorescent and photoactive proteins. Trends Biochem. Sci. 42, 111–129 (2017).

    Article  CAS  PubMed  Google Scholar 

  8. Cranfill, P. J. et al. Quantitative assessment of fluorescent proteins. Nat. Methods 13, 557–562 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Shu, X., Shaner, N. C., Yarbrough, C. A., Tsien, R. Y. & Remington, S. J. Novel chromophores and buried charges control color in mFruits. Biochemistry 45, 9639–9647 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. Oliinyk, O. S., Shemetov, A. A., Pletnev, S., Shcherbakova, D. M. & Verkhusha, V. V. Smallest near-infrared fluorescent protein evolved from cyanobacteriochrome as versatile tag for spectral multiplexing. Nat. Commun. 10, 279 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Baddeley, D. & Bewersdorf, J. Biological insight from super-resolution microscopy: what we can learn from localization-based images. Annu. Rev. Biochem. 87, 965–989 (2018).

    Article  CAS  PubMed  Google Scholar 

  12. Choi, H. S. et al. Targeted zwitterionic near-infrared fluorophores for improved optical imaging. Nat. Biotechnol. 31, 148–153 (2013).

    Article  CAS  PubMed  Google Scholar 

  13. Hyun, H. et al. Structure-inherent targeting of near-infrared fluorophores for parathyroid and thyroid gland imaging. Nat. Med. 21, 192–197 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Wang, L. et al. Far-red and near-infrared seminaphthofluorophores for targeted pancreatic cancer imaging. ACS Omega 2, 154–163 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Wang, L. G. et al. Near-infrared nerve-binding fluorophores for buried nerve tissue imaging. Sci. Transl. Med. 12, eaay0712 (2020).

    Article  CAS  PubMed  Google Scholar 

  16. Cosco, E. D. et al. Shortwave infrared polymethine fluorophores matched to excitation lasers enable non-invasive, multicolour in vivo imaging in real time. Nat. Chem. 12, 1123–1130 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Dempsey, G. T., Vaughan, J. C., Chen, K. H., Bates, M. & Zhuang, X. Evaluation of fluorophores for optimal performance in localization-based super-resolution imaging. Nat. Methods 8, 1027–1036 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Fölling, J. et al. Fluorescence nanoscopy by ground-state depletion and single-molecule return. Nat. Methods 5, 943–945 (2008).

    Article  PubMed  Google Scholar 

  19. Heilemann, M., van de Linde, S., Mukherjee, A. & Sauer, M. Super-resolution imaging with small organic fluorophores. Angew. Chem. Int. Ed. 48, 6903–6908 (2009).

    Article  CAS  Google Scholar 

  20. Beija, M., Afonso, C. A. M. & Martinho, J. M. G. Synthesis and applications of Rhodamine derivatives as fluorescent probes. Chem. Soc. Rev. 38, 2410–2433 (2009).

    Article  CAS  PubMed  Google Scholar 

  21. Grimm, J. B. et al. A general method to improve fluorophores for live-cell and single-molecule microscopy. Nat. Methods 12, 244–250 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Grimm, J. B. et al. Bright photoactivatable fluorophores for single-molecule imaging. Nat. Methods 13, 985–988 (2016).

    Article  CAS  PubMed  Google Scholar 

  23. Grimm, J. B. et al. A general method to fine-tune fluorophores for live-cell and in vivo imaging. Nat. Methods 14, 987–994 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lukinavičius, G. et al. A near-infrared fluorophore for live-cell super-resolution microscopy of cellular proteins. Nat. Chem. 5, 132–139 (2013).

    Article  PubMed  Google Scholar 

  25. Wang, L. et al. A general strategy to develop cell permeable and fluorogenic probes for multicolour nanoscopy. Nat. Chem. 12, 165–172 (2020).

    Article  CAS  PubMed  Google Scholar 

  26. Pressman, D., Day, E. D. & Blau, M. The use of paired labeling in the determination of tumor-localizing antibodies. Cancer Res. 17, 845–850 (1957).

