We describe a clearing method for enhanced visualization of cell morphology and connections in neuronal and non-neuronal tissue. Using ClearT or ClearT2, which are composed of formamide or formamide/polyethylene glycol, respectively, embryos, whole mounts and thick brain sections can be rapidly cleared with minimal volume changes. Unlike other available clearing techniques, these methods do not use detergents or solvents, and thus preserve lipophilic dyes, fluorescent tracers and immunohistochemical labeling, as well as fluorescent-protein labeling.

Appreciation of neural circuitry and single-cell morphology has benefited from new labeling methods, including fluorescent tracers and genetically encoded fluorescent proteins (Luo et al., 2008). Although these methods produce superb detail of labeled cells and pathways, tissue opacity limits the depth of imaging, necessitating imaging sectioned material in order to attain high microscopic resolution. However, because images must be reconstructed in three dimensions (3D) post-acquisition, imaging and reconstructing sections is neither as efficient nor as accurate as imaging thicker tissue samples.

New reagents that clear or render tissue transparent include Scale, benzyl-alcohol and benzyl-benzoate (BABB), and a combination of tetrahydrofuran and BABB, all of which preserve genetically expressed fluorescent signal, allowing deep imaging of neural circuitry in 3D (Dodt et al., 2007; Hama et al., 2011; Ertürk et al., 2012). However, these reagents change tissue volume and require several days to weeks to fully clear the tissue (Hama et al., 2011; Ertürk et al., 2012). More importantly, owing to their reliance on detergents or organic solvents, Scale and BABB disrupt the fluorescent signal of immunohistochemistry, of conventional lipophilic carbocyanine dyes [such as 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine perchlorate (DiI)] and of fluorescent tracers such as cholera toxin subunit B (CTB). Here, we describe a rapid clearing method that maintains tissue volume and preserves fluorescent signal from tracers, immunohistochemistry and genetically expressed fluorescent proteins.

ClearT and ClearT2 solutions

For ClearT, 20%, 40%, 80% and 95% formamide solutions were made by adding formamide (99.6%, considered 100%) (Fisher) to PBS (pH 7.4) (vol/vol).

For ClearT2, a 50% formamide/20% polyethylene glycol (PEG) solution was made by mixing formamide (99.6%, considered 100%, as made for ClearT) with 40% PEG/H2O (wt/vol) at a ratio of 1:1 (vol/vol). A 25% formamide/10% PEG solution was made by mixing 50% formamide plus 20% PEG/H2O (wt/vol) at a ratio of 1:1 (vol/vol). A 40% PEG solution was made by stirring powdered PEG 8000 MW (Sigma-Aldrich) in warm H2O for 30 minutes, and is stable at room temperature for several months.

Preparation of specimens and clearing procedures

Procedures for the care and breeding of mice follow regulatory guidelines of the Columbia University Institutional Animal Care and Use Committee. Noon of the day on which a plug was found was considered to be E0.5. C57BL/6J wild-type and actin-GFP mouse embryos were removed from mothers anesthetized with ketamine-xylazine (100 and 10 mg/kg, respectively, in 0.9% saline); postnatal wild-type, Thy1-GFP (M-line) (a gift from J. A. Gogos, Columbia University, NY, USA) and adult Tcf/Lef:H2B-GFP mice (a gift from E. Laufer, Columbia University, NY, USA) were anesthetized with ketamine-xylazine (100 and 10 mg/kg, respectively, in 0.9% saline), fixed in 4% paraformaldehyde (PFA)/PBS (pH 7.4) overnight, or perfused and washed with PBS at 4°C. Embryos were perfused transcardially for optimal clearing. All clearing protocols took place at room temperature.

ClearT and ClearT2 tissue-clearing method

Incubation times in each solution vary according to tissue thickness for the desired transparency (see Table 1 for details).

ScaleA2 has been described previously (Hama et al., 2011). E14.5 embryos were cleared with ScaleA2 for 14 days; DiI-labeled embryos or CTB-labeled sections were treated overnight or 2 hours, respectively.

