Current Biology 23, 1746–1755, September 23, 2013 ª2013 Elsevier Ltd All rights reserved http://dx.doi.org/10.1016/j.cub.2013.07.033 Article Tracing Inputs to Inhibitory or Excitatory Neurons of Mouse and Cat Visual Cortex with a Targeted Virus

Yong-Jun Liu,1,4 Markus U. Ehrengruber,1,2,4 Moritz Negwer,1 is essential for several basic functional characteristics, Han-Juan Shao,1 Ali H. Cetin,3 and David C. Lyon1,* including a neuron’s preference for stimulus contrast, size, 1Department of Anatomy & Neurobiology, School of Medicine, and orientation (see e.g. [5–7]). Although inhibitory neurons University of California, Irvine, Irvine, CA 92697, USA serve to control excitation, it is not known whether local pop- 2Department of Biology, Kantonsschule Hohe Promenade, ulations of inhibitory and excitatory cell types are mediated 8001 Zurich, Switzerland through the same set of cortical circuits, or whether the func- 3Callaway Lab, Systems Neurobiology Laboratories, Salk tional selectivity of these circuits differs. This is largely due to Institute for Biological Studies, 10010 North Torrey Pines technical limitations, since inhibitory and excitatory neurons Road, La Jolla, CA 92037, USA are intermingled and the study of one population over the other is difficult to achieve. Circuits related to specific cell types have been identified in part using various neurophysiological Summary techniques in slice preparations [8–10] and in vivo [11, 12], yet these approaches are unable to provide a full-scale Background: Cortical inhibition plays a critical role in con- pattern of connectivity throughout the intact brain. On the trolling and modulating cortical excitation, and a more other hand, traditional neuronal tracers can reveal widespread detailed understanding of the neuronal circuits contributing connection patterns yet are unable to discriminate between to each will provide more insight into their roles in complex inputs to inhibitory and excitatory neurons unless the ultra- cortical computations. Traditional neuronal tracers lack a structure of is examined under electron microscopy means for easily distinguishing between circuits of inhibi- [13–15], where only a fraction of the connections can be tory and excitatory neurons. To overcome this limitation, studied. we have developed a technique for retrogradely labeling in- To overcome these limitations, we developed a technique puts to local clusters of inhibitory or excitatory neurons, but for labeling presynaptic inputs to inhibitory or excitatory neu- not both, using neurotropic adenoassociated and lentiviral rons that takes advantage of the neurotropism of recombinant vectors, cell-type-specific promoters, and a modified rabies viral vectors, cell-type-specific promoters, and a modified virus. (RV). The modified rabies virus can be made to Results: Applied to primary visual cortex (V1) in mouse, the retrogradely trace the direct presynaptic inputs to a particular cell-type-specific tracing technique labeled thousands of population of neurons through delivery of two key genes [16]. presynaptically connected neurons and revealed that the The most successful approaches thus far have used trans- dominant source of input to inhibitory and excitatory neu- genic mouse lines to target and enhance the necessary gene rons is local in origin. Neurons in other visual areas are expression [17–20]. However, key to our new technique is also labeled; the percentage of these intercortical inputs to the design of two helper viruses that enable cell-type-specific excitatory neurons is somewhat higher (w20%) than to gene delivery without relying on transgenic animals. inhibitory neurons (<10%), suggesting that intercortical con- Tests in V1 of wild-type mice confirmed the cell-type speci- nections have less direct control over inhibition. The inputs ficity of gene transduction of our helper viruses and estab- to inhibitory neurons were also traced in cat V1, and when lished the efficiency of modified rabies virus infection and aligned with the orientation preference map revealed for retrograde spread. At the same time, these experiments the first time that long-range inputs to inhibitory neurons allowed unprecedented comparison of wide-scale input pat- are well tuned to orientation. terns to inhibitory and excitatory V1 neurons. The technique Conclusions: These novel findings for inhibitory and excit- was also used to target inhibitory neurons in cat V1, where, atory circuits in the visual cortex demonstrate the efficacy in combination with intrinsic signal optical imaging to reveal of our new technique and its ability to work across species, the orientation preference map, we show for the first time including larger-brained mammals such as the cat. This that orientation preference of long-range lateral inputs to paves the way for a better understanding of the roles of spe- inhibitory neurons is well tuned. The technique’s ability to cific cell types in higher-order perceptual and cognitive work in nontransgenic animals provides the first opportunity processes. to explore cell-type-specific connections in neocortex of higher-order mammals, paving the way for a better under- Introduction standing of the roles of excitatory and inhibitory neurons in complex perceptual and cognitive processes. The neocortex is comprised of a dense population of excit- atory neurons that are balanced by a diverse and more Results sparsely distributed array of inhibitory neurons [1–4]. Interac- tions between local inhibitory and excitatory neurons provide Design of the Cell-Type-Specific Tracing Technique gain control that works under myriad conditions throughout Our new method was developed to label the widespread the cortex. In primary visual cortex (V1), for example, inhibition presynaptic inputs of local clusters of inhibitory or excitatory neurons in neocortex of any mammalian species. To do this, we designed two helper viruses (Figure 1A) to optimize for 4These authors contributed equally to this work cell-type-specific transduction of the three-gene construct *Correspondence: [email protected] YTB [22], which allows for expression of two key proteins, Cell-Type-Specific 1747

A and 2D), enabling virions to spread from the host neuron to infect presynaptic terminals and be retrogradely transported to the (Figures 2C and 2E). The DG-RV cannot spread additional synapses, because the RabG gene will not be avail- able in presynaptically connected neurons, only in starter cells [16]. Importantly, the helper viruses express YFP and EnvA- DG-RV expresses mCherry, so that initially infected starter cells appear double labeled with both YFP and mCherry, whereas presynaptically connected neurons can be distin- guished as being labeled by mCherry-only (see Figures 2A B [right], 2C, and 2E).

