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.

  • Brief Communication
  • Published:

RETRACTED ARTICLE: A fast, high-affinity fluorescent serotonin biosensor engineered from a tick lipocalin

A Retraction to this article was published on 23 April 2021

This article has been updated

Abstract

Serotonin (5-HT) is an important signaling monoamine and neurotransmitter. We report structure-guided engineering of a green fluorescent, genetically encoded serotonin sensor (G-GESS) from a 5-HT-binding lipocalin in the soft tick Argas monolakensis. G-GESS shows fast response kinetics and high affinity, specificity, brightness and photostability. We used G-GESS to image 5-HT dynamics in cultured cells, brain slices and behaving mice.

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: Design and characterization of G-GESS.
Fig. 2: Imaging of endogenous 5-HT dynamics with G-GESS in mice.

Similar content being viewed by others

Data availability

The gene sequence for G-GESS has been deposited with GenBank under the accession number MW027675. The plasmids for pDisplay-G-GESS-M (plasmid 154273), pAAV-hSyn-G-GESS-M (plasmid 154274), pLenti-hSyn-G-GESS-M (plasmid 154275), pDisplay-G-dGESS-M (plasmid 154276), pAAV-hSyn-G-dGESS-M (plasmid 154277) and pLenti-hSyn-G-GESS-M (plasmid 154278) and their sequence information have been deposited with Addgene. Source data are provided with this paper.

Change history

References

  1. Berger, M., Gray, J. A. & Roth, B. L. The expanded biology of serotonin. Annu. Rev. Med. 60, 355–366 (2009).

    Article  CAS  Google Scholar 

  2. Sabatini, B. L. & Tian, L. Imaging neurotransmitter and neuromodulator dynamics in vivo with genetically encoded indicators. Neuron 108, 17–32 (2020).

    Article  CAS  Google Scholar 

  3. Chefer, V. I., Thompson, A. C., Zapata, A. & Shippenberg, T. S. Overview of brain microdialysis. Curr. Protoc. Neurosci. Chapter 7, Unit 7.1 (2009).

    Google Scholar 

  4. Puthongkham, P. & Venton, B. J. Recent advances in fast-scan cyclic voltammetry. Analyst 145, 1087–1102 (2020).

    Article  CAS  Google Scholar 

  5. Jeong, S. et al. High-throughput evolution of near-infrared serotonin nanosensors. Sci. Adv. 5, eaay3771 (2019).

    Article  CAS  Google Scholar 

  6. Hettie, K. S. & Glass, T. E. Turn-on near-infrared fluorescent sensor for selectively imaging serotonin. ACS Chem. Neurosci. 7, 21–25 (2016).

    Article  CAS  Google Scholar 

  7. Borden, P. M. et al. A fast genetically encoded fluorescent sensor for faithful in vivo acetylcholine detection in mice, fish, worms and flies. Preprint at bioRxiv https://doi.org/10.1101/2020.04.18.048397 (2020).

  8. Tubio, M. R. et al. Expression of a G protein-coupled receptor (GPCR) leads to attenuation of signaling by other GPCRs: experimental evidence for a spontaneous GPCR constitutive inactive form. J. Biol. Chem. 285, 14990–14998 (2010).

    Article  CAS  Google Scholar 

  9. Mans, B. J., Ribeiro, J. M. & Andersen, J. F. Structure, function, and evolution of biogenic amine-binding proteins in soft ticks. J. Biol. Chem. 283, 18721–18733 (2008).

    Article  CAS  Google Scholar 

  10. Mans, B. J., Calvo, E., Ribeiro, J. M. & Andersen, J. F. The crystal structure of D7r4, a salivary biogenic amine-binding protein from the malaria mosquito Anopheles gambiae. J. Biol. Chem. 282, 36626–36633 (2007).

    Article  CAS  Google Scholar 

  11. Hofherr, A., Fakler, B. & Klöcker, N. Selective Golgi export of Kir2.1 controls the stoichiometry of functional Kir2.x channel heteromers. J. Cell Sci. 118, 1935–1943 (2005).

    Article  CAS  Google Scholar 

  12. Cataldo Bascuñan, L. R., Lyons, C., Bennet, H., Artner, I. & Fex, M. Serotonergic regulation of insulin secretion. Acta Physiol. (Oxf.) 225, e13101 (2019).