BABB has been described (Dodt et al., 2007). DiI-labeled embryos or CTB-labeled sections were treated with BABB overnight or for 2 hours, respectively, after dehydration with 30%, 50%, 70% and 100% ethanol for 1 hour each and with hexane for 1 hour.

Retinal axon labeling with DiI and CTB

Anterograde DiI labeling has been described previously (Plump et al., 2002). The eye was placed back into the optic cup and heads were incubated in PBS containing 0.1% sodium azide as follows: E14-E16, 5-7 days at room temperature; E17-P0, 5-7 days at 37°C. The retinogeniculate projection was labeled with CTB as described previously (Jaubert-Miazza et al., 2005; Rebsam et al., 2009), and single neuron labeling in the CTB-labeled dLGN was performed as described previously (Krahe et al., 2011).

Immunohistochemistry

Vibratome and cryosections were blocked in 5% BSA/1% Tween (Sigma-Aldrich) in PBS (pH 7.4) for 1 hour at room temperature. Mouse monoclonal anti-RC2 (IgM) antibody (Developmental Hybridoma Bank) (1:4) and mouse monoclonal anti-neurofilament (IgG) antibody (2H3) (a gift from T. Jessell and S. Morton, Columbia University, NY, USA) (1:5) were incubated in 1% BSA/1% Tween in PBS overnight at 4°C. After washes with 1% Tween in PBS, Cy3-conjugated anti-mouse IgM and Cy5-conjugated anti-mouse IgG (1:500) secondary antibodies (Jackson) were applied, incubated in 1% BSA/1% Tween in PBS overnight at 4°C. Hoechst 33258 (Molecular Probes) was used for nuclear staining. Whole-mount immunolabeling of embryos with anti-neurofilament antibody has been described previously (Huber et al., 2005).

Imaging

Whole brains or sections with DiI, CTB or immunolabeling, or sections of GFP-labeled mice were imaged on a Zeiss AxioImager M2 microscope with Apotome, AxioCam MRm camera, Neurolucida software (V10.40, MicroBrightField Systems); with a 5× objective lens (FLUAR, NA=0.25, working distance=12.5 mm), a 20× objective lens (PLAN-APOCHROMAT, NA=0.8, working distance=550 μm) or a 40× objective lens (PLAN-NEOFLUAR, NA=0.75, working distance=710 μm) (Fig. 2B,C; Fig 2D, bottom; Fig. 3C,D,F; supplementary material Fig. S3B, Fig. S4). Using the principle of structured illumination, the Apotome provides confocal-like resolution with epifluorescence imaging. The Apotome improves the signal to noise ratio by acquiring three images of an optical section and subtracting background fluorescence signal. Imaging of whole heads and brains was performed using a Zeiss dissecting microscope StemiSV11, Axiovision software, AxioCam camera (Fig. 1; Fig. 2A; Fig. 3A,B; supplementary material Fig. S1; Fig. S2A). Imaging of whole embryos with immunolabeling was performed using Nikon SMZ 1500 zoom stereomicroscope and DS-Qi1Mc camera (Fig. 3E). A Zeiss Axioplan 2 microscope, AxioCam camera and Axiovision software was used to image thin brain sections using a 10× objective lens (PLAN-NEOFLUAR, NA=0.3) or a 20× objective lens (PLAN-NEOFLUAR, NA=0.5) (supplementary material Fig. S2B, Fig. S3A, Fig. S5). Thick samples were imaged using a home-made slide to keep tissue submerged in formamide solutions and covered with a regular glass coverslip: a square rim of plastic or silicone elastomer was super-glued to a regular glass slide.

Statistical analysis

All experiments were performed three or more times with similar results. Data were analyzed and graphs constructed using Metamorph or Microsoft Excel. Error bars represent s.e.m. and statistical analysis was performed using Student’s t-test; P>0.05 indicates non-significance.