Cell-Type Specificity of the AAV/GAD1/YTB and LV/aCaMKII/YTB Helper Viruses Following helper virus production, we next determined whether each would lead to detectable levels of transgene expression, and whether expression would be cell-type specific. Two weeks after injections of each helper virus, histo- logical examination revealed strong native YFP expression (Figure 3). AAV/GAD1/YTB yielded a more sparsely distributed Figure 1. Design of the Cell-Type-Specific Vectors and Modified Rabies array of cells (Figures 3A, 3C, and 3F; see also Figure S1A avail- Virus able online) characteristic of cortical inhibitory neurons [1, 3]. (A) Schematic representation of the transfer vectors for recombinant Importantly, every YFP-positive cell (Figures 3C and 3F) was adenoassociated virus AAV/GAD1/YTB (top), and lentivirus LV/aCaMKII/ YTB (bottom). Each vector includes a cell-type-specific promoter also positive for the inhibitory neuron marker GABA (Figures (magenta), GAD1 (glutamic acid decarboxylase 1) or aCaMKII (a-calcium/ 3D and 3G), verifying the selectivity of this helper virus (see calmodulin-dependent protein kinase II), cis-regulatory sequences also Figures S1B–S1D). (hatched; SD/SA = splice donor and acceptor sequence from human YFP expression from LV/aCaMKII/YTB (green in Figures 3I b-globin, BGH = bovine growth hormone polyadenylation signal, WPRE = and 3L), in contrast, was distinctly absent from GABA- woodchuck hepatitis virus posttranscriptional regulatory element), and immunopositive neurons (red in Figures 3I and 3L), verifying the YTB transgene. YTB codes for three gene products using 2A peptide- mediated cleavage [21]: the yellow fluorescent protein reporter (YFP, or Y; excitatory neuron selectivity of this helper virus. More- green), the artificial TVA 950 receptor (TVA, or T; blue), and the glycoprotein over, LV-transduced neurons formed a denser cluster along from the B-19 strain of rabies virus (RabG, or B; purple). Arrows indicate the the injection track (Figure 3H), consistent with excitatory start codons for each gene. Inverted terminal repeats (ITR) and long terminal neurons comprising 80% of the neuronal population in repeats (LTR) are indicated in black. neocortex [1, 3]. (B) The rabies virus (RV) was modified in two ways based on [16]: the glyco- D protein gene (G) was deleted ( G) and replaced with the gene for mCherry, D creating DG-RV-mCherry, and it was subsequently pseudotyped with the Cell-Type-Specific Infection and Spread of EnvA- G-RV- envelope A glycoprotein, creating EnvA-DG-RV-mCherry. mCherry Tracing Presynaptic Inputs to Inhibitory Neurons Having established the cell-type specificity of our helper TVA (T) and RabG (B for the B-19 strain of rabies virus), neces- viruses, we next tested whether expression of TVA and sary for initial infection and retrograde spread of the modified RabG is sufficient to allow for EnvA-DG-RV-mCherry infection rabies virus (RV), as well as a third gene, the YFP reporter (Y). A and monosynaptic retrograde spread (see schematics in recombinant adenoassociated virus (AAV) was constructed to Figure 2). Data from three mice using AAV/GAD1/YTB and express YTB specifically in inhibitory neurons under the con- EnvA-DG-RV-mCherry are presented in Figure 4. In one case trol of a 3.1 kb fragment of the GAD1 promoter (Figure 1A, (M11-16; Figures 4A–4G), a relatively large, 1.0 ml injection of top), whereas a lentivirus (LV; Figure 1A, bottom) with the AAV/GAD1/YTB was made into a single location in V1 and aCaMKII promoter [23, 24] was used to transduce excitatory followed three weeks later by a single 0.4 ml injection of cortical neurons with YTB. EnvA-DG-RV-mCherry to within w300 mm of the same location The modified RV, EnvA-DG-RV (Figure 1B), has been pseu- (Figure 4A). In the two other cases, smaller AAV injections of dotyped with the envelope A glycoprotein and can thus only 0.3 ml (M12-17) and 0.5 ml (M11-20) were made, also followed infect cells containing the receptor TVA [16]. Normally, TVA is three weeks later by 0.4 ml injections of EnvA-DG-RV-mCherry only expressed in avian cells (see [16]), yet through selective (Figure 4H; Figures S2A–S2C). After post-EnvA injection sur- delivery of the TVA gene to inhibitory or excitatory mammalian vival times of 8–14 days, we found that AAV infection led to neurons, we can target EnvA-DG-RV infection (Figure 2). As robust YFP expression (e.g., Figure 4A) in a relatively sparse indicated in Figures 2C and 2E, EnvA infection occurs at the array consistent with the distribution of inhibitory neurons cell body where the TVA receptor becomes embedded. (see close-up images in Figures 4C, 4F, 4I, and S2F). GABA Furthermore, EnvA-DG-RV has been genetically modified so immunofluorescence was also used to confirm inhibitory cell that the gene coding for RabG has been removed (DG) specificity in every fourth section, as already demonstrated and replaced with the sequence coding for mCherry (Fig- (Figures 3A–3G). In addition, in one case a series of sections ure 1B). Without RabG, newly produced DG-RV virions in the were processed for the calcium-binding protein parvalbumin starter cells cannot retrogradely spread and infect other neu- (PV), a marker of a large subpopulation of inhibitory cells, rons. The critical component for retrograde tracing using showing a high, though not exclusive, overlap (Figures S2E– EnvA-DG-RV, therefore, is the delivery beforehand of RabG S2G). AAV spread as observed through YFP expression was via our helper viruses into starter cells (Figures 2A [left], 2B, restricted to within w500 mm for the two smaller injections Current Biology Vol 23 No 18 1748

A Figure 2. Schematic for Targeting Rabies Virus to Trace the Inputs to Inhibitory or Excitatory Neurons A simplified diagram of our tracing technique is shown in (A) and described in more detail in (B)–(E). In the left panel of (A), helper virus is used to deliver three genes (YFP, TVA, and RabG; see Figure 1A) to a local population of neurons in mouse V1 at the site of injection. As shown in (B) and (D), the helper viruses infect directly through the cell soma (magenta lines). The cell-type specificity of gene expression depends on which helper virus is used: AAV/ GAD67/YTB (B) and LV/aCaMKII/YTB (D) vectors are designed to introduce the genes to inhibitory or excitatory neurons, respectively. The three gene products are produced by the single cell- type-specific promoter (GAD1 or aCaMKII) using 2A peptide-mediated cleavage [21]. Helper-virus- infected neurons are depicted as green due to B C YFP expression. Following a 14–21 day survival period to allow for sufficient gene expression, the modified rabies virus, EnvA-DG-RV-mCherry (Figure 1B), is injected in the same V1 location (middle panel in A). The TVA receptor now embedded in the membrane of the cell soma (blue rectangles on cell bodies in B–E) enables direct EnvA infection at the soma as indicated in (C) and (E). These EnvA-DG-RV-mCherry-super- infected neurons are depicted as yellow because they will now begin to express mCherry as well as YFP. Because RabG is also coexpressed via the helper virus, rabies virions produced over the next 10 days in starter cells will incorporate RabG and spread retrogradely from the yellow ‘‘starter cells’’ via the synaptic terminals of pre- synaptically connected neurons (orange arrows in C and E). Resulting ‘‘connected cells’’ that can be found locally within V1 and at longer range in other brain regions such as V2 (right panel in A) are depicted as red because they will express only mCherry delivered by the rabies virus. Black D E crosses (X) in (C) and (E) illustrate that the G-RV D virus cannot spread beyond these directly con- nected cells because they were not transduced with RabG.

