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.

  • Technical Report
  • Published:

A genetically encoded sensor for measuring serotonin dynamics

Abstract

Serotonin (5-HT) is a phylogenetically conserved monoamine neurotransmitter modulating important processes in the brain. To directly visualize the release of 5-HT, we developed a genetically encoded G-protein-coupled receptor (GPCR)-activation-based 5-HT (GRAB5-HT) sensor with high sensitivity, high selectivity, subsecond kinetics and subcellular resolution. GRAB5-HT detects 5-HT release in multiple physiological and pathological conditions in both flies and mice and provides new insights into the dynamics and mechanisms of 5-HT signaling.

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, optimization and characterization of a new genetically encoded 5-HT sensor.
Fig. 2: GRAB5-HT can report the release of endogenous 5-HT in acute mouse brain slices and Drosophila.
Fig. 3: GRAB5-HT can report endogenous serotonergic activity in freely behaving mice.

Similar content being viewed by others

Data availability

Plasmids for expressing the sensors used in this study were deposited at Addgene (https://www.addgene.org/140552/). The human GPCR cDNA library was obtained from the hORFeome database 8.1 (http://horfdb.dfci.harvard.edu/index.php?page=home). Source data are provided with this paper.

Code availability

The EZcalcium algorithm and BEADS baseline estimation and denoising with sparsity algorithm are available at https://github.com/porteralab/EZcalcium and https://www.mathworks.com/matlabcentral/fileexchange/49974-beads-baseline-estimation-and-denoising-with-sparsity.

References

  1. Lesch, K. P. et al. Association of anxiety-related traits with a polymorphism in the serotonin transporter gene regulatory region. Science 274, 1527–1531 (1996).

    Article  CAS  PubMed  Google Scholar 

  2. Li, Y. et al. Serotonin neurons in the dorsal raphe nucleus encode reward signals. Nat. Commun. 7, 10503 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Portas, C. M. et al. On-line detection of extracellular levels of serotonin in dorsal raphe nucleus and frontal cortex over the sleep/wake cycle in the freely moving rat. Neuroscience 83, 807–814 (1998).

    Article  CAS  PubMed  Google Scholar 

  4. Vaswani, M., Linda, F. K. & Ramesh, S. Role of selective serotonin reuptake inhibitors in psychiatric disorders: a comprehensive review. Prog. Neuropsychopharmacol. Biol. Psychiatry 27, 85–102 (2003).

    Article  CAS  PubMed  Google Scholar 

  5. Fuller, R. W. Uptake inhibitors increase extracellular serotonin concentration measured by brain microdialysis. Life Sci. 55, 163–167 (1994).

    Article  CAS  PubMed  Google Scholar 

  6. Bunin, M. A., Prioleau, C., Mailman, R. B. & Wightman, R. M. Release and uptake rates of 5-hydroxytryptamine in the dorsal raphe and substantia nigra reticulata of the rat brain. J. Neurochem. 70, 1077–1087 (1998).

    Article  CAS  PubMed  Google Scholar 

  7. Candelario, J. & Chachisvilis, M. Mechanical stress stimulates conformational changes in 5-hydroxytryptamine receptor 1B in bone cells. Cell. Mol. Bioeng. 5, 277–286 (2012).

    Article  CAS  Google Scholar 

  8. Jing, M. et al. A genetically encoded fluorescent acetylcholine indicator for in vitro and in vivo studies. Nat. Biotechnol. 36, 726–737 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Patriarchi, T. et al. Ultrafast neuronal imaging of dopamine dynamics with designed genetically encoded sensors. Science 360, eaat4422 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Sun, F. et al. A genetically encoded fluorescent sensor enables rapid and specific detection of dopamine in flies, fish, and mice. Cell 174, 481–496 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Feng, J. et al. A genetically encoded fluorescent sensor for rapid and specific in vivo detection of norepinephrine. Neuron 102, 745–761 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Bajar, B. T. et al. Improving brightness and photostability of green and red fluorescent proteins for live cell imaging and FRET reporting. Sci. Rep. 6, 20889 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Pedelacq, J. D., Cabantous, S., Tran, T., Terwilliger, T. C. & Waldo, G. S. Engineering and characterization of a superfolder green fluorescent protein. Nat. Biotechnol. 24, 79–88 (2006).