    Article  Google Scholar 

  13. Rohm, B. et al. Nonivamide, a capsaicin analog, increases dopamine and serotonin release in SH-SY5Y cells via a TRPV1-independent pathway. Mol. Nutr. Food Res. 57, 2008–2018 (2013).

    Article  CAS  Google Scholar 

  14. Fletcher, P. J., Korth, K. M., Robinson, S. R. & Baker, G. B. Multiple 5-HT receptors are involved in the effects of acute MDMA treatment: studies on locomotor activity and responding for conditioned reinforcement. Psychopharmacol. (Berl.) 162, 282–291 (2002).

    Article  CAS  Google Scholar 

  15. Bauer, E. P. Serotonin in fear conditioning processes. Behav. Brain Res. 277, 68–77 (2015).

    Article  CAS  Google Scholar 

  16. Fujino, K. et al. Increased serotonin release in mice frontal cortex and hippocampus induced by acute physiological stressors. Neurosci. Lett. 320, 91–95 (2002).

    Article  CAS  Google Scholar 

  17. Bindels, D. S. et al. mScarlet: a bright monomeric red fluorescent protein for cellular imaging. Nat. Methods 14, 53–56 (2017).

    Article  CAS  Google Scholar 

  18. Paxinos, G., Burt, J., Atrens, D. M. & Jackson, D. M. 5-hydroxytryptamine depletion with para-chlorophenylalanine: effects on eating, drinking, irritability, muricide and copulation. Pharmacol. Biochem. Behav. 6, 439–447 (1977).

    Article  CAS  Google Scholar 

  19. Unger, E. K. et al. Directed evolution of a selective and sensitive serotonin sensor via machine learning. Cell 183, 1986–2002.e1926 (2020).

    Article  CAS  Google Scholar 

  20. Wan, J. et al. A genetically encoded GRAB sensor for measuring serotonin dynamics. Preprint at bioRxiv https://doi.org/10.1101/2020.02.24.962282 (2020).

  21. Flower, D. R., North, A. C. & Sansom, C. E. The lipocalin protein family: structural and sequence overview. Biochim. Biophys. Acta 1482, 9–24 (2000).

    Article  CAS  Google Scholar 

  22. Skerra, A. Alternative binding proteins: anticalins - harnessing the structural plasticity of the lipocalin ligand pocket to engineer novel binding activities. FEBS J. 275, 2677–2683 (2008).

    Article  CAS  Google Scholar 

  23. Marvin, J. S. et al. An optimized fluorescent probe for visualizing glutamate neurotransmission. Nat. Methods 10, 162–170 (2013).

    Article  CAS  Google Scholar 

  24. Hochbaum, D. R. et al. All-optical electrophysiology in mammalian neurons using engineered microbial rhodopsins. Nat. Methods 11, 825–833 (2014).

    Article  CAS  Google Scholar 

  25. Lin, J. Y., Knutsen, P. M., Muller, A., Kleinfeld, D. & Tsien, R. Y. ReaChR: a red-shifted variant of channelrhodopsin enables deep transcranial optogenetic excitation. Nat. Neurosci. 16, 1499–1508 (2013).

    Article  CAS  Google Scholar 

  26. Zhao, Y. et al. An expanded palette of genetically encoded Ca(2)(+) indicators. Science 333, 1888–1891 (2011).

    Article  CAS  Google Scholar 

  27. Marvin, J. S., Schreiter, E. R., Echevarría, I. M. & Looger, L. L. A genetically encoded, high-signal-to-noise maltose sensor. Proteins 79, 3025–3036 (2011).

    Article  CAS  Google Scholar 

  28. Chen, S., Chen, Z. J., Ren, W. & Ai, H. W. Reaction-based genetically encoded fluorescent hydrogen sulfide sensors. J. Am. Chem. Soc. 134, 9589–9592 (2012).

    Article  CAS  Google Scholar 

  29. Fan, Y., Chen, Z. & Ai, H. W. Monitoring redox dynamics in living cells with a redox-sensitive red fluorescent protein. Anal. Chem. 87, 2802–2810 (2015).

    Article  CAS  Google Scholar 

  30. Kardash, E., Bandemer, J. & Raz, E. Imaging protein activity in live embryos using fluorescence resonance energy transfer biosensors. Nat. Protoc. 6, 1835–1846 (2011).

    Article  CAS  Google Scholar 

  31. Chen, Z. & Ai, H. W. Single fluorescent protein-based indicators for zinc ion (Zn2+). Anal. Chem. 88, 9029–9036 (2016).

