SnapShot: Optical Control and Imaging of Brain Activity X. Richard Sun, Andrea Giovannucci, Allyson E. Sgro, and Samuel S.-H. Wang Princeton Neuroscience Institute and Departments of Molecular Biology and Physics, Princeton University, Princeton, NJ 08544, USA Timescales of recording techniques 100 µs 1 ms 10 ms 100 ms 1 s

Ion Channels Action Potentials Synaptic Potentials Spike Trains Field Potentials

OPTICAL METHODS Optical principles microscopy Pulsed infrared laser Thin specimens In vivo two-photon (e.g. brain slices) microscopy

Detection Objective (one- or two-photon)

TREADMILL One-photon Two-photon Detection (conventional and (two-photon only) confocal microscopy)

Fluorescent and photoactivatable probes

Glutamate Cl-, Ca2+ CFP Ca2+ EXTRACELLULAR Voltage Sensors CFP Glutamate sensors (e.g. SuperGluSnFR) ∆V ∆V YFP

FP Caged compounds One FP, Arch and MNI-Glu CFP multiple FPs, Arch(D95N) CDNI-Glu

GFP and FRET-based GFP YFP CFP CDNI-GABA Ca2+ Light-activated channels Ions Single fluorophore indicator 2+ ChR2 and C1V1 IP3, cAMP, cGMP, Ca FRET-based indicators Light-activated pumps Arch and eNpHR3.0 INTRACELLULAR Ions

Methods for labeling

Synthetic probes 4 2 TVA Genetically encodable probes 5

GP

1 Bolus loading of synthetic probes 2 In utero electroporation 3 3 Transgenic expression 4 Pseudotyped rabies virus 1 5 Pseudorabies virus (PRV) 6 6 rAAV and lentivirus

1650 Cell 149, June 22, 2012 ©2012 Elsevier Inc. DOI 10.1016/j.cell.2012.06.009 See online version for legend and references. SnapShot: Optical Control and Imaging of Brain Activity X. Richard Sun, Andrea Giovannucci, Allyson E. Sgro, and Samuel S.-H. Wang Princeton Neuroscience Institute and Departments of Molecular Biology and Physics, Princeton University, Princeton, NJ 08544, USA

Optical methods have assumed a leading role in the study of intact nervous system function. In comparison with traditional electrical recordings, optical imaging enables the measurement of activity in many structures at once, including subcellular domains. For observing function at the cellular level, multiphoton fluorescence microscopy allows tracking to be done with spatiotemporal resolution in the micron and millisecond range. Light can also be used to activate brain tissue by using “caged” and second messengers, and, more recently, by using light-sensitive ion channels, pumps, and receptors to allow optogenetic activation.

Two-Photon Fluorescence Microscopy In confocal fluorescence microscopy, single-photon excitation is typically delivered at blue and green wavelengths, and the penetration depth is limited to less than 100 µm. For imaging at deeper distances, two-photon microscopy uses pulsed infrared laser light, which is scattered less by cellular structures and therefore penetrates further into a sample than visible light. The depth limit depends on laser wavelength, pulse energy, width, and tissue properties. The depth limit of two-photon microscopy is up to 500–800 µm but is often reduced in highly scattering tissues such as brain slices or myelinated or cell-dense structures. Two-photon excitation also restricts photobleaching and photodamage to the focal point. Two-photon imaging has been applied to brain slices and intact brains, including head-fixed behaving mice. For brain slice imaging, fluorescence can be collected both above and below the specimen. Imaging is complemented by electrophysiological recording, which can reach submillisecond time domains.