    Article  CAS  PubMed  Google Scholar 

  14. Peng, Y. et al. 5-HT2C receptor structures reveal the structural basis of GPCR polypharmacology. Cell 172, 719–730 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ballesteros, J. A. & Weinstein, H. [19] Integrated methods for the construction of three-dimensional models and computational probing of structure-function relations in G protein-coupled receptors. Methods in Neurosciences 25, 366–428 (1995).

  16. Harada, K. et al. Red fluorescent protein-based cAMP indicator applicable to optogenetics and in vivo imaging. Sci. Rep. 7, 7351 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Wan, Q. et al. Mini G protein probes for active G protein-coupled receptors (GPCRs) in live cells. J. Biol. Chem. 293, 7466–7473 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Barnea, G. et al. The genetic design of signaling cascades to record receptor activation. Proc. Natl Acad. Sci. USA 105, 64–69 (2008).

    Article  CAS  PubMed  Google Scholar 

  19. Ishimura, K. et al. Quantitative analysis of the distribution of serotonin-immunoreactive cell bodies in the mouse brain. Neurosci. Lett. 91, 265–270 (1988).

    Article  CAS  PubMed  Google Scholar 

  20. Waddell, S., Armstrong, J. D., Kitamoto, T., Kaiser, K. & Quinn, W. G. The amnesiac gene product is expressed in two neurons in the Drosophila brain that are critical for memory. Cell 103, 805–813 (2000).

    Article  CAS  PubMed  Google Scholar 

  21. Lee, P. T. et al. Serotonin–mushroom body circuit modulating the formation of anesthesia-resistant memory in Drosophila. Proc. Natl Acad. Sci. USA 108, 13794–13799 (2011).

    Article  CAS  PubMed  Google Scholar 

  22. Keene, A. C. et al. Diverse odor-conditioned memories require uniquely timed dorsal paired medial neuron output. Neuron 44, 521–533 (2004).

    Article  CAS  PubMed  Google Scholar 

  23. Yu, D. et al. Drosophila DPM neurons form a delayed and branch-specific memory trace after olfactory classical conditioning. Cell 123, 945–957 (2005).

    Article  CAS  PubMed  Google Scholar 

  24. Xu, M. et al. Basal forebrain circuit for sleep–wake control. Nat. Neurosci. 18, 1641–1647 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ren, J. et al. Anatomically defined and functionally distinct dorsal raphe serotonin sub-systems. Cell 175, 472–487 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Oikonomou, G. et al. The serotonergic raphe promote sleep in zebrafish and mice. Neuron 103, 686–701 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ren, J. et al. Single-cell transcriptomes and whole-brain projections of serotonin neurons in the mouse dorsal and median raphe nuclei. eLife 8, e49424 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Rudnick, G. & Wall, S. C. The molecular mechanism of ‘ecstasy’ [3,4-methylenedioxy-methamphetamine (MDMA)]: serotonin transporters are targets for MDMA-induced serotonin release. Proc. Natl Acad. Sci. USA 89, 1817–1821 (1992).

  29. Bicks, L. K., Koike, H., Akbarian, S. & Morishita, H. Prefrontal cortex and social cognition in mouse and man. Front. Psychol. 6, 1805 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Liechti, M. E., Saur, M. R., Gamma, A., Hell, D. & Vollenweider, F. X. Psychological and physiological effects of MDMA (‘ecstasy’) after pretreatment with the 5-HT2 antagonist ketanserin in healthy humans. Neuropsychopharmacology 23, 396–404 (2000).

