Abstract
In this photo essay, we present a sampling of technologies from laboratories at the forefront of whole-brain clearing and imaging for high-resolution analysis of cell populations and neuronal circuits. The data presented here were provided for the eponymous Mini-Symposium presented at the Society for Neuroscience's 2018 annual meeting.
Introduction
Microscopic analysis of tissues reveals that intricate organization of cells underlies biological function. Tissues are not translucent against visible light, so it is impossible to image far beyond the surface. Thus, conventional tissue imaging applies a microtome to cut samples into thin sections before staining with dyes and antibodies to visualize cells. Inferring 3D structure from thin sections is often problematic, and requires minimal sample distortion and precision alignment of serial sections.
The analysis of large 3D volumes is necessary for mapping the connections of far-reaching neurons inside the brain and determining the nature of cellular interactions underlying proper function and behavior. In recent years, several techniques have emerged to achieve optical transparency and enable high-resolution microscopy of thick tissue sections and whole organs. These techniques use different strategies to reduce light scattering in tissues and improve image sharpness.
Scattering occurs in tissues due to light's heterogeneous interaction with different molecules, subcellular structures, membranes, and cell populations inside the tissue. For example, the interface between a cell's lipid membrane and the cytoplasm causes a significant drop in refractive index (RI). Heterogeneity at the scale of molecules, cells, and tissues contribute to light scattering and requires homogenization via tissue clearing to increase overall light penetration (Tainaka et al., 2016; Treweek and Gradinaru, 2016). Combining tissue clearing with light-sheet fluorescence microscopy (LSFM) has paved the road for current whole-brain imaging by eliminating out-of-focus excitation (hence reduced background level and greatly preventing photo-bleaching), and by accelerating image acquisition.
Tissue clearing originated in the early 20th century, when Werner Spalteholz experimented with high RI organic solvents (Spalteholz, 1914). Organic solvent-based clearing homogenizes a tissue's RI by the removal of highly scattering lipids and the displacement of water by high RI solvents. Building on this approach, Dodt's group revitalized organic solvent-based tissue clearing for the modern era of neuroscience by coupling it with a new optical imaging method that improves the resolution and field-of-view using LSFM (Fig. 1; Dodt et al., 2007; Ertürk et al., 2012; Saghafi et al., 2014). This technique allows rapid imaging of larger volumes than with conventional point scanning microscopy. Erturk's group developed uDISCO method (Fig. 2) to image whole-mouse bodies (Pan et al., 2016) and recently vDISCO method, a whole-mouse immunolabeling method to amplify the signal two orders of magnitude for detecting single cells in cleared mouse bodies through intact bones and muscles (Cai et al., 2018). vDISCO enabled construction of the first whole-mouse neuronal connectivity map, study brain trauma effects in peripheral nerves, and image meningeal vessels and their cellular contest through intact skull.
Some techniques have replaced organic solvents with aqueous solutions to improve preservation of endogenous reporter proteins and simplicity of experimental handling, since a urea-based clearing reagent ScaleA2 was developed (Hama et al., 2011). To date, various mixtures and protocols have emerged that use aqueous chemicals for whole-tissue clearing. Miyawaki's group has additionally developed a urea/sorbitol-based clearing method called ScaleS (Hama et al., 2015) to achieve mild clarification where synaptic ultrastructure, observed by electron microscopy, is well preserved. ScaleS was capable of imaging Aβ plaques in rodent models of Alzheimer's disease and human clinical samples (Fig. 3). Ueda's group has developed a clearing method called CUBIC (clear, unobstructed brain/body imaging cocktails and computational analysis; Susaki et al., 2014, 2015; Tainaka et al., 2014; Kubota et al., 2017) and has recently reported controlled expansion of cleared mouse brains using CUBIC-X (Fig. 4; Murakami et al., 2018). Physical expansion of a sample (or swelling) enables imaging of subcellular structures across the entire mouse brain. Using LSFM, authors generated a point-based mouse brain atlas (called CUBIC-Atlas) with single-cell annotation, and tracked postnatal subregion development in mice.
