Nanoscale Imaging of Proteins and Nucleic Acids via Expansion Microscopy

Information

  • Patent Application
  • 20170067096
  • Publication Number
    20170067096
  • Date Filed
    August 05, 2016
    7 years ago
  • Date Published
    March 09, 2017
    7 years ago
Abstract
The invention enables in situ genomic and transcriptomic assessment of nucleic acids to be conducted in biological specimens that have been physically expanded. The invention leverages the techniques for expansion microscopy (ExM) to provide new methods for in situ genomic and transcriptomic assessment of nucleic in a new process referred to herein as “expansion fluorescent in situ hybridization” (ExFISH).
Description
BACKGROUND OF THE INVENTION

Nanoscale-resolution imaging of RNA throughout cells, tissues, and organs is key for an understanding of local RNA processing, mapping structural roles of RNA, and defining cell types and states. However, it has remained difficult to image RNA in intact tissues with the nanoscale precision required to pinpoint associations with cellular compartments or proteins important for RNA function.


Expansion microscopy (ExM) enables imaging of thick preserved specimens with ˜70 nm lateral resolution. Using ExM the optical diffraction limit is circumvented by physically expanding a biological specimen before imaging, thus bringing sub-diffraction limited structures into the size range viewable by a conventional diffraction-limited microscope. ExM can image biological specimens at the voxel rates of a diffraction limited microscope, but with the voxel sizes of a super-resolution microscope. Expanded samples are transparent, and index-matched to water, as the expanded material is >99% water. The original ExM protocol worked by labeling biomolecules of interest with a gel-anchorable fluorophore. Then, a swellable polyelectrolyte gel was synthesized in the sample, so that it incorporated the labels. Finally, the sample was treated with a nonspecific protease to homogenize its mechanical properties, followed by dialysis in water to mediate uniform physical expansion of the polymer-specimen composite. All of the chemicals required for ExM can be purchased except for the gel-anchorable label, which requires custom synthesis and raises the barrier for researchers to adopt the method. Another drawback of the ExM protocol is that genetically encoded fluorophores cannot be imaged without antibody labeling. Additionally, ExM was unable to retain native proteins in the gel and used custom made reagents not widely available. Thus, it would be desirable to leverage ExM to devise new methods for in situ retention and imaging of nucleic acids and proteins within a sample.


SUMMARY OF THE INVENTION

A small molecule linker is synthesized that enables RNA to be covalently attached to the ExM gel. This method, referred to as ExFISH, enables RNA fluorescent in situ hybridization (FISH), which enables identification of transcripts in situ with single molecule precision. In RNA FISH, a set of fluorescent probes complementary to a target strand of mRNA are delivered2,3. Single molecule FISH (smFISH) can be performed with multiple fluorophores delivered to a single mRNA via oligonucleotide probes4. In intact tissues, amplification strategies, such as hybridization chain reaction (HCR)5,6, and branched DNA amplification7,8, can enable a large number of fluorophores to be targeted to a single mRNA. ExFISH can support smFISH in cell culture, and HCR-amplified FISH in intact mouse brain tissues. ExFISH can reveal nanoscale structures of long non-coding RNAs (lncRNAs), as well as for localizing neural mRNAs to individual dendritic spines. ExFISH will be useful for a diversity of questions relating the structure and location of RNA to biological functions.





BRIEF DESCRIPTION OF THE DRAWINGS

The foregoing and other objects, features and advantages of the invention will be apparent from the following more particular description of preferred embodiments of the invention, as illustrated in the accompanying drawings in which like reference characters refer to the same parts throughout the different views. The drawings are not necessarily to scale, emphasis instead being placed upon illustrating the principles of the invention.


The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawings will be provided to the Office upon request and payment of the necessary fee.



FIG. 1A-1I: Design and validation of ExFISH chemistry. (1A) Acryloyl-X SE (top left) is reacted to LABEL-IT® amine (top right) via NETS-ester chemistry to form LabelX (middle), which serves to make RNA gel-anchorable by alkylating its bases (e.g., the N7 position of guanines) (bottom). (1B) Workflow for ExFISH: biological specimens are treated with LabelX (left), which enables RNA to be anchored to the ExM gel (middle). Anchored RNA can be probed via hybridization (right), after gelation, digestion, and expansion. (1C) smFISH image of ACTB before expansion. Inset shows zoomed-in region, highlighting transcription sites in nucleus. (1D) As in (1C), using ExFISH. (1E) smFISH counts before versus after expansion for seven different transcripts (n=59 cells; each symbol represents one cell). (1F) smFISH image of XIST long non-coding RNA (1ncRNA) in the nucleus of a HEK293 cell before expansion (white line denotes nuclear envelope in 1F-1H). (1G) As in (1F), using ExFISH. (1H) smFISH image before expansion (top), and using ExFISH (bottom), of NEAT1 lncRNA in the nucleus of a HeLa cell. Magenta and green indicate probesets binding to different parts of the 5′ (1-3756 nts) of NEAT1 (see Methods). (1I) Insets showing a NEAT1 cluster (boxed region of (1H)) with smFISH (left) and ExFISH (right). Scale bars (white, in pre-expansion units; blue scale bars are divided by the expansion factor noted))): (1C, 1D) 10 μm (expansion factor, 3.3×), inset 2 μm; (1F, 1G) 2 μm (3.3×), Z scale represented by color coding in pre-expansion units; (1H) 2 μm (3.3×); (1I) 200 nm (3.3×).