    Article  CAS  PubMed  Google Scholar 

  31. Hagino, Y. et al. Effects of MDMA on extracellular dopamine and serotonin levels in mice lacking dopamine and/or serotonin transporters. Curr. Neuropharmacol. 9, 91–95 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Wang, Q., Shui, B., Kotlikoff, M. I. & Sondermann, H. Structural basis for calcium sensing by GCaMP2. Structure 16, 1817–1827 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kroeze, W. K. et al. PRESTO-Tango as an open-source resource for interrogation of the druggable human GPCRome. Nat. Struct. Mol. Biol. 22, 362–369 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Pfeiffer, B. D., Truman, J. W. & Rubin, G. M. Using translational enhancers to increase transgene expression in Drosophila. Proc. Natl Acad. Sci. USA 109, 6626–6631 (2012).

    Article  CAS  PubMed  Google Scholar 

  35. Dana, H. et al. Sensitive red protein calcium indicators for imaging neural activity. eLife 5, e12727 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  37. Yusa, K. et al. Targeted gene correction of α1-antitrypsin deficiency in induced pluripotent stem cells. Nature 478, 391–394 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Shin, M. & Venton, B. J. Electrochemical measurements of acetylcholine-stimulated dopamine release in adult Drosophila melanogaster brains. Anal. Chem. 90, 10318–10325 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Cantu, D. A. et al. EZcalcium: open-source toolbox for analysis of calcium imaging data. Front. Neural Circuits 14, 25 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Y. Rao for providing the two-photon microscope and X. Lei at PKU-CLS and the National Center for Protein Sciences at Peking University for support and assistance with the Opera Phenix high-content screening system. We thank D. Lin and X. Xu for critical reading of the manuscript. This work was supported by the Beijing Municipal Science & Technology Commission (Z181100001318002), the Beijing Brain Initiative of the Beijing Municipal Science & Technology Commission (Z181100001518004), a Guangdong grant, ‘Key Technologies for Treatment of Brain Disorders’ (2018B030332001), the General Program of National Natural Science Foundation of China (projects 31671118, 31871087 and 31925017), the Science Fund for Creative Research Groups of the National Natural Science Foundation of China (81821092), the NIH BRAIN Initiative (NS103558), grants from the Peking-Tsinghua Center for Life Sciences and the State Key Laboratory of Membrane Biology at Peking University School of Life Sciences (to Y.L.), the Shanghai Municipal Science and Technology Major Project (2018SHZDZX05 to M.X.) and the Shanghai Pujiang Program (18PJ1410800 to M.X.), a Peking University Postdoctoral Fellowship (J.F.), an Alzheimer’s Association Postdoctoral Research Fellowship (AARF 19 619387 to P.Z.), a Peking-Tsinghua Center Excellence Postdoctoral Fellowship (Y.Z.) and the Beijing Nova Program (Z201100006820100 to M.J.).

Author information

Authors and Affiliations

Authors

Contributions

Y.L. conceived and supervised the project. J.W., M.J., F.D., S.H., J.F., J. Zou and S.P. performed the experiments related to developing, optimizing and characterizing the sensor in cultured HEK293T cells and neurons. T.Q. performed the experiments using AAVs in slices. P.Z., Y.Z. and M.S. performed the simultaneous imaging and FSCV experiments using the Sindbis virus in slices under the supervision of J.J.Z. and B.J.V. X.L. and J. Zeng performed the Drosophila experiments. W.P. and K.S. performed the fiber-photometry recordings in behaving mice under the supervision of M.X. J.W. performed two-photon imaging of head-fixed mice. All authors contributed to data interpretation and analysis. Y.L. and J.W. wrote the manuscript with input from all other authors.

Corresponding author

Correspondence to Yulong Li.

Ethics declarations

Competing interests

The authors declare competing financial interests. J.W., M.J., J.F. and Y.L. have filed patent applications, the value of which might be affected by this publication.

Additional information

Peer review information Nature Neuroscience thanks Adam Cohen and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Characterization of the membrane trafficking for 5-HT receptor-based chimeras.

a, Representative fluorescence images of HEK293T cells co-expressing the indicated 5-HT receptors fused with cpGFP (green) and RFP-CAAX (red); EGFP-CAAX was used as a positive control. Similar results were observed for more than 100 cells. Scale bar, 10 μm. b, Normalized fluorescence intensity measured at the white dashed lines shown in (a) for each candidate sensor.

