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.

  • Article
  • Published:

Large-scale, in-cell photocrosslinking at single-residue resolution reveals the molecular basis for glucocorticoid receptor regulation by immunophilins

Abstract

The Hsp90 co-chaperones FKBP51 and FKBP52 play key roles in steroid-hormone-receptor regulation, stress-related disorders, and sexual embryonic development. As a prominent target, glucocorticoid receptor (GR) signaling is repressed by FKBP51 and potentiated by FKBP52, but the underlying molecular mechanisms remain poorly understood. Here we present the architecture and functional annotation of FKBP51-, FKBP52-, and p23-containing Hsp90–apo-GR pre-activation complexes, trapped by systematic incorporation of photoreactive amino acids inside human cells. The identified crosslinking sites clustered in characteristic patterns, depended on Hsp90, and were disrupted by GR activation. GR binding to the FKBPFK1, but not the FKBPFK2, domain was modulated by FKBP ligands, explaining the lack of GR derepression by certain classes of FKBP ligands. Our findings show how FKBPs differentially interact with apo-GR, help to explain the differentiated pharmacology of FKBP51 ligands, and provide a structural basis for the development of improved FKBP ligands.

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: Large-scale in-cell photocrosslinking reveals large and defined interaction interfaces of FKBPs with GR.
Fig. 2: Large-scale photocrosslinking of the full-length GR in mammalian cells confirms the interaction of the GRLBD and co-chaperones FKBP51, FKBP52, and p23.
Fig. 3: FKBP51, FKBP52, and p23 interact with the apo-GR in a Hsp90-dependent manner.
Fig. 4: Inhibition of FKBPs only partially disrupts FKBP–GR interactions.
Fig. 5: Co-chaperones preferentially engage apo-GR in pre-activation complexes in human cells.

Similar content being viewed by others

Data availability

The data supporting the findings of this study are available within the paper and its Supplementary Information. Should any raw data files be needed in another format, they are available from the corresponding author upon reasonable request. The following protein structures were used in this paper, accessed via the PDB: 5OMP, 7L7I, 7KRJ, 7KW7, 3O5R, 6TXX, and 5OBK. Source data are provided with this paper.

References

  1. Prodromou, C. & Bjorklund, D. M. Advances towards understanding the mechanism of action of the Hsp90 complex. Biomolecules 12, 600 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Schopf, F. H., Biebl, M. M. & Buchner, J. The HSP90 chaperone machinery. Nat. Rev. Mol. Cell Biol. 18, 345–360, (2017).

    Article  CAS  PubMed  Google Scholar 

  3. Sanchez, E. R. Chaperoning steroidal physiology: lessons from mouse genetic models of Hsp90 and its cochaperones. Biochim. Biophys. Acta 1823, 722–729 (2012).

    Article  CAS  PubMed  Google Scholar 

  4. Maeda, K. et al. FKBP51 and FKBP52 regulate androgen receptor dimerization and proliferation in prostate cancer cells. Mol. Oncol. 16, 940–956 (2022).

    Article  CAS  PubMed  Google Scholar 

  5. Habara, M. et al. FKBP52 and FKBP51 differentially regulate the stability of estrogen receptor in breast cancer. Proc. Natl Acad. Sci. USA 119, e2110256119 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Smedlund, K. B., Sanchez, E. R. & Hinds, T. D. Jr FKBP51 and the molecular chaperoning of metabolism. Trends Endocrinol. Metab. 32, 862–874 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Balsevich, G. et al. Stress-responsive FKBP51 regulates AKT2-AS160 signaling and metabolic function. Nat. Commun. 8, 1725 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Maiarù, M. et al. The stress regulator FKBP51: a novel and promising druggable target for the treatment of persistent pain states across sexes. Sci. Transl. Med. 8, 325ra19 (2016).

    PubMed  PubMed Central  Google Scholar 

  9. Touma, C. et al. FK506 binding protein 5 shapes stress responsiveness: modulation of neuroendocrine reactivity and coping behavior. Biol. Psychiatry 70, 928–936 (2011).