Critically, in all three cases, injections of EnvA-DG-RV-mCherry led to 100 or more AAV-infected (YFP-positive) neu- rons coexpressing mCherry (see starter cells in Table 1). This confirms sufficient AAV expression of the TVA receptor, since EnvA infection cannot occur in its absence. The observed EnvA superin- fection rate of TVA-expressing neurons was high, over 75%, particularly in re- gions where the EnvA and AAV injection sites were highly overlapping (see ex- amples in Figures 4B–4D, 4I, 4J, and S2A–S2C). In this regard, despite the variability in spread of AAV/GAD1/YTB- infected neurons, double-labeled starter cells were confined to within a 250 mm radius of the EnvA-DG-RV- (see examples in Figures 4I and S2B) but extended more mCherry injection track (red arrow in Figures 4A and S2A), than 2 mm in superficial layers 1–3 for the larger injection likely due to the more limited extracellular spread of EnvA- (Figure 4A). DG-RV-mCherry. Cell-Type-Specific Neuronal Tracing 1749

Figure 3. Cell-Type Specificity of Helper Virus Trans- duction in Mouse Neocortex A B C D Low-magnification images show expression of YFP (green) in cortical neurons following AAV/GAD1/YTB (A; AAV) and LV/aCaMKII/YTB (H; LV) helper virus injections in mouse cortex. Higher-magnification black-and-white images of AAV-infected neurons (C and F) colocalize with neurons labeled via GABA immunofluorescence (D and G), as can be seen E F G when shown together in color (B and E; AAV is shown in green and GABA in red). In contrast, higher-magni- fication black-and-white images of LV-infected neu- rons (J and M) do not colocalize with neurons positive for GABA immunofluorescence (K and N), as can be seen when shown together in color (I and L; LV is shown in green and GABA in red). Scale bars repre- sent 100 mm. See also Figure S1.

H I J K (>75%; see examples in Figures 5A–5L). In addition, thousands of presynaptically con- nected neurons expressing mCherry-only were also found, indicating adequate expression of RabG in starter cells (see red-only neurons in Figure 5). As found for M N L inhibitory starter cells, the majority of neu- rons projecting to excitatory starter cells were found locally within V1 (Figure 5M), with only a small percentage in LGN, LP (Figure 5M, right inset), RS, Cg, and the opposite cortical hemisphere. However, in comparison to V1 inhibitory starter cells, where only 3%–8% of the inputs come Finally, several thousand presynaptically connected neu- from higher visual areas, the percentage of inputs to V1 excit- rons expressing mCherry-only were identified locally within atory neurons is higher, averaging over 20% (Table 1; see V1 (Figures 4A and 4H) and throughout the brain (Figure 4K), examples in Figures 5I and 5M). Although factors such as dif- confirming that RabG expression was sufficient to allow retro- ferences in the concentration of starter cells by cortical layer grade spread of DG-RV from starter cells. Counts showed that could affect these percentages and are not accounted for in the vast majority of connected neurons are within V1, ranging our current analysis, the higher percentage of ‘‘feedback- from 86% to 97% (Table 1). Feedback-like inputs from neurons like’’ projections to excitatory neurons is nevertheless consis- in higher visual areas (V2L, V2ML, and V2M) were far fewer in tent with ultrastructural analyses in rat [14, 25, 26] and monkey number but represented the next largest source of inputs to [13, 15] showing that a greater percentage of V2 feedback V1 inhibitory neurons, ranging from 2.5% to 8.4% (Table 1; synapses are onto excitatory neurons in V1. left image in Figure 4K; Figure S2D). Inputs from the dorsal lateral geniculate nucleus (LGN; Figure 4K, center inset), lateral Comparison of EnvA-DG-RV Complementation posterior nucleus (LP), retrosplenial cortex (RS), cingulate cor- Techniques with Unpseudotyped DG-RV tex (Cg; Figure 4K, right inset), hypothalamus (HT), and the To help evaluate the efficacy of our helper-virus-mediated opposite cortical hemisphere made up the remainder of the EnvA-DG-RV-mCherry tracing technique, we made small V1 mCherry-labeled inputs (Table 1). injections of the unpseudotyped DG-RV in separate mice. These results demonstrate that our AAV-EnvA vector-medi- The results are summarized in Table 1 and discussed in detail ated technique is effective at labeling the inputs to GABAergic in the Supplemental Information (see Figure S3 and Additional neurons. Moreover, the sensitivity of this technique reveals Histology and Data Analysis in the Supplemental Experimental that the dominant source of inputs to V1 inhibitory cells is local Procedures). in origin, with only a small percentage coming from higher visual areas and subcortical thalamic nuclei. Tracing Presynaptic Inputs to Inhibitory Neurons in Cat V1 Tracing Presynaptic Inputs to Excitatory Neurons We next applied our AAV helper virus to evaluate the functional We next evaluated the efficacy of EnvA-DG-RV-mCherry input to inhibitory neurons in cat V1 by correlating the pattern complementation in neurons transduced by the LV/aCaMKII/ of long-range horizontal connections to the orientation prefer- YTB helper virus. In three mice, LV/aCaMKII/YTB injection ence map. Orientation preference maps are only present in volumes of 1.0 ml (M12-15, Figures 5A–5D) and 0.5 ml (M12- highly visual mammals such as carnivores [27], tree shrews 09, Figures 5E–5H; M12-10, Figures 5I–5L) were used, followed [28], and primates [29], and long-range horizontal connections three weeks later by a 0.4 ml injection of EnvA-DG-RV-mCherry across these maps have been shown to link regions, or do- at or near the same location. After an additional 11- to 14-day mains, of V1 neurons preferring the same orientation. Further- survival time, TVA expression was confirmed, as all three more, in cat and monkey these tuned long-range intrinsic cases exhibited a high rate of superinfected starter cells inputs have been indirectly implicated in the orientation tuning Current Biology Vol 23 No 18 1750

