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.

A light-gated potassium channel for sustained neuronal inhibition

Abstract

Currently available inhibitory optogenetic tools provide short and transient silencing of neurons, but they cannot provide long-lasting inhibition because of the requirement for high light intensities. Here we present an optimized blue-light-sensitive synthetic potassium channel, BLINK2, which showed good expression in neurons in three species. The channel is activated by illumination with low doses of blue light, and in our experiments it remained active over (tens of) minutes in the dark after the illumination was stopped. This activation caused long periods of inhibition of neuronal firing in ex vivo recordings of mouse neurons and impaired motor neuron response in zebrafish in vivo. As a proof-of-concept application, we demonstrated that in a freely moving rat model of neuropathic pain, the activation of a small number of BLINK2 channels caused a long-lasting (>30 min) reduction in pain sensation.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Engineering and characterization of BLINK2.
Fig. 2: BLINK2 expression in rat hippocampal neurons.
Fig. 3: BLINK2-mediated silencing of tonic firing activity in mouse DRN neurons.
Fig. 4: BLINK2 expression and functional silencing in zebrafish.
Fig. 5: BLINK2-mediated reversal of chemotherapy-induced neuropathic pain in rats.

Similar content being viewed by others

Data availability

Raw data generated and analyzed during the current study are available from the corresponding author on reasonable request. Data have been deposited under the following accession codes: AddGene 117075; GenBank submission MH937726. Source data for Fig. 1 and Supplementary Fig. 9 are available online.

References

  1. Han, X. & Boyden, E. S. Multiple-color optical activation, silencing, and desynchronization of neural activity, with single-spike temporal resolution. PLoS One 2, e299 (2007).

    PubMed  PubMed Central  Google Scholar 

  2. Chow, B. Y. et al. High-performance genetically targetable optical neural silencing by light-driven proton pumps. Nature 463, 98–102 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Zhang, F. et al. Multimodal fast optical interrogation of neural circuitry. Nature 446, 633–639 (2007).

    CAS  PubMed  Google Scholar 

  4. Chuong, A. S. et al. Noninvasive optical inhibition with a red-shifted microbial rhodopsin. Nat. Neurosci. 17, 1123–1129 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Wietek, J. et al. Conversion of channelrhodopsin into a light-gated chloride channel. Science 344, 409–412 (2014).

    CAS  PubMed  Google Scholar 

  6. Berndt, A., Lee, S. Y., Ramakrishnan, C. & Deisseroth, K. Structure-guided transformation of channelrhodopsin into a light-activated chloride channel. Science 344, 420–424 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Govorunova, E. G., Sineshchekov, O. A., Janz, R., Liu, X. & Spudich, J. L. Natural light-gated anion channels: a family of microbial rhodopsins for advanced optogenetics. Science 349, 647–650 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Wietek, J. et al. An improved chloride-conducting channelrhodopsin for light-induced inhibition of neuronal activity in vivo. Sci. Rep. 5, 14807 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Inoue, K., Kato, Y. & Kandori, H. Light-driven ion-translocating rhodopsins in marine bacteria. Trends Microbiol. 23, 91–98 (2015).

    CAS  PubMed  Google Scholar 

  10. Mattis, J. et al. Principles for applying optogenetic tools derived from direct comparative analysis of microbial opsins. Nat. Methods 9, 159–172 (2011).

    PubMed  PubMed Central  Google Scholar 

  11. Alfonsa, H. et al. The contribution of raised intraneuronal chloride to epileptic network activity. J. Neurosci. 35, 7715–7726 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Raimondo, J. V., Kay, L., Ellender, T. J. & Akerman, C. J. Optogenetic silencing strategies differ in their effects on inhibitory synaptic transmission. Nat. Neurosci. 15, 1102–1104 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Mahn, M., Prigge, M., Ron, S., Levy, R. & Yizhar, O. Biophysical constraints of optogenetic inhibition at presynaptic terminals. Nat. Neurosci. 19, 554–556 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Berndt, A. et al. Structural foundations of optogenetics: determinants of channelrhodopsin ion selectivity. Proc. Natl. Acad. Sci. USA 113, 822–829 (2016).