    CAS  PubMed  Google Scholar 

  27. Tichauer, K. M. et al. Microscopic lymph node tumor burden quantified by macroscopic dual-tracer molecular imaging. Nat. Med. 20, 1348–1353 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Baeten, J., Haller, J., Shih, H. & Ntziachristos, V. In vivo investigation of breast cancer progression by use of an internal control. Neoplasia 11, 220–227 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Liu, J. T. et al. Quantifying cell-surface biomarker expression in thick tissues with ratiometric three-dimensional microscopy. Biophys. J. 96, 2405–2414 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Pogue, B. W. et al. Imaging targeted-agent binding in vivo with two probes. J. Biomed. Opt. 15, 030513 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Barth, C. W., Schaefer, J. M., Rossi, V. M., Davis, S. C. & Gibbs, S. L. Optimizing fresh specimen staining for rapid identification of tumor biomarkers during surgery. Theranostics 7, 4722–4734 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Boyarskiy, V. P. et al. Photostable, amino reactive and water-soluble fluorescent labels based on sulfonated rhodamine with a rigidized xanthene fragment. Chemistry 14, 1784–1792 (2008).

    Article  CAS  PubMed  Google Scholar 

  33. Kolmakov, K. et al. Red-emitting rhodamines with hydroxylated, sulfonated, and phosphorylated dye residues and their use in fluorescence nanoscopy. Chemistry 18, 12986–12998 (2012).

    Article  CAS  PubMed  Google Scholar 

  34. Kolmakov, K. et al. Polar red-emitting rhodamine dyes with reactive groups: synthesis, photophysical properties, and two-color STED nanoscopy applications. Chemistry 20, 146–157 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Keppler, A., Arrivoli, C., Sironi, L. & Ellenberg, J. Fluorophores for live cell imaging of AGT fusion proteins across the visible spectrum. BioTechniques 41, 167–175 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Orsi, M. & Essex, J. W. in Molecular Simulations and Biomembranes (eds Sansom, M. S. P. & Biggin, P. C.) Ch. 4, 76–90 (RSC, 2010).

  37. Tsien, R. Y. A non-disruptive technique for loading calcium buffers and indicators into cells. Nature 290, 527–528 (1981).

    Article  CAS  PubMed  Google Scholar 

  38. Kim, E., Yang, K. S., Giedt, R. J. & Weissleder, R. Red Si-rhodamine drug conjugates enable imaging in GFP cells. Chem. Commun. 50, 4504–4507 (2014).

    Article  CAS  Google Scholar 

  39. Su, D. D. et al. The development of a highly photostable and chemically stable zwitterionic near-infrared dye for imaging applications. Chem. Commun. 51, 3989–3992 (2015).

    Article  CAS  Google Scholar 

  40. Choi, H. S. et al. Synthesis and in vivo fate of zwitterionic near-infrared fluorophores. Angew. Chem. Int. Ed. 50, 6258–6263 (2011).

    Article  CAS  Google Scholar 

  41. de Valk, K. S. et al. A zwitterionic near-infrared fluorophore for real-time ureter identification during laparoscopic abdominopelvic surgery. Nat. Commun. 10, 3118 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Numasawa, K. et al. A fluorescent probe for rapid, high-contrast visualization of folate-receptor-expressing tumors in vivo. Angew. Chem. Int. Ed. 59, 6015–6020 (2020).

    Article  CAS  Google Scholar 

  43. Jo, A. et al. Near-IR fluorescent tracer for glucose-uptake monitoring in live cells. Bioconjug. Chem. 29, 3394–3401 (2018).

    Article  CAS  PubMed  Google Scholar 

  44. Sato, R. et al. Intracellular protein-labeling probes for multicolor single-molecule imaging of immune receptor–adaptor molecular dynamics. J. Am. Chem. Soc. 139, 17397–17404 (2017).

    Article  CAS  PubMed  Google Scholar 

  45. Kim, E. et al. Optimized near-IR fluorescent agents for in vivo imaging of Btk expression. Bioconjug. Chem. 26, 1513–1518 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kim, E., Yang, K. S., Giedt, R. J. & Weissleder, R. Red Si-rhodamine drug conjugates enable imaging in GFP cells. Chem. Commun. 50, 4504–4507 (2014).

    Article  CAS  Google Scholar 

  47. Sung, J. et al. A new infrared probe targeting mitochondria via regulation of molecular hydrophobicity. Bioconjug. Chem. 30, 210–217 (2019).

    Article  CAS  PubMed  Google Scholar 

  48. Chu, Y. et al. Development of theragnostic tool using NIR fluorescence probe targeting mitochondria in glioma cells. Bioconjug. Chem. 30, 1642–1648 (2019).

    Article  CAS  PubMed  Google Scholar 

  49. Oda, K., Matsuoka, Y., Funahashi, A. & Kitano, H. A comprehensive pathway map of epidermal growth factor receptor signaling. Mol. Syst. Biol. 1, 2005.0010 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Scaltriti, M. & Baselga, J. The epidermal growth factor receptor pathway: a model for targeted therapy. Clin. Cancer Res. 12, 5268–5272 (2006).