Figure 4. Targeting EnvA-DG Rabies Virus Infection to and Monosynaptic Retrograde Spread from Inhibitory Neurons in Mouse V1 using the AAV/GAD1/ YTB Helper Virus (A–J) The local pattern of RV-infected inputs to AAV-infected inhibitory neurons is shown in coronal sections through V1 in three mice, M11-16 (A–G), M12-17 (H–J), and M11-20 (K). In (A), the AAV and EnvA-DG-RV-mCherry (RV) injection sites are indicated by green and red arrowheads, respectively. In (H), injection sites overlap. Starter cells appear yellow due to transduction of the cell first with AAV, leading to YFP expression (green), and subsequent infection by EnvA- DG-RV-mCherry due to the presence of the TVA receptor, leading to mCherry expression (red; see schematic in Figures 2B and 2C). Neurons presynaptically connected to starter cells that were retrogradely infected by RV following RabG complementation within starter cells are identified by expression of mCherry-only (red; see Figure 2C). Higher-magnification images of the regions outlined by white and black rectangles in (A) and (H) are shown in (B)–(G), (I), and (J). AAV-infected neurons expressing YFP (green; C, F and I) and RV-infected neurons expressing mCherry (red; D, G, and J) are shown together in (B) and (E). (K) A representative reconstruction of the brain-wide pattern of RV-infected neurons (mCherry-expressing; red) providing direct inputs to inhibitory starter cells (green) found in layers 1–6 of V1 is shown for mouse M11-20. The AAV and RV injection sites are located between sections 65 and 76 (see Figures S2A– S2C). Sections are ordered from posterior to anterior. Inset images show mCherry expression in long-range connected neurons in V2L, LGN, and cingulate cortex (Cg). For all sections, left is lateral and up is dorsal. Scale bars represent 200 mm (A and H), 50 mm (E, I, and K inset), and 1 mm (K). See also Figure S2. of the suppressive surround [30–32], which largely depends on Moreover, in three cats the orientation preference map of the ‘‘near’’ component of the surround [30], a relatively narrow area 17 (V1) was first obtained through intrinsic signal optical region nearest to the classical receptive field that can be imaging and used to guide the AAV injection. For example, in accounted for by monosynaptic intrinsic connections ranging the left hemisphere of C12-02, the large ‘‘orange’’ domain from 0.5 to 2.0 mm within carnivore and monkey V1 [33–35]. near the center of the imaged V1 region was identified (white However, even though much of the evidence from intracellular circle in Figure 6E), and a small, w0.4 ml injection of AAV/ recording indicates that inhibitory and excitatory inputs to cat GAD1/YTB was followed three weeks later by an w0.4 ml injec- V1 neurons are similarly tuned to orientation (e.g., [6, 7, 36]), it tion of EnvA-DG-RV-mCherry. Resulting YFP/mCherry starter has not been shown directly that inhibitory neurons, which can cells were confined to the domain, and several hundred provide the local source of suppression, receive tuned long- mCherry-only connected neurons within V1 were found locally range horizontal inputs. Adding to this uncertainty, other (<250 mm radius) and at long range (>250 mm; see Figures 6 and studies report that a subgroup of cat V1 inhibitory neurons S5). Notably, the pattern of connected cells in superficial lack orientation tuning [37, 38]. Moreover, there is mounting cortex (<600 mm from the pial surface) was quite patchy in evidence from mouse V1, where transgenic lines allow for appearance (Figure 6D) and, when aligned with the orientation clearer identification of inhibitory neurons, that orientation preference map (Figure 6E), showed tuning to the preferred tuning of many inhibitory neurons is much broader than for orientation (112.5; orange in Figure 6F). This observed orien- excitatory neurons (e.g., [39–41]). Thus, an examination of tation tuning preference of inputs to inhibitory neurons was the orientation preference of inputs to inhibitory neurons in found in two additional cases (Figures S5A–S5H). cat is needed. Quantitatively, the normalized and averaged orientation The inhibitory cell-type specificity of AAV/GAD1/YTB in cat preference of long-range intrinsic connections for the three visual cortex was first confirmed (Figure S2), followed by cases indicates that although inputs come mainly from the demonstration that it allowed for initial infection and subse- preferred orientation, there are substantial inputs from non- quent spread of EnvA-DG-RV-mCherry (Figure S4; Table 1). preferred orientations, which leads to a moderate orientation Cell-Type-Specific Neuronal Tracing 1751

Table 1. Number of Cells Providing Inputs to Inhibitory or Excitatory Neurons as Labeled through Targeted Rabies Virus Tracing