    Article  CAS  Google Scholar 

  32. Molina, R. S. et al. Understanding the fluorescence change in red genetically encoded calcium ion indicators. Biophys. J. 116, 1873–1886 (2019).

    Article  CAS  Google Scholar 

  33. Drobizhev, M., Molina, R. S. & Hughes, T. E. Characterizing the two-photon absorption properties of fluorescent molecules in the 680-1300 nm spectral range. Bio-Protoc. 10, e3498 (2020).

    Article  Google Scholar 

  34. Gibson, D. G. et al. Enzymatic assembly of DNA molecules up to several hundred kilobases. Nat. Methods 6, 343–345 (2009).

    Article  CAS  Google Scholar 

  35. Chen, M. et al. Genetically encoded, photostable indicators to image dynamic Zn(2+) secretion of pancreatic islets. Anal. Chem. 91, 12212–12219 (2019).

    Article  CAS  Google Scholar 

  36. Burroughs, S. L. et al. Plate reader-based assays for measuring cell viability, neuroprotection and calcium in primary neuronal cultures. J. Neurosci. Methods 203, 141–145 (2012).

    Article  CAS  Google Scholar 

  37. Rego, M. et al. Improved yield of AAV2 and rAAV2-retro serotypes following sugar supplementation during the viral production phase. bioRxiv https://doi.org/10.1101/488585 (2018).

  38. Ding, B. & Kilpatrick, D. L. Lentiviral vector production, titration, and transduction of primary neurons. Methods Mol. Biol. 1018, 119–131 (2013).

    Article  CAS  Google Scholar 

  39. Tetteh, H., Lee, J., Lee, J., Kim, J. G. & Yang, S. Investigating long-term synaptic plasticity in interlamellar hippocampus CA1 by electrophysiological field recording. J. Vis. Exp. https://doi.org/10.3791/59879 (2019).

  40. Otis, J. M. et al. Prefrontal cortex output circuits guide reward seeking through divergent cue encoding. Nature 543, 103–107 (2017).

    Article  CAS  Google Scholar 

  41. Holtmaat, A. et al. Long-term, high-resolution imaging in the mouse neocortex through a chronic cranial window. Nat. Protoc. 4, 1128–1144 (2009).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. Chen (Weill Cornell) for sharing MIN6 cells, Z. Chen for early exploration of this project, R. Molina for technical help with analyzing two-photon spectra and Y. Wang, Z. Li and other members in the Ai group for helpful discussion and assistance with experiments. Research reported in this publication was supported by funding to H.-w.A. (University of Virginia Start-up Package and NIH grant nos R01 DK122253, R01 GM118675 and R01 GM129291) and to M.D. (NIH grant nos U01 NS094246 and U24 NS109107).

Author information

Authors and Affiliations

Authors

Contributions

H.-w.A. conceived and supervised the project. X.L. and S. Zhao performed experiments involving brain slices and live mice. M.D. determined photophysical parameters and recorded two-photon excitation spectra. S. Zhang performed all other experiments, including protein engineering, characterization in vitro and in cultured cells, and preparation of viral vectors. All authors analyzed the data and prepared the manuscript.

Corresponding author

Correspondence to Hui-wang Ai.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Methods thanks Bryan Roth, Jing Ren and Oliver Griesbeck for their contribution to the peer review of this work. Nina Vogt was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

>This article has been retracted. Please see the retraction notice for more detail: https://dx.doi.org/10.1038/s41592-021-01148-w

Extended data

Extended Data Fig. 1 Structural analysis to identify potential cpGFP insertion sites.

a, Cartoon presentation of AM10. Loops showing ligand-dependent changes in dihedral angle (defined by the Cα atoms spanning every four sequential residues) are highlighted in red. b, Cα dihedral differences calculated from dihedrals between the ligand-bound (PDB 3BU1) and apo (PDB 3BS2) states of AM10 plotted against residue numbers. c, Dihedral angles for residue 113 of AM10 in the ligand-bound and apo states. d, Cartoon presentation of D7r4. Loop 2 showing a 5-HT-dependent change in dihedral angle is highlighted in red. e, Cα dihedral differences calculated from dihedrals between the ligand-bound (PDB 2QEH) and apo (PDB 2QEV) states of D7r4 plotted against residue numbers. f, Dihedral angles for residue 29 of D7r4 in the ligand-bound and apo states.