    Article  CAS  PubMed  Google Scholar 

  10. O’Leary, J. C. III et al. A new anti-depressive strategy for the elderly: ablation of FKBP5/FKBP51. PLoS ONE 6, e24840 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Jääskeläinen, T., Makkonen, H. & Palvimo, J. J. Steroid up-regulation of FKBP51 and its role in hormone signaling. Curr. Opin. Pharmacol. 11, 326–331 (2011).

    Article  PubMed  Google Scholar 

  12. Zannas, A. S., Wiechmann, T., Gassen, N. C. & Binder, E. B. Gene–stress–epigenetic regulation of FKBP5: clinical and translational implications. Neuropsychopharmacology 41, 261–274 (2016).

    Article  CAS  PubMed  Google Scholar 

  13. Ratajczak, T. Steroid receptor-associated immunophilins: candidates for diverse drug-targeting approaches in disease. Curr. Mol. Pharmacol. 9, 66–95 (2016).

    Article  CAS  Google Scholar 

  14. Hähle, A., Merz, S., Meyners, C. & Hausch, F. The many faces of FKBP51. Biomolecules 9, 35 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Wang, R. Y.-R. et al. Structure of Hsp90–Hsp70–Hop–GR reveals the Hsp90 client-loading mechanism. Nature 601, 460–464 (2022).

    Article  CAS  PubMed  Google Scholar 

  16. Noddings, C. M., Wang, R. Y.-R., Johnson, J. L. & Agard, D. A. Structure of Hsp90–p23–GR reveals the Hsp90 client-remodelling mechanism. Nature 601, 465–469 (2022).

    Article  CAS  PubMed  Google Scholar 

  17. Biebl, M. M. et al. Structural elements in the flexible tail of the co-chaperone p23 coordinate client binding and progression of the Hsp90 chaperone cycle. Nat. Commun. 12, 828 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Biebl, M. M. et al. NudC guides client transfer between the Hsp40/70 and Hsp90 chaperone systems. Mol. Cell 82, 555–569 (2022).

    Article  CAS  PubMed  Google Scholar 

  19. Gruszczyk, J. et al. Cryo-EM structure of the agonist-bound Hsp90-XAP2-AHR cytosolic complex. Nat. Commun. 13, 7010 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Smith, D. F. & Toft, D. O. Minireview: the intersection of steroid receptors with molecular chaperones: observations and questions. Mol. Endocrinol. 22, 2229–2240 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kirschke, E., Goswami, D., Southworth, D., Griffin, P. R. & Agard, D. A. Glucocorticoid receptor function regulated by coordinated action of the Hsp90 and Hsp70 chaperone cycles. Cell 157, 1685–1697 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Coin, I. Application of non-canonical crosslinking amino acids to study protein–protein interactions in live cells. Curr. Opin. Chem. Biol. 46, 156–163 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Nguyen, T. A., Cigler, M. & Lang, K. Expanding the genetic code to study protein–protein interactions. Angew. Chem. Int. Ed. Engl. 57, 14350–14361 (2018).

    Article  CAS  PubMed  Google Scholar 

  24. Lee, K. et al. The structure of an Hsp90–immunophilin complex reveals cochaperone recognition of the client maturation state. Mol. Cell 81, 3496–3508 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kumar, R., Moche, M., Winblad, B. & Pavlov, P. F. Combined X-ray crystallography and computational modeling approach to investigate the Hsp90 C-terminal peptide binding to FKBP51. Sci. Rep. 7, 14288 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Püllmann, P. et al. Golden mutagenesis: an efficient multi-site-saturation mutagenesis approach by Golden Gate cloning with automated primer design. Sci. Rep. 9, 10932 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Backe, S. J. et al. A specialized Hsp90 co-chaperone network regulates steroid hormone receptor response to ligand. Cell Rep. 40, 111039 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Biebl, M. M. & Buchner, J. Structure, function, and regulation of the Hsp90 machinery. Cold Spring Harb. Perspect. Biol. 11, a034017 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Davies, T. H., Ning, Y.-M. & Sánchez, E. R. A new first step in activation of steroid receptors: hormone-induced switching of FKBP51 and FKBP52 immunophilins. J. Biol. Chem. 277, 4597–4600 (2002).

