multiplex, quantitative cellular analysis in large tissue ...multiplex, quantitative cellular...

10
Multiplex, quantitative cellular analysis in large tissue volumes with clearing-enhanced 3D microscopy (C e 3D) Weizhe Li a , Ronald N. Germain a,1 , and Michael Y. Gerner a,b,1 a Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases/NIH, Bethesda, MD 20892; and b Department of Immunology, University of Washington, Seattle, WA 98109 Contributed by Ronald N. Germain, July 19, 2017 (sent for review May 31, 2017; reviewed by Marc K. Jenkins and Mark J. Miller) Organ homeostasis, cellular differentiation, signal relay, and in situ function all depend on the spatial organization of cells in complex tissues. For this reason, comprehensive, high-resolution mapping of cell positioning, phenotypic identity, and functional state in the context of macroscale tissue structure is critical to a deeper understanding of diverse biological processes. Here we report an easy to use method, clearing-enhanced 3D (C e 3D), which generates excellent tissue transparency for most organs, preserves cellular morphology and protein fluorescence, and is robustly com- patible with antibody-based immunolabeling. This enhanced sig- nal quality and capacity for extensive probe multiplexing permits quantitative analysis of distinct, highly intermixed cell populations in intact C e 3D-treated tissues via 3D histo-cytometry. We use this technology to demonstrate large-volume, high-resolution micros- copy of diverse cell types in lymphoid and nonlymphoid organs, as well as to perform quantitative analysis of the composition and tissue distribution of multiple cell populations in lymphoid tissues. Combined with histo-cytometry, C e 3D provides a comprehensive strategy for volumetric quantitative imaging and analysis that bridges the gap between conventional section imaging and disassociation-based techniques. tissue clearing | quantitative microscopy | histo-cytometry | immune system M ajor physiological processes rely on the precise positioning of diverse cell types in specific anatomical locations. Such organization allows exposure of cells to appropriate tissue mi- croenvironments that shape their differentiation, promote ap- propriate cellcell communication events, and collectively define the global properties of the whole organ. Understanding these structurefunction relationships requires a detailed mapping of both the large-scale organization and fine-grained molecular and cellular composition of complex tissues. The majority of information on such processes comes from mi- croscopic imaging of relatively thin (520 μm) 2Dtissue cross- sections, examining several markers of interest to visualize a limited number of cell populations with respect to a tissues representative structural elements. Although providing an excellent framework for understanding general features and the respective positioning of well-represented cell types, such data lack information on 3D or- ganization, being particularly limiting for irregular structures such as the vasculature, airways, nervous tissue, inflamed sites, tumors, or reactive lymph nodes. Furthermore, detection and analysis of rare cellular events requires imaging of a large number of disconnected sections, which introduces possible image selection bias and suffers from the potential omission of key physiological landmarks located just outside of the sampled area. Finally, many cell types require simultaneous visualization of multiple phenotypic markers for cor- rect subset identification, making interpretation of cell composition within tissues problematic without the use of highly multiplexed imaging panels. Recently, several tissue clearing methodologies have been developed that render organs transparent and allow section-free imaging of significant volumes, thereby improving our capacity to study the relationships between cell positioning and 3D tissue architecture (111). These techniques work by reducing light scattering in tissues through minimizing refractive index mis- matches between the immersion medium and the various protein, aqueous, and lipid tissue constituents (12). However, each of the currently reported clearing techniques suffers to a greater or lesser extent from various method-specific limitations. For example, techniques that excel at brain tissue clearing through lipid extrac- tion suffer from limited compatibility with immunolabeling, while solvent-based methods that permit useful antibody-based immu- nofluorescence microscopy induce substantial tissue shrinkage and are associated with suboptimal signal quality (1, 2, 6, 7, 9). These limitations prevent assessment of a tissues large-scale structural organization in concert with high-fidelity information on single cell morphology and complex molecular phenotype, while also limiting the combined use of fluorescent genetic markers and antibody- based staining. One application that requires highly optimized acquisition of such information is histo-cytometry (13). This imaging/analysis pipeline allows multiplex phenotypic identification and quanti- fication of cells directly in tissues, akin to in situ flow cytometry. This technique has been used to study the composition, distri- bution, and function of densely packed cells with complex phe- notypes and morphology in tissue sections (1321). Even for cell types with relatively simple morphology, such as T and B lym- phocytes, histo-cytometry requires a high degree of spatial signal resolution, achieved with high numerical aperture (N.A.) ob- jectives and deconvolution. The ability to simultaneously detect Significance Major biological processes rely on the precise positioning of di- verse cell types in specific anatomical locations. Existing tech- niques for studying cellular spatial positioning in tissues, especially with robust identification of densely packed cells, have substantial time, cost, resolution, and multiplexing limitations. Here, we describe an easy-to-use and inexpensive tissue clearing technique for attaining high-quality images of cells and diverse molecules of interest in substantial tissue volumes, enabling si- multaneous quantitative analysis of 3D organ structure and fine- grained cellular composition. This technology will enhance our capacity for acquiring a quantitative understanding of the rela- tionships between cells and their microenvironments in the context of broader tissue organization and is directly applicable to diverse biological disciplines as well as diagnostic medicine. Author contributions: W.L., R.N.G., and M.Y.G. designed research; W.L. and M.Y.G. per- formed research; W.L. and M.Y.G. contributed new reagents/analytic tools; W.L., R.N.G., and M.Y.G. analyzed data; and W.L., R.N.G., and M.Y.G. wrote the paper. Reviewers: M.K.J., University of Minnesota; and M.J.M., Washington University in St. Louis. Conflict of interest statement: The initial patent for the methodology described in this paper was filed with the US Patent Office, E-Numbers: E-168-2016/0-US-01 Appl. Serial No.: 62/380,593, Title: Clearing-enhanced 3d (ce3d): a novel tissue clearing method preserving cellular morphology, reporter fluorescence, epitope.1 To whom correspondence may be addressed. Email: [email protected] or gernermy@ uw.edu. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1708981114/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1708981114 PNAS | Published online August 14, 2017 | E7321E7330 IMMUNOLOGY AND INFLAMMATION PNAS PLUS Downloaded by guest on April 6, 2020

Upload: others

Post on 28-Mar-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Multiplex, quantitative cellular analysis in large tissue ...Multiplex, quantitative cellular analysis in large tissue volumes with clearing-enhanced 3D microscopy (C e3D) Weizhe Lia,

Multiplex, quantitative cellular analysis in large tissuevolumes with clearing-enhanced 3D microscopy (Ce3D)Weizhe Lia, Ronald N. Germaina,1, and Michael Y. Gernera,b,1

aLymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases/NIH, Bethesda, MD 20892; andbDepartment of Immunology, University of Washington, Seattle, WA 98109

Contributed by Ronald N. Germain, July 19, 2017 (sent for review May 31, 2017; reviewed by Marc K. Jenkins and Mark J. Miller)