Vector Species Case Starters Connected V1 Feedback Callosal RS Cg LGN LP OT HT AAV Mouse M11-16 537 7,851 7,620 193 (2.5%) 6 8 0 16 6 2 0 M11-20 70 1,752 1,510 148 (8.4%) 10 14 8 22 9 8 23 M12-17 390 9,359 8,433 667 (7.9%) 4 244 0 8 0 2 1 LV Mouse M12-09 404 4,647 3,225 1,057 (23%) 57 8 1 7 3 7 0 M12-10 300 3,727 2,140 1,314 (35%) 13 268 4 0 0 0 0 M12-15 1,032 21,358 15,749 5,029 (24%) 344 221 3 4 1 6 1 DG-RV Mouse M12-13mCh NA 3,456 3,315 756 (22%) 40 6 12 3 2 1 2 M12-13GFP NA 4,188 3,102 832 (20%) 24 220 2 3 1 1 3 M12-21GFP NA 6,919 5,367 1,274 (18%) 21 95 26 52 19 13 51 M12-22GFP NA 1,975 1,755 181 (9.2%) 2 4 4 16 2 3 8 AAV Cat C12-02R 336 9,174 8,916 258 (2.8%) NA NA NA NA NA NA NA C12-02L 12 4,149 3,945 192 (2.2%) NA NA 0 0 12 0 0 Numbers of EnvA-DG-RV-Infected starter cells in V1 and presynaptically connected cells located in V1 (intrinsic V1), higher visual cortex (Feedback), cortex in the opposite hemisphere (Callosal), restrosplenial cortex (RS), cingulate cortex (Cg), lateral geniculate nucleus (LGN), lateral posterior nucleus (LP), other thalamic (OT), and hypothalamic (HT) nuclei. See also Figure S4. selectivity index (OSI) of 0.25 6 0.12 (Figure S5I, top right; for of long-range inputs from higher visual cortex is larger for more detailed analysis, see Additional Histology and Data excitatory neurons, suggesting that excitation is controlled Analysis in the Supplemental Experimental Procedures). more directly through intercortical connections than inhibition Thus, if involved in the suppressive surround of V1 neurons is. Moreover, in combination with intrinsic signal optical as suggested above [30–33], such inputs should provide sup- imaging of the V1 orientation map in the cat, we provide the pression that is tuned but not exclusive to the preferred orien- first anatomical evidence that the orientation preference of tation. This is what has been reported. In fact, the average OSI intrinsic inputs to inhibitory neurons is well tuned. in cat for the suppressive surround of V1 domain neurons is Although our demonstrations were in visual cortex, this 0.26 6 0.19 [30], quite similar to the OSI for our long-range method can be used to investigate the connections to inhibi- inputs. tory or excitatory neurons of any neocortical system due to Qualitatively, the observed patchy, orientation-tuned con- the flexibility of the viral vector-based delivery of YTB. More- nectivity is similar to that in cat, monkey, and tree shrew that over, as demonstrated, the cell-type-specific tracing method has been repeatedly revealed through traditional neuronal can be applied to multiple mammalian species because our tracers (e.g., [7, 27–29, 42]). However, traditional tracers could viral vector design is not cre dependent, opening the door to not distinguish between inputs to excitatory and inhibitory animal models with more complex cortical organization that neurons. Thus, our results provide the first anatomical evi- may be better suited for studying higher-order perceptual dence that long-range intrinsic inputs to V1 inhibitory neurons and cognitive processes. can be tuned to the preferred orientation, and they may explain recent results that indirectly link long-range intrinsic connec- Cell-Type Specificity of the Helper Viruses tions to the orientation tuning of surround suppression [30, 31]. In order for our technique to work, it was imperative that our vector/promoter combinations selectively transduce either Discussion inhibitory or excitatory neurons, but not both. To optimize cell-type specificity, we used AAV with an inhibitory cell pro- Here we demonstrate a novel method for independent moter to target inhibitory neurons and LV with an excitatory anatomical tracing of inputs to inhibitory or excitatory neurons cell promoter to target excitatory neurons. In mice, inhibitory in mammalian neocortex. The technique takes advantage of cell specificity was confirmed by showing complete overlap neurotropic adenoassociated and lentiviral vectors, cell- with GABA immunostaining; likewise, excitatory cell specificity type-specific promoters, and a genetically modified and pseu- was confirmed through an absence of GABA labeling (Figure 3). dotyped rabies virus. Our viral vector/promoter design drove In cat, inhibitory cell specificity was further confirmed through cell-type-specific expression of a three-gene construct (YTB) an absence of double labeling with the antibody for aCaMKII that enabled targeted initial infection and subsequent spread (Figures S1E–S1G). The observed selectivity of our LV/ of a custom-made EnvA pseudotyped and glycoprotein- aCaMKII/YTB vector is consistent with published reports deleted rabies virus (EnvA-DG-RV) [16] that labeled thousands showing that the same LV/aCaMKII vector/promoter combina- of presynaptically connected neurons with a fluorescent tion leads to excitatory cell-specific expression of enhanced reporter (mCherry). Our AAV/GAD1/YTB helper virus led to GFP in somatosensory cortex of mouse [23] and of channelr- cell-type-specific RV tracing of inputs to inhibitory cells, hodopsin-2 fused to GFP in frontal cortex of monkey [24]. whereas the LV/aCaMKII/YTB vector led to RV tracing of Furthermore, our inhibitory cell-type-specific results observed inputs to excitatory neurons. Because traditional neuronal for AAV/GAD1/YTB are consistent with recent work by Nathan- tracers cannot distinguish between inputs to these two types son and colleagues [43] showing that AAV used in conjunction of neocortical neurons, this new technique allows for un- with a 2.6 kb fragment of the somatostatin inhibitory cell precedented exploration of cell-type-specific circuitry. For promoter leads to selective expression of GFP in inhibitory example, we have demonstrated here how the method can neurons of somatosensory and motor cortex in mouse, rat, be used to compare the local and long-range connections of and monkey. inhibitory and excitatory neurons in primary visual cortex Importantly, we chose not to use AAV with an excitatory (V1), showing that although both cell types receive the domi- cell promoter or LV with an inhibitory cell promoter because nant share of their inputs locally from within V1, the percentage the same Nathanson et al. study [43] showed that such Current Biology Vol 23 No 18 1752

Figure 5. Targeting EnvA-DG-RV-mCherry Infection to and Retrograde Spread from Excitatory Neurons in Mouse V1 Using the LV/aCaMKII/YTB Helper Virus (A–L) Examples of the local and long-range pattern of RV-infected neurons (red) providing inputs to excitatory starter neurons (yellow) are shown in coronal sections through V1 in three mice, M12-15 (A–D), M12-09 (E–H), and M12-10 (I–L). In (A), (E), and (I), overlapping injection sites of LV/aCaMKII/YTB (green) and EnvA-DG-RV-mCherry (red) are shown at low magnification; higher-magnification images of the regions outlined by black and white rectangles are shown in (B), (F), and (J). Double-labeled starter cells (yellow in B, F, and J) coinfected by LV (C, G, and K) and RV (D, H, and L) express YFP from LV infection and mCherry from RV infection. (M) A representative reconstruction of the brain-wide pattern of RV-infected neurons (mCherry-expressing; red) providing direct inputs to excitatory starter cells (green) found in layers 2–6 of V1 is shown for mouse M12-09. The LV and RV injection sites are marked by thick black lines in sections 40 and 45. Inset images show RV-infected neurons in V2L (section 49) and the lateral posterior nucleus (LP) (section 78). Conventions are as in Figure 4. Scale bars represent 200 mm (A, E and I), 50 mm (B, F, L, and M inset), and 1 mm (M). See also Figure S3. combinations led to non-cell-type-specific transduction. to presynaptically connected neurons, was also deemed Moreover, in a second study [44], the same authors found adequate, as several thousand connected neurons were iden- that LV pseudotyped with vesicular stomatitis virus (VSV)-G tified all over the brain in regions known to project to V1. has an endogenous tropism for excitatory cortical neurons With our new helper-virus-based approach, the great major- and AAV a tropism for inhibitory cortical neurons, the latter ity of RV-connected neurons are found locally within V1 (>80%; particularly being the case when AAV titers were low. In this Table 1). The implication here that local V1 circuits provide the regard, it is worth noting that because our GAD1 promoter dominant input to both inhibitory and excitatory neurons is and YTB insert totaled w7.4 kb (Figure 1A, top), there is likely consistent with several other lines of evidence (see [2, 47]). an overall reduction in effective titer of our AAV virus. This is Moreover, results for inhibitory neurons showing that only because the packaging capacity of AAV is only w5kb[45]. 3%–8% of inputs arise from extrinsic cortical connections Recent evidence indicates that genomes as large as 8.7 kb are similar to one example shown for mouse somatosensory are packaged in AAV as smaller fragments that can be cortex using a PV-cre line to target TVA and RabG expression reassembled following transduction into the full transgene; to the inhibitory cell subtype and subsequent infection and however, this process invariably reduces the effective titer of spread with EnvA-DG-RV [19]. There, Wall et al. found that the virus [46]. only w3% of RV-connected neurons were not local (20 out of 620 in counts of every sixth section). Given that our Tracing Efficacy of the Helper-Virus-Mediated Approach AAV/GAD1/YTB-infected inhibitory neurons included the PV- In addition to the cell-type specificity of our helper viruses, the positive subpopulation (Figures S2E–S2H), these results transgene expression driven by the promoters must be high corroborate our findings. enough to visualize the transduced neurons and to interact effectively with EnvA-DG-RV, enabling initial infection and Conclusion subsequent spread. In this regard, YFP expression for both Understanding the connectivity of neocortical networks is helper viruses was high enough to be visualized without anti- essential for explaining complex cortical computations. body amplification. In addition, expression of TVA, the EnvA Decades of research using ‘‘traditional’’ neuronal tracers receptor, was sufficient to enable EnvA-DG-RV-mCherry have been spent piecing together the connectivity, but without infection in w75% of YTB-transduced cells. Furthermore, a means for easily distinguishing between circuits of inhibitory expression of RabG, which is necessary for DG-RV spread and excitatory cell types. Cortical inhibition plays a critical role Cell-Type-Specific Neuronal Tracing 1753