    Article  CAS  PubMed  Google Scholar 

  30. Kauppi, B. et al. The three-dimensional structures of antagonistic and agonistic forms of the glucocorticoid receptor ligand-binding domain: RU-486 induces a transconformation that leads to active antagonism. J. Biol. Chem. 278, 22748–22754 (2003).

    Article  CAS  PubMed  Google Scholar 

  31. Riggs, D. L. et al. Noncatalytic role of the FKBP52 peptidyl-prolyl isomerase domain in the regulation of steroid hormone signaling. Mol. Cell. Biol. 27, 8658–8669 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Riggs, D. L. et al. The Hsp90-binding peptidylprolyl isomerase FKBP52 potentiates glucocorticoid signaling in vivo. EMBO J. 22, 1158–1167 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wochnik, G. M. et al. FK506-binding proteins 51 and 52 differentially regulate dynein interaction and nuclear translocation of the glucocorticoid receptor in mammalian cells. J. Biol. Chem. 280, 4609–4616 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Gaali, S. et al. Selective inhibitors of the FK506-binding protein 51 by induced fit. Nat. Chem. Biol. 11, 33 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Kolos, J. M. et al. Picomolar FKBP inhibitors enabled by a single water-displacing methyl group in bicyclic [4.3.1] aza-amides. Chem. Sci. 12, 14758–14765 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Gnatzy, M. T. et al. Development of NanoBRET‐binding assays for FKBP‐ligand profiling in living cells. ChemBioChem 22, 2257–2261 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Bracher, A., Kozany, C., Thost, A.-K. & Hausch, F. Structural characterization of the PPIase domain of FKBP51, a cochaperone of human Hsp90. Acta Crystallogr. D Biol. Crystallogr. 67, 549–559 (2011).

    Article  CAS  PubMed  Google Scholar 

  38. Draxler, S. W. et al. Hybrid screening approach for very small fragments: X-ray and computational screening on FKBP51. J. Med. Chem. 63, 5856–5864 (2020).

    Article  CAS  PubMed  Google Scholar 

  39. Silverstein, A. M. et al. Different regions of the immunophilin FKBP52 determine its association with the glucocorticoid receptor, Hsp90, and cytoplasmic dynein. J. Biol. Chem. 274, 36980–36986 (1999).

    Article  CAS  PubMed  Google Scholar 

  40. Davies, T. H., Ning, Y.-M. & Sánchez, E. R. Differential control of glucocorticoid receptor hormone-binding function by tetratricopeptide repeat (TPR) proteins and the immunosuppressive ligand FK506. Biochemistry 44, 2030–2038 (2005).

    Article  CAS  PubMed  Google Scholar 

  41. Cluning, C. et al. The helix 1–3 loop in the glucocorticoid receptor LBD is a regulatory element for FKBP cochaperones. Mol. Endocrinol. 27, 1020–1035 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sinars, C. R. et al. Structure of the large FK506-binding protein FKBP51, an Hsp90-binding protein and a component of steroid receptor complexes. Proc. Natl Acad. Sci. USA 100, 868–873 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Bracher, A. et al. Crystal structures of the free and ligand-bound FK1–FK2 domain segment of FKBP52 reveal a flexible inter-domain hinge. J. Mol. Biol. 425, 4134–4144 (2013).

    Article  CAS  PubMed  Google Scholar 

  44. LeMaster, D. M. et al. Coupling of conformational transitions in the N-terminal domain of the 51-kDa FK506-binding protein (FKBP51) near its site of interaction with the steroid receptor proteins. J. Biol. Chem. 290, 15746–15757 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Reynolds, P. D., Ruan, Y., Smith, D. F. & Scammell, J. G. Glucocorticoid resistance in the squirrel monkey is associated with overexpression of the immunophilin FKBP51. J. Clin. Endocrinol. Metab. 84, 663–669 (1999).