    CAS  PubMed  Google Scholar 

  15. Kaila, K., Price, T. J., Payne, J. A., Puskarjov, M. & Voipio, J. Cation-chloride cotransporters in neuronal development, plasticity and disease. Nat. Rev. Neurosci. 15, 637–654 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Szabadics, J. et al. Excitatory effect of GABAergic axo-axonic cells in cortical microcircuits. Science 311, 233–235 (2006).

    CAS  PubMed  Google Scholar 

  17. Christie, J. M. Phototropin blue-light receptors. Annu. Rev. Plant. Biol. 58, 21–45 (2007).

    CAS  PubMed  Google Scholar 

  18. Christie, J. M., Gawthorne, J., Young, G., Fraser, N. J. & Roe, A. J. LOV to BLUF: flavoprotein contributions to the optogenetic toolkit. Mol. Plant 5, 533–544 (2012).

    PubMed  Google Scholar 

  19. Plugge, B. et al. A potassium channel protein encoded by chlorella virus PBCV-1. Science 287, 1641–1644 (2000).

    CAS  PubMed  Google Scholar 

  20. Cosentino, C. et al. Engineering of a light-gated potassium channel. Science 348, 707–710 (2015).

    CAS  PubMed  Google Scholar 

  21. Gradinaru, V. et al. Molecular and cellular approaches for diversifying and extending optogenetics. Cell 141, 154–165 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Zuzarte, M. et al. Intracellular traffic of the K+ channels TASK-1 and TASK-3: role of N- and C-terminal sorting signals and interaction with 14-3-3 proteins. J. Physiol. (Lond.) 587, 929–952 (2009).

    CAS  Google Scholar 

  23. Sottocornola, B. et al. The potassium channel KAT1 is activated by plant and animal 14-3-3 proteins. J. Biol. Chem. 281, 35735–35741 (2006).

    CAS  PubMed  Google Scholar 

  24. Sottocornola, B. et al. 14-3-3 proteins regulate the potassium channel KAT1 by dual modes. Plant Biol. (Stuttg.) 10, 231–236 (2008).

    CAS  Google Scholar 

  25. Saponaro, A. et al. Fusicoccin activates KAT1 channels by stabilizing their interaction with 14-3-3 proteins. Plant Cell 29, 2570–2580 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Pagliuca, C. et al. Molecular properties of Kcv, a virus encoded K+ channel. Biochemistry 46, 1079–1090 (2007).

    CAS  PubMed  Google Scholar 

  27. Marcello, E., Gardoni, F., Di Luca, M. & Pérez-Otaño, I. An arginine stretch limits ADAM10 exit from the endoplasmic reticulum. J. Biol. Chem. 285, 10376–10384 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhao, S. et al. Cell type–specific channelrhodopsin-2 transgenic mice for optogenetic dissection of neural circuitry function. Nat. Methods 8, 745–752 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Giorgi, A. et al. Brain-wide mapping of endogenous serotonergic transmission via chemogenetic fMRI. Cell Rep. 21, 910–918 (2017).

    CAS  PubMed  Google Scholar 

  30. Mlinar, B., Montalbano, A., Piszczek, L., Gross, C. & Corradetti, R. Firing properties of genetically identified dorsal raphe serotonergic neurons in brain slices. Front. Cell. Neurosci. 10, 195 (2016).

    PubMed  PubMed Central  Google Scholar 

  31. Perkins, K. L. Cell-attached voltage-clamp and current-clamp recording and stimulation techniques in brain slices. J. Neurosci. Methods 154, 1–18 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Pudasaini, A., El-Arab, K. K. & Zoltowski, B. D. LOV-based optogenetic devices: light-driven modules to impart photoregulated control of cellular signaling. Front. Mol. Biosci. 2, 18 (2015).

    PubMed  PubMed Central  Google Scholar 

  33. Baier, H. & Scott, E. K. Genetic and optical targeting of neural circuits and behavior—zebrafish in the spotlight. Curr. Opin. Neurobiol. 19, 553–560 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Böhm, U. L. et al. CSF-contacting neurons regulate locomotion by relaying mechanical stimuli to spinal circuits. Nat. Commun. 7, 10866 (2016).

    PubMed  PubMed Central  Google Scholar 

  35. Flanagan-Steet, H., Fox, M. A., Meyer, D. & Sanes, J. R. Neuromuscular synapses can form in vivo by incorporation of initially aneural postsynaptic specializations. Development 132, 4471–4481 (2005).