    Article  CAS  PubMed  Google Scholar 

  51. Whirl-Carrillo, M. et al. Pharmacogenomics knowledge for personalized medicine. Clin. Pharmacol. Ther. 92, 414–417 (2012).

    Article  CAS  PubMed  Google Scholar 

  52. Bakker, J., Spits, M., Neefjes, J. & Berlin, I. The EGFR odyssey—from activation to destruction in space and time. J. Cell Sci. 130, 4087–4096 (2017).

    CAS  PubMed  Google Scholar 

  53. Park, J. H., Liu, Y., Lemmon, Mark, A. & Radhakrishnan, R. Erlotinib binds both inactive and active conformations of the EGFR tyrosine kinase domain. Biochem. J. 448, 417–423 (2012).

    Article  CAS  PubMed  Google Scholar 

  54. Wu, P., Nielsen, T. E. & Clausen, M. H. FDA-approved small-molecule kinase inhibitors. Trends Pharmacol. Sci. 36, 422–439 (2015).

    Article  CAS  PubMed  Google Scholar 

  55. Stamos, J., Sliwkowski, M. X. & Eigenbrot, C. Structure of the epidermal growth factor receptor kinase domain alone and in complex with a 4-anilinoquinazoline inhibitor. J. Biol. Chem. 277, 46265–46272 (2002).

    Article  CAS  PubMed  Google Scholar 

  56. Gruber, A. et al. Monitoring of erlotinib in pancreatic cancer patients during long-time administration and comparison to a physiologically based pharmacokinetic model. Cancer Chemother. Pharmacol. 81, 763–771 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. O’Bryant, C. L. et al. An open-label study to describe pharmacokinetic parameters of erlotinib in patients with advanced solid tumors with adequate and moderately impaired hepatic function. Cancer Chemother. Pharmacol. 69, 605–612 (2012).

    Article  PubMed  Google Scholar 

  58. Peters, S., Zimmermann, S. & Adjei, A. A. Oral epidermal growth factor receptor tyrosine kinase inhibitors for the treatment of non-small cell lung cancer: comparative pharmacokinetics and drug-drug interactions. Cancer Treat. Rev. 40, 917–926 (2014).

    Article  CAS  PubMed  Google Scholar 

  59. Li, J., Zhao, M., He, P., Hidalgo, M. & Baker, S. D. Differential metabolism of gefitinib and erlotinib by human cytochrome P450 enzymes. Clin. Cancer Res. 13, 3731–3737 (2007).

    Article  CAS  PubMed  Google Scholar 

  60. McMahon, N. P. et al. TRIPODD: a novel fluorescence imaging platform for in situ quantification of drug distribution and therapeutic response. Mol. Imaging Biol. 23, 650–664 (2021).

    Article  CAS  PubMed  Google Scholar 

  61. Lee, Y. et al. High-throughput, single-particle tracking reveals nested membrane domains that dictate KRas(G12D) diffusion and trafficking. eLife 8, e46393 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Nan, X. et al. Ras-GTP dimers activate the mitogen-activated protein kinase (MAPK) pathway. Proc. Natl Acad. Sci. USA 112, 7996–8001 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Perkins, A. et al. A structural switch between agonist and antagonist bound conformations for a ligand-optimized model of the human aryl hydrocarbon receptor ligand binding domain. Biology 3, 645–669 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Sun, L. et al. Gut microbiota and intestinal FXR mediate the clinical benefits of metformin. Nat. Med. 24, 1919–1929 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. McMahon, N. P. In Situ Therapeutic Response Imaging Using a Novel Fluorescence Imaging PlatformTRIPODD (Oregon Health & Science Univ., 2021).

Download references

Acknowledgements

This work was supported by an ASPIRE Award from the Mark Foundation for Cancer Research (S.L.G.) as well as the National Cancer Institute (R21CA257942; S.L.G.). We thank M. Vescio (OHSU) and S. Kumarapeli (OHSU) for experimental assistance, and the OHSU Advanced Light Microscopy Core for imaging assistance.