Source data

Extended Data Fig. 2 Process to engineer G-GESS.

a, Flowchart to illustrate our integrated, multi-step process to derive G-GESS. ΔF/F0 for each sensor generation is also presented. From G-GESS0.3 to G-GESS0.4, several N-terminal residues uninvolved in ligand binding were deleted, resulting in increased fluorescence change. However, it was found later in G-GESS0.5 that these residues were important for achieving good cell surface localization, so they were added back to G-GESS0.6. From G-GESS0.6 to G-GESS0.7, superfolder mutations (previously reported for sfGFP) were introduced into the cpGFP fragment of the sensor, resulting in improved brightness and fluorescence change. b, Sequence alignment of G-GESS with two earlier variants.

Extended Data Fig. 3 Two-photon fluorescence characterization of G-GESS.

a, Two-photon fluorescence excitation spectra in the presence (green) and absence (black) of 5-HT. b, Ratio of two-photon excitation in the presence over absence of 5-HT plotted against wavelength. These experiments were repeated twice with similar results.

Source data

Extended Data Fig. 4 Characterization of G-GESS-M for membrane localization in HEK 293 T cells.

a, Representative fluorescence images of HEK 293 T cells co-transfected with pDisplay-G-GESS-M and pCMV-R-GECO1. Red fluorescent Ca2+ sensor R-GECO1 was co-expressed as a whole-cell label. Scale bar, 20 µm. b, Fluorescence intensity measured over the white line shown in the ‘overlay’ image of panel a. These experiments were repeated three times with similar results using independent cultures.

Source data

Extended Data Fig. 5 Construction of G-dGESS, an unresponsive variant of G-GESS.

a, Cartoon presentation of AM182 (PDB 3BRN), highlighting the 5-HT ligand (magenta) and two residues (F58 and D106; colored in cyan and numbered according to PDB 3BRN) important for ligand binding. Mutations corresponding to F58A and D106L were introduced into G-GESS to create an unresponsive variant named ‘G-dGESS’. b, Representative fluorescence images of G-dGESS-M at the surface of HEK 293 T cells, in the presence or absence of 5-HT. This experiment was repeated three times with similar results using independent cell cultures. Scale bar, 20 µm.

Extended Data Fig. 6 Imaging of 5-HT release from β-cells and neuroblastoma cells.

a, Representative images of G-GESS-M at the surface of mouse pancreatic MIN6 β-cells. Scale bar, 20 µm. b, Fluorescence traces of G-GESS-M in response to high glucose (25 mM). n = 7 cells for G-GESS-M and 6 cells for G-dGESS-M from 3 independent repeats. c, Representative images of G-GESS-M at the surface of SH-SY5Y neuroblastoma cells. Scale bar, 20 µm. d, Fluorescence traces of G-GESS-M in response to nonivamide (0.1 µM). n = 13 cells from 3 repeats for each group. Data are presented as mean ± s.d. of indicated replicates.

Source data

Extended Data Fig. 7 Imaging of 5-HT release in primary mouse neuron cultures.

a, Representative images of G-GESS-M at the surface of primary mouse neurons in response to 5-HT (10 µM). Also shown is a pseudocolor ratiometric image of the neuron in the presence over absence of 5-HT. Scale bar, 20 µm. This experiment was repeated three times with similar results using independent cultures. b, Fluorescence traces of G-GESS-M at the surface of primary mouse neurons, in response to high potassium induced membrane polarization. n = 19 cells for G-GESS-M and 12 cells for G-dGESS-M from 4 repeats. Data are presented as mean ± s.d. overlaid on the top of the traces of individual cells.

Source data

Extended Data Fig. 8 Evaluation of G-GESS-M cytotoxicity in primary mouse neurons.

a, MTT assay of primary neurons transfected with pDisplay-G-GESS-M or pDisplay-sfGFP-M in the absence or presence of the oxidative stress inducer, tert-butyl hydroperoxide (tBHP). P values determined by unpaired, two-tailed Student’s t-test. n = 6 cultures each group. b, Ca2+ mobilization assay of primary neurons transfected with pDisplay-G-GESS-M or pDisplay-sfGFP-M in response to depolarization with 100 mM KCl at the indicated time point. Intracellular Ca2+ was monitored with an orange fluorescent Ca2+ indicator (Enzo GFP-Certified Fluoforte Calcium Assay Kit). P values determined by unpaired, two-tailed Student’s t-test. n = 4 cultures each group. Individual data points and mean ± s.d. are presented. n.s., not significant.