    CAS  PubMed  Google Scholar 

  46. Denny, W. B., Valentine, D. L., Reynolds, P. D., Smith, D. F. & Scammell, J. G. Squirrel monkey immunophilin FKBP51 is a potent inhibitor of glucocorticoid receptor binding. Endocrinology 141, 4107–4113 (2000).

    Article  CAS  PubMed  Google Scholar 

  47. Noddings, C. M., Johnson, J. L. & Agard, D. A. Cryo-EM reveals how Hsp90 and FKBP immunophilins co-regulate the glucocorticoid receptor. Nat. Struct. Mol. Biol. https://doi.org/10.1038/s41594-023-01128-y

  48. Grunbeck, A., Huber, T., Sachdev, P. & Sakmar, T. P. Mapping the ligand-binding site on a G protein-coupled receptor (GPCR) using genetically encoded photocrosslinkers. Biochemistry 50, 3411–3413 (2011).

    Article  CAS  PubMed  Google Scholar 

  49. Rannversson, H. et al. Genetically encoded photocrosslinkers locate the high-affinity binding site of antidepressant drugs in the human serotonin transporter. Nat. Commun. 7, 11261 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Rudolf, S. et al. Binding of natural peptide ligands to the neuropeptide Y5 receptor. Angew. Chem. Int. Ed. Engl. 61, e202108738 (2022).

    Article  CAS  PubMed  Google Scholar 

  51. Coin, I. et al. Genetically encoded chemical probes in cells reveal the binding path of urocortin-I to CRF class B GPCR. Cell 155, 1258–1269 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Wilkins, B. J. et al. A cascade of histone modifications induces chromatin condensation in mitosis. Science 343, 77–80 (2014).

    Article  CAS  PubMed  Google Scholar 

  53. Kashiwagi, K. et al. Crystal structure of eukaryotic translation initiation factor 2B. Nature 531, 122–125 (2016).

    Article  CAS  PubMed  Google Scholar 

  54. Serfling, R. & Coin, I. Incorporation of unnatural amino acids into proteins expressed in mammalian cells. Methods Enzymol. 580, 89–107 (2016).

Download references

Acknowledgements

We thank M. Weissenborn, P. Püllmann, and C. Ulpinnis (University of Halle) for suggestions and training on the Golden Gate mutagenesis protocol, I. Coin (University of Leipzig) for plasmids and suggestions for pBpa incorporation in mammalian cells, and J. Kolos and T. Heymann for samples of 18(S)-Me and SAFit2, respectively. We are indebted to M. Wilfinger and J.-P. Kahl for cloning GR mutants and helping to establish the ELISA, and to F. Halloy for preliminary work on reporter gene assays. This work was supported by funding from the HMWK (LOEWE-Schwerpunkt TRABITA) and the BMBF (16GW0290K) to F.H.

Author information

Authors and Affiliations

Authors

Contributions

A.B. and S.E. designed and executed all photocrosslinking experiments and subsequent analysis. T.M.G. performed and analyzed the reporter gene assays. M.C.T. performed and analyzed translocation and control experiments. F.H. conceived the project. All authors interpreted the results. A.B., S.E., and F.H. wrote the manuscript, which was approved by all authors.

Corresponding author

Correspondence to Felix Hausch.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Structural & Molecular Biology thanks Francis O’Reilly and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Katarzyna Ciazynska, in collaboration with the Nature Structural & Molecular Biology team. Peer reviewer reports are available.