ClearT is a rapid tissue clearing method

After observing that 20 μm cryosections of embryonic mouse brain became transparent in the hybridization buffer used for in situ hybridization, we found that a component of the buffer, formamide, could clear thick tissue samples. Here, we demonstrate the versatility of our method, named ClearT for neuronal and non-neuronal tissue, and compare its clarity, rapidity and tissue expansion/shrinkage to existing clearing methods.

Intact embryos, embryonic and postnatal dissected heads, brains, and thick (up to 1000 μm) brain sections, were fixed and sequentially immersed in graded concentrations of formamide (Table 1A, Fig. 1A). The ClearT procedure rendered embryonic brains as transparent as with ScaleA2, but did so significantly faster (1 day versus 14 days) (Fig. 1B). Completely cleared postnatal day 0 (P0) brain sections were similar to their original size (before clearing=1.0±0 versus ClearT=1.04±0.02, not significant, n=6 sections) (Fig. 1C). Even after prolonged treatment with ClearT, sample volume only increased slightly, significantly less than in ScaleA2 [1 day, ClearT=1.33±0.09 versus ScaleA2=1.81±0.05, P<0.01; 2 days, ClearT=1.27±0.09 versus ScaleA2=1.83±0.06, P<0.01, n=5 (ClearT), 4 (ScaleA2) sections] (supplementary material Fig. S1). Although formamide is not harmful to tissue in the short term, it is unsuitable for long-term tissue storage. Therefore, we transferred samples treated with ClearT into PBS, where they became opaque within 30 minutes and could be safely stored for at least 1 month (Fig. 1D).

Fig. 1.

Rapid tissue clearing with ClearT. (A) Fixed whole embryos (E14.5) and dissected postnatal brains (P0) were cleared overnight. The grid is visible through tissue cleared by ClearT. (B) E14.5 embryos cleared with ClearT or ScaleA2 reach full transparency in 1 day or 14 days, respectively. (C) ClearT does not lead to volume changes. P0 sections (800 μm), surface area measured: pre-cleared, red line; ClearT, blue line. (D) Clearing is reversible with PBS (30 minutes). Scale bars: 1 mm.

Fig. 1.

Rapid tissue clearing with ClearT. (A) Fixed whole embryos (E14.5) and dissected postnatal brains (P0) were cleared overnight. The grid is visible through tissue cleared by ClearT. (B) E14.5 embryos cleared with ClearT or ScaleA2 reach full transparency in 1 day or 14 days, respectively. (C) ClearT does not lead to volume changes. P0 sections (800 μm), surface area measured: pre-cleared, red line; ClearT, blue line. (D) Clearing is reversible with PBS (30 minutes). Scale bars: 1 mm.

Table 1.

Clearing procedures

Clearing procedures
Clearing procedures

Visualization of DiI- or CTB-labeled axons in tissue cleared with ClearT

The projections, connections, and growth cone (GC) morphology of developing axons can be visualized by anterograde labeling with lipophilic dyes (Little et al., 2009; Bielle et al., 2011). Here, we use the mouse visual system, a classic model for studying neural circuitry development, to demonstrate the advantages of clearing the mouse brain with ClearT in preserving lipophilic fluorescent dye labeling. We anterogradely labeled retinal ganglion cell (RGC) axons in embryonic day (E) 14.5 embryos with DiI and treated embryos with ClearT, ScaleA2 or BABB for 1 day. DiI labeling of retinal axons in the optic nerve and chiasm was preserved after treatment with ClearT, but treatment with either ScaleA2 or BABB degraded the fluorescent signal (supplementary material Fig. S2A).