Source data

Extended Data Fig. 9 Imaging of 5-HT release in acute mouse brain slices.

a, Schematic diagram illustrating the preparation of acute mouse brain slices with viral expression of G-GESS-M. b, Representative fluorescence images of G-GESS-M-expressing mouse brain slices. Scale bar, 200 µm. This experiment was repeated five times with three slice preparations and similar results were obtained. c, Representative fluorescence images of G-GESS-M-expressing hippocampal neurons in mouse brain slices in response to electrical stimuli (20 pulses at 64 Hz). Also shown is a pseudocolor ratiometric image (peak F/F0) of a representative neuron. Scale bar, 20 µm. This experiment was repeated six times with three slice preparations and similar results were obtained. d, Fluorescence traces of G-GESS-M in response to 20 pulses of electric stimuli at 50 V and the indicated frequencies. e, Fluorescence traces of G-GESS-M in response to 20 pulses of electric stimuli at 30 V and the indicated frequencies. f, Maximal fluorescence ratio change (peak F/F0) plotted against stimulation frequency at 50 V or 30 V. P values determined by 2-way ANOVA with Sidak’s multiple comparisons test. n = 6 cells on 3 slices from 2 mice for each group stimulated at 50 V, and 3 cells on 2 slices from 2 mice for each group stimulated at 30 V. Data are presented as mean ± s.d in all panels.

Source data

Extended Data Fig. 10 Bicistronic co-expression of G-GESS-M with a red fluorescent protein mScarlet-I to detect pharmacological modulation of 5-HT by p-chlorophenylalanine methyl ester (PCPA), a tryptophan hydroxylase inhibitor.

a, Schematic diagram illustrating the co-expression construct including G-GESS-M, a P2A peptide self-cleaving site, and mScarlet-I. b, Representative fluorescence images of the co-expression construct expressed in HEK 293 T cells in the absence and presence of 5-HT acquired with a Leica DMi8 SPE confocal microscope. Scale bar, 20 µm. c, Quantitative analysis of the green-to-red intensity ratios of HEK 293 T cells, in the absence or presence of 5-HT for G-dGESS-M-P2A-mScarlet-I and G-GESS-M-P2A-mScarlet-I. P values determined by unpaired, two-tailed Student’s t-test. n = 12 random cells from three independent cultures. d, Schematic diagram illustrating the mouse treatment timeline, including intracranial injection of AAV on Day 0 and PCPA injection from Day 7 to Day 11. Acute brain slices were prepared and imaged on Day 12. e, Representative fluorescence images of the co-expression construct expressed in the hippocampus of mice with or without PCPA treatment. Images were acquired with a Scientifica SliceScope Pro 1000. Scale bar, 20 µm. f, Quantitative analysis of the green-to-red intensity ratios of mouse brain slices, in the presence or absence of PCPA. P values determined by one-way ANOVA with Tukey’s multiple comparisons test. n = 7 cells on one 5-HT-treated slice expressing G-GESS-M-P2A-mScarlet-I, and 35 cells on 5 slices from two mice for all other groups. Individual data points and mean ± s.d. are presented. n.s., not significant.

Source data

Supplementary information

Supplementary Information

Supplementary Table 1 and Figs. 1 and 2.

Reporting Summary

Source data

Source Data Fig. 1

Figure and statistical source data.

Source Data Fig. 2

Figure and statistical source data.

Source Data Extended Data Fig. 1

Figure and statistical source data.

Source Data Extended Data Fig. 3

Figure and statistical source data.

Source Data Extended Data Fig. 4

Figure and Statistical source data.

Source Data Extended Data Fig. 6

Figure and statistical source data.

Source Data Extended Data Fig. 7

Figure and statistical source data.

Source Data Extended Data Fig. 8

Figure and statistical source data.

Source Data Extended Data Fig. 9

Figure and statistical source data.

Source Data Extended Data Fig. 10

Figure and statistical source data.

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, S., Li, X., Zhao, S. et al. RETRACTED ARTICLE: A fast, high-affinity fluorescent serotonin biosensor engineered from a tick lipocalin. Nat Methods 18, 258–261 (2021). https://doi.org/10.1038/s41592-021-01078-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-021-01078-7

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