    CAS  PubMed  Google Scholar 

  36. Yoo, S. K., Starnes, T. W., Deng, Q. & Huttenlocher, A. Lyn is a redox sensor that mediates leukocyte wound attraction in vivo. Nature 480, 109–112 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Li, Y. et al. Dorsal root ganglion neurons become hyperexcitable and increase expression of voltage-gated T-type calcium channels (Cav3.2) in paclitaxel-induced peripheral neuropathy. Pain 158, 417–429 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Moutal, A. et al. Blocking CRMP2 SUMOylation reverses neuropathic pain. Mol. Psychiatry https://doi.org/10.1038/mp.2017.117 (2017).

    Google Scholar 

  39. Wiegert, J. S., Mahn, M., Prigge, M., Printz, Y. & Yizhar, O. Silencing neurons: tools, applications, and experimental constraints. Neuron 95, 504–529 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Finnerup, N. B. et al. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol. 14, 162–173 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Stachniak, T. J., Ghosh, A. & Sternson, S. M. Chemogenetic synaptic silencing of neural circuits localizes a hypothalamus→midbrain pathway for feeding behavior. Neuron 82, 797–808 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Bryksin, A. V. & Matsumura, I. Overlap extension PCR cloning: a simple and reliable way to create recombinant plasmids. Biotechniques 48, 463–465 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Piccoli, G. et al. Proteomic analysis of activity-dependent synaptic plasticity in hippocampal neurons. J. Proteome Res. 6, 3203–3215 (2007).

    CAS  PubMed  Google Scholar 

  44. Romani, G. et al. A virus-encoded potassium ion channel is a structural protein in the chlorovirus Paramecium bursaria chlorella virus 1 virion. J. Gen. Virol. 94, 2549–2556 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Westerfield, M. The Zebrafish Book: A Guide for the Laboratory Use of Zebrafish (Danio rerio) 4th ed. (Univ. of Oregon Press, Eugene, OR, 2000).

  46. Malinverno, M. et al. Synaptic localization and activity of ADAM10 regulate excitatory synapses through N-cadherin cleavage. J. Neurosci. 30, 16343–16355 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Suster, M. L., Sumiyama, K. & Kawakami, K. Transposon-mediated BAC transgenesis in zebrafish and mice. BMC Genomics 10, 477 (2009).

    PubMed  PubMed Central  Google Scholar 

  48. Polomano, R. C., Mannes, A. J., Clark, U. S. & Bennett, G. J. A painful peripheral neuropathy in the rat produced by the chemotherapeutic drug, paclitaxel. Pain 94, 293–304 (2001).

    CAS  PubMed  Google Scholar 

  49. Yaksh, T. L. & Rudy, T. A. Chronic catheterization of the spinal subarachnoid space. Physiol. Behav. 17, 1031–1036 (1976).

    CAS  PubMed  Google Scholar 

  50. Chaplan, S. R., Bach, F. W., Pogrel, J. W., Chung, J. M. & Yaksh, T. L. Quantitative assessment of tactile allodynia in the rat paw. J. Neurosci. Methods 53, 55–63 (1994).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank S. Guazzi and M. Festa for technical help with cloning and zebrafish expression. We acknowledge M. Pesce and A. Gino for help with immunohistochemistry. pENN-AAV-hSyn-Cre-WPRE-hGH was a gift from J.M. Wilson (Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA). This work was supported by the 2016 Schaefer Research Scholars Program of Columbia University (to A. Moroni), MIUR PRIN (Programmi di Ricerca di Rilevante Interesse Nazionale; 494 2015, 2015795S5W to A. Moroni), the European Research Council (ERC; 2015 Advanced Grant 495 (AdG) n. 695078 noMAGIC to A. Moroni and G.T.), DFG priority program SPP1926 (to G.T.), the Fondazione Istituto Italiano di Tecnologia (to A.L., A.C., A.J.B. and R.T.), and AIRAlzh Onlus-COOP Italia (fellowship to S.P.).

