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.

All-optical electrophysiology in mammalian neurons using engineered microbial rhodopsins

Abstract

All-optical electrophysiology—spatially resolved simultaneous optical perturbation and measurement of membrane voltage—would open new vistas in neuroscience research. We evolved two archaerhodopsin-based voltage indicators, QuasAr1 and QuasAr2, which show improved brightness and voltage sensitivity, have microsecond response times and produce no photocurrent. We engineered a channelrhodopsin actuator, CheRiff, which shows high light sensitivity and rapid kinetics and is spectrally orthogonal to the QuasArs. A coexpression vector, Optopatch, enabled cross-talk–free genetically targeted all-optical electrophysiology. In cultured rat neurons, we combined Optopatch with patterned optical excitation to probe back-propagating action potentials (APs) in dendritic spines, synaptic transmission, subcellular microsecond-timescale details of AP propagation, and simultaneous firing of many neurons in a network. Optopatch measurements revealed homeostatic tuning of intrinsic excitability in human stem cell–derived neurons. In rat brain slices, Optopatch induced and reported APs and subthreshold events with high signal-to-noise ratios. The Optopatch platform enables high-throughput, spatially resolved electrophysiology without the use of conventional electrodes.

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

Figure 1: Nonpumping Arch-derived voltage indicators with improved speed, sensitivity and brightness.
Figure 2: CheRiff is a fast and sensitive blue-shifted channelrhodopsin.
Figure 3: Optopatch enables high-fidelity optical stimulation and recording in cultured neurons.
Figure 4: Homeostatic plasticity of intrinsic excitability in hiPSC-derived neurons probed via Optopatch2.
Figure 5: Optopatch2 in organotypic brain slice.

Accession codes

Accessions

Protein Data Bank

References

  1. Peron, S. & Svoboda, K. From cudgel to scalpel: toward precise neural control with optogenetics. Nat. Methods 8, 30–34 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Petreanu, L., Mao, T., Sternson, S.M. & Svoboda, K. The subcellular organization of neocortical excitatory connections. Nature 457, 1142–1145 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Scanziani, M. & Hausser, M. Electrophysiology in the age of light. Nature 461, 930–939 (2009).

    Article  CAS  PubMed  Google Scholar 

  4. Boulting, G.L. et al. A functionally characterized test set of human induced pluripotent stem cells. Nat. Biotechnol. 29, 279–286 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Furuta, T. et al. Brominated 7-hydroxycoumarin-4-ylmethyls: photolabile protecting groups with biologically useful cross-sections for two photon photolysis. Proc. Natl. Acad. Sci. USA 96, 1193–1200 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kramer, R.H., Fortin, D.L. & Trauner, D. New photochemical tools for controlling neuronal activity. Curr. Opin. Neurobiol. 19, 544–552 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Boyden, E.S., Zhang, F., Bamberg, E., Nagel, G. & Deisseroth, K. Millisecond-timescale, genetically targeted optical control of neural activity. Nat. Neurosci. 8, 1263–1268 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. Looger, L.L. & Griesbeck, O. Genetically encoded neural activity indicators. Curr. Opin. Neurobiol. 22, 18–23 (2012).

    Article  CAS  PubMed  Google Scholar 

  9. Miller, E.W. et al. Optically monitoring voltage in neurons by photo-induced electron transfer through molecular wires. Proc. Natl. Acad. Sci. USA 109, 2114–2119 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Yan, P. et al. Palette of fluorinated voltage-sensitive hemicyanine dyes. Proc. Natl. Acad. Sci. USA 109, 20443–20448 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Vogt, K.E., Gerharz, S., Graham, J. & Canepari, M. Combining membrane potential imaging with l-glutamate or GABA photorelease. PLoS ONE 6, e24911 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Tsuda, S. et al. Probing the function of neuronal populations: combining micromirror-based optogenetic photostimulation with voltage-sensitive dye imaging. Neurosci. Res. 75, 76–81 (2013).

    Article  PubMed  Google Scholar 

  13. Lim, D.H. et al. In vivo large-scale cortical mapping using channelrhodopsin-2 stimulation in transgenic mice reveals asymmetric and reciprocal relationships between cortical areas. Front. Neural Circuits 6, 11 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Gong, Y., Li, J.Z. & Schnitzer, M.J. Enhanced archaerhodopsin fluorescent protein voltage indicators. PLoS ONE 8, e66959 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Klapoetke, N.C. et al. Independent optical excitation of distinct neural populations. Nat. Methods 11, 338–346 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Jin, L. et al. Single action potentials and subthreshold electrical events imaged in neurons with a fluorescent protein voltage probe. Neuron 75, 779–785 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kralj, J.M., Douglass, A.D., Hochbaum, D.R., Maclaurin, D. & Cohen, A.E. Optical recording of action potentials in mammalian neurons using a microbial rhodopsin. Nat. Methods 9, 90–95 (2012).