in controlling and modulating cortical excitation, and a more detailed understanding of the neuronal circuits contributing to each will provide more insight into the interplay between these two major classes of neurons. Our viral vector-mediated technique provides a new way for addressing inhibitory and excitatory circuits through retrograde labeling of presynaptic inputs and, when paired with global measures of functional selectivity such as intrinsic signal optical imaging, can be used to infer functional properties of these circuits. Beyond this, genes that allow for cell monitoring through calcium imag- ing [48] or cell manipulation through light-gated ion channels [49, 50] can also be delivered with the DG-RV virus [51]. Thus, not only can inputs to excitatory and inhibitory neurons now be distinguished anatomically by our new approach, future applications using this method should provide the opportunity to observe and manipulate the function of each circuit in vivo.

Experimental Procedures

Generation of AAV/GAD1/YTB A four-step cloning procedure (see Supplemental Experimental Procedures) was used to generate our final plasmid, AAV/GAD1/YTB (11.0 kb). The viral genome to be packed into infectious particles encompasses 7,382 bp (including 50- and 30-inverted terminal repeat; Figure 1, top). Sero- type 9 AAV particles were prepared and purified with iodixanol by the Viral Vector Core Facility at the Salk Institute (La Jolla, CA, USA), yielding a titer of 9 3 109 genome copies/ml.

Generation of LV/aCaMKII/YTB For details on LV subcloning, see Supplemental Experimental Procedures. The resulting LV/aCaMKII/YTB (12.3 kb) plasmid has a viral genome to be packaged of 7.5 kb (including 50- and 30-long terminal repeat; Figure 1, bot- tom). Purified VSV-G pseudotyped lentiviral particles were prepared by the Viral Vector Core Facility at the Salk Institute, yielding a titer of 2 3 1010 transducing units/ml.

Rabies Viruses The modified RV, EnvA-DG-RV expressing mCherry, as well as the DG-RVs expressing either mCherry or GFP were produced and concentrated following protocols described previously [16, 52].

Animal Procedures All animal procedures were approved by the University of California, Irvine Institutional Animal Care and Use Committee and Institutional Biosafety Committee and followed National Institutes of Health guidelines. Pressure injections of viruses were made directly into V1 of anesthetized mouse and cat using sterile procedures. Intrinsic signals in cat were imaged by a 12-bit video camera with a 50 mm/50 mm tandem-lens combination feeding into the Optical Imager 3001 system [30, 31]. See Supplemental Experi- mental Procedures for more details.

Histology After the full survival period, animals were deeply anesthetized with Euthasol and perfused transcardially, first with saline and then followed

connected neurons (red). AAV/GAD1/YTB- and EnvA-DG-RV-mCherry-in- fected neurons are shown separately in (B) and (C), respectively. In (D), the pattern of EnvA-DG-RV-mCherry-infected neurons is shown in a low-magni- fication image of a superficial section of flattened V1 cortex adjacent to that shown in (A)–(C). A reconstruction of this pattern is overlaid on the orienta- tion preference map in (E). The numbers of cells present in each colored domain in (E) were counted for short-range (within the white circle) and long-range intrinsic distances and are displayed as histograms in (F). Corre- Figure 6. Correlating Orientation Preference of Inputs to V1 Inhibitory sponding orientation selectivity index values of 0.54 and 0.35 calculated us- Neurons in Cat ing the formula provided in Supplemental Experimental Procedures are also As shown in the high-magnification image of a superficial section of flattened given. The red line in (D) points to the location of the EnvA-DG-RV-mCherry V1 cortex (A), AAV/GAD1/YTB transduction (green) allowed for initial infec- injection site. The white circle in (E) represents a 250 mm radius around the tion with EnvA-DG-RV-mCherry, creating a double-labeled starter cell (yel- approximate center of the location of the starter cell identified in (A). Scale low; indicated by white line) and leading to RV spread to presynaptically bars represent 100 mm (A) and 500 mm (D and E). See also Figure S5. Current Biology Vol 23 No 18 1754