We then examined DiI-labeled RGC axons in cleared tissue at the optic chiasm at E15.5 (Fig. 2A). The DiI-labeled retinal projection was not visible prior to clearing, but could be seen through both dorsal and ventral aspects of the cleared head, with jaw and tongue removed but skin and skull intact (Fig. 2A). We examined the resolution of fine morphological detail of DiI-labeled axons and GCs in the proximal ipsilateral optic tract at E14.5 before and after clearing (Fig. 2B). The number and resolution of DiI-labeled axons and GC processes (e.g. filopodia and lamellopodia) were markedly increased after clearing with ClearT (Fig. 2B). Furthermore, E18.5 DiI-labeled RGC axons in the thalamus and superior colliculus were not visible before clearing when imaged from the midline of parasagittal hemisections, but the full tract was distinctly visible after clearing with ClearT, even through a depth of ∼1 mm (Fig. 2C).

Fig. 2.

Retinal axon projections in brain tissue cleared with ClearT. (A) E15.5 eye was labeled with DiI, the jaw and tongue were cut away and the head was cleared with ClearT. DiI-labeled contralateral (C) and ipsilateral (I) retinal axons and optic chiasm are detected in both dorsal and ventral views after clearing with ClearT. (B) Merged stack (41 images, 5 μm steps) of E14.5 DiI-labeled growth cones (GCs) (arrows) and axons (arrowheads) of the ipsilateral optic tract; imaged from the ventral surface of 200 μm brain section, before and after clearing. (C) DiI-labeled contralateral RGC projection to the thalamus and superior colliculus at E18.5. Brains were cut sagittally at the midline and cleared with ClearT. Merged stack (51 images, 20 μm steps), viewed from the midline. DiI-labeled RGC axons in the dLGN in the thalamus (TH) and superior colliculus (SC) were undetectable in pre-cleared tissue, but easily visible after clearing. (D) CTB conjugated to Alexa Fluor 488 or 594 was injected into each eye and a 700 μm frontal section of P5 brain was cleared with ClearT. Optical slices at 250 μm, 450 μm and 600 μm below the tissue section surface are shown (from 71 images, 10 μm steps). Both CTB labels were observable, though deeper, in cleared dLGN compared with the same tissue before clearing. Scale bars: 1 mm in C (top); 100 μm in A and bottom of C,D (bottom); 10 μm in B.

Fig. 2.

Retinal axon projections in brain tissue cleared with ClearT. (A) E15.5 eye was labeled with DiI, the jaw and tongue were cut away and the head was cleared with ClearT. DiI-labeled contralateral (C) and ipsilateral (I) retinal axons and optic chiasm are detected in both dorsal and ventral views after clearing with ClearT. (B) Merged stack (41 images, 5 μm steps) of E14.5 DiI-labeled growth cones (GCs) (arrows) and axons (arrowheads) of the ipsilateral optic tract; imaged from the ventral surface of 200 μm brain section, before and after clearing. (C) DiI-labeled contralateral RGC projection to the thalamus and superior colliculus at E18.5. Brains were cut sagittally at the midline and cleared with ClearT. Merged stack (51 images, 20 μm steps), viewed from the midline. DiI-labeled RGC axons in the dLGN in the thalamus (TH) and superior colliculus (SC) were undetectable in pre-cleared tissue, but easily visible after clearing. (D) CTB conjugated to Alexa Fluor 488 or 594 was injected into each eye and a 700 μm frontal section of P5 brain was cleared with ClearT. Optical slices at 250 μm, 450 μm and 600 μm below the tissue section surface are shown (from 71 images, 10 μm steps). Both CTB labels were observable, though deeper, in cleared dLGN compared with the same tissue before clearing. Scale bars: 1 mm in C (top); 100 μm in A and bottom of C,D (bottom); 10 μm in B.