Author information

Authors and Affiliations

Authors

Contributions

L.G.W. and S.L.G. designed the study. L.G.W., A.R.M. and J.R.C. performed the probe synthesis. L.G.W. and A.R.M. completed all spectral measurements. L.G.W. performed confocal microscopy. W.H.B. completed the molecular docking studies. M.M.G. and K.T. completed the BLI studies. N.P.M. and A.S. conducted intracellular paired agent imaging studies. D.A.S. conducted in vivo biodistribution studies. K.S.S., K.M.T. and S.L.G. completed intracellular paired-agent imaging data analysis and interpretation. L.G.W. and S.L.G. wrote and edited the paper. All authors reviewed and approved the paper. S.L.G. supervised the project.

Corresponding author

Correspondence to Summer L. Gibbs.

Ethics declarations

Competing interests

L.G.W., A.R.M. and S.L.G. are inventors on patent application PCT/US21/53806, ‘Zwitterionic cell-permeant and water-soluble rhodamine dyes for quantitative imaging applications’, submitted to the World Intellectual Property Organization and held by Oregon Health and Science University. This covers the composition and methods of use of the ORFluor compounds discussed herein. All other authors declare no competing interests.

Peer review

Peer review information

Nature Chemistry thanks Hak Soo Choi, Lin Yuan and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Fluorescence live-cell images of SiR, SiR-Me and OF650.

Representative images of U2OS cells stained with a fluorescent on/off switchable rhodamine fluorophore SiR, b fluorescent always-on fluorophore SiR-Me, and c ORFluor OF650 at 30 nM for 30 min. Fluorescence images are shown normalized to one another for the three rhodamine dyes. Positive intracellular localization was observed in cells that were stained with SiR-Me, while negligible fluorescence signal was observed in the cells stained with either SiR or OF650. d Co-localization imaging showed SiR-Me preferentially localized to the mitochondria with high affinity and specificity, confirmed by co-staining with commercial mitochondria probe, MitoTracker Green (green). All cells were co-stained with Hoechst (blue) and examined from a minimum of three independent staining experiments. Scale bar, 100 μm.

Extended Data Fig. 2 Synthesis of ORFluor OF650 and its conjugatable versions.

ORFluor OF650 was prepared in six steps with an averaged chemical yield of 68%. A Vilsmeier–Haack reaction of dimethylbromoaniline precursor 23 gave benzaldehyde intermediate 24 in 88% yield. This was then followed by the addition of pre-lithiated 2-bromo-toluene (25), giving diaryl methanol intermediate 26 in decent yield (81%). The allyl-protected N-methyl bromoaniline 27 was activated by Lewis acid, ZnCl2, to participate in a nucleophilic substitution reaction with secondary alcohol 26, providing heterodimer 28 in reasonable yield (56%). Subsequent lithiation of heterodimeric bromide 28 using tert-BuLi and addition of dichlorodimethylsilane yielded the leuco-basic rhodamine 29 (60%). Deprotection (that is, allyl group removal) of 29, catalyzed by a Pd(PPh3)4 and dimethylbarbituric acid (DMBA) system, provided leuco-base rhodamine 30 in 68% yield. Introduction of a sulfonate group to the silicon-substituted rhodamine was achieved through alkylation of intermediate 30 using excess propane-1,3-sultone, followed by direct oxidation with p-chloranil, affording the final ORFluor OF650 (2) in 55% yield. To demonstrate that our synthetic strategy can be generalized to other asymmetrical silicon-substituted rhodamines and provide access to bioconjugatable derivatives, we modified the synthetic sequence to prepare carboxylic acid OF650COOH and N-hydroxysuccinimide (NHS) ester OF650NHS versions (Fig. 1d). Intermediate 31, bearing a functional handle, was metalated before reaction with compound 24, giving the diaryl methanolic intermediate 32 with a protected carboxyl group in 74% yield. The reaction protocols used to synthesize OF650 – until removal of the allylic protecting group – were again utilized to afford compound 35. Initial attempts at alkylation using propane-1,3-sultone followed directly by hydrolysis gave very poor yields. However, when an acid-catalyzed hydrolysis was performed first, releasing the carboxyl group, the free carboxylic acid intermediate (36) was given in 46% yield and subsequent alkylation and oxidation products were furnished by the aforementioned protocols. The free acid OF650COOH (6), treated with TSTU, was transformed to NHS ester OF650NHS (37) in 69% yield.

Extended Data Fig. 3 Extension of zwitterionic structural design strategy to the TMR analog.