    Article  CAS  Google Scholar 

  18. Maclaurin, D., Venkatachalam, V., Lee, H. & Cohen, A.E. Mechanism of voltage-sensitive fluorescence in a microbial rhodopsin. Proc. Natl. Acad. Sci. USA 110, 5939–5944 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sakai, R., Repunte-Canonigo, V., Raj, C.D. & Knöpfel, T. Design and characterization of a DNA-encoded, voltage-sensitive fluorescent protein. Eur. J. Neurosci. 13, 2314–2318 (2001).

    Article  CAS  PubMed  Google Scholar 

  20. Bean, B.P. The action potential in mammalian central neurons. Nat. Rev. Neurosci. 8, 451–465 (2007).

    Article  CAS  PubMed  Google Scholar 

  21. Schoenenberger, P., Grunditz, Å., Rose, T. & Oertner, T.G. Optimizing the spatial resolution of Channelrhodopsin-2 activation. Brain Cell Biol. 36, 119–127 (2008).

    Article  PubMed  Google Scholar 

  22. Johnson, M.T.J. et al. Evaluating methods for isolating total RNA and predicting the success of sequencing phylogenetically diverse plant transcriptomes. PLoS ONE 7, e50226 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Melkonian, M. & Preisig, H.R. A light and electron microscopic study of Scherffelia dubia, a new member of the scaly green flagellates (Prasinophyceae). Nord. J. Bot. 6, 235–256 (1986).

    Article  Google Scholar 

  24. Mattis, J. et al. Principles for applying optogenetic tools derived from direct comparative analysis of microbial opsins. Nat. Methods 9, 159–172 (2012).

    Article  CAS  Google Scholar 

  25. Lin, J.Y., Lin, M.Z., Steinbach, P. & Tsien, R.Y. Characterization of engineered channelrhodopsin variants with improved properties and kinetics. Biophys. J. 96, 1803–1814 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Takahashi, H. et al. Light-addressed single-neuron stimulation in dissociated neuronal cultures with sparse expression of ChR2. Biosystems 107, 106–112 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Fitzsimonds, R.M., Song, H.-j. & Poo, M.-m. Propagation of activity-dependent synaptic depression in simple neural networks. Nature 388, 439–448 (1997).

    Article  CAS  PubMed  Google Scholar 

  28. Foust, A., Popovic, M., Zecevic, D. & McCormick, D.A. Action potentials initiate in the axon initial segment and propagate through axon collaterals reliably in cerebellar Purkinje neurons. J. Neurosci. 30, 6891–6902 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Popovic, M.A., Foust, A.J., McCormick, D.A. & Zecevic, D. The spatio-temporal characteristics of action potential initiation in layer 5 pyramidal neurons: a voltage imaging study. J. Physiol. (Lond.) 589, 4167–4187 (2011).

    Article  CAS  Google Scholar 

  30. Kole, M.H. & Stuart, G.J. Signal processing in the axon initial segment. Neuron 73, 235–247 (2012).

    Article  CAS  PubMed  Google Scholar 

  31. Turrigiano, G., Abbott, L. & Marder, E. Activity-dependent changes in the intrinsic properties of cultured neurons. Science 264, 974–977 (1994).

    Article  CAS  PubMed  Google Scholar 

  32. Desai, N.S., Rutherford, L.C. & Turrigiano, G.G. Plasticity in the intrinsic excitability of cortical pyramidal neurons. Nat. Neurosci. 2, 515–520 (1999).

    Article  CAS  PubMed  Google Scholar 

  33. Grubb, M.S. & Burrone, J. Activity-dependent relocation of the axon initial segment fine-tunes neuronal excitability. Nature 465, 1070–1074 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lambo, M.E. & Turrigiano, G.G. Synaptic and intrinsic homeostatic mechanisms cooperate to increase L2/3 pyramidal neuron excitability during a late phase of critical period plasticity. J. Neurosci. 33, 8810–8819 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Trounson, A., Shepard, K.A. & DeWitt, N.D. Human disease modeling with induced pluripotent stem cells. Curr. Opin. Genet. Dev. 22, 509–516 (2012).