Organ homeostasis, cellular differentiation, signal relay, and insitu function all depend on the spatial organization of cells incomplex tissues. For this reason, comprehensive, high-resolutionmapping of cell positioning, phenotypic identity, and functionalstate in the context of macroscale tissue structure is critical to adeeper understanding of diverse biological processes. Here wereport an easy to use method, clearing-enhanced 3D (Ce3D), whichgenerates excellent tissue transparency for most organs, preservescellular morphology and protein fluorescence, and is robustly com-patible with antibody-based immunolabeling. This enhanced sig-nal quality and capacity for extensive probe multiplexing permitsquantitative analysis of distinct, highly intermixed cell populationsin intact Ce3D-treated tissues via 3D histo-cytometry. We use thistechnology to demonstrate large-volume, high-resolution micros-copy of diverse cell types in lymphoid and nonlymphoid organs, aswell as to perform quantitative analysis of the composition andtissue distribution of multiple cell populations in lymphoid tissues.Combined with histo-cytometry, Ce3D provides a comprehensivestrategy for volumetric quantitative imaging and analysis thatbridges the gap between conventional section imaging anddisassociation-based techniques.

tissue clearing | quantitative microscopy | histo-cytometry | immune system

Major physiological processes rely on the precise positioningof diverse cell types in specific anatomical locations. Such

organization allows exposure of cells to appropriate tissue mi-croenvironments that shape their differentiation, promote ap-propriate cell–cell communication events, and collectively definethe global properties of the whole organ. Understanding thesestructure–function relationships requires a detailed mapping ofboth the large-scale organization and fine-grained molecular andcellular composition of complex tissues.The majority of information on such processes comes from mi-

croscopic imaging of relatively thin (5–20 μm) “2D” tissue cross-sections, examining several markers of interest to visualize a limitednumber of cell populations with respect to a tissue’s representativestructural elements. Although providing an excellent framework forunderstanding general features and the respective positioning ofwell-represented cell types, such data lack information on 3D or-ganization, being particularly limiting for irregular structures such asthe vasculature, airways, nervous tissue, inflamed sites, tumors, orreactive lymph nodes. Furthermore, detection and analysis of rarecellular events requires imaging of a large number of disconnectedsections, which introduces possible image selection bias and suffersfrom the potential omission of key physiological landmarks locatedjust outside of the sampled area. Finally, many cell types requiresimultaneous visualization of multiple phenotypic markers for cor-rect subset identification, making interpretation of cell compositionwithin tissues problematic without the use of highly multiplexedimaging panels.Recently, several tissue clearing methodologies have been

developed that render organs transparent and allow section-freeimaging of significant volumes, thereby improving our capacity tostudy the relationships between cell positioning and 3D tissuearchitecture (1–11). These techniques work by reducing light

scattering in tissues through minimizing refractive index mis-matches between the immersion medium and the various protein,aqueous, and lipid tissue constituents (12). However, each of thecurrently reported clearing techniques suffers to a greater or lesserextent from various method-specific limitations. For example,techniques that excel at brain tissue clearing through lipid extrac-tion suffer from limited compatibility with immunolabeling, whilesolvent-based methods that permit useful antibody-based immu-nofluorescence microscopy induce substantial tissue shrinkage andare associated with suboptimal signal quality (1, 2, 6, 7, 9). Theselimitations prevent assessment of a tissue’s large-scale structuralorganization in concert with high-fidelity information on single cellmorphology and complex molecular phenotype, while also limitingthe combined use of fluorescent genetic markers and antibody-based staining.One application that requires highly optimized acquisition of

such information is histo-cytometry (13). This imaging/analysispipeline allows multiplex phenotypic identification and quanti-fication of cells directly in tissues, akin to in situ flow cytometry.This technique has been used to study the composition, distri-bution, and function of densely packed cells with complex phe-notypes and morphology in tissue sections (13–21). Even for celltypes with relatively simple morphology, such as T and B lym-phocytes, histo-cytometry requires a high degree of spatial signalresolution, achieved with high numerical aperture (N.A.) ob-jectives and deconvolution. The ability to simultaneously detect

Significance

Major biological processes rely on the precise positioning of di-verse cell types in specific anatomical locations. Existing tech-niques for studying cellular spatial positioning in tissues,especially with robust identification of densely packed cells, havesubstantial time, cost, resolution, and multiplexing limitations.Here, we describe an easy-to-use and inexpensive tissue clearingtechnique for attaining high-quality images of cells and diversemolecules of interest in substantial tissue volumes, enabling si-multaneous quantitative analysis of 3D organ structure and fine-grained cellular composition. This technology will enhance ourcapacity for acquiring a quantitative understanding of the rela-tionships between cells and their microenvironments in thecontext of broader tissue organization and is directly applicableto diverse biological disciplines as well as diagnostic medicine.

Author contributions: W.L., R.N.G., and M.Y.G. designed research; W.L. and M.Y.G. per-formed research; W.L. and M.Y.G. contributed new reagents/analytic tools; W.L., R.N.G.,and M.Y.G. analyzed data; and W.L., R.N.G., and M.Y.G. wrote the paper.

Reviewers: M.K.J., University of Minnesota; and M.J.M., Washington University inSt. Louis.

Conflict of interest statement: The initial patent for the methodology described in thispaper was filed with the US Patent Office, E-Numbers: E-168-2016/0-US-01 Appl. Serial No.:62/380,593, Title: “Clearing-enhanced 3d (ce3d): a novel tissue clearing method preservingcellular morphology, reporter fluorescence, epitope.”1To whom correspondence may be addressed. Email: [email protected] or [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1708981114/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1708981114 PNAS | Published online August 14, 2017 | E7321–E7330

IMMUNOLO

GYAND

INFLAMMATION

PNASPL

US

Dow

nloa

ded

by g

uest

on

Apr

il 6,

202

0

Page 2: Multiplex, quantitative cellular analysis in large tissue ...Multiplex, quantitative cellular analysis in large tissue volumes with clearing-enhanced 3D microscopy (C e3D) Weizhe Lia,

multiple distinct probes, achieved by multiplex labeling with di-rectly conjugated antibodies, is critical for this technique, as itallows in-depth in situ phenotypic analysis, including definitionof cellular lineages, analysis of ongoing signaling events throughphosphoprotein detection, and delineation of functional statessuch as cytokine production (16). Furthermore, this analyticalimaging approach can achieve more accurate cellular enumera-tion compared with disassociation-based flow cytometry, espe-cially for highly adherent cell types (13, 22). However, ourattempts to use histo-cytometry on organs cleared with existingtechniques that permit modest multiplex antibody stainingproved challenging due to relatively poor signal-to-noise char-acteristics and lack of clear-cut cellular definition (14).Here, we describe a tissue clearing method, clearing-enhanced

3D (Ce3D). This technically nondemanding and inexpensivetechnique simultaneously achieves: excellent tissue transparency,maintenance of fluorescence from diverse reporter proteins andorganic fluorophores, retention of cellular morphology, and acapacity for highly multiplexed antibody-based immunolabeling.Ce3D allows rapid clearing of most tissues, other than erythrocyte-dense organs rich in light-absorbing heme, and works well withlipid-rich brain tissues using additional mild detergent treatment.Here, we applied Ce3D to multiplexed confocal imaging of variouslymphoid and nonlymphoid organs and cell types. The high signalquality and spatial resolution achieved with Ce3D allowed us toperform accurate quantitative analysis of cellular phenotypic pro-files in dense lymphoid tissues using histo-cytometry, usingoptimized algorithms for cellular segmentation in 3D volumes.Collectively, this clearing technique and analysis pipeline pro-vides a significant advance, attaining nearly lossless tissue trans-parency that supports large volumetric multiplexed microscopyand quantitative tissue cytometry.