One-Photon Two-Photon Depth penetration Shallow; limited by scattering Deeper; longer wavelengths scatter less Spatial resolution Higher Slightly coarser Photodamage Throughout the beam volume Near focal point only Detection Above specimen only (i.e., epifluorescence) Above and, if specimen is thin, below specimen

Fluorescent Activity Probes The monitoring of biological activity requires probes that are sensitive to specific biological signals. Neurons and glia can be imaged by using synthetic or genetically encodable fluorescent molecules. The revolution in fluorescent (FP) imaging has permitted not just labeling but also the design of that change their fluorescence in response to a biological signal. FPs can also be used to label other proteins and/or be photoactivated at discrete moments in time. Synthetic probes are in wide use for imaging , pH, voltage, and other biological signals. A particularly powerful application is the monitoring of cytoplasmic calcium, which is around 0.1 µM at baseline and is elevated by action potentials or synaptic activity. Synthetic calcium indicators such as Oregon Green BAPTA-1, fluo-4, and X-rhod-1 are available over a range of colors and calcium affinities. A number of genetically encodable calcium indicators (GECIs) are available or under development. GECIs can be expressed in specific cell types and are retained for long peri- ods of time, allowing extended imaging. GCaMP3 is based on a single enhanced green FP fluorophore, generates large fluorescence increases upon binding calcium, and has been used to detect calcium transients in C. elegans and small numbers of action potentials in Drosophila and mouse. Other single-fluorophore GECIs include the red-emitting R-GECO and the blue-emitting B-GECO. A second GECI design principle is based on Förster resonance energy transfer (FRET), where two FPs (for example cyan FP [CFP] and yellow FP [YFP]) are brought into proximity upon binding, leading to weaker CFP emission and stronger YFP emission. This color shift enables ratiometric imaging and correction of movement artifacts. FRET-based GECIs are exemplified by D3cpV and YC3.60/YC-Nano, which use calmodulin as the sensor, and TN-XXL, which uses troponin. Recurring challenges in GECI design include low signal-to-noise ratios, slow response kinetics, a narrow dynamic range because of high cooperativity, and probe degradation or instability over extended periods. Although the aforementioned GECIs have been successfully applied in neural tissue, most GECIs developed during the past decade have not been shown to be useful for diverse biological applications. FRET-based glutamate sensors include FLIPE, which is sensitive to glutamate concentrations from 100 nM to 1 mM, the higher signal-to-noise and faster kinetics Super- − GluSnFR, and EOS, which is conjugated with the synthetic fluorophore Oregon Green. Probes have also been developed for chloride (Clomeleon and Cl Sensor), IP3 (LIBRA and fretino), cAMP (Epac), and cGMP (cGi), Membrane voltage indicators are proteins containing a voltage-sensitive membrane channel domain fused to one FP, multiple FPs, or a FRET pair (VSFP). Archaerhodopsin 3 (Arch) is a voltage-sensitive fluorescent proton pump that demonstrates higher sensitivity and faster response time than older generations of voltage sensors. Even though voltage sensors have been widely used for the readout of population activity, the monitoring of single- voltage changes is not yet routine and is an area of ongoing devel- opment. The plethora of FP variants exhibit emission profiles covering the entire visible spectrum and, more recently, entering the infrared spectrum. Understanding of intracellular protein dynamics can be aided by a class of reengineered FPs known as optical highlighters which, when excited at specific wavelengths, enables high-contrast photoactiva- tion, color-shifting photoconversion, and reversible photoswitching. Another valuable tool is the split-GFP and its optimized variant GRASP, which allows for genetically targeted synaptic tracing in neuronal populations.

Caged Neurotransmitters and Optogenetic Manipulation Photoactivation has been performed with synthetic light-sensitive groups, of which many types have now been developed. In order to perturb or control biological activity with temporal and spatial specificity, optical approaches may be both convenient and effective. One type of light-sensitive molecule is the caged compound, in which a biologically active molecule is rendered inactive with a photolabile group. A wide variety of caged compounds have been developed, most frequently glutamate and GABA, but other neu- rotransmitters and second messengers as well. Uncaging light is usually in the 350–400 nm range (700–800 nm for two-photon excitation). Uncaging can be performed at single points or in complex spatial patterns. Optogenetic probes (light-sensitive genetically encodable proteins) reach a potentially much broader audience than caged compounds. Excitatory light-activated cation channels such as ChR2(H134R) have been applied in vivo in mice, primates, flies, and zebrafish. C1V1 is a red-shifted cation channel that can be suitable when combined with commonly used green calcium indicators. The inhibitory light-activated chloride pump eNpHR is one of the first optogenetic tools used for the study of loss-of-function in behav- ing mammals and has been applied in a variety of subcortical locations. Arch is an inhibitory proton pump with high fidelity of inactivation (?100% inhibition). Optogenetic probes are under development for improved cellular processing, use at a variety of wavelengths, and multiple targets.