CTB is widely used for the analysis of postnatal RGC axon targeting in the dLGN (Jaubert-Miazza et al., 2005; Rebsam et al., 2009). To test the compatibility of CTB with ClearT, we anterogradely labeled each eye of P5 pups with CTB conjugated to either Alexa Fluor 488 or 594. CTB labeling was preserved after ClearT and BABB treatments, but BABB reduced tissue size by half (before clearing=1±0 versus cleared=0.50±0.02, P<0.05, n=4 sections), while labeling was diffuse following ScaleA2 treatment (supplementary material Fig. S2B). CTB labeling was visible through the entire depth of a 700 μm section of P5 brain treated with ClearT, whereas fluorescence could not be seen beyond 250 μm before clearing (Fig. 2D). Moreover, it is possible to successively clear, unclear (in PBS) and re-clear DiI- or CTB-labeled samples without compromising tissue or label integrity (supplementary material Fig. S3A,B).

ClearT2 clears tissue with fluorescent protein and with immunohistochemical label

Our original ClearT protocol diminished green fluorescent protein (GFP) intensity in E14.5 actin-GFP embryos (Ikawa et al., 1995). Because polyethylene glycol (PEG) stabilizes protein conformation (Rawat et al., 2010), we investigated whether PEG would stabilize GFP expression in formamide. Whereas 50% formamide failed to clear brains, a 20% PEG/50% formamide mixture successfully cleared brain tissue and preserved fluorescence. This modified method, named ClearT2, also requires immersion in a graded series of formamide/PEG solutions (25% formamide/10% PEG then 50% formamide/20% PEG) (Table 1B, Fig. 3A). Although tissue transparency with ClearT2 was less complete than with ClearT, application of ClearT2 induced robust transparency of thick P0 brain sections without volume changes (before clearing=1.0 versus ClearT2=0.98±0.02, n=6, not significant) (Fig. 3B). Sections treated with ClearT2 for 1 or 2 days were slightly larger than pre-cleared sections, but these changes were significantly less than with ScaleA2 [1 day, ClearT2=1.30±0.02 versus ScaleA2=1.81±0.05, P<0.01; 2 days, ClearT2=1.30±0.01 versus ScaleA2=1.83±0.06, P<0.01, n=6 (ClearT2), n=4 sections (ScaleA2)] (supplementary material Fig. S1). ClearT2 also maintained DiI and CTB labeling in axons as successfully as ClearT (supplementary material Fig. S2A,B).

Fig. 3.

ClearT2 clears tissue with fluorescent proteins or immunohistochemistry. (A) ClearT cleared E14.5 actin-GFP embryos, but reduced GFP fluorescence. Formamide (50%) maintained fluorescence, but failed to clear embryos. ClearT2 cleared embryos and maintained fluorescence. (B) P0 sections (800 μm) were transparent after ClearT2, with no volume change. (C) P11 Thy1-GFP (M-line) hippocampus section (800 μm), before and after clearing with ClearT2; 38 images, 20 μm steps (top and middle). GFP+ pyramidal neurons (arrows) and dendrites (arrowheads) in CA1 region are markedly more visible after clearing; 52 images, 2.5 μm steps (bottom). GCL, granule cell layer; ML, molecular layer. (D) Sections of E14.5 optic chiasm (200 μm), immunolabeled with the radial glial marker RC2, cleared with ClearT2; 51 images, 3 μm steps; three optical slices shown. RC2+ staining was observed deeper in cleared compared with pre-cleared tissue. Blue indicates Hoechst staining. (E) E11.5 whole embryos, immunolabeled with neurofilament antibody (NF) and treated with ClearT2. NF+ axons were much more visible in cleared embryos (top); magnification of trigeminal axons reaching epithelial targets (bottom). (F) Section (300 μm) of postnatal mouse brain, dLGN anterogradely labeled with CTB conjugated to Alexa Fluor 594. A single neuron was filled with biocytin and immunostained with streptavidin-Alexa Fluor 647. Clearing with ClearT2 enhanced resolution and visibility of the dendritic arbor of the neuron. Merged stack, 55 images, 2 μm steps. CTB label is in red; biocytin-filled neuron is pseudo-colored green. Scale bars: 1 mm in A,B,E; 40 μm in C; 20 μm in D,F.

Fig. 3.