    Article  CAS  PubMed  Google Scholar 

  36. Shcheglovitov, A. et al. SHANK3 and IGF1 restore synaptic deficits in neurons from 22q13 deletion syndrome patients. Nature 503, 267–271 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Akemann, W. et al. Imaging neural circuit dynamics with a voltage-sensitive fluorescent protein. J. Neurophysiol. 108, 2323–2337 (2012).

    Article  CAS  PubMed  Google Scholar 

  38. Cao, G. et al. genetically targeted optical electrophysiology in intact neural circuits. Cell 154, 904–913 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Huys, Q.J., Ahrens, M.B. & Paninski, L. Efficient estimation of detailed single-neuron models. J. Neurophysiol. 96, 872–890 (2006).

    Article  PubMed  Google Scholar 

  40. Williams, J.C. et al. Computational optogenetics: empirically-derived voltage-and light-sensitive channelrhodopsin-2 model. PLoS Comput. Biol. 9, e1003220 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hou, J.H., Venkatachalam, V. & Cohen, A.E. Temporal dynamics of microbial rhodopsin fluorescence reports absolute membrane voltage. Biophys. J. 106, 639–648 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Wainger, B.J. et al. Intrinsic membrane hyperexcitability of amyotrophic lateral sclerosis patient-derived motor neurons. Cell Rep. 7, 1–11 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Higurashi, N. et al. A human Dravet syndrome model from patient induced pluripotent stem cells. Mol. Brain 6, 19 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Badger, J.L., Cordero-Llana, O., Hartfield, E.M. & Wade-Martins, R. Parkinson's disease in a dish–using stem cells as a molecular tool. Neuropharmacology 76, 88–96 (2014).

    Article  CAS  PubMed  Google Scholar 

  45. Marchetto, M.C. et al. A model for neural development and treatment of Rett syndrome using human induced pluripotent stem cells. Cell 143, 527–539 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Auerbach, B.D., Osterweil, E.K. & Bear, M.F. Mutations causing syndromic autism define an axis of synaptic pathophysiology. Nature 480, 63–68 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhao, H., Giver, L., Shao, Z., Affholter, J.A. & Arnold, F.H. Molecular evolution by staggered extension process (StEP) in vitro recombination. Nat. Biotechnol. 16, 258–261 (1998).

    Article  CAS  PubMed  Google Scholar 

  48. Zhao, Y. et al. An expanded palette of genetically encoded Ca2+ indicators. Science 333, 1888–1891 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Cheng, Z. & Campbell, R.E. Assessing the structural stability of designed β-hairpin peptides in the cytoplasm of live cells. ChemBioChem 7, 1147–1150 (2006).

    Article  CAS  PubMed  Google Scholar 

  50. Lanyi, J.K. Proton translocation mechanism and energetics in the light-driven pump bacteriorhodopsin. Biochim. Biophys. Acta 1183, 241–261 (1993).

    Article  CAS  PubMed  Google Scholar 

  51. Lanyi, J.K. Bacteriorhodopsin. Annu. Rev. Physiol. 66, 665–688 (2004).

    Article  CAS  PubMed  Google Scholar 

  52. Kolodner, P., Lukashev, E.P., Ching, Y. & Rousseau, D.L. Electric-field-induced Schiff-base deprotonation in D85N mutant bacteriorhodopsin. Proc. Natl. Acad. Sci. USA 93, 11618–11621 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Ma, D. et al. Role of ER export signals in controlling surface potassium channel numbers. Science 291, 316–319 (2001).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kirkton, R.D. & Bursac, N. Engineering biosynthetic excitable tissues from unexcitable cells for electrophysiological and cell therapy studies. Nat. Commun. 2, 300 (2011).

    Article  CAS  PubMed  Google Scholar 

  56. Park, J. et al. Screening fluorescent voltage indicators with spontaneously spiking HEK cells. PLoS ONE 8, e85221 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Pucihar, G., Kotnik, T. & Miklavčič, D. Measuring the induced membrane voltage with di-8-ANEPPS. J. Vis. Exp. 33, e1659 (2009).

    Google Scholar 

  58. Enami, N. et al. Crystal structures of archaerhodopsin-1 and-2: common structural motif in archaeal light-driven proton pumps. J. Mol. Biol. 358, 675–685 (2006).