ResultsOptimization of a Clearing Protocol. Recently described tissueclearing techniques have been valuable for visualization of mac-roscale 3D tissue architecture, suggesting that such approachesmay also allow examination of the fine-grained relationships be-tween various cell populations and their microanatomical sur-roundings. However, consistent with reports by others (2, 3, 8, 12),in our hands all major clearing methodologies (CUBIC, CLARITY,PACT, SeeDB, ClearT, ClearT2, Scale, AbScale, SWITCH,DISCO-based methods, BABB) failed to simultaneously yieldoptimal tissue transparency, bright reporter protein fluorescence,strong immunolabeling via directly conjugated antibodies, as wellas minimally perturbed tissue and cellular morphology (SI Ap-pendix, Figs. S1 and S2 A and B and Table S1). For some tech-niques, immunolabeling could be partially rescued via secondaryreagent amplification, although this is problematic for extensiveprobe multiplexing with antibody clones derived from the samehost species (SI Appendix, Fig. S2C). Considering the importanceof all these parameters for accurate characterization of pheno-typically and morphologically complex cells in large 3D volumes,we performed a chemical screen based on previous methods tofind a clearing method that would satisfy the following criteria:tissue clarity, conservation of reporter protein fluorescence, opti-mal preservation of antibody-based staining for multiplex imaging,overall signal quality, morphological integrity at the cell and tissuelevel, minimal clearing time, and low reagent costs.We began our search using 1% paraformaldehyde (PFA)-fixed

murine lymph nodes (LNs). These tissues are ideal for suchscreening as they are (i) composed of diverse hematopoietic andstromal cell types with discrete phenotypic, morphologic, andspatial distribution properties; (ii) have cells that are denselypacked, requiring a high degree of signal-to-cell spatial fidelityfor quantitative image analysis; and (iii) are ≈1–2 mm3, allowingrapid treatment followed by full volume imaging with conven-tional confocal objectives with reasonable N.A. One percent

PFA, instead of 4%, was used to minimize epitope loss andimprove tissue transparency. To further facilitate our search, weused tissues from Itgax-Venus (commonly known as CD11c-YFP) mice, which were fixed and then stained for 1–3 d withantibodies against various lymphocyte and stromal cell markersthat have been well characterized to work with PFA-fixed tissues.This approach allowed us to ascertain a reagent’s clearingproperties together with its ability to retain signal from fluores-cent proteins and permit antibody-based staining. It also pro-vided information on preservation of fine cellular structuresbased on examination of morphologically complex dendrites ofCD11c-YFP–expressing cells.Solvent-based clearing approaches, such as BABB and different

DISCO techniques, induce clearance at the cost of significanttissue shrinkage and loss of reporter protein fluorescence (1, 2, 9).Therefore, our primary screening was based on published aqueouspassive immersion reagents, which on their own can reduce tissuelight scattering. For this we compared various concentrations ofHistodenz and fructose, previously described inexpensive immer-sion reagents that provide the ability to modulate the final re-fractive index through concentration adjustment (7, 11). Asexpected, immersion media alone only partially cleared the tissues.However, Histodenz was noted to be of substantially lower vis-cosity compared with fructose, allowing improved equilibrationwith the tissues, and was thus used for further screening of primaryclearing reagents.Comparison of various published aqueous primary clearing re-

agents demonstrated that formamide, a urea-like chemical used inthe ClearT methodology (10), when combined with Histodenz in-duced exceptional transparency in treated tissues, while preservingYFP fluorescence and cell morphology. However, in our hands,antibody-based staining was poor with this treatment and did notpermit imaging using the majority of tested antibodies, especiallywith directly conjugated probes (SI Appendix, Fig. S2 A and B). Asecondary screen based on formamide’s molecular structure wasperformed (SI Appendix, Table S2). Of the tested compounds,N-methylacetamide, 2-chloro-N-(hydroxymethyl)acetamide and N,N-dimethylacetamide yielded excellent optical tissue transparencywithin 12 h of tissue incubation at room temperature. However,only N-methylacetamide (22% wt/vol) in Histodenz (86% wt/vol)yielded tissue transparency with retention of reporter protein fluo-rescence and an ability to perform direct multiplex antibody-basedimmunolabeling (Fig. 1A and SI Appendix, Fig. S3). As commonlyobserved with other clearing protocols, some yellow/brownish tissuediscoloration was noted, which was minimized by addition of1-thioglycerol (0.5% vol/vol) (11). Finally, a mild detergent, 0.1%Triton X-100, was added to the final Ce3D clearing solution toexpedite the clearing process.Once optimal LN clearing was obtained with Ce3D treatment,

we performed confocal imaging of both reporter proteins andvarious epitopes labeled with directly conjugated antibody probes.The ∼650-μm working distance of a 20× 0.75-N.A. objectivecoupled with motorized stage tiling allowed us to image the en-tirety of smaller LNs using a conventional confocal microscope.Various hematopoietic and nonhematopoietic cell types wereeasily visualized throughout the whole ∼650-μm imaging rangewith excellent signal fidelity (Fig. 1B and SI Appendix, Fig. S3).Clear-cut demarcation of B cells, dendritic cells, T cells, andlymphatic endothelium was seen, with the appropriate tissue-levelcompartmentalization (SI Appendix, Fig. S3). These results sug-gested that Ce3D achieves LN tissue transparency, preserves YFPreporter protein fluorescence, and is generally compatible withantibody-based immunofluorescence microscopy.We next examined the performance of Ce3D for clarification

and imaging of other major mouse organs. We observed robustclearing of lung, intestine, liver, muscle, and thymus, as well aspreservation of reporter protein fluorescence in these organs(Fig. 1 C and D and see below). Similarly, Ce3D treatment of

E7322 | www.pnas.org/cgi/doi/10.1073/pnas.1708981114 Li et al.

Dow

nloa

ded

by g

uest

on

Apr

il 6,

202

0

Page 3: Multiplex, quantitative cellular analysis in large tissue ...Multiplex, quantitative cellular analysis in large tissue volumes with clearing-enhanced 3D microscopy (C e3D) Weizhe Lia,

bone samples demonstrated rapid gain of transparency for entirefemurs without necessitating decalcification, suggesting thatdense connective tissues are also compatible with this technique.Kidneys could also be cleared and imaged with Ce3D, but re-quired extensive portal vein perfusion (SI Appendix, Fig. S4A).Only the spleen that is exceptionally rich in erythrocytes did notachieve optical transparency with Ce3D in our studies, pre-sumably due to the preservation of heme’s molecular structureand its light absorbing characteristics. Brain tissues rich in lipidsalso gained transparency, but required substantially longer in-cubation times in the clearing medium (≈2 wk per 1 mm of tis-sue). To minimize this lag time, we explored various detergentsfor enhancing lipid removal and found that commercial saponin-

based fixation and permeabilization flow cytometry buffers pro-moted dramatically faster rates of brain tissue clearing (∼24 hper millimeter of tissue), while also maintaining the capacity forantibody-based immunolabeling (Fig. 1C, see below).To quantitatively assess the effective light transmittance

gained with Ce3D treatment, we measured the fluorescence inYFP-expressing cells in various tissues using a fixed laser/detector power setting throughout the working range of theobjective. As expected, uncleared organ imaging resulted in rapiddecay of signal detection within the first 50 μm of imaging. Incontrast, minimal signal attenuation was observed over the first200 μm in Ce3D-treated tissues, although beyond that depth, re-sidual light scatter did result in decreased signal intensity (Fig. 1D).