ClearT2 clears tissue with fluorescent proteins or immunohistochemistry. (A) ClearT cleared E14.5 actin-GFP embryos, but reduced GFP fluorescence. Formamide (50%) maintained fluorescence, but failed to clear embryos. ClearT2 cleared embryos and maintained fluorescence. (B) P0 sections (800 μm) were transparent after ClearT2, with no volume change. (C) P11 Thy1-GFP (M-line) hippocampus section (800 μm), before and after clearing with ClearT2; 38 images, 20 μm steps (top and middle). GFP+ pyramidal neurons (arrows) and dendrites (arrowheads) in CA1 region are markedly more visible after clearing; 52 images, 2.5 μm steps (bottom). GCL, granule cell layer; ML, molecular layer. (D) Sections of E14.5 optic chiasm (200 μm), immunolabeled with the radial glial marker RC2, cleared with ClearT2; 51 images, 3 μm steps; three optical slices shown. RC2+ staining was observed deeper in cleared compared with pre-cleared tissue. Blue indicates Hoechst staining. (E) E11.5 whole embryos, immunolabeled with neurofilament antibody (NF) and treated with ClearT2. NF+ axons were much more visible in cleared embryos (top); magnification of trigeminal axons reaching epithelial targets (bottom). (F) Section (300 μm) of postnatal mouse brain, dLGN anterogradely labeled with CTB conjugated to Alexa Fluor 594. A single neuron was filled with biocytin and immunostained with streptavidin-Alexa Fluor 647. Clearing with ClearT2 enhanced resolution and visibility of the dendritic arbor of the neuron. Merged stack, 55 images, 2 μm steps. CTB label is in red; biocytin-filled neuron is pseudo-colored green. Scale bars: 1 mm in A,B,E; 40 μm in C; 20 μm in D,F.

We next examined whether neurons genetically labeled with fluorescent proteins, such as in Thy1-GFP (M-line) mice (Feng et al., 2000), could be visualized with ClearT2 (Fig. 3C). After clearing thick hippocampal sections with ClearT2, Thy1-GFP+ neurons were visible deeper within the granule cell layer (Fig. 3C, top) and details of GFP+ pyramidal neuron dendrites in the CA1 region were more distinct than without clearing (Fig. 3C, bottom). To determine whether ClearT2 could be applied to adult or non-neuronal tissue, we used Tcf/Lef:H2B-GFP mice, in which reporter expression is detected in neuronal and non-neuronal tissues from early embryonic to adult stages (Ferrer-Vaquer et al., 2010). H2B-GFP nuclear labeling in neurons of the cerebral cortex, cells within the granule cell and molecular layers of the hippocampus, progenitor cells of the small intestine and satellite cells of skeletal muscle were more apparent after clearing with ClearT2 (supplementary material Fig. S4).

Immunohistochemistry is used to visualize protein expression, but labeling is usually visible only superficially in thick tissue sections. To examine whether immunohistochemistry labeling is compatible with tissue clearing, we immunostained E14.5 cryosections through the optic chiasm with an antibody to the radial glia marker RC2 and treated sections with ClearT, ClearT2, ScaleA2 or BABB (supplementary material Fig. S5A,B). ClearT and ScaleA2 disrupted RC2 immunolabeling, and BABB maintained fluorescent signal but produced labeling artifacts in bone and cell nuclei that should not express RC2. As ClearT2 successfully preserved immunolabeling (supplementary material Fig. S5A-E), we applied it to 200 μm RC2-immunolabeled vibratome sections of the optic chiasm at E14.5 (Fig. 3D). RC2+ glial processes were visible as deep as ∼120 μm in cleared tissue, twice as deep as in pre-cleared tissue (Fig. 3D). Finally, ClearT2 treatment of whole mouse embryos immunostained with an antibody to neurofilament (NF) provided a complete view of axon tracts and arbors in the CNS and PNS in distal appendages (Fig. 3E).