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 In-cell photocrosslinking confirms the interaction between TPR domain of FKBP51 and the C-terminal domain of Hsp90.

a, Western blots of exemplary FKBP51 pBpa mutants expressed and photocrosslinked in HEK293 cells co-expressing HA-tagged Hsp90. UV light-induced HA-reactive bands at a size of approx. 160 kDa are indicative of the mutated position being in proximity to Hsp90. b+c, Hsp90-photoreactive positions (highlighted in green) in the TPR domain of FKBP51 (pale pink) identified by Western blotting as in a were mapped on the structure of the Hsp90-FKBP51 complex (PDB: 7J7I, p23 omitted for clarity). Hsp90 dimers are shown in blue, the C-terminal MEEVD peptide of Hsp90 is depicted as blue spheres, and the unresolved linker between the MEEVD motif and one of the C-terminal domains of the Hsp90 dimers is indicated by a dotted blue line. c, Closeup view of crosslinks between positions K385, S392, A398, and M412 of FKBP51 with Hsp90. d+e, Close-up view of the predicted interaction of A398 and M412 of FKBP51 with Hsp90. points. All western blots shown are only performed once.

Source data

Extended Data Fig. 2 Sequence alignment of FKBP51 and FKBP52 showing FKBP→GR interaction.

Structural elements were marked on the sequence. Crosslinks to HA-GR are highlighted in green and no crosslinks are indicated in red.

Extended Data Fig. 3 Photocrosslinking of the full-length GR in mammalian cells confirms the interaction of the GR ligand binding domain and Hsp90 and the co-chaperones FKBP51 and p23.

a, Western blots of full length GR pBpa mutants expressed, photocrosslinked in HEK293 cells co-overexpressing HA-tagged FKBP51, and concentrated by FLAG-IP. UV light-induced HA-reactive bands at a size of approx. 180 kDa are indicative of the mutated position being in proximity to FKBP51. b, Tested position of GR→FKBP51 interaction are shown on GRLBD (PDB: 7KRJ), green indicates crosslink detected, red indicates no crosslink detected at this position. c, Structure of the GRLBD (523–777, PDB: 7krj) with α-helices shown in red and yellow and key loops highlighted in blue. d Overview of GR→FKBP51 and GR→FKBP52 Crosslinks e, GR→P23 crosslinks were highlighted on the GRLBD structure (523–777, PDB: 7krj), with indication of secondary structure. f, Western blots of full length GR pBpa mutants expressed, photocrosslinked in HEK293 cells without HA-FKBP51 overexpression, and concentrated by FLAG-IP. UV light-induced FLAG-reactive bands and FLAG-reactive bands at a size of 130 kDa are indicative of the mutated position being in proximity to p23. points. All western blots shown are only performed once.

Source data

Extended Data Fig. 4 GR agonist/antagonist disrupt FKBPs interaction with GR.

GR antagonist (Mifepristone, Mif) treatment for 1 h disrupts FKBP51→GR (a) and FKBP52→GR crosslinks (b). c, GR agonist (Dexamethasone, Dex), GR antagonist (Mifepristone, Mif) and Hsp90 inhibitor (Geldanamycin, GA) treatment for 1 h disrupts GR→FKBP51 crosslinks. points. All western blots shown are only performed once.

Source data

Extended Data Fig. 5 GR agonist disrupts FKBPs interaction with GR.

a, GR agonist (Dexamethasone, Dex) treatment for 1 h disrupts GRK743pBpa → FKBP51 and GRN768pBpa → FKBP51 crosslinks in a dose-dependent manner. Individual data points are shown. (n = 3 independent biological replicates). b, Amber suppression control of Extended Data Fig. 6a, Individual data points are shown. (n = 3 biological replicates). c + d, Treatment with Dexamethasone for 1 h, after transfection with double amount of GR plasmid compared to Fig. 3h. HA signal (c) and FLAG signal (d) measured from the same samples. (n = 3 biological replicates). e + f, Treatment with Dexamethasone for 10 min, after transfection with double amount of GR plasmid compared to Fig. 3h. HA signal (e) and FLAG signal (f) measured from the same samples. (n = 3 biological replicates). g, Time course experiment with varying Dex treatment times, 125 nM or 2 µM Dex was used h + j, cell fractionation study, stimulation with 1 µM Dex, except no treatment control (NT), with varying treatment times. Cells lysates were fractionated into; WC- whole cell lysate, C-cytosolic and N-nuclear fractions. i, Dex stimulation of GR results in loss of Hsp90 binding (sample concentrated by FLAG-IP). points. All western blots shown are only performed once.