Author information

Authors and Affiliations

Authors

Contributions

L.A. designed and prepared channel constructs, performed whole-cell patch-clamp experiments in vitro and analyzed the data; A.S. contributed to the design of the final BLINK2 clone; A.P. conducted and analyzed some electrophysiological recordings in vitro; G.R. produced the anti-BLINK2 antibody; G.T. and H.M.C. performed the single-channel in vitro patch experiments and analyzed the data; S.P., E.M. and M.D.L. designed, conducted and analyzed the immunolocalization experiments in rat primary neurons; A.L. designed, performed and analyzed the ex vivo mouse patch-clamp experiments; A.J.B. and A.C. designed and produced the BLINK2 viral constructs; A.L., N.B. and A.J.B. performed intracerebral viral injections; N.B. and M.P. carried out immunofluorescence analysis; E.R., V.B., S.A., F.S., S.M., M.B. and F.D.B. designed, performed and analyzed the zebrafish experiments; K.K. and G.T. designed, performed and analyzed the artificial bilayer measurements; S.L., A. Moutal, Y.J. and R.K. designed, performed and analyzed the pain experiments in rats; R.T. designed and supervised the electrophysiological ex vivo experiments and the production of BLINK2 and GFP-control viral constructs; A. Moroni conceived the study, coordinated research and wrote the manuscript; and G.T., F.D.B., E.M., M.B., R.K. and R.T. contributed to the writing.

Corresponding authors

Correspondence to Raffaella Tonini or Anna Moroni.

Ethics declarations

Competing interests

The authors declare no competing interests

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Comparison of dark current levels in BLINK2-transfected and control COS7 cells.

Currents recorded in the dark at –60 mV (N = 13 cells each) are shown normalized for cell capacitance (pA/pF) in the following conditions: untransfected, + 5 mM BaCl2, GFP-transfected and BLINK2-transfected cells. The mean values of the experimental groups Untransfected, GFP, and BLINK2 show no significant differences (P = 0.64). Only in barium-treated cells the current values were significantly smaller than in untransfected, GFP and BLINK2 (P = 0.001, 0.0001 and 0.003, respectively). Significance was calculated by one-way ANOVA and Tukey post hoc test.

Supplementary Figure 2 Spectral sensitivity of BLINK2 transfected in HEK293T cells.

(a) Representative whole-cell current traces recorded in HEK293T cells expressing BLINK2 channel. Voltage steps from + 80 to –100 mV, in the dark, 3 min after exposing the cell to 630-nm red light, 530-nm green light, 500-nm green light, 455-nm blue light and after the addition of 5 mM BaCl2 in the external solution. (b) Current–voltage relationship of measurement in (a) in the dark (filled black circles), in 630-nm light (filled red circles), in 530-nm light (filled green circles), in 500-nm light (filled light-green circles), in 455-nm light (filled blue circles) and after the addition of 5 mM BaCl2 in the external solution (open blue circles). Light of all wavelengths was provided at 90 μW/mm2 intensity. The effect of each wavelength was tested versus the effect of blue light (455 nm) in 3 independent experiments.

Supplementary Figure 3 Single-channel i/V curve of BLINK2.

BLINK2 single-channel currents (blue symbols) plotted as a function of voltage. Currents for BLINK1 (white circle) and KcvPBCV1 (black circle) are shown for comparison. Each data point is the average of n = 3 measurements performed in cell-attached configuration (BLINK2) or after insertion of purified proteins in planar lipid bilayers (BLINK1 and KcvPBCV1) in n = 3 independent experiments. Standard deviations are within the dimensions of the symbols. Recordings were performed in the following conditions: 103 mM K+ in the pipette solution for BLINK2 and symmetrical 100 mM K+ (BLINK1 and KcvPBCV1).

Supplementary Figure 4 Validation of the immunofluorescence-based antibody assay and targeting of BLINK2 to the synapses.

(a) Representative staining of BLINK2 surface and total expression. After fixation, rat hippocampal neurons, infected with AAV1/2-hSyn-BLINK2-IRES-eGFP expressing BLINK2 and GFP, were stained with the MAP2 (microtubule-associated protein 2) antibody and the 8D6 monoclonal antibody for BLINK2, without any permeabilization (upper panels). In the lower panels the staining of BLINK2 and MAP2 is shown after permeabilization with Triton X-100. Scale bar, 10 μm. (b) Representative dendrites showing the staining of BLINK2 and synaptic markers. BLINK2-expressing hippocampal neurons were stained with the BLINK2 8D6 monoclonal antibody (magenta) and Bassoon (turquoise), a presynaptic protein, or PSD-95 (turquoise), a postsynaptic marker. In merged images the red arrowheads indicate partial colocalization (white) of BLINK2 signal with the synaptic markers in a few synapses. Scale bar, 5 μm.