Source data

Extended Data Fig. 2 Sequence alignment of cpGFP from 5-HT1.0 sensor, sfGFP, and mClover3.

a, The sequence of cpGFP from the 5-HT1.0 sensor, sfGFP, and mClover3 are aligned. Amino acids in the cpGFP chose for optimization are labeled with light green color, and the mutations adopted by the 5-HT1.0 sensor are indicated with red stars.

Extended Data Fig. 3 The amino acid sequence of 5-HT1.0.

a, Schematic representation of the 5-HT1.0 structure. For simplicity, TM1-4, TM7, and H8 are not shown. b, The amino acid sequence of the 5-HT1.0 sensor after three steps of evolution. The mutated amino acids in cpGFP (cpGFP from GCaMP6s, see Chen, T.W., et al. 2013.) are indicated with red stars.

Extended Data Fig. 4 Further characterization of GRAB5-HT in cultured HEK293T cells and rat cortical neurons.

a, Representative fluorescence and pseudocolor images of HEK293T cells expressing 5-HT1.0 or 5-HTmut before (left) and after (right) application of 10 μM 5-HT. Similar results were observed for more than 10 cells. Scale bar, 20 μm. b,c, Representative fluorescence traces and group summary of the peak response in HEK293T cells expressing 5-HT1.0 or 5-HTmut; n = 14 and 15 cells from 3 cultures for 5-HT1.0 and 5-HTmut group. Two-tailed Student’s t-test was performed. P = 8.18 × 10−12 between 5-HT1.0 and 5-HTmut group. d, 5-HT dose-response curves measured in cells expressing 5-HT1.0 or 5-HTmut, the EC50 for 5-HT1.0 is shown. n = 3 wells per group with 300–500 cells per well. e, Representative normalized fluorescence measured in HEK293T cells expressing 5-HT1.0, EGFP-CAAX, or iGluSnFR during continuous exposure to 488-nm laser (power: 350 μW). f, Summary of the decay time constant calculated from the photobleaching curves shown in (e). n = 10/3, 14/3, and 12/3 for 5-HT1.0, EGFP-CAAX, and iGluSnFR, respectively. Two-tailed Student’s t-test was performed. P = 2.45 × 10−9, 1.90 × 10−9, 3.05 × 10−8, and 7.22 × 10−7 between EGFP-CAAX and iGluSnFR without or with Glu, and 5-HT1.0 without or with 5-HT. P = 4.43 × 10−8 and 7.78 × 10−6 between iGluSnFR without or with Glu and 5-HT1.0 without 5-HT. P = 4.62 × 10−8 and 7.05 × 10−6 between iGluSnFR without or with Glu and 5-HT1.0 with 5-HT. g, Summary of the brightness measured in HEK293T cells expressing 5-HT1.0 or 5-HT2C-EGFP in the absence or presence of 10 μM 5-HT, normalized to the 5-HT2C-EGFP + 5-HT group; n = 3 wells per group with 300–500 cells per well. h,i, Intracellular calcium was measured in cells expressing 5-HT1.0 or the 5-HT2C receptor and loaded with the red fluorescent calcium dye Cal590. Representative traces are shown in (h), and the peak responses are plotted against 5-HT concentration in (i); n = 15/3 for each