    Article  CAS  PubMed  Google Scholar 

  59. Barondeau, D.P., Putnam, C.D., Kassmann, C.J., Tainer, J.A. & Getzoff, E.D. Mechanism and energetics of green fluorescent protein chromophore synthesis revealed by trapped intermediate structures. Proc. Natl. Acad. Sci. USA 100, 12111–12116 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. McCarthy, K.D. & de Vellis, J. Preparation of separate astroglial and oligodendroglial cell cultures from rat cerebral tissue. J. Cell Biol. 85, 890–902 (1980).

    Article  CAS  PubMed  Google Scholar 

  61. Banker, G. & Goslin, K. Culturing Nerve Cells (MIT Press, 1998).

  62. Chen, G., Harata, N.C. & Tsien, R.W. Paired-pulse depression of unitary quantal amplitude at single hippocampal synapses. Proc. Natl. Acad. Sci. USA 101, 1063–1068 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Jiang, M. & Chen, G. High Ca2+ -phosphate transfection efficiency in low-density neuronal cultures. Nat. Protoc. 1, 695–700 (2006).

    Article  CAS  PubMed  Google Scholar 

  64. Stoppini, L., Buchs, P.-A. & Muller, D. A simple method for organotypic cultures of nervous tissue. J. Neurosci. Methods 37, 173–182 (1991).

    Article  CAS  PubMed  Google Scholar 

  65. Mukamel, E.A., Nimmerjahn, A. & Schnitzer, M.J. Automated analysis of cellular signals from large-scale calcium imaging data. Neuron 63, 747–760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank N. Anand, L. Rosenbaum, T. Shen and V. Nathan for technical assistance; A. Douglass, A. Ting, F. Zhang, L. Looger and D. Kim for helpful discussions; and Cellular Dynamics Inc. for technical assistance with hiPSC neurons. This work was supported by the Harvard Center for Brain Science, PECASE award N00014-11-1-0549, US National Institutes of Health (NIH) grant 1-R01-EB012498-01 and New Innovator grant 1-DP2-OD007428, a National Science Foundation (NSF) Graduate Fellowship (D.R.H. and S.L.F.), the Natural Sciences and Engineering Research Council of Canada (Discovery grants to R.E.C. and D.J.H.), the Canadian Institutes of Health Research (R.E.C.) and graduate scholarships from the University of Alberta and Alberta Innovates (Y.Z.). E.S.B. was supported by Defense Advanced Research Projects Agency (DARPA) Living Foundries HR0011-12-C-0068, a New York Stem Cell Foundation (NYSCF) Robertson Neuroscience Investigator Award, an Institution of Engineering and Technology (IET) A.F. Harvey Prize, NIH grants 1R01NS075421, 1R01NS067199, 1DP2OD002002 and 1R01DA029639, the Human Frontiers Science Program, and NSF CAREER Award CBET 1053233 and EFRI 0835878. Work in V.N.M.'s lab was supported by NIH grants R01DC011291 and R01DC013329.

Author information

Authors and Affiliations

Authors

Contributions

D.R.H. designed the Optopatch construct and system. Y.Z. and D.R.H. engineered the QuasArs. D.R.H., N.K. and Y.K.C. engineered CheRiff. D.R.H. and S.L.F. acquired the optical electrophysiology data. C.A.W. developed the low-magnification imaging system. V.K. and J.L.S. designed and prepared the slice experiments. D.R.H., D.M. and A.E.C. analyzed data. P.Z. assisted with measurements of rhodopsin photophysics. J.M.K. screened ion channel blockers on hiPSC-derived neurons. N.S.-M. performed cell culture and sample preparation. G.L.B. performed immunostaining in hiPSC-derived neurons. C.S. assisted with measurements of ArcLight in slice. M.M. and G.K.-S.W. provided the transcriptomic data from which sdChR was mined and phenotyped. D.R.H. and A.E.C. wrote the paper with input from S.L.F., Y.Z. and R.E.C. D.J.H., V.N.M., B.L.S., E.S.B. and R.E.C. supervised aspects of the research. Correspondence regarding directed evolution of Arch should be addressed to R.E.C. (robert.e.campbell@ualberta.ca). A.E.C. conceived of and oversaw the project.

Corresponding author

Correspondence to Adam E Cohen.