Supplementary Figure 5 Light exposure did not silence tonic firing activity of neurons from mouse DRN injected with a GFP control virus.

(a) (Left) Diagram of injection site in the DRN. (Right) Sample confocal image showing expression of the AAV1/2-hSyn-eGFP virus in the DRN (green, GFP; gray, DAPI) (image representative of n = 3). Scale bars, 200 µm or 40 µm (inset). AQ, aqueduct. (b) (Top, left) Representative cell-attached voltage clamp recording of firing response before and after 60-s blue light illumination (blue bar) (n = 7 independent recordings). (Top right) Expanded view of the recording period defined by the red dotted box. (Bottom left) Time course of the effect of 60-s blue light stimulation (blue bar) on discharge firing rate (5-s binning). (Bottom right) Summary plots indicate mean firing discharge rate 2 min prior to light (Beforelight) and 2 min after lights-off (Afterlight 0–2′) (Beforelight, 3.4 ± 0.8 Hz; Afterlight 0–2′, 4.1 ± 1.0 Hz n = 7, P = 0.46, t = 0.78, df = 6, two-sided paired t-test). Data are presented as mean ± s.e.m.

Supplementary Figure 6 Passive and active membrane properties in mouse DRN BLINK2-expressing and GFP-expressing neurons.

(a) (Left) Representative current traces. (Right) Membrane resistance (Rm) was measured in response to –5-mV steps in voltage clamp configuration, and did not significantly change between BLINK2 and CTRL group (Rm, BLINK2 386 ± 35 MΩ, n = 14; CTRL, 308 ± 35 MΩ, n = 15; P = 0.12, t = 1.59, df = 27, two-sided unpaired t-test). (b) (Left) Representative current clamp traces in response to fixed current injections. Action potential firing was generated by 2-ms current pulses at 1.2 nA. (Right) resting membrane potential (R.M.P.) and the action potential threshold did not differ between BLINK2 and CTRL groups (R.M.P.: BLINK2, –50.6 ± 1.8 mV n = 7; CTRL, –50.9 ± 1,8 mV, n = 15, P = 0.90, t = 0.13, df = 20, two-sided unpaired t-test; Threshold: BLINK2, –31.1 ± 2.3 mV, n = 8; CTRL, –35.4 ± 1.9 mV, n = 15, P = 0.18, t = 1.39, df = 21, two-sided unpaired t-test). Action potential threshold was determined from the second derivative of the spike waveform. Data are presented as mean ± s.e.m. obtained from: BLINK2, independent recordings, n = 14, n = 6 mice; CTRL, independent recordings, n = 15, n = 4 mice.

Supplementary Figure 7 Effect of long-term expression of BLINK2 in neurons.

(Left) Representative confocal images from coronal brain sections obtained from AAV1/2-hSyn-BLINK2-IRES-eGFP injected animals and immunoreacted with anti-GFP antibody. (Right) Plot indicates the number of GFP-expressing neurons scored in the region of the DRN surrounding the injection site 2 (n = 12), 4 (n = 18), and 8 (n = 12) weeks after injection. Points represent the number of GFP-expressing neurons in each volume analyzed. Quantification analysis was performed in a blinded manner and sample identity was not revealed until correlation was completed. The number of virus-infected BLINK2-expressing cells was scored assessing the number of GFP-positive (GFP+) cells in 10 × confocal image sections. GFP-positive neurons were counted on 3 consecutive 50 μm coronal sections within two distinct volumes of 200 μm × 200 μm × 15 μm chosen in the DRN region surrounding the injection site (n = 2 mice at 2 and 8 weeks post-injection; n = 3 mice at 4 weeks post-injection). Statistical significance was calculated with one-way ANOVA with multiple comparison and Tukey’s P value correction (ns: P > 0.05). Scale bar, 200 µm. AQ, aqueduct. Data are presented as mean ± s.d.

Supplementary Figure 8 Optogenetic activation of eNpHR3.0 silences tonic firing activity of mouse DRN neurons.