FIG. 2A-2E: Serially hybridized and multiplexed ExFISH. (2A) Widefield fluorescence image of ExFISH targeting GAPDH. (2B) Boxed region of (2A), showing 5 repeated re-stainings following probe removal (see Methods); lower right panel, overlay of the 5 images (with each a different color, red, green, blue, magenta, yellow), showing co-localization. (2C) ExFISH RNA counts for each round, normalized to the round 1 count; plotted is mean±standard error; n=3 regions of (2A). (2D) Signal-to-noise ratio (SNR) of ExFISH across the five rounds of staining of (2A), computed as the mean puncta brightness divided by the standard deviation of the background. (2E) Composite image showing ExFISH with serially delivered probes against six RNA targets in a cultured HeLa cell (raw images in FIG. 9); colors are as follows: NEAT1, blue; EEF2, orange; GAPDH, yellow; ACTB, purple; UBC, green; USF2, light blue. Scale bars (expanded coordinates): (2A) 20 μm; (2B) 10 μm; (2E) 20 μm.



FIG. 3A-3K: Nanoscale imaging of RNA in mammalian brain. (3A) Widefield fluorescence image of Thyl-YFP mouse brain. (3B) Post-expansion widefield image of (3A). (3C) Widefield fluorescence showing HCR-ExFISH of YFP mRNA in the sample of (3B). (3D) As in (3C), but for Gadl mRNA. (3E) Composite of (3B-3D), highlighting distribution of Gad1 versus Thy1-YFP mRNAs. (3F) Confocal image of mouse hippocampal tissue from (e) showing single RNA puncta. Inset, one plane of the boxed region (red, YFP protein; cyan, YFP mRNA; magenta, Gad1 mRNA). (3G(i)) Confocal image and (3G(ii)) processed image of HCR-ExFISH using a missense D1g4 probe, in Thyl-YFP mouse tissue (green, YFP protein). The raw image (3G(i)) uses alternating probes in two colors (red, D1g4 missense even; blue, D1g4 missense odd). The processed image (3G(ii)) shows zero co-localized spots (magenta). (3H, 3I) As in (3G(i) and 3G(ii)), but for HCR-ExFISH targeting Actb in Thy1-YFP mouse brain (green, YFP protein; red, Actb even, and blue, Actb odd in (3H(i)); co-localized spots in magenta (3H(ii))). (3I) Confocal image of hippocampal tissue showing co-localized D1g4 puncta (magenta) overlaid on YFP (green). (3J(i), 3J(ii)) Two representative examples of dendrites with D1g4 mRNA localized to spines (arrows). (3K(i), 3K(ii)) As in (3J), but with HCR-ExFISH of Camk2a mRNA showing transcripts in dendritic spines and processes. Scale bars (white, in pre-expansion units; blue scale bars are divided by the expansion factor noted): (3A) 500 μm; (3B-3E) 500 μm (expansion factor 2.9×); (3F) 50 μm (2.9×), inset 10 μm; (3G-3I) 10 μm (3×); (3J, 3K) 2 μm (3×). (3E, 3I) maximum-intensity projection (MIP) 27 μm thick (pre-expanded units); (3G, 3H, 3J, 3K) MIPs ˜1.6 μm thick.



FIG. 4A-4B: (4A) Epi-fluorescence image of single molecule FISH (smFISH) against GAPDH on HeLa cells expanded without LabelX treatment. (4B) Epi-fluorescence image of smFISH performed against GAPDH on expanded HeLa cells treated with LabelX. Images are maximum intensity projections of 3-D stacks. Nuclei stained with DAPI (shown in blue). Scale bars: 20 μm (post-expanded units).