group. j,k, Fluorescence response of 5-HT1.0 expressing cells to 5-HT perfusion for two hours. Representative fluorescence images (j) and the summary data (k) showing the response to 10 μM 5-HT applied at 30 min intervals to cells expressing 5-HT1.0; n = 3 wells per group with 100-300 cells per well. Scale bar, 20 μm. F4,10 = 0.888, P = 0.505 for 0 min, 30 min, 60 min, 90 min and 120 min by one-way ANOVA. l, Left, the Gs-coupled cAMP level was detected by pink-Flamindo with or without 5-HT1.0 sensor expression. The exemplar fluorescence response traces of pink-Flamindo without (top) or with 5-HT1.0 sensor (bottom) expression, when treated with 50 μM 5-HT or 50 μM 5-HT + 10 μM Forskolin. Right, quantification data for left. n = 23/3, 23 cells from 3 cultures for each group. Two-tailed Student’s t-test was performed. P = 0.084 and P = 0.488 for 5-HT and 5-HT + FSK group. m, Buffering effects of the 5-HT1.0 sensor by luciferase complementation assay. Luminescence signals were measured when treated with different concentrations of 5-HT (left) or 5-HT2C receptor specific agonist CP809101 (right) with or without co-expression of 5-HT1.0 sensor with 5-HT2C receptor. The luminescence signal of cells treated with the control buffer is normalized to 1. Data of 5-HT induced G-protein signaling in 5-HT2C receptor expression group were re-plotted from Fig. 1 f. n = 3 wells per group with 100-300 cells per well. Two-tailed Student’s t-test was performed. P = 0.693, 0.0402, 0.993, 0.340, 0.0618, 0.0691 and 0.127 between 5-HT1.0 and 5-HT1.0 + 5-HT2C with 10−4, 10−5, 10−6, 10−7, 10−8, 10−9, and 10−10 M 5-HT. P = 0.733, 0.801, 0.346, 0.998, 0.304 and 0.380 between 5-HT1.0 and 5-HT1.0 + 5-HT2C with 10−4, 10−5, 10−6, 10−7, 10−8 and 10−9 M CP809101. n, Cultured rat cortical neurons expressing the 5-HTmut sensor were imaged before (left) and after (middle) 5-HT application. These insets in the left and middle fluorescence images show the region with increased contrast. The pseudocolor image on the right shows the change in fluorescence of 5-HTmut in response to 10 μM 5-HT. Similar results were observed for more than 10 neurons. Scale bar, 20 μm. o,p, Representative trace (o) and group summary (p) of cultured neurons expressing 5-HT1.0 in response to indicated compounds at 10 μM each; in (p), Met was applied where indicated; n = 9/3. Two-tailed Student’s t-test was performed. P = 6.74 × 10−22, 1.09 × 10−22, 1.27 × 10−21, 3.33 × 10−22, and 0.0939 between 5-HT1st and DA, NE, His, ACh and 5-HT2nd. P = 1.97 × 10−11 between 5-HT2nd and Met. Data are shown as the mean±s.e.m. in b-d, f, g, i, k-m, p, with the error bars or shaded regions indicating s.e.m., *p < 0.05, ** p < 0.01, ***p < 0.001, and n.s., not significant.