Fig. 1. Ce3D attains tissue transparency while retaining reporter protein fluorescence and capacity for immunolabeling. (A) Images of fixed LNs were ac-quired before (PBS) and after Ce3D treatment. (B) LNs were stained with the indicated antibodies, cleared with Ce3D, and imaged by confocal microscopy,with the zoom-in panel (Bottom) demonstrating the capacity to resolve individual cells. (C) Images of various murine organs were acquired before andafter Ce3D treatment. (D) Average cellular fluorescence for CD11c-YFP–expressing cells across indicated imaging depths was quantified for PBS (black) or Ce3D(red) -treated tissues. Laser power ramping and signal attenuation correction was not used for quantitative comparison of fluorescence detection. Imagesrepresent at least two independent experiments.

Li et al. PNAS | Published online August 14, 2017 | E7323

IMMUNOLO

GYAND

INFLAMMATION

PNASPL

US

Dow

nloa

ded

by g

uest

on

Apr

il 6,

202

0

Page 4: Multiplex, quantitative cellular analysis in large tissue ...Multiplex, quantitative cellular analysis in large tissue volumes with clearing-enhanced 3D microscopy (C e3D) Weizhe Lia,

In practice, however, such attenuation could be easily correctedwith a minor laser power ramp (∼2–3× laser intensity increaseacross 650 μm), as well as further optimized by software-basedattenuation correction algorithms after image acquisition, allow-ing for productive, high-quality imaging throughout the entire650-μm working distance range of the objective (Fig. 1B and SIAppendix, Fig. S3).Tissue deformation and morphological distortions are common

artifacts observed with clearing protocols (12). Ce3D inducedminimal change in volume for many tested organs, although someshrinkage (10–20%) was observed for select tissues, such as brain,gut, and muscle (SI Appendix, Fig. S4B). To examine whether suchshrinkage would influence cellular morphology, we compared themorphology of astrocytes, cells with fine processes, in untreatedbrain slices vs. in tissues cleared with the different methodologies.This approach was taken because brains demonstrated the mostdiscernible volumetric contraction and the astrocyte-specificGFAP antibody has been previously reported to work with a va-riety of clearing techniques, allowing direct method comparison.Indeed, astrocyte labeling was observed with all tested techniques,although iDISCO treatment resulted in reduced fluorescent signaldetection (SI Appendix, Fig. S4C). Importantly, Ce3D led to anoverall preservation of astrocyte integrity and morphologicalcomplexity compared with cells in noncleared control thin sec-tions. In contrast, substantial shrinkage and rounding, as well asnoticeable serration of cellular processes, was observed withiDISCO-based clearing. Some serration was also noticeable withPACT clearing. To further assess the effect of Ce3D on cellmorphology, we visualized neuronal cell bodies using brain tissuesisolated from Confetti reporter animals (see below). Althoughshrinkage of the cell bodies (12–15%) was observed, consistentwith the change in organ volume, the overall neuronal morphologyappeared intact, while the improved fluorescent reporter signaldetection due to optical clearing led to enhanced visualization offine axonal processes (SI Appendix, Fig. S4D). Furthermore, weexamined the morphology of myeloid dendritic cells in LNs andsmall intestines, and observed no significant alterations in cellshape and volume before and after Ce3D clearing (SI Appendix,Fig. S4E). Finally, we observed no evidence of differential regionaldeformation within the cleared tissues, as the relative diameter ofcells remained unchanged within different tissue sites, suggestingoverall uniformity of volumetric contraction during the clearingprocess. Collectively, these data indicate that although Ce3D doeslead to modest tissue shrinkage in certain organs, it preservesexcellent morphological integrity, and due to improved signaldetection, allows enhanced visualization of fine cellular processes.To test the compatibility of Ce3D with various commonly used

fluorescent reporter proteins, we examined tissues from Confettireporter mice, which express GFP, RFP, YFP, and CFP in dis-tinct cells. Ce3D treatment led to preservation of fluorescencefor these reporters, as well as to improved detection of theseproteins, with tissues stored in Ce3D medium retaining fluores-cence for more than 4 wk (SI Appendix, Fig. S5A). Similar resultswere observed for DsRed-expressing tissues (SI Appendix, Fig.S4A). Next, we tested how well Ce3D enables combined use ofantibody-based and reporter protein imaging by comparing thestaining intensity of labeled antibodies against distinct B- andT-cell markers before and after Ce3D treatment of thin CD11c-YFP tissue sections. Both lymphocyte populations were detectedin the physiologically appropriate compartments, with antibody-stained and YFP-expressing cells showing the same or betterfluorescence signal detection after Ce3D treatment (SI Appendix,Fig. S5B). Further testing of multiple fluorophore-conjugatedantibodies against diverse biological targets and in various tissuesalso demonstrated similar performance to that seen with PFA-fixed, noncleared tissue sections, suggesting good preservation ofantibody–antigen complexes with Ce3D (SI Appendix, Table S3).Moreover, all fluorophores commonly used in immunolabeling

that we tested were compatible with the Ce3D protocol, retainingstable fluorescence for several weeks after clearing (SI Appendix,Table S3).Volumetric imaging of reporter protein expressing and

antibody-stained tissues after Ce3D processing revealed at highresolution the complex relationships of diverse cell types withtissue-specific anatomical landmarks. As an example, small in-testines from CD11c-YFP reporter animals allowed visualizationof the spatial associations between the densely packed epithelialcells, intraepithelial lymphocytes, lymphatic vessels, and den-dritic cells (Fig. 2A and SI Appendix, Fig. S6A and Movie S1).Cleared 1-mm brain slices from Confetti animals allowed 3Dvisualization of neurons (Fig. 2B and Movie S2), while GFAP-stained CD11c-YFP brain tissues demonstrated fine morpho-logical processes for astrocytes and dendritic cells, respectively,and imaging of cleared Cx3CR1-GFP reporter brains revealedfine branching of ramified microglial cells (Fig. 2C and SI Ap-pendix, Fig. S6 B and C and Movie S3). Furthermore, complex3D networks of blood vessels in the brain were revealed usingDsRed reporter animals (Fig. 2D and Movie S3). Large volu-metric scans of diverse CD11c-YFP tissues permitted visualiza-tion of additional cellular anatomical relationships, such as closeassociations of dendritic cells with lung alveolar epithelium, asrevealed by either confocal or light sheet imaging (Fig. 2 E and Fand SI Appendix, Fig. S6D and Movie S4), dense dendritic cellaggregates in thymic medullary regions, and close associations ofYFP-expressing Kupffer cells with liver sinusoids (SI Appendix,Fig. S6 E and F). Similarly, cleared dsRed femurs allowed high-resolution confocal imaging of blood vessels within the calcium-rich periosteum (Fig. 2G). Collectively, these results demon-strate that Ce3D achieves tissue transparency with preservationof overall tissue and cellular integrity in a variety of organ sys-tems and is compatible with fluorescent reporter proteins andantibody-based labeling, thus allowing comprehensive high-resolution imaging across large tissue volumes.