1650.e1 Cell 149, June 22, 2012 ©2012 Elsevier Inc. DOI 10.1016/j.cell.2012.06.009 SnapShot: Optical Control and Imaging of Brain Activity X. Richard Sun, Andrea Giovannucci, Allyson E. Sgro, and Samuel S.-H. Wang Princeton Neuroscience Institute and Departments of Molecular Biology and Physics, Princeton University, Princeton, NJ 08544, USA

Delivering and Expressing Probes Bulk Loading of AM-Conjugated Synthetic Probes The covalently-bound acetoxymethyl ester (AM-ester) renders charged molecules nonpolar and can cross plasma membranes, where they are hydrolyzed and trapped inside cells. Bulk-loading labels tissue indiscriminately and tends to emphasize large cellular structures. In Utero Electroporation Electroporation is an efficient method for expressing transgenes carried by plasmid DNA in neural tissue with cell-type specificity. Although this method has low adverse effects on brain tissue, labeling may be sparse. Transgenic Animals with Integrated Exogenous Genes Transgenes are typically inserted into the Rosa26 locus to avoid interference with endogenous or expressed conditionally, for instance by using the Cre or Tet system. In addition, BAC transgenesis may be used if the transgene expression pattern must replicate that of a specific endogenous gene. Viral Vectors Pseudotyped rabies virus containing a glycoprotein (GP) deletion and modified envelope proteins (TVA) can label genetically targeted neurons and their immediate presynaptic partners, at which point viral spread is terminated. Attenuated pseudorabies virus (PRV) can cross multiple synapses, in retrograde or anterograde directions depending on the strain, allowing long-distance pathway tracing. PRV entry occurs preferentially through axon terminals. Certain recombinant adeno-associated virus (rAAV) and lentiviral vectors serotypes have high tropism for neurons in rodents and can also target neurons with cell specificity when combined with the appropriate promoters. The size of exogenous DNA is typically limited to <5 kb for rAAV and <10 kb for lentiviral vectors.

References Card, J.P., and Enquist, L.W. (2001). Transneuronal circuit analysis with pseudorabies viruses. Curr Protoc Neurosci 1.5.1–1.5.28.

Dombeck, D.A., and Tank, D.W. (2011). Two-photon imaging of neural activity in awake mobile mice. Imaging in Neuroscience: A Laboratory Manual (Cold Spring Harbor, New York: Cold Spring Harbor Laboratory Press), pp. 827–838.

Helmchen, F., and Denk, W. (2005). Deep tissue two-photon microscopy. Nat. Methods 2, 932–940.

Kerr, J.N.D., and Denk, W. (2008). Imaging in vivo: watching the brain in action. Nat. Rev. Neurosci. 9, 195–205.

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

Scanziani, M., and Häusser, M. (2009). Electrophysiology in the age of light. Nature 461, 930–939.

Shaner, N.C., Patterson, G.H., and Davidson, M.W. (2007). Advances in fluorescent protein technology. J. Cell Sci. 120, 4247–4260.

Thompson, S.M., Kao, J.P., Kramer, R.H., Poskanzer, K.E., Silver, R.A., Digregorio, D., and Wang, S.S.-H. (2005). Flashy science: controlling neural function with light. J. Neurosci. 25, 10358–10365.

Wickersham, I.R., Lyon, D.C., Barnard, R.J.O., Mori, T., Finke, S., Conzelmann, K.-K., Young, J.A.T., and Callaway, E.M. (2007). Monosynaptic restriction of transsynaptic tracing from single, genetically targeted neurons. Neuron 53, 639–647.

Yizhar, O., Fenno, L.E., Davidson, T.J., Mogri, M., and Deisseroth, K. (2011). in neural systems. Neuron 71, 9–34.

1650.e2 Cell 149, June 22, 2012 ©2012 Elsevier Inc. DOI 10.1016/j.cell.2012.06.009