Ethics declarations

Competing interests

A.E.C. and J.M.K. are founders of Q-State Biosciences, a company focused on combining all-optical electrophysiology with cell-based models of disease. D.R.H., Y.Z., S.L.F., N.K., J.M.K., D.M., Y.K.C., D.J.H., R.E.C., E.S.B. and A.E.C. have filed patents related to Optopatch.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–23 and Supplementary Tables 1–6 (PDF 3634 kb)

Supplementary Software

The Sub-Nyquist Action Potential Timing (SNAPT) algorithms enable pixel-by-pixel determination of the timing of an action potential. The input is a movie of a neuron repetitively triggered to fire, where the neuron contains a high-speed fluorescent voltage reporter. The output is a movie at a higher frame-rate showing the propagation of the action potential through the neuron. The implementation given here is written for Matlab R2012a, and has been confirmed to run on a personal computer with 14 GB of RAM, running Windows 7 (64-bit). Several of the sub-routines are memory intensive. There are many adjustable parameters in the code, so it is important to run the code one piece at a time, looking at the output at each step and checking that it makes sense. (ZIP 368 kb)

Optopatch single neuron raw data

Raw QuasAr2 fluorescence from a neuron expressing Optopatch2, imaged at 1 kHz frame rate. Fluorescence was excited by whole-field red illumination (640 nm, 300 W/cm2). Activity was induced via whole-field blue light illumination (488 nm, 20 mW/cm2, 10 ms pulses, repeated at 5 Hz). Portions of the intervals between action potentials have been elided to maintain manageable movie size. Movie acquired on an sCMOS camera. (AVI 2020 kb)

Optopatch single neuron trial averaged AP

Same data as in Movie 1, averaged over 98 temporally registered action potentials. The average fluorescence intensity is shown in grayscale in the background, and the change in fluorescence is shown as a colormap. Movie acquired on an sCMOS camera. (AVI 288 kb)

Movie of AP initiation and propagation 1

Movie of an action potential (averaged over n = 203 optically evoked action potentials) in a neuron expressing Optopatch1. Region receiving blue light stimulus is shown in blue. The subthreshold voltage spreads during stimulation, followed by a spike in cell-wide fluorescence which peaked within two frames. Movie acquired on an EMCCD camera. (AVI 836 kb)

Sub-frame movie of AP initiation and propagation 1

Sub-frame interpolated movie showing action potential initiation and propagation in the neuron shown in Movie 3. Region receiving blue light stimulus is shown in blue. Action potential initiation occurs at the distal end of the axon initial segment. The movie was constructed from an average of n = 203 optically evoked action potentials. Movie acquired on an EMCCD camera. (AVI 2208 kb)

Sub-frame movie of AP initiation and propagation 2

Sub-frame interpolated movie showing action potential initiation and propagation in a neuron expressing Optopatch1, with blue light stimulation targeted to the soma. The movie was acquired using the same parameters as in Supplementary Movie 3. The movie was constructed from an average of n = 383 optically evoked action potentials. Movie acquired on an EMCCD camera. (AVI 992 kb)

Sub-frame movie of AP initiation and propagation 3

Sub-frame interpolated movie showing action potential initiation and propagation in a neuron expressing Optopatch1, with blue light stimulation targeted to a small group of dendrites. The movie was acquired using the same parameters as in Supplementary Movie 3. The movie was constructed from an average of n = 338 optically evoked action potentials. Movie acquired on an EMCCD camera. (AVI 1605 kb)

Wide-field spontaneous activity

Spontaneous network activity in neurons expressing Optopatch2, monitored with red illumination (640 nm, 100 W/cm2) in a field of view 1 mm × 3 mm. The movie was acquired at a 500 Hz frame rate. Movie acquired on an sCMOS camera. (AVI 11247 kb)

Wide-field evoked activity

Simultaneous excitation and recording of action potentials in many neurons expressing Optopatch2. A pulse of blue light (500 ms, 5 mW/cm2) was applied to stimulate neural activity via excitation of CheRiff, and activity was monitored with red illumination (640 nm, 100 W/cm2) in a field of view 1 mm × 3 mm. The blue dot next to the time stamp indicates when the blue light is on. Synaptic blockers were added to prevent network activity. The movie was acquired at a 500 Hz frame rate. Movie acquired on an sCMOS camera. (AVI 3205 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hochbaum, D., Zhao, Y., Farhi, S. et al. All-optical electrophysiology in mammalian neurons using engineered microbial rhodopsins. Nat Methods 11, 825–833 (2014). https://doi.org/10.1038/nmeth.3000

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nmeth.3000

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