Quantitative Volumetric Cell-Level Analysis Using Ce3D and Histo-Cytometry. This simultaneous high degree of signal quality, ca-pacity for extensive probe multiplexing, and preservation of cel-lular morphology obtained with Ce3D imaging suggested that 3Dquantitative histo-cytometry might also be feasible. This techniqueextracts statistical information for imaged cells, such as the asso-ciated fluorescent intensity, cellular morphological properties, andposition (13). This in turn allows software-based comparison andpopulation gating of cells, akin to in situ flow cytometry, with themajor added benefit of retaining information on cellular spatialdistribution in tissues. Previous cell segmentation techniques,necessary for identifying individual cells within an image, usedeither more restrictive segmentation of a single cell type (e.g.,CD11c-YFP signal-based segmentation), or nuclear stain-basedsegmentation for all nucleated cells (13). As the latter approacheffectively provides quantitative information on all imaged nu-cleated cells and not just select subpopulations, we first attemptedto perform whole-mount nuclear staining for nuclear-based seg-mentation. However, substantial inhomogeneity in nuclear label-ing was observed, with significantly greater staining in peripheralregions of dye-incubated tissues. Modifications to dye concentra-tions, the choice of probe (e.g., DAPI, Hoechst, Yoyo-1, Jojo-1,and ToPro-5), or labeling time failed to normalize staining in-tensity across tissues. Nuclear segmentation of tissues from histoneGFP-tagged reporter animals also failed to produce accurate re-sults due to significant cellular heterogeneity in nuclear fluores-cence. To circumvent these issues, we explored various imageprocessing pipelines to accurately segment all imaged cells in-dependent of nuclear staining. Effective cell object creation wasachieved via seeded watershed segmentation of a calculatedchannel representing the inverse sum of all membrane signals (Fig.3A). Additional enhancements of cell separation and segmentation

E7324 | www.pnas.org/cgi/doi/10.1073/pnas.1708981114 Li et al.

Dow

nloa

ded

by g

uest

on

Apr

il 6,

202

0

Page 5: Multiplex, quantitative cellular analysis in large tissue ...Multiplex, quantitative cellular analysis in large tissue volumes with clearing-enhanced 3D microscopy (C e3D) Weizhe Lia,

500um 100um

Lung - Cytokeratin CD11c-YFP

Lung - Cytokeratin

500um

A

B

C

F G

D E

Fig. 2. Large volumetric Ce3D microscopy of diverse organs. (A) Segments of the proximal small intestine were stained with the indicated antibodies, clearedand imaged, allowing visualization of the spatial relationships between the mucosal epithelium (EpCAM+, cyan), T cells (CD3+, red), and lymphatics (Lyve-1+,yellow). (B) Ce3D-processed 1-mm Confetti reporter brain slices were imaged, allowing visualization of neuronal processes and blood vessels. (C) GFAP-stainedCD11c-YFP brain slices demonstrate compatibility of reporter and immunolabeling-based fluorescence, as well as preservation of cell morphology with Ce3D.(D) Cleared DsRed reporter brain tissues allow visualization of complex vascular networks. Cytokeratin-stained lungs from CD11c-YFP mice allow 3D visu-alization of bronchial epithelial cells and dendritic cells via light-sheet (E) or confocal (F) microscopy. (G) Visualization of periosteal blood vessels within actin-DsRed Ce3D-cleared femurs. Images represent at least two independent experiments.

Li et al. PNAS | Published online August 14, 2017 | E7325

IMMUNOLO

GYAND

INFLAMMATION

PNASPL

US

Dow

nloa

ded

by g

uest

on

Apr

il 6,

202

0

Page 6: Multiplex, quantitative cellular analysis in large tissue ...Multiplex, quantitative cellular analysis in large tissue volumes with clearing-enhanced 3D microscopy (C e3D) Weizhe Lia,

were attained by creating binary skeleton channels derived fromthe sum of membrane signals, and then by subtracting these fromthe inverse sum channel. Exporting statistics data for the seg-mented cell objects into flow cytometry analysis software (FlowJo)and plotting mean voxel fluorescence intensity allowed robustdiscrimination of various lymphocyte populations based on markerexpression (Fig. 3B). Importantly, analysis of the cellular positionof the gated cell types revealed physiologically appropriate local-ization in LN tissues (23, 24), with positioning of B cells in theperipheral follicular regions and with T cells concentrated in thecentral T-cell zone (Fig. 3C).To further examine whether Ce3D volume imaging would al-

low histo-cytometric analysis of multiple cell populations, LNswere stained with a multiplexed panel of antibodies against di-verse hematopoietic and stromal cell types, cleared, and imagedwith a 20× 0.75 N.A. objective. Collected images were com-pensated for fluorescence spillover between channels, decon-volved to enhance spatial signal allocation, and corrected forsignal attenuation in the axial plane. Three-dimentional imagevisualization revealed the highly branched organization of theblood and lymphatic vessel networks, along with their relation-ships to the B-cell follicles and T-cell zones within LNs (Fig. 4Aand SI Appendix, Fig. S7A and Movie S5). We next performedcellular segmentation and exported cell object statistics toFlowJo software for quantitative cellular analysis (Fig. 4B and SIAppendix, Fig. S7B). Large, irregular events or objects lackingcell-associated signal (sum of cell membrane channel) wereeliminated from further evaluation. CD31+ blood endothelialcells (BECs), Lyve1+ lymphatic endothelial cells (LECs), CD169+

subcapsular sinus macrophages (Macs), B cells, and T cellswere readily identified in a hierarchical gating analysis (Fig.4C). Importantly, XYZ positioning for all gated cell pop-ulations precisely mirrored the original image dataset and theexpected physiological compartmentalization (23, 24), suggestingaccurate cellular segmentation, analysis, and spatial allocation(Fig. 4D and Movie S6).One area of concern was that many LECs appeared to show

expression of CD169, a marker typically associated with LN mac-rophages (25) (Fig. 4C). Closer image examination revealed ex-tremely tight association of medullary macrophages and LECs, withthe CD169 and Lyve-1 signals not readily separable using the 0.75N.A. optics, especially in the axial plane (SI Appendix, Fig. S7A,cross-section zoom). Such signal misallocation would thus lead toinaccurate phenotypic characterization of LECs with histo-cytometry. These results suggested that higher N.A. objectivesmay be necessary for accurate phenotypic characterization ofclosely juxtaposed cells, as was previously observed in section-based histo-cytometry analysis of dendritic cells and T cells (13).Indeed, use of 40× 1.3 N.A. optics, with further enhancement bydeconvolution, readily separated juxtaposed macrophages andLECs in the axial and lateral planes and allowed accuratequantitative histo-cytometry of LECs and macrophages (SI Ap-pendix, Fig. S7 C and D).To further probe the fidelity of Ce3D histo-cytometry for

closely proximal cells, we examined whether phenotypic char-acterization of CD4+ and CD8+ T cells could be achieved. TheseT-cell populations are tightly packed within the LN T-cell zoneand do not share expression of the CD4 and CD8 coreceptormarkers, thereby providing an excellent opportunity for testingthe Ce3D histo-cytometry pipeline (Fig. 5A and SI Appendix, Fig.S8). Recent section-based image analysis has revealed a morefine-grained microanatomical organization of the T-cell zone, withimmunologically experienced CD44+ memory T-cell populations

A

B

C

Fig. 3. Volumetric segmentation pipeline for quantitative tissue analysis.(A) Cell membrane stains were normalized by intensity and summed to-gether to yield the sum channel. This channel was then inverted to create aninverse sum channel and used to create 2D skeletons in the XY, XZ, and YZplanes. These skeletons were then subtracted from the inverse sum channelto enhance the separation between cells. This enhanced image was thensegmented to create individual cell objects. (B) The statistics for the cellobjects were exported into FlowJo for cellular phenotyping and populationgating. (C) Positional information on gated cell populations was used to

quantitatively visualize cellular position. Data are representative of at leastthree independent experiments.