by 4% paraformaldehyde in phosphate buffer (PB; pH 7.4). For most 16. Wickersham, I.R., Lyon, D.C., Barnard, R.J., Mori, T., Finke, S., animals, 1.5% glutaraldehyde was also included. were removed Conzelmann, K.K., Young, J.A., and Callaway, E.M. (2007). and cryoprotected in 30% sucrose for w48 hr prior to sectioning. Sectioned Monosynaptic restriction of transsynaptic tracing from single, geneti- brains were processed for GABA, aCaMKII, or DAPI. See Supplemental cally targeted neurons. Neuron 53, 639–647. Experimental Procedures for more details. 17. Haubensak, W., Kunwar, P.S., Cai, H., Ciocchi, S., Wall, N.R., Ponnusamy, R., Biag, J., Dong, H.W., Deisseroth, K., Callaway, E.M., Supplemental Information et al. (2010). Genetic dissection of an amygdala microcircuit that gates conditioned fear. Nature 468, 270–276. Supplemental Information includes five figures and Supplemental Experi- 18. Miyamichi, K., Amat, F., Moussavi, F., Wang, C., Wickersham, I., Wall, mental Procedures and can be found with this article online at http://dx. N.R., Taniguchi, H., Tasic, B., Huang, Z.J., He, Z., et al. (2011). Cortical doi.org/10.1016/j.cub.2013.07.033. representations of olfactory input by trans-synaptic tracing. Nature 472, 191–196. Acknowledgments 19. Wall, N.R., Wickersham, I.R., Cetin, A., De La Parra, M., and Callaway, E.M. (2010). Monosynaptic circuit tracing in vivo through Cre- We thank Ed Callaway for the YTB construct, cell lines for RV production, dependent targeting and complementation of modified rabies virus. and access to facilities for cloning the GAD1 promoter; Daniel Gibbs for Proc. Natl. Acad. Sci. USA 107, 21848–21853. helpful discussions regarding AAV vectors; Maziar Hashemi-Nezhad for 20. Weible, A.P., Schwarcz, L., Wickersham, I.R., Deblander, L., Wu, H., initial assistance with cat surgery and intrinsic signal optical imaging; and Callaway, E.M., Seung, H.S., and Kentros, C.G. (2010). Transgenic tar- the Whitehall Foundation (2009-12-44; D.C.L.) and NIH (NS072948; D.C.L.) geting of recombinant rabies virus reveals monosynaptic connectivity for support. of specific neurons. J. Neurosci. 30, 16509–16513. 21. Szymczak, A.L., Workman, C.J., Wang, Y., Vignali, K.M., Dilioglou, S., Received: February 27, 2013 Vanin, E.F., and Vignali, D.A. (2004). Correction of multi-gene deficiency Revised: June 7, 2013 in vivo using a single ‘self-cleaving’ 2A peptide-based retroviral vector. Accepted: July 5, 2013 Nat. Biotechnol. 22, 589–594. Published: August 29, 2013 22. Marshel, J.H., Mori, T., Nielsen, K.J., and Callaway, E.M. (2010). Targeting single neuronal networks for gene expression and cell label- References ing in vivo. Neuron 67, 562–574. 23. Dittgen, T., Nimmerjahn, A., Komai, S., Licznerski, P., Waters, J., 1. Beaulieu, C. (1993). Numerical data on neocortical neurons in adult Margrie, T.W., Helmchen, F., Denk, W., Brecht, M., and Osten, P. rat, with special reference to the GABA population. Brain Res. 609, (2004). Lentivirus-based genetic manipulations of cortical neurons and 284–292. their optical and electrophysiological monitoring in vivo. Proc. Natl. 2. Burkhalter, A. (2008). Many specialists for suppressing cortical excita- Acad. Sci. USA 101, 18206–18211. tion. Front. Neurosci. 2, 155–167. 24. Han, X., Qian, X., Bernstein, J.G., Zhou, H.H., Franzesi, G.T., Stern, P., 3. DeFelipe, J. (1993). Neocortical neuronal diversity: chemical heteroge- Bronson, R.T., Graybiel, A.M., Desimone, R., and Boyden, E.S. (2009). neity revealed by colocalization studies of classic neurotransmitters, Millisecond-timescale optical control of neural dynamics in the neuropeptides, calcium-binding proteins, and cell surface molecules. nonhuman primate brain. Neuron 62, 191–198. 3 Cereb. Cortex , 273–289. 25. Gonchar, Y., and Burkhalter, A. (1999). Differential subcellular localiza- 4. Markram, H., Toledo-Rodriguez, M., Wang, Y., Gupta, A., Silberberg, G., tion of forward and feedback interareal inputs to parvalbumin ex- and Wu, C. (2004). Interneurons of the neocortical inhibitory system. pressing GABAergic neurons in rat visual cortex. J. Comp. Neurol. 5 Nat. Rev. Neurosci. , 793–807. 406, 346–360. 5. Adesnik, H., Bruns, W., Taniguchi, H., Huang, Z.J., and Scanziani, M. 26. Gonchar, Y., and Burkhalter, A. (2003). Distinct GABAergic targets of (2012). A neural circuit for spatial summation in visual cortex. Nature feedforward and feedback connections between lower and higher areas 490 , 226–231. of rat visual cortex. J. Neurosci. 23, 10904–10912. 6. Anderson, J.S., Carandini, M., and Ferster, D. (2000). Orientation tuning 27. Gilbert, C.D., and Wiesel, T.N. (1989). Columnar specificity of intrinsic of input conductance, excitation, and inhibition in cat primary visual horizontal and corticocortical connections in cat visual cortex. cortex. J. Neurophysiol. 84, 909–926. J. Neurosci. 9, 2432–2442. 7. Marin˜ o, J., Schummers, J., Lyon, D.C., Schwabe, L., Beck, O., Wiesing, 28. Bosking, W.H., Zhang, Y., Schofield, B., and Fitzpatrick, D. (1997). P., Obermayer, K., and Sur, M. (2005). Invariant computations in Orientation selectivity and the arrangement of horizontal connections local cortical networks with balanced excitation and inhibition. Nat. in tree shrew striate cortex. J. Neurosci. 17, 2112–2127. Neurosci. 8, 194–201. 29. Malach, R., Amir, Y., Harel, M., and Grinvald, A. (1993). Relationship 8. Fino, E., and Yuste, R. (2011). Dense inhibitory connectivity in between intrinsic connections and functional architecture revealed by neocortex. Neuron 69, 1188–1203. optical imaging and in vivo targeted biocytin injections in primate striate 9. Xu, X., and Callaway, E.M. (2009). Laminar specificity of functional cortex. Proc. Natl. Acad. Sci. USA 90, 10469–10473. input to distinct types of inhibitory cortical neurons. J. Neurosci. 29, 70–85. 30. Hashemi-Nezhad, M., and Lyon, D.C. (2012). Orientation tuning of the 10. Yoshimura, Y., Dantzker, J.L., and Callaway, E.M. (2005). Excitatory suppressive extraclassical surround depends on intrinsic organization 22 cortical neurons form fine-scale functional networks. Nature 433, of V1. Cereb. Cortex , 308–326. 868–873. 31. Liu, Y.J., Hashemi-Nezhad, M., and Lyon, D.C. (2013). Sharper orienta- 11. Bock, D.D., Lee, W.C., Kerlin, A.M., Andermann, M.L., Hood, G., Wetzel, tion tuning of the extraclassical suppressive-surround due to a neuron’s A.W., Yurgenson, S., Soucy, E.R., Kim, H.S., and Reid, R.C. (2011). location in the V1 orientation map emerges late in time. 229 Network anatomy and in vivo physiology of visual cortical neurons. , 100–117. Nature 471, 177–182. 32. Shushruth, S., Mangapathy, P., Ichida, J.M., Bressloff, P.C., Schwabe, 12. Ko, H., Hofer, S.B., Pichler, B., Buchanan, K.A., Sjo¨ stro¨ m, P.J., and L., and Angelucci, A. (2012). Strong recurrent networks compute the Mrsic-Flogel, T.D. (2011). Functional specificity of local synaptic con- orientation tuning of surround modulation in the primate primary visual 32 nections in neocortical networks. Nature 473, 87–91. cortex. J. Neurosci. , 308–321. 13. Anderson, J.C., and Martin, K.A. (2009). The synaptic connections 33. Angelucci, A., Levitt, J.B., Walton, E.J., Hupe, J.M., Bullier, J., and Lund, between cortical areas V1 and V2 in macaque monkey. J. Neurosci. J.S. (2002). Circuits for local and global signal integration in primary 29, 11283–11293. visual cortex. J. Neurosci. 22, 8633–8646. 14. Johnson, R.R., and Burkhalter, A. (1996). Microcircuitry of forward and 34. Cantone, G., Xiao, J., McFarlane, N., and Levitt, J.B. (2005). Feedback feedback connections within rat visual cortex. J. Comp. Neurol. 368, connections to ferret striate cortex: direct evidence for visuotopic 383–398. convergence of feedback inputs. J. Comp. Neurol. 487, 312–331. 15. Rockland, K.S. (1997). Elements of cortical architecture: Hierarchy re- 35. Salin, P.A., Girard, P., Kennedy, H., and Bullier, J. (1992). Visuotopic visited. In Cerebral Cortex, Volume 12, K.S. Rockland, J.H. Kaas, and organization of corticocortical connections in the visual system of the A. Peters, eds. (New York: Plenum Publishing), pp. 243–293. cat. J. Comp. Neurol. 320, 415–434. Cell-Type-Specific Neuronal Tracing 1755