Another important innovation in tissue imaging and phenotyping is the development of technologies that engineer tissue physicochemical properties, tissue-molecular interaction, and molecular transport within intact tissue. Chung's group has developed a suite of technologies that engineer tissue properties to maximally preserve tissue information using synthetic polymer (e.g., CLARITY, MAP) or crosslinker-based tissue reinforcement strategies (e.g., SWITCH, Fig. 5; Chung et al., 2013; Murray et al., 2015; Ku et al., 2016). They have also developed MAP (magnified analysis of proteome) that expands entire brains fourfold linearly while preserving 3D proteome for super-resolution molecular imaging. When the transport of reagents into a large volume is diffusion-limited, chemical reactions are slow and uneven. To achieve uniform tissue preservation and staining, the Chung group has developed SWITCH to control a wide range of chemical reaction kinetics. In addition, they have developed stochastic electro-transport (Kim et al., 2015) that accelerates lipid removal and tissue staining to substantially improve the throughput of intact tissue phenotyping approaches. Together, these technologies enable rapid, integrated, multiscale phenotyping of neuronal circuits.
Gradinaru's group has developed hydrogel-tissue chemistry (HTC)-based strategies to stabilize endogenous fluorescent proteins and tissue biomolecules during successive rounds of clearing, RNA labeling via single-molecule fluorescence in situ hybridization or hybridization chain reaction (HCR), immunohistochemistry, and imaging at depth (Fig. 6; Yang et al., 2014; Treweek et al., 2015; Shah et al., 2016; Greenbaum et al., 2017a,b). Recent work on viral vector engineering has led to the generation of novel AAV capsids with enhanced tropism for specific cell types, and with the ability to cross the blood–brain barrier when delivered systemically (Deverman et al., 2016; Chan et al., 2017). These vectors permit noninvasive gene transfer and tunable multicolor labeling of discrete cell populations across the CNS and PNS.
This sampling of current work demonstrates how the breathtaking complexity of the human brain and brains of other preclinical models continues to inspire technological advancements in whole-tissue clearing, fluorescent labeling, microscope design, and analysis of large information-rich datasets. These technological breakthroughs are paving the road to whole-brain single-cell atlases and connectomes (maps of connected neurons) for future neuroscientists.
Footnotes
The Dodt laboratory thanks Dr. Saiedeh Saghafi and Dr. Klaus Becker for aspheric optics-based LSFM work, and the Austrian science funding agency FWF and the German Hertie foundation for support; the Ertürk laboratory thanks Ruiyao Cai for uDISCO figure and Synergy Excellence Cluster Munich (SyNergy), Fritz Thyssen Stiftung, and DFG for support; the Miyawaki laboratory thanks Dr. Hiroshi Hama for ScaleS figure and Grant-in-Aid for Scientific Research on Priority Areas (JSPS KAKENHI), the Human Frontier Science Program, and the Brain Mapping by Integrated Neurotechnologies for Disease Studies (Brain/MINDS) for support; the Ueda laboratory thanks Tatsuya Murakami for offering picture materials for CUBIC figure, and Brain/MINDS (AMED/MEXT), the Basic Science and Platform Technology Program for Innovative Biological Medicine (AMED/MEXT)and Grant-in-Aid for Scientific Research (S) (JSPS KAKENHI) for support; the Chung laboratory thanks The Packard Award, the McKnight Foundation and the NIH (1-DP2-ES027992) for support; the Gradinaru laboratory thanks Sripriya Ravindra Kumar, Gerard M. Coughlin, Rosemary Challis, and Collin Challis for CREATE and VAST images, Min Jee Jang for HCR images, and J. Ryan Cho for GCaMP imaging, and the NIH BRAIN Initiative and NIH Director's Office and NSF Neuronex for support.
The authors declare no competing financial interests.