(Top) Diagram represents virus injection site (AAV1-hSyn-Cre + AAV5-EF1α-DIO-eNpHR3.0-eYFP). (Bottom) Confocal image showing expression of eNpHR3.0-eYFP in the mouse DRN (green, YFP; gray, DAPI; scale bar, 200 µm; AQ, aqueduct) (n = 3 mice). (b) (Top) Representative cell-attached voltage clamp recording of firing response before and after 60 s of yellow light illumination (yellow bar) (independent recordings, n = 9, n = 3 mice). (Bottom) Time course of the effect on the discharge rate of 60 s of yellow light stimulation (yellow bar) on this representative recording; red horizontal bar represents the threshold (Th) defined as the mean discharge rate minus two times the s.d.; mean firing rate is calculated on values (5-s binning) computed over 1 min prior to light illumination (see also Methods). (c) (Left) Average time course of the effect of 60 s of yellow light stimulation (yellow bar) on firing discharge rate (5-s binning). (Right) Summary plots indicate mean firing discharge rate 2 min prior to light (Beforelight), during 1 min of light (Light) and 2 min after light (Afterlight 0–2′) (n = 9; Repeated Measures 1 Way ANOVA (RM1WA), F8,2 = 6, P = 0.019; post hoc, Beforelight versus Light, P = 0.047; Beforelight versus Afterlight 0–2′, P = 0.62; with multiple comparison and Dunnett’s P value correction). (d) Bar graph indicating the duration of neuronal silencing (time below threshold; Timeth) induced by eNpHR3.0 or BLINK2 activation (Timeth: BLINK2 versus eNpHR3.0, P = 0.03, Mann–Whitney U = 18, two-sided Mann–Whitney test). The TimeTh of BLINK2 has been calculated from the dataset included in Fig. 3c. Data are presented as mean ± s.e.m.

Supplementary Figure 9 Light controls the behavior of zebrafish embryos transiently expressing BLINK2: comparison with BLINK1 and BLINK2 Q513D.

(a) Altered escape response in 2-d-old zebrafish embryos expressing BLINK2 (squares) or GFP (circles) (both RNAs injected at 200 pg/embryo), measured in embryos kept in the dark (black) or after 60 min of exposure to blue light (465 nm, 85 μW/mm2) (blue). The escape response was considered altered when one or two touches were not sufficient to elicit it. BLINK2 Q513D RNA was used for the experiments shown in this graph. Similar results were obtained when BLINK2 RNA was injected (data not shown); mean and s.e.m. were calculated on three (GFP) and four (BLINK2) different experiments. Total number of embryos (n) is 49 and 119, respectively. (b) BLINK120 versus BLINK2. The graph shows the number of stimulations required in order to elicit an escape response in each embryo, injected with either BLINK1 (dots) or BLINK2 (squares) RNA. BLINK2 Q513D RNA was used for the experiments shown in this graph. Measurements were performed after 30 min of exposure to blue light. The blue dashed line highlights the maximum number of tactile stimuli (8 touches) required to elicit an escape response in BLINK1 injected embryos. (c) BLINK2 versus BLINK2 Q513Q. Reversibility in dark and kinetics of the light effect on the escape response of 2-d-old embryos expressing BLINK2 (continuous line) and BLINK2 Q513D (dashed line) Blue light was turned on at time 0, after the first measurement. After 60 min of exposure, the light was turned off for 30 min and then turned on again (as indicated by blue and black bars above the graph). The response to mechanical stimulation was checked and recorded every 15 min. The total number of embryos in each group was n = 86 (BLINK2) and n = 98 (BLINK2 Q513D) from 3 independent experiments. For a, we performed two-tailed t-tests comparing GFP versus BLINK2 in the dark, GFP versus BLINK2 in blue light, GFP dark versus blue light, BLINK2 dark versus blue light. Statistically significant results are shown with asterisks (P = 0.028 and 0.0023 for BLINK2 dark/light and GFP/BLINK2 light, respectively).

Source data.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9, Supplementary Table 1 and Supplementary Methods

Reporting Summary

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Alberio, L., Locarno, A., Saponaro, A. et al. A light-gated potassium channel for sustained neuronal inhibition. Nat Methods 15, 969–976 (2018). https://doi.org/10.1038/s41592-018-0186-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-018-0186-9

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