Source data

Extended Data Fig. 6 Inhibition of FKBPs only partially disrupts FKBP-GR interactions.

a, Treatment with the FKBP inhibitor (18(S)-Me) for 1 h disrupts FKBP51→GR crosslinks in the FK1 (D68, E75) but not the FK2 domain (Q210, Y159). b, Treatment with the FKBP inhibitor (18(S)-Me) for 1 h disrupts FKBP52→GR crosslinks in the FK1 (D68, D75) but not the FK2 domain (R210, Y161). Western blots of FKBP51 (a) and FKBP52 (b) pBpa mutants expressed and photocrosslinked in HEK293 cells co-overexpressing HA-tagged GR. UV light-induced HA-reactive bands at a size of approx. 180 kDa are indicative of the mutated position being in proximity to GR. c, Treatment with the FKBP inhibitor (SAFit2) for 1 h disrupts FKBP52→GR crosslinks in the FK1 (D68, D75) but not the FK2 domain (R210, Y161). Western blots of exemplary FKBP52 pBpa mutants expressed and photocrosslinked in HEK293 cells co-overexpressing HA-tagged GR. UV light-induced HA-reactive bands at a size of approx. 180 kDa are indicative of the mutated position being in proximity to GR. d, SAFit2-sensitivity and resistance of exemplary GR pBpa mutants, detected by Western blots after expression and photocrosslinked in HEK293 cells with co-overexpressing HA-tagged FKBP51. points. All western blots shown are only performed once.

Source data

Extended Data Fig. 7 Reporter Gene Assays.

a, GR transactivation was measured by reporter gene assays in HEK293 cells transiently co-transfected with the dual reporters pGL4.36 (MMTV-luc2p), pGL4.74 (TK-hRluc) as well as expression plasmids for human GR and optionally FKBP51 and/or a 3-fold excess of FKBP52. Luminescence of the dual reporters luc2p and hRluc was measured 24 h after treatment with Dex and optionally 18(S)-Me (a). The FKBP ligand 18(S)-Me does not block FKBP51-induced GR suppression. Each bar (mean ± SD) represents data from biological replicates (n = 6, grey dots). b, FK506 (cyan, from PDB-ID: 3O5R), SAFit2 (green, from PBD-ID: 6TXX), and compound 18 (yellow, a close homolog of 18(S)-Me, from PDB-ID: 5OBK) are overlayed in the structure of FKBP51FK1 (PDB-ID: 5OBK), the close-up is rotated for a better view insight the binding pocket.

Source data

Extended Data Fig. 8 Differential GR interactions for the FK1 domains of FKBP51 and FKBP52.

a, Close-up view of FKBP51FK1→GR interactions, shown from three different angles. Crosslink (green) and inactive positions (red) were mapped on the structure of FKBP51FK1 (PDB: 3O5Q). b, Close-up view of FKBP52FK1→GR interactions, shown from three different angles. Crosslink (green) and inactive positions (red) GR were mapped on the structure of FKBP52FK1 (4LAV). c, Sequence alignment of the FK1 domains of FKBP51 and FKBP52, with GR-photoreactive positions highlighted in green and tested inactive positions in red. (·) different residue, (I) same residue, (!) crosslinking behavior different, (* or *) crosslinking behavior similar.

Supplementary information

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 3

Raw data graph.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 4

Raw data graph.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 5

Raw data graph.

Source Data Extended Data Fig. 5

Raw data graph.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 7

Raw data graph.

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

Baischew, A., Engel, S., Taubert, M.C. et al. Large-scale, in-cell photocrosslinking at single-residue resolution reveals the molecular basis for glucocorticoid receptor regulation by immunophilins. Nat Struct Mol Biol 30, 1857–1866 (2023). https://doi.org/10.1038/s41594-023-01098-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-023-01098-1

This article is cited by

Search

Quick links

Nature Briefing

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

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