ORFluor OF550 was prepared in three steps with an averaged chemical yield of 48%. Typically, an asymmetric rhodamine dye is prepared through condensation between a hydroxy benzophenone and an aniline derivative together in a one-to-one ratio at high temperature, under acidic conditions. In our case, we sought to efficiently prepare asymmetrical zwitterionic fluorophore derivatives by introducing required structural variables (that is, the phenyl ring motifs) at a later stage in the synthetic sequence. An Ullmann cross-coupling reaction of the commercially available dimethylaminophenol 38 and aryl bromide 39, catalyzed by CuI and 2-picolinic acid under basic conditions, afforded the diaryl ether intermediate 40. Alkylation of compound 40 with an excess amount of propane-1,3-sultone gave the sulfonated intermediate 41 (70%). Because the diaryl ether derivative 41 is prone to electronic activation, an acid catalyzed reaction should readily provide the oxidized fluorophore product via the Friedel–Crafts acylation–cyclization reaction. To ease the product recovery and purification process, a catalytic system consisting of ZnCl2 in ethanol was introduced to the condensation step. However, cyclization of compound 41 with a benzaldehyde 42 did not proceed until the temperature rose above 100 °C. A subsequent DDQ-mediated oxidation reaction was performed to push the reaction to completion in 33% yield over two steps. A bioconjugatable version OF550NHS was prepared using the aforementioned key intermediate 41. Using this same protocol and the orthogonal protecting group-bearing intermediate 43 allowed for the introduction of a functional handle (for example, protected carboxyl group). Deprotection under acidic conditions, followed by DDQ-mediated oxidation provided the OF550COOH (8) as a single regioisomer. The free acid OF550COOH (8) was then transformed into NHS ester OF550NHS (44) via treatment with TSTU in 48% yield.

Extended Data Fig. 4 Fast Airyscan confocal fluorescence imaging with SNAP-tag and HaloTag ligands.

Intracellular localization of OF650 (red) and OF550 (green) labeled SNAP-tag and HaloTag substrates in doxycycline inducible U2OS TetR-Mito-SNAP-tag expressing cells and TetR-ER-HaloTag expressing cells. All cells were co-stained with Hoechst (blue) prior to fixation, and examined from a minimum of three independent staining experiments. Scale bar = 10 μm. Single focal plane.

Extended Data Fig. 5 BLI binding determination of erlotinib and SiR and TMR labeled iPAI probes.

Representative BLI binding sensorgrams of erlotinib, SiR and TMR labeled targeted and untargeted probes, as well as untagged fluorophores SiR and TMR, binding to biotinylated recombinant EGFR-TKD immobilized on Super Streptavidin (SSA) biosensors. BLI sensorgrams show the association of each probe (0–90 s) at the indicated concentrations ranging from 37–3000 nM, followed by the subsequent dissociation in binding buffer without analytes (90–180 s). Kinetic data (Colored curve: observed experimental data) were fit globally using a simple Langmuir 1:1 binding model (Black dashed curve: fitted experimental data) in ForteBio Data Analysis HT 10.0 evaluation software to obtain the association rate constant ka, dissociation rate constant kd and the equilibrium dissociation constant KD.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–27 and full synthetic details of all compounds, their structural and optical characterization, as well as imaging experiments.

Reporting Summary

Real-time three-colour fast Airyscan confocal microscopy of U2OS TetR- ER-Halo-Mito-SNAP expressing cells labelled with OF650HALO, OF550SNAP and Hoechst 33342.

Real-time two-colour fast Airyscan confocal microscopy of U2OS TetR- ER-HaloTag expressing cells labelled with OF650HALO and Hoechst 33342.

Real-time two-colour fast Airyscan confocal microscopy of U2OS TetR- ER-HaloTag expressing cells labelled with OF550HALO and Hoechst 33342.

Real-time two-colour fast Airyscan confocal microscopy of U2OS TetR- Mito-SNAP-tag expressing cells labelled with OF650SNAP and Hoechst 33342.

Real-time two-colour fast Airyscan confocal microscopy of U2OS TetR- Mito-SNAP-tag expressing cells labelled with OF550SNAP and Hoechst 33342.

Source data

Source Data Fig. 1

Source data for Fig. 1.

Source Data Fig. 3

Source data for Fig. 3.

Source Data Fig. 5

Source data for Fig. 5.

Source Data Extended Data Fig. 5

Source data for Extended Data Fig. 5.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, L.G., Montaño, A.R., Combs, J.R. et al. OregonFluor enables quantitative intracellular paired agent imaging to assess drug target availability in live cells and tissues. Nat. Chem. 15, 729–739 (2023). https://doi.org/10.1038/s41557-023-01173-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41557-023-01173-6

  • Springer Nature Limited

Navigation