Source data

Extended Data Fig. 5 Probing endogenous 5-HT release in mouse brain slices.

a, Schematic diagram depicting the acute mouse brain slice preparation, with AAV-mediated expression of 5-HT1.0 in the hippocampus. b, Representative fluorescence images of the 5-HT1.0 sensor expressed in the mouse hippocampal neurons of brain slices in ACSF (left) and 50 μM 5-HT (right). Similar results were observed from 4 slices. Scale bar, 50 μm. c, A magnified view of the rectangular region in (b) showing the 5-HT1.0 sensor response to exogenously applied 50 μM 5-HT; left, fluorescence image; right, corresponding pseudocolor image indicating ΔF/F0. The arrowheads indicate somata. Scale bar, 15 μm. d, Representative traces (left) and quantification (right) of peak ΔF/F0 of the 5-HT1.0 sensor in response to 50 μM 5-HT from a single soma or neurite (n = 4 slices from 1 mouse). Two-tailed Student’s t-test was performed. P = 0.0226 between soma and neurite. e, Left, schematic diagram depicting the acute mouse brain slice preparation, with AAV-mediated expression of 5-HT1.0 in the DRN. Middle and right, fluorescence traces (middle) and group data (right) of the change in 5-HT1.0 fluorescence in response to 10 electrical stimuli applied at the indicated frequencies; n = 7 slices from 5 mice. f, Summary of the change in 5-HT1.0 fluorescence in response to 6 trains of electrical stimuli (20 pulses at 20 Hz) delivered at 5-min intervals. The responses are normalized to the first train; n = 8 slices from 5 mice. F5,42 = 1.18, P = 0.335 for 0 min, 5 min, 10 min, 15 min, 20 min, and 25 min by one-way ANOVA. g,h, Representative fluorescence image, pseudocolor images (g), fluorescence traces (h, left), and group data (h, right) of 5-HT1.0 fluorescence in response to perfusion of 5-HT, 5-HT + Halo, and 5-HT + Met; n = 4 slices from 3 mice for each group. Two-tailed Student’s t-test was performed. P = 0.0816 between 5-HT and Halo. P = 0.00297 between 5-HT and Met. i, Left, representative FSCV data of 5-HT release in DRN. A specific 5-HT waveform (0.2 V to 1.0 V and ramped down to −0.1 V, and back to 0.2 V at a scan rate of 1000 V/s) was applied to the CFME at a frequency of 10 Hz. Right, current vs time traces are extracted at a horizontal white dashed line shows an immediate increase in 5-HT response after electrical stimulation (20 pulses, 2 ms pulse width, 64 Hz). A cyclic voltammogram (inset) is extracted at the vertical black dashed line shows oxidation and reduction peaks at 0.8 V and 0 V, respectively. j, Left, group data of fluorescence response in 5-HT1.0-expressing DRN neurons to electrical stimuli with varied frequencies delivered at 20 pulses. Right, average data of peak 5-HT concentration measured by FSCV at varied stimulating frequencies delivered at 20 pulses; n = 11 neurons from 9 mice. Data are shown as the mean±s.e.m. in d-f, h, j, with the error bars or shaded regions indicating s.e.m., *p < 0.05, ** p < 0.01, ***p < 0.001, and n.s., not significant.

Source data

Extended Data Fig. 6 Probing endogenous 5-HT release in Drosophila in vivo.

a, Schematic drawing showing in vivo two-photon imaging of a Drosophila, with the stimulating electrode positioned near the mushroom body (MB). b,c, Representative pseudocolor images (b), fluorescence traces, and group summary (c) of the change in 5-HT1.0 fluorescence in the MB horizontal lobe in response to 40 electrical stimuli at 15 Hz in control (saline) or 10 μM Met; n = 9 flies for each group. Two-tailed Student’s t-test was performed. P = 2.36 × 10-5 between saline and Met. Scale bar, 10 μm. d, Fluorescence images measured in the MB of flies expressing 5-HT1.0 or 5-HTmut; the β’ lobe is indicated. Scale bar, 10 μm. e-i, Representative pseudocolor images (e), fluorescence traces (f–h), and group summary (i) of 5-HT1.0 and 5-HTmut in the MB β’ lobe measured in response to a 1-s odor application, a 0.5-s body shock, and application of 100 μM 5-HT; n = 14, 12 and 10 flies for 5-HT1.0 group under odor, body shock and perfusion conditions; n = 9, 5 and 9 flies for 5-HTmut group under odor, body shock and perfusion conditions. Two-tailed Student’s t-test was performed. P = 1.14 × 10−5, P = 0.00273, P = 8.93 × 10−5 between 5-HT1.0 and 5-HTmut under odor, body shock and perfusion conditions. j,k, Quantification data of area under the calcium transient curves (k) and the τon, τoff (j) in the main Fig. 2r,s; n = 11 and 10 flies for 5-HT1.0+ and 5-HT1.0- group. Two-tailed Student’s t-test was performed. P = 0.497 for calcium signal between two groups. P = 0.710 for τon and P = 0.307 for τoff. Data are shown as the mean ± s.e.m. in c, i-k, with the error bars or shaded regions indicating s.e.m., *p < 0.05, ** p < 0.01, ***p < 0.001, and n.s., not significant.

Source data

Supplementary information

Reporting Summary

Supplementary Video 1

GRAB5-HT reports sensory-relevant 5-HT release in Drosophila.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wan, J., Peng, W., Li, X. et al. A genetically encoded sensor for measuring serotonin dynamics. Nat Neurosci 24, 746–752 (2021). https://doi.org/10.1038/s41593-021-00823-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-021-00823-7

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