E7326 | www.pnas.org/cgi/doi/10.1073/pnas.1708981114 Li et al.

Dow

nloa

ded

by g

uest

on

Apr

il 6,

202

0

Page 7: Multiplex, quantitative cellular analysis in large tissue ...Multiplex, quantitative cellular analysis in large tissue volumes with clearing-enhanced 3D microscopy (C e3D) Weizhe Lia,

predominantly residing in the interfollicular regions, and withregulatory CD4+CD25+ T cells dispersed throughout the T-cellzone but also occasionally localized in tight clusters in the pe-

ripheral regions of the LN paracortex (16, 26, 27). Staining of LNtissues with the appropriate antibody panels followed by Ce3Dimaging and histo-cytometric analysis allowed discrimination of

A

B

D

C

Fig. 4. Ce3D permits volumetric multiplex imaging and quantitative histo-cytometry. (A) LNs were stained with the indicated antibodies, cleared, and imaged with a 20×0.7 5 N.A. objective. (B) Cell membrane signals were used to create cell objects, which were then exported into FlowJo. The 3D organ reconstruction in FlowJo is presentedand compared with the original image. (C) Cell object statistics were used for population phenotypic analysis and gating of blood endothelial cells (BECs), lymphaticendothelial cells (LECs), macrophages (Macs), as well as B and T cells. (D) Gated cell populations from Cwere displayed on an XY positional plot for the Z-dimension gatesdefined in B (red rectangles) and compared with the same Z planes from the original image. Images are representative of at least two independent experiments.

Li et al. PNAS | Published online August 14, 2017 | E7327

IMMUNOLO

GYAND

INFLAMMATION

PNASPL

US

Dow

nloa

ded

by g

uest

on

Apr

il 6,

202

0

Page 8: Multiplex, quantitative cellular analysis in large tissue ...Multiplex, quantitative cellular analysis in large tissue volumes with clearing-enhanced 3D microscopy (C e3D) Weizhe Lia,

all cellular populations of interest, such as B cells, and im-portantly CD4+ and CD8+ T cells, with little marker overlap,suggesting that the 1.3 N.A. optics allowed accurate signal–cell

allocation during image segmentation (Fig. 5 B and C). CD44+

memory T cells and CD25+ regulatory CD4+ T cells were alsodetected at the expected frequencies and displayed previously

A B

C

D

Fig. 5. Ce3D histo-cytometry of closely adjoining cells in large tissue volumes. (A) LNswere stainedwith the indicated antibodies, cleared, and imagedwith a 40×1.3 N.A.objective (Top). A zoom-in image, corresponding to a single virtual Z section within the area highlighted by the white box (Top), demonstrates the visual clarity of variouslymphocyte subpopulations (Bottom). (B) All imaged cells were segmented and their statistical information was exported into FlowJo for quantitative tissue visualization,as well as (C) population phenotypic analysis and gating. (D) Indicated T-cell populations were displayed as pseudocolor density XY positional plots, with the B cellssuperimposed (gray density gradients) to highlight regionalized cell distribution. Zoom-in image demonstrates an area enriched in CD4+CD25+ cell clusters, with theoriginal microscopy image presented below. Images are representative of at least two independent experiments.

E7328 | www.pnas.org/cgi/doi/10.1073/pnas.1708981114 Li et al.

Dow

nloa

ded

by g

uest

on

Apr

il 6,

202

0

Page 9: Multiplex, quantitative cellular analysis in large tissue ...Multiplex, quantitative cellular analysis in large tissue volumes with clearing-enhanced 3D microscopy (C e3D) Weizhe Lia,

described nonhomogeneous distribution, with more predominantlocalization of memory T cells in the interfollicular zones, andwith dispersed but sometimes clustered regulatory T-cell posi-tioning in peripheral regions (Fig. 5 C and D and SI Appendix,Fig. S8).One limitation of conventional confocal objectives for obtaining

high-quality image datasets is the substantially shorter workingdistances, which restrict extensive volumetric microscopy. Recentdevelopments of new objectives designed for cleared tissues haveyielded optics with both high N.A. and large working distances. Aclose correspondence between the refractive index of Ce3D me-dium and the optimal immersion medium of the 25× 1.0 N.A.motCORR objective allowed us to examine the ability of Ce3Dcleared organs to generate high-quality datasets across muchlarger tissue volumes. Indeed, using this objective to image Ce3Dcleared mouse lung tissues, stained with an anti-cytokeratin anti-body, we effectively visualized lung epithelial cells with clear-cutdefinition of cellular membranes to a depth of 3 mm (SI Appendix,Fig. S9). These data indicate that use of such specialized objectiveswith Ce3D cleared tissues will permit much larger volumetricquantitative microscopy of tissue samples from diverse species andorgans. Collectively, these results indicate that the Ce3D imagingand histo-cytometry pipeline allows robust quantitative analysis ofcellular states and distribution directly in 3D organs, includingtissues with exceptionally high cellular densities and phenotypiccomplexities (SI Appendix, Fig. S10).

DiscussionValuable imaging of large tissues, such as entire mouse brains, hasbeen achieved using available clearing methods (12). However,these analyses typically are conducted with low power, low N.A.objectives, thus producing images of modest resolution unsuited tofine-grained, highly-multiplex dissection of densely packed cells,such as the diverse cell populations of the innate and adaptiveimmune system (13, 28). Here, we describe a tissue clearingtechnique, Ce3D, that results in rapid and robust tissue trans-parency for most organs and is fully compatible with a wide as-sortment of reporter proteins and antibody-associated fluorophores,allowing multiplexed imaging of diverse analytes of interest in large(>2–3 mm thick) tissue volumes at high resolution. Furthermore,we provide an enhanced pipeline for cell segmentation andquantitative analysis, Ce3D histo-cytometry, which supports de-tailed exploration of cellular phenotypic and functional states insuch 3D datasets (SI Appendix, Fig. S10). Collectively, these toolsallow comprehensive dissection of the structure–function rela-tionships involving diverse cell types with their surrounding mi-croenvironments, effectively bridging the gap between imagingand quantitative cytometry.Ce3D is conceptually based on several recent tissue clearing