References
- Barretto RP, Ko TH, Jung JC, Wang TJ, Capps G, Waters AC, Ziv Y, Attardo A, Recht L, Schnitzer MJ (2011) Time-lapse imaging of disease progression in deep brain areas using fluorescence microendoscopy. Nat Med 17:223–228. 10.1038/nm.2292 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bedbrook CN, Deverman BE, Gradinaru V (2018) Viral strategies for targeting the central and peripheral nervous systems. Annu Rev Neurosci 41:323–348. 10.1146/annurev-neuro-080317-062048 [DOI] [PubMed] [Google Scholar]
- Cai R, Pan C, Ghasemigharagoz A, Todorov MI, Foerstera B, Zhao S, Bhatia HS, Mrowka L, Theodorou D, Rempfler M, Xavier A, Kress BT, Benakis C, Liesz A, Menze B, Kerschensteiner M, Nedergaard M, Erturk A (2018) Panoptic vDISCO imaging reveals neuronal connectivity, remote trauma effects and meningeal vessels in intact transparent mice. bioRxiv 374785. Advance online publication. Retrieved July 23, 2018. doi: 10.1101/374785. [DOI] [Google Scholar]
- Challis RC, Kumar SR, Chan KY, Challis C, Jang MJ, Rajendran PS, Tompkins JD, Shivkumar K, Deverman BE, Gradinaru V (2018) Widespread and targeted gene expression by systemic AAV vectors: production, purification, and administration. bioRxiv 246405. Advance onile publication. Retrieved Jan 19, 2018. doi: 10.1101/246405. [DOI] [Google Scholar]
- Chan KY, Jang MJ, Yoo BB, Greenbaum A, Ravi N, Wu WL, Sánchez-Guardado L, Lois C, Mazmanian SK, Deverman BE, Gradinaru V (2017) Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat Neurosci 20:1172–1179. 10.1038/nn.4593 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choi HM, Beck VA, Pierce NA (2014) Next-generation in situ hybridization chain reaction: higher gain, lower cost, greater durability. ACS Nano 8:4284–4294. 10.1021/nn405717p [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chung K, Wallace J, Kim SY, Kalyanasundaram S, Andalman AS, Davidson TJ, Mirzabekov JJ, Zalocusky KA, Mattis J, Denisin AK, Pak S, Bernstein H, Ramakrishnan C, Grosenick L, Gradinaru V, Deisseroth K (2013) Structural and molecular interrogation of intact biological systems. Nature 497:332–337. 10.1038/nature12107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deverman BE, Pravdo PL, Simpson BP, Kumar SR, Chan KY, Banerjee A, Wu WL, Yang B, Huber N, Pasca SP, Gradinaru V (2016) Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nat Biotechnol 34:204–209. 10.1038/nbt.3440 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dodt HU, Leischner U, Schierloh A, Jährling N, Mauch CP, Deininger K, Deussing JM, Eder M, Zieglgänsberger W, Becker K (2007) Ultramicroscopy: three-dimensional visualization of neuronal networks in the whole mouse brain. Nat Methods 4:331–336. 10.1038/nmeth1036 [DOI] [PubMed] [Google Scholar]
- Ertürk A, Becker K, Jährling N, Mauch CP, Hojer CD, Egen JG, Hellal F, Bradke F, Sheng M, Dodt HU (2012) Three-dimensional imaging of solvent-cleared organs using 3DISCO. Nat Protoc 7:1983–1995. 10.1038/nprot.2012.119 [DOI] [PubMed] [Google Scholar]
- Gradinaru V, Treweek J, Overton K, Deisseroth K (2018) Hydrogel-tissue chemistry: principles and applications. Annu Rev Biophys 47:355–376. 10.1146/annurev-biophys-070317-032905 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greenbaum A, Chan KY, Dobreva T, Brown D, Balani DH, Boyce R, Kronenberg HM, McBride HJ, Gradinaru V (2017a) Bone CLARITY: clearing, imaging, and computational analysis of osteoprogenitors within intact bone marrow. Sci Transl Med 9:eaah6518. 10.1126/scitranslmed.aah6518 [DOI] [PubMed] [Google Scholar]