Finally, we examined whether ClearT2 is compatible with multiple fluorescent labels. After applying ClearT2 to a thick brain section with CTB-labeled dLGN, a biocytin-filled relay neuron was visualized more deeply and at higher resolution than before clearing, with both labels successfully maintained (Fig. 3F).

ClearT and ClearT2 are solvent- and detergent-free rapid tissue clearing methods

We have developed two clearing methods, ClearT and ClearT2, which aid analysis of fluorescent labeling in embryonic and mature neuronal and non-neuronal tissue. ClearT2 clears specimens while effectively maintaining the fluorescent signal of genetically encoded proteins, immunohistochemistry labeling, and dye tracers such as DiI and CTB. Whereas ClearT is incompatible with immunohistochemistry and genetically encoded fluorescence proteins (supplementary material Table S1), transparency of whole brains treated with ClearT is better than with ClearT2 (Fig. 3A). Therefore, tissue samples labeled with DiI or CTB alone are best cleared by ClearT.

ClearT and ClearT2 provide several advantages over other available clearing methods. Clearing time for thick sections, whole brains or embryos is significantly faster than with ScaleA2 or BABB. In addition, ClearT and ClearT2 produce minimal tissue volume changes, significantly less than ScaleA2 or BABB. Most importantly, our methods maintain DiI- and CTB-labeling in axons, unlike ScaleA2 and BABB (supplementary material Table S1). ClearT and ClearT2 successfully clear postnatal and adult brain and other tissues. ClearT2 provides a final important advantage over ScaleA2 and BABB, in that it can clear immunolabeled tissue. Thus, ClearT and ClearT2 provide improved clearing of embryonic and adult neuronal and non-neuronal tissue for viewing fluorescent labeling of cells and fiber tracts by high-resolution optical imaging.

We thank members of the Mason lab and Columbia University colleagues, Wes Grueber for advice and reading the manuscript, Joseph Gogos for Thy1-GFP (M-line) mice, Ed Laufer for Tcf/Lef:H2B-GFP mice, and Tom Jessell and Susan Morton for the anti-neurofilament antibody. Thomas E. Krahe in William Guido’s laboratory produced additional slice preparations of biocytin-filled cells in the dLGN (not shown).

Funding

This work was supported by the National Institues of Health [R01 EY012736 and EY015290 to C.M., T32 EY013933 to A.A.S. and R01 EY012716 to W.G.] and the Uehara Foundation (T.K.). Deposited in PMC for release after 12 months.