technologies, which have reignited interest in volumetric organimaging, but have also been associated with certain limitations(12) (SI Appendix, Table S1). For example, solvent-based clearingquenches reporter protein fluorescence and leads to tissueshrinkage (1, 2, 9). On the other hand, urea-based hyperhydrationcauses tissue swelling and disruption of antibody–antigen inter-actions (4, 5). The newest generation urea-based method mini-mizes the latter issues, but necessitates use of a complex series ofextended incubation steps, while still resulting in suboptimal epi-tope labeling and quenched protein fluorescence (3). In contrast,passive clearing techniques use incubations in simple high re-fractive index media, but do not result in optimal tissue trans-parency (10, 11). Hydrogel-embedding methods, which removetissue lipids, can result in extensive protein cross-linking and poorepitope-based immunolabeling (6, 7). A recent glutaraldehyde-based method successfully demonstrated volumetric multiplexedimaging, but uses suboptimal fixation reagents and extreme pHtreatments that can compromise antibody-based labeling and in-crease tissue autofluorescence, while also requiring multiple time-

intensive incubation steps and complex 3D image registration al-gorithms (8). The Ce3D technology reported here is largely devoidof these limitations, inducing rapid optical transparency in mosttissues using simple and inexpensive reagents and permitting high-quality, multiplexed, large volumetric microscopy of cells withvaried morphology and phenotypic complexity in structurally in-tricate organs. While we did observe some decrease in tissuevolume after Ce3D treatment of certain organs, most notably thebrain, this shrinkage was not associated with apparent loss ofmorphological detail, and actually promoted discrimination of thefine cellular processes. Importantly, the obtained image qualitywas similar to gold-standard section-based imaging, with the majorbenefit of allowing volumetric tissue reconstruction or examina-tion of samples in any preferred plane of interest. The latter isimportant as it reduces the undersampling errors associated withconventional thin sections taken at arbitrary angles through sam-ples with asymmetric cell distributions, for example, a tumor withan immune infiltrate.The compatibility of Ce3D with immunolabeling and a wide

array of fluorophores provides a high degree of flexibility in re-agent choice, and more importantly permits routine probe mul-tiplexing using traditional confocal instruments equipped withspectral detectors. Considering the necessity for robust quanti-tation of such multidimensional datasets, we also developed anenhanced technique for 3D image segmentation and quantitativecellular analysis. The described Ce3D histo-cytometry pipeline isbased on the previously described methodology for section-basedimaging, with enhancements for segmentation of all imaged cellsin large 3D datasets, irrespective of nuclear staining (13). It isbased on commercial and open source software platforms andthus can be readily applied in diverse settings. One caveat is thateven with this improved algorithm, some errors in enumerationcan occur with cells of complex shape; we estimate the error tobe on the order of 10–15% for cell types like myeloid dendriticcells, but this error is substantially smaller than that accompa-nying incomplete tissue extraction and enumeration by flowcytometry (13, 22).Several roadblocks currently hinder large-scale adoption of

volumetric quantitative imaging. The high molecular weights ofantibodies and certain fluorophores necessitate long incubationtimes and often result in inhomogeneous tissue staining. Use ofFab antibody fragments, aptamers, or camelid-derived single chainantibodies conjugated to small organic fluorophores should im-prove tissue penetration and staining homogeneity (12). Pulsedvibrational energy with microwave technology may also promotereagent penetration into tissues (29). Furthermore, althoughconventional confocal microscopes allow high-resolution imagingof tissues, point-by-point raster scanning with high magnificationoptics necessitates significant time for acquisition of large volu-metric datasets. For example, imaging of relatively small ∼1-mm3

LNs with a 20× objective, with multiple tiled stacks and severalsequential scans for detection of multiplexed fluorescent probes,necessitates hours of microscope time per tissue sample. Currentdevelopments in light-sheet microscopy are likely to resolve thislimitation in the near future, although a combination of high-speed and high-resolution imaging across such large tissue vol-umes is yet to be demonstrated with this technology (12, 30) (Fig.2E). Further, the use of conventional high N.A. objectives is as-sociated with significantly shorter working distances, thereby lim-iting the overall accessible image volume. The 20× objective usedfor the majority of imaging in this study allowed an effective im-aging depth of up to 650 μm, but was suboptimal for accuratesignal–cell allocation in the axial plane for closely adjoining,morphologically complex cells. Although these issues were re-solved with the 40× 1.3 N.A. objective, its working distance waslimited to less than 250 μm. Of note, even such a relatively limiteddepth provides an effective 10- to 30-fold increase in the amountof acquired information over traditional thin-section microscopy.

Li et al. PNAS | Published online August 14, 2017 | E7329

IMMUNOLO

GYAND

INFLAMMATION

PNASPL

US

Dow

nloa

ded

by g

uest

on

Apr

il 6,

202

0

Page 10: Multiplex, quantitative cellular analysis in large tissue ...Multiplex, quantitative cellular analysis in large tissue volumes with clearing-enhanced 3D microscopy (C e3D) Weizhe Lia,

Importantly, recently developed microscope objectives designedfor cleared tissues attain both high N.A. and large working dis-tances (>5 mm), although such optics are currently limited tospecific immersion media. We were able to examine the perfor-mance of one of these objectives with Ce3D-cleared tissues andobtained images with high signal-to-noise characteristics at 3-mmtissue depths, suggesting that such optics will aid in large volume,high-resolution imaging.In addition to imaging constraints, computational requirements for

quantitative analysis of large volumetric datasets are currently de-manding, necessitating use of dedicated workstations with largeamounts of RAM, as well as of specialized software tools, which whencommercial, are quite expensive. Although successful cellular seg-mentation was achieved with the Ce3D histo-cytometry pipeline,limitations in parallel processing in the segmentation software avail-able to us necessitated processing of very large image datasets asseparate fragments. Such limitations are likely to be temporary, assubstantial interest in large 4D datasets obtained with light-sheetmicroscopy has promoted development of novel open-source solu-tions for data handling and analysis of extremely large datasets (31,32). Finally, commercially available fluorophores with distinctly sep-arable excitation or emission spectra currently limit the number ofprobes to a maximum of 8–15. Although certain thin-section imagingapproaches entailing repeated antibody labeling have demonstratedmore extensive multiplexing, these will likely not be directly applicablewithout modification to large tissue volumes due to temporal re-quirements for reagent penetration (33, 34). Even with these limita-tions, powerful statistical or machine learning methods will be neededfor better understanding of the complex relationships between cellsand their surroundings as revealed by these imaging methods.

MethodsMice. CD11c-YFP [B6.Cg-Tg(Itgax-Venus)1Mnz/J], actin-DsRed [B6.Cg-Tg(CAG-DsRed*MST)1Nagy], Cxcl12-DsRed (Cxcl12tm2.1Sjm/J), Cx3cr1-GFP (B6.129P-

Cx3cr1tm1Litt/J), and histone GFP-tagged reporters [B6.Cg-Tg(HIST1H2BB/EGFP)1Pa] were obtained from The Jackson Laboratory. Fluorescent Confettianimals were generated by crossing B6.129P2-Gt(ROSA)26Sortm1(CAG-Brain-bow2.1)Cle/J × B6.Cg-Tg(UBC-cre/ERT2)1Ejb/2J (The Jackson Laboratory). Miceheterozygous for both transgenes were injected i.p. with tamoxifen 100 μg pergram of body weight in peanut oil (Sigma-Aldrich) for 5 consecutive days, withtissues collected on the fifth day for processing. Two- to 6-mo-old male/femalemice were used for all experiments and were randomly allocated into treat-ment groups. The investigators were not blinded to allocation during experi-ments and outcome assessment. All mice were maintained in specificpathogen-free conditions at an Association for Assessment and Accredita-tion of Laboratory Animal Care-accredited animal facility at the National In-stitute of Allergy and Infectious Diseases (NIAID). All procedures wereapproved by the NIAID Animal Care and Use Committee (NIH).