- Greenbaum A, Jang MJ, Challis C, Gradinaru V (2017b) Q&A: how can advances in tissue clearing and optogenetics contribute to our understanding of normal and diseased biology? BMC Biol 15:87. 10.1186/s12915-017-0421-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. 10.1038/nn.2928 [DOI] [PubMed] [Google Scholar]
- Hama H, Hioki H, Namiki K, Hoshida T, Kurokawa H, Ishidate F, Kaneko T, Akagi T, Saito T, Saido T, Miyawaki A (2015) ScaleS: an optical clearing palette for biological imaging. Nat Neurosci 18:1518–1529. 10.1038/nn.4107 [DOI] [PubMed] [Google Scholar]
- Jung JC, Mehta AD, Aksay E, Stepnoski R, Schnitzer MJ (2004) In vivo mammalian brain imaging using one- and two-photon fluorescence microendoscopy. J Neurophysiol 92:3121–3133. 10.1152/jn.00234.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim SY, Cho JH, Murray E, Bakh N, Choi H, Ohn K, Ruelas L, Hubbert A, McCue M, Vassallo SL, Keller PJ, Chung K (2015) Stochastic electrotransport selectively enhances the transport of highly electromobile molecules. Proc Natl Acad Sci U S A 112:E6274–6283. 10.1073/pnas.1510133112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ku T, Swaney J, Park JY, Albanese A, Murray E, Cho JH, Park YG, Mangena V, Chen J, Chung K (2016) Multiplexed and scalable super-resolution imaging of three-dimensional protein localization in size-adjustable tissues. Nat Biotechnol 34:973–981. 10.1038/nbt.3641 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kubota SI, Takahashi K, Nishida J, Morishita Y, Ehata S, Tainaka K, Miyazono K, Ueda HR (2017) Whole-body profiling of cancer metastasis with single-cell resolution. Cell Rep 20:236–250. 10.1016/j.celrep.2017.06.010 [DOI] [PubMed] [Google Scholar]
- Marshall JD, Li JZ, Zhang Y, Gong Y, St-Pierre F, Lin MZ, Schnitzer MJ (2016) Cell-type-specific optical recording of membrane voltage dynamics in freely moving mice. Cell 167:1650–1662.e15. 10.1016/j.cell.2016.11.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murakami TC, Mano T, Saikawa S, Horiguchi SA, Shigeta D, Baba K, Sekiya H, Shimizu Y, Tanaka KF, Kiyonari H, Iino M, Mochizuki H, Tainaka K, Ueda HR (2018) A three-dimensional single-cell-resolution whole-brain atlas using CUBIC-X expansion microscopy and tissue clearing. Nat Neurosci 21:625–637. 10.1038/s41593-018-0109-1 [DOI] [PubMed] [Google Scholar]
- Murray E, Cho JH, Goodwin D, Ku T, Swaney J, Kim SY, Choi H, Park YG, Park JY, Hubbert A, McCue M, Vassallo S, Bakh N, Frosch MP, Wedeen VJ, Seung HS, Chung K (2015) Simple, scalable proteomic imaging for high-dimensional profiling of intact systems. Cell 163:1500–1514. 10.1016/j.cell.2015.11.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan C, Cai R, Quacquarelli FP, Ghasemigharagoz A, Lourbopoulos A, Matryba P, Plesnila N, Dichgans M, Hellal F, Ertürk A (2016) Shrinkage-mediated imaging of entire organs and organisms using uDISCO. Nat Methods 13:859–867. 10.1038/nmeth.3964 [DOI] [PubMed] [Google Scholar]
- Saghafi S, Becker K, Hahn C, Dodt HU (2014) 3D-ultramicroscopy utilizing aspheric optics. J Biophotonics 7:117–125. 10.1002/jbio.201300048 [DOI] [PubMed] [Google Scholar]