Bielle
F.
,
Marcos-Mondéjar
P.
,
Leyva-Díaz
E.
,
Lokmane
L.
,
Mire
E.
,
Mailhes
C.
,
Keita
M.
,
García
N.
,
Tessier-Lavigne
M.
,
Garel
S.
, et al. 
. (
2011
).
Emergent growth cone responses to combinations of Slit1 and Netrin 1 in thalamocortical axon topography
.
Curr. Biol.
21
,
1748
1755
.
Dodt
H. U.
,
Leischner
U.
,
Schierloh
A.
,
Jährling
N.
,
Mauch
C. P.
,
Deininger
K.
,
Deussing
J. M.
,
Eder
M.
,
Zieglgänsberger
W.
,
Becker
K.
(
2007
).
Ultramicroscopy: three-dimensional visualization of neuronal networks in the whole mouse brain
.
Nat. Methods
4
,
331
336
.
Ertürk
A.
,
Mauch
C. P.
,
Hellal
F.
,
Förstner
F.
,
Keck
T.
,
Becker
K.
,
Jährling
N.
,
Steffens
H.
,
Richter
M.
,
Hübener
M.
, et al. 
. (
2012
).
Three-dimensional imaging of the unsectioned adult spinal cord to assess axon regeneration and glial responses after injury
.
Nat. Med.
18
,
166
171
.
Feng
G.
,
Mellor
R. H.
,
Bernstein
M.
,
Keller-Peck
C.
,
Nguyen
Q. T.
,
Wallace
M.
,
Nerbonne
J. M.
,
Lichtman
J. W.
,
Sanes
J. R.
(
2000
).
Imaging neuronal subsets in transgenic mice expressing multiple spectral variants of GFP
.
Neuron
28
,
41
51
.
Ferrer-Vaquer
A.
,
Piliszek
A.
,
Tian
G.
,
Aho
R. J.
,
Dufort
D.
,
Hadjantonakis
A. K.
(
2010
).
A sensitive and bright single-cell resolution live imaging reporter of Wnt/ß-catenin signaling in the mouse
.
BMC Dev. Biol.
10
,
121
.
Hama
H.
,
Kurokawa
H.
,
Kawano
H.
,
Ando
R.
,
Shimogori
T.
,
Noda
H.
,
Fukami
K.
,
Sakaue-Sawano
A.
,
Miyawaki
A.
(
2011
).
Scale: a chemical approach for fluorescence imaging and reconstruction of transparent mouse brain
.
Nat. Neurosci.
14
,
1481
1488
.
Huber
A. B.
,
Kania
A.
,
Tran
T. S.
,
Gu
C.
,
De Marco Garcia
N.
,
Lieberam
I.
,
Johnson
D.
,
Jessell
T. M.
,
Ginty
D. D.
,
Kolodkin
A. L.
(
2005
).
Distinct roles for secreted semaphorin signaling in spinal motor axon guidance
.
Neuron
48
,
949
964
.
Ikawa
M.
,
Kominami
K.
,
Yoshimura
Y.
,
Tanaka
K.
,
Nishimune
Y.
,
Okabe
M.
(
1995
).
A rapid and non-invasive selection of transgenic embryos before implantation using green fluorescent protein (GFP)
.
FEBS Lett.
375
,
125
128
.
Jaubert-Miazza
L.
,
Green
E.
,
Lo
F. S.
,
Bui
K.
,
Mills
J.
,
Guido
W.
(
2005
).
Structural and functional composition of the developing retinogeniculate pathway in the mouse
.
Vis. Neurosci.
22
,
661
676
.
Krahe
T. E.
,
El-Danaf
R. N.
,
Dilger
E. K.
,
Henderson
S. C.
,
Guido
W.
(
2011
).
Morphologically distinct classes of relay cells exhibit regional preferences in the dorsal lateral geniculate nucleus of the mouse
.
J. Neurosci.
31
,
17437
17448
.
Little
G. E.
,
López-Bendito
G.
,
Rünker
A. E.
,
García
N.
,
Piñon
M. C.
,
Chédotal
A.
,
Molnár
Z.
,
Mitchell
K. J.
(
2009
).
Specificity and plasticity of thalamocortical connections in Sema6A mutant mice
.
PLoS Biol.
7
,
e98
.
Luo
L.
,
Callaway
E. M.
,
Svoboda
K.
(
2008
).
Genetic dissection of neural circuits
.
Neuron
57
,
634
660
.
Plump
A. S.
,
Erskine
L.
,
Sabatier
C.
,
Brose
K.
,
Epstein
C. J.
,
Goodman
C. S.
,
Mason
C. A.
,
Tessier-Lavigne
M.
(
2002
).
Slit1 and Slit2 cooperate to prevent premature midline crossing of retinal axons in the mouse visual system
.
Neuron
33
,
219
232
.
Rawat
S.
,
Raman Suri
C.
,
Sahoo
D. K.
(
2010
).
Molecular mechanism of polyethylene glycol mediated stabilization of protein
.
Biochem. Biophys. Res. Commun.
392
,
561
566
.
Rebsam
A.
,
Petros
T. J.
,
Mason
C. A.
(
2009
).
Switching retinogeniculate axon laterality leads to normal targeting but abnormal eye-specific segregation that is activity dependent
.
J. Neurosci.
29
,
14855
14863
.

Competing interests statement

The authors declare no competing financial interests.

Supplementary information