Microscopy and Ce3D Pipeline. Detailed methods for Ce3D-tissue clearing,imaging, segmentation and quantitation, as well as other non-Ce3D meth-ods examined in this report are provided in SI Appendix, SI Methods.

Statistical Analysis. Statistical tests were selected based on appropriate as-sumptions with respect to data distribution and variance characteristics. Nostatistical methods were used to predetermine sample size. The statisticalsignificance of differences in mean values was analyzed by a two-tailedStudent’s t test. Paired t tests were conducted for comparison of the sametissue samples before and after Ce3D treatment. ***P < 0.0001, **P < 0.005,and *P < 0.05.

ACKNOWLEDGMENTS. We thank Dr. Andrea Radtke for providing tissuesfrom Confetti reporter animals; Dr. Kairui Mao for helping with gut tissuepreparation; all members of the Lymphocyte Biology Section, Laboratory ofSystems Biology, National Institute of Allergy and Infectious Diseases(NIAID), NIH for many helpful comments during the course of these exper-iments; and Drs. Kerry Casey, Andrea Radtke, Kerry Mao, Antonio PiresDa Silva Baptista, and Stefan Uderhardt for critical review of the manu-script. This research was supported by the Intramural Research Program,NIAID, NIH.

1. Ertürk A, et al. (2012) Three-dimensional imaging of solvent-cleared organs using3DISCO. Nat Protoc 7:1983–1995.

2. Renier N, et al. (2014) iDISCO: A simple, rapid method to immunolabel large tissuesamples for volume imaging. Cell 159:896–910.

3. Hama H, et al. (2015) ScaleS: An optical clearing palette for biological imaging. NatNeurosci 18:1518–1529.

4. Hama H, et al. (2011) Scale: A chemical approach for fluorescence imaging and re-construction of transparent mouse brain. Nat Neurosci 14:1481–1488.

5. Tainaka K, et al. (2014) Whole-body imaging with single-cell resolution by tissuedecolorization. Cell 159:911–924.

6. Chung K, et al. (2013) Structural and molecular interrogation of intact biologicalsystems. Nature 497:332–337.

7. Yang B, et al. (2014) Single-cell phenotyping within transparent intact tissue throughwhole-body clearing. Cell 158:945–958.

8. Murray E, et al. (2015) Simple, scalable proteomic imaging for high-dimensionalprofiling of intact systems. Cell 163:1500–1514.

9. Dodt HU, et al. (2007) Ultramicroscopy: Three-dimensional visualization of neuronalnetworks in the whole mouse brain. Nat Methods 4:331–336.

10. Kuwajima T, et al. (2013) ClearT: A detergent- and solvent-free clearing method forneuronal and non-neuronal tissue. Development 140:1364–1368.

11. Ke MT, Fujimoto S, Imai T (2013) SeeDB: A simple and morphology-preserving opticalclearing agent for neuronal circuit reconstruction. Nat Neurosci 16:1154–1161.

12. Richardson DS, Lichtman JW (2015) Clarifying tissue clearing. Cell 162:246–257.13. Gerner MY, Kastenmuller W, Ifrim I, Kabat J, Germain RN (2012) Histo-cytometry: A

method for highly multiplex quantitative tissue imaging analysis applied to dendriticcell subset microanatomy in lymph nodes. Immunity 37:364–376.

14. Gerner MY, Torabi-Parizi P, Germain RN (2015) Strategically localized dendritic cellspromote rapid T cell responses to lymph-borne particulate antigens. Immunity 42:172–185.

15. Fonseca DM, et al. (2015) Microbiota-dependent sequelae of acute infection com-promise tissue-specific immunity. Cell 163:354–366.

16. Liu Z, et al. (2015) Immune homeostasis enforced by co-localized effector and regu-latory T cells. Nature 528:225–230.

17. Radtke AJ, et al. (2015) Lymph-node resident CD8α+ dendritic cells capture antigensfrom migratory malaria sporozoites and induce CD8+ T cell responses. PLoS Pathog11:e1004637.

18. Petrovas C, et al. (2012) CD4 T follicular helper cell dynamics during SIV infection.J Clin Invest 122:3281–3294.

19. Torabi-Parizi P, et al. (2014) Pathogen-related differences in the abundance of pre-

sented antigen are reflected in CD4+ T cell dynamic behavior and effector function in

the lung. J Immunol 192:1651–1660.20. Lee YJ, et al. (2015) Tissue-specific distribution of iNKT cells impacts their cytokine

response. Immunity 43:566–578.21. Tsujikawa T, et al. (2017) Quantitative multiplex immunohistochemistry reveals

myeloid-inflamed tumor-immune complexity associated with poor prognosis. Cell Rep

19:203–217.22. Steinert EM, et al. (2015) Quantifying memory CD8 T cells reveals regionalization of

immunosurveillance. Cell 161:737–749.23. Qi H, Kastenmüller W, Germain RN (2014) Spatiotemporal basis of innate and

adaptive immunity in secondary lymphoid tissue. Annu Rev Cell Dev Biol 30:141–167.24. Mueller SN, Germain RN (2009) Stromal cell contributions to the homeostasis and

functionality of the immune system. Nat Rev Immunol 9:618–629.25. Gray EE, Cyster JG (2012) Lymph node macrophages. J Innate Immun 4:424–436.26. Kastenmüller W, et al. (2013) Peripheral prepositioning and local CXCL9 chemokine-

mediated guidance orchestrate rapid memory CD8+ T cell responses in the lymph

node. Immunity 38:502–513.27. Sung JH, et al. (2012) Chemokine guidance of central memory T cells is critical for

antiviral recall responses in lymph nodes. Cell 150:1249–1263.28. Woodruff MC, Herndon CN, Heesters BA, Carroll MC (2013) Contextual analysis of

immunological response through whole-organ fluorescent imaging. Lymphat Res Biol

11:121–127.29. Ferris AM, Giberson RT, Sanders MA, Day JR (2009) Advanced laboratory techniques

for sample processing and immunolabeling using microwave radiation. J Neurosci

Methods 182:157–164.30. Mertz J (2011) Optical sectioning microscopy with planar or structured illumination.

Nat Methods 8:811–819.31. Amat F, et al. (2015) Efficient processing and analysis of large-scale light-sheet mi-

croscopy data. Nat Protoc 10:1679–1696.32. Stegmaier J, et al. (2016) Real-time three-dimensional cell segmentation in large-scale

microscopy data of developing embryos. Dev Cell 36:225–240.33. Gerdes MJ, et al. (2013) Highly multiplexed single-cell analysis of formalin-fixed,

paraffin-embedded cancer tissue. Proc Natl Acad Sci USA 110:11982–11987.34. Schubert W, et al. (2006) Analyzing proteome topology and function by automated

multidimensional fluorescence microscopy. Nat Biotechnol 24:1270–1278.

E7330 | www.pnas.org/cgi/doi/10.1073/pnas.1708981114 Li et al.

Dow

nloa

ded

by g

uest

on

Apr

il 6,

202

0