- Shah S, Lubeck E, Schwarzkopf M, He TF, Greenbaum A, Sohn CH, Lignell A, Choi HM, Gradinaru V, Pierce NA, Cai L (2016) Single-molecule RNA detection at depth by hybridization chain reaction and tissue hydrogel embedding and clearing. Development 143:2862–2867. 10.1242/dev.138560 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spalteholz W. (1914) Uber das durchsichtigmachen von menschlichen und tierischen präparaten und seine theoretischen bedingungen, nebst anhang: Uber Knochenfärbung. Leipzig: S. Hirzel. [Google Scholar]
- Susaki EA, Tainaka K, Perrin D, Kishino F, Tawara T, Watanabe TM, Yokoyama C, Onoe H, Eguchi M, Yamaguchi S, Abe T, Kiyonari H, Shimizu Y, Miyawaki A, Yokota H, Ueda HR (2014) Whole-brain imaging with single-cell resolution using chemical cocktails and computational analysis. Cell 157:726–739. 10.1016/j.cell.2014.03.042 [DOI] [PubMed] [Google Scholar]
- Susaki EA, Tainaka K, Perrin D, Yukinaga H, Kuno A, Ueda HR (2015) Advanced CUBIC protocols for whole-brain and whole-body clearing and imaging. Nat Protoc 10:1709–1727. 10.1038/nprot.2015.085 [DOI] [PubMed] [Google Scholar]
- Tainaka K, Murakami TC, Susaki EA, Shimizu C, Saito R, Takahashi K, Hayashi-Takagi A, Sekiya H, Arima Y, Nojima S, Ikemura M, Ushiku T, Shimizu Y, Murakami M, Tanaka KF, Iino M, Kasai H, Sasaoka T, Kobayashi K, Miyazono K, et al. (2018) Chemical landscape for tissue clearing based on hydrophilic reagents. Cell Rep 24:2196–2210.e9. 10.1016/j.celrep.2018.07.056 [DOI] [PubMed] [Google Scholar]
- Tainaka K, Kubota SI, Suyama TQ, Susaki EA, Perrin D, Ukai-Tadenuma M, Ukai H, Ueda HR (2014) Whole-body imaging with single-cell resolution by tissue decolorization. Cell 159:911–924. 10.1016/j.cell.2014.10.034 [DOI] [PubMed] [Google Scholar]
- Tainaka K, Kuno A, Kubota SI, Murakami T, Ueda HR (2016) Chemical principles in tissue clearing and staining protocols for whole-body cell profiling. Annu Rev Cell Dev Biol 32:713–741. 10.1146/annurev-cellbio-111315-125001 [DOI] [PubMed] [Google Scholar]
- Tomer R, Ye L, Hsueh B, Deisseroth K (2014) Advanced CLARITY for rapid and high-resolution imaging of intact tissues. Nat Protoc 9:1682–1697. 10.1038/nprot.2014.123 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Treweek JB, Gradinaru V (2016) Extracting structural and functional features of widely distributed biological circuits with single cell resolution via tissue clearing and delivery vectors. Curr Opin Biotechnol 40:193–207. 10.1016/j.copbio.2016.03.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Treweek JB, Chan KY, Flytzanis NC, Yang B, Deverman BE, Greenbaum A, Lignell A, Xiao C, Cai L, Ladinsky MS, Bjorkman PJ, Fowlkes CC, Gradinaru V (2015) Whole-body tissue stabilization and selective extractions via tissue-hydrogel hybrids for high-resolution intact circuit mapping and phenotyping. Nat Protoc 10:1860–1896. 10.1038/nprot.2015.122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang B, Treweek JB, Kulkarni RP, Deverman BE, Chen CK, Lubeck E, Shah S, Cai L, Gradinaru V (2014) Single-cell phenotyping within transparent intact tissue through whole-body clearing. Cell 158:945–958. 10.1016/j.cell.2014.07.017 [DOI] [PMC free article] [PubMed] [Google Scholar]