36. Cardin, J.A., Palmer, L.A., and Contreras, D. (2007). Stimulus feature selectivity in excitatory and inhibitory neurons in primary visual cortex. J. Neurosci. 27, 10333–10344. 37. Hirsch, J.A., Martinez, L.M., Pillai, C., Alonso, J.M., Wang, Q., and Sommer, F.T. (2003). Functionally distinct inhibitory neurons at the first stage of visual cortical processing. Nat. Neurosci. 6, 1300–1308. 38. Nowak, L.G., Sanchez-Vives, M.V., and McCormick, D.A. (2008). Lack of orientation and direction selectivity in a subgroup of fast-spiking inhib- itory interneurons: cellular and synaptic mechanisms and comparison with other electrophysiological cell types. Cereb. Cortex 18, 1058–1078. 39. Kerlin, A.M., Andermann, M.L., Berezovskii, V.K., and Reid, R.C. (2010). Broadly tuned response properties of diverse inhibitory neuron sub- types in mouse visual cortex. Neuron 67, 858–871. 40. Li, Y.T., Ma, W.P., Li, L.Y., Ibrahim, L.A., Wang, S.Z., and Tao, H.W. (2012). Broadening of inhibitory tuning underlies contrast-dependent sharpening of orientation selectivity in mouse visual cortex. J. Neurosci. 32, 16466–16477. 41. Sohya, K., Kameyama, K., Yanagawa, Y., Obata, K., and Tsumoto, T. (2007). GABAergic neurons are less selective to stimulus orientation than excitatory neurons in layer II/III of visual cortex, as revealed by in vivo functional Ca2+ imaging in transgenic mice. J. Neurosci. 27, 2145–2149. 42. Buza´ s, P., Kova´ cs, K., Ferecsko´ , A.S., Budd, J.M., Eysel, U.T., and Kisva´ rday, Z.F. (2006). Model-based analysis of excitatory lateral con- nections in the visual cortex. J. Comp. Neurol. 499, 861–881. 43. Nathanson, J.L., Jappelli, R., Scheeff, E.D., Manning, G., Obata, K., Brenner, S., and Callaway, E.M. (2009). Short promoters in viral vectors drive selective expression in mammalian inhibitory neurons, but do not restrict activity to specific inhibitory cell-types. Front. Neural Circuits 3, 19. 44. Nathanson, J.L., Yanagawa, Y., Obata, K., and Callaway, E.M. (2009). Preferential labeling of inhibitory and excitatory cortical neurons by endogenous tropism of adeno-associated virus and lentivirus vectors. Neuroscience 161, 441–450. 45. Bu¨eler, H. (1999). Adeno-associated viral vectors for gene transfer and gene therapy. Biol. Chem. 380, 613–622. 46. Hirsch, M.L., Agbandje-McKenna, M., and Samulski, R.J. (2010). Little vector, big gene transduction: fragmented genome reassembly of adeno-associated virus. Mol. Ther. 18, 6–8. 47. Douglas, R.J., and Martin, K.A. (2007). Recurrent neuronal circuits in the neocortex. Curr. Biol. 17, R496–R500. 48. Akerboom, J., Chen, T.W., Wardill, T.J., Tian, L., Marvin, J.S., Mutlu, S., Caldero´ n, N.C., Esposti, F., Borghuis, B.G., Sun, X.R., et al. (2012). Optimization of a GCaMP calcium indicator for neural activity imaging. J. Neurosci. 32, 13819–13840. 49. Boyden, E.S., Zhang, F., Bamberg, E., Nagel, G., and Deisseroth, K. (2005). Millisecond-timescale, genetically targeted optical control of neural activity. Nat. Neurosci. 8, 1263–1268. 50. Zhang, F., Wang, L.P., Brauner, M., Liewald, J.F., Kay, K., Watzke, N., Wood, P.G., Bamberg, E., Nagel, G., Gottschalk, A., and Deisseroth, K. (2007). Multimodal fast optical interrogation of neural circuitry. Nature 446, 633–639. 51. Osakada, F., Mori, T., Cetin, A.H., Marshel, J.H., Virgen, B., and Callaway, E.M. (2011). New rabies virus variants for monitoring and manipulating activity and gene expression in defined neural circuits. Neuron 71, 617–631. 52. Wickersham, I.R., Sullivan, H.A., and Seung, H.S. (2010). Production of glycoprotein-deleted rabies viruses for monosynaptic tracing and high-level gene expression in neurons. Nat. Protoc. 5, 595–606.