FIG. 5A-5E: To assess the effect of LabelX on fluorescent in situ hybridization, fixed HeLa cells were stained with smFISH probe-sets, followed by DNAse I treatment to remove the staining. The cells were then treated with LabelX and stained again with the same smFISH probe-sets. (5A) UBC staining before LabelX treatment and (5B) UBC staining after probe removal and LabelX treatment. (5C) EEF2 staining before LabelX treatment. (5D) EEF2 staining after probe removal and LabelX treatment. (5E) Comparison of smFISH spots counted for individual cells before LabelX, and after probe removal and application of LabelX. The number of RNA molecules detected in a given cell was quantified using an automated spot counting algorithm (n=7 cells for each bar). Plotted are mean±standard error; no significant difference in spot counts before vs after LabelX (p>0.5 for before vs. after for UBC, p>0.5 for before vs. after for EEF2; t-test, unpaired, two-tailed). Images in 5A-5D are maximum intensity projections of 3-D stacks; scale bars: 10 μm (pre-expanded units).



FIG. 6A-6G: Different RNA species spanning 3 orders of magnitude in abundance were detected via single molecule RNA fluorescent in situ hybridization (FISH) in HeLa cells before and after ExM with LabelX treatment (shown in FIG. 1E). (1A) Ratio of FISH spots detected after expansion to spots detected before expansion for single cells. Representative before vs. after ExFISH images shown: (1B,1C) TFRC; (1D,1E) GAPDH; (1F, 1G) ACTB. Scale bars, 10 μm (pre-expanded units) in 1B, 1D, 1F; 1C, 1E, 1G, expanded physical size 21 μm (imaged in PBS).



FIG. 7A-7E: (7A) Pre-expansion widefield image of a cultured HeLa cell stained with DAPI to visualize the nucleus (top panel) and smFISH probes against ACTB (bottom panel). (7B) Post-expansion widefield image of the same cell as in (a). (7C) Pre-expansion widefield image of LabelX treated Thyl-YFP brain slice (left panel, YFP protein) stained with DAPI (right panel) (MIP, 4 μm z-depth). (7D) Post-expansion image of the same region as in (c) (MIP, 12 μm). (7E) Ratio of the expansion factor of cell bodies for individual cells to the expansion factor of their respective nuclei. smFISH stain is used to outline the boundaries of the cell bodies of cultured cells while the endogenous YFP protein is used to demarcate the cell bodies of neurons in Thy1-YFP brain slices. Plotted are mean±standard error. The ratio for both cultured cells and brain slices did not significantly deviate from one (p>0.05 for both, 1-sample t-test; n=6, cultured HeLa cells; n=7, cells in 1 brain slice). Scale bars, 10 μm.



FIG. 8A-8D: (8A) Representative FISH image of TOP2A in a single HeLa cell before expansion (MIP of cell thickness). (8B) ExFISH image of cell in (8A) taken with the same optical parameters. (8C) Merged image of (8A) and (8B) (red and green for before and after expansion respectively); distance measurements between pairs of mRNA spots before (L, red line) and after (L′, green line; note that these lines overlap nearly completely) expansion were used to quantify expansion isotropy. (8D) Mean of the absolute value of the measurement error (i.e., |L-L′|) plotted against measurement length (L) for all pairs of mRNA spots (mean±standard deviation, N=4 samples, 6.8×105 measurements). Scale bars: white, 10 μm pre-expansion units; blue, white scale bar divided by expansion factor. Orange line indicates diffraction limit of the microscope used (see methods for details).



FIG. 9A-9B: (9A) Five consecutive widefield fluorescence images (top to bottom, then left to right) of GAPDH, applied to the cell of FIG. 2a. (9B) Widefield fluorescence images showing ExFISH with serially delivered probes against six RNA targets (right to left, then top to bottom: NEAT1, EEF2, ACTB, UBC, GAPDH, and USF2) in a cultured HeLa cell (raw images of composite shown in FIG. 2E). Scale bars: 20 μm in expanded units.



FIG. 10: FISH probes bearing HCR initiators are hybridized to a target mRNA. During amplification, metastable DNA hairpins bearing fluorophores assemble into polymer chains onto the initiators, thus amplifying signal downstream of the FISH probe hybridization event.



FIG. 11A-11C: (11A) Widefield image of a LabelX treated Thy1-YFP brain slice (YFP protein, green) stained with probes against YFP (red) and Gad1 (magenta) followed by HCR amplification. Probes against YFP transcripts were amplified with the B1 amplifier set (see Methods) while probes against Gad1 transcripts were amplified with the B2 amplifier set (MIP, 59 μm). (11B) Widefield image of LabelX treated Thy1-YFP brain slice (YFP protein, green) treated with the same HCR amplifiers as in (a) (namely B1 (red) and B2 (magenta)) without the addition of probes (MIP, 50 μm). (11C) HCR spots detected per volume of expanded sample. Analysis was performed on samples which were either treated or not treated with FISH probes followed by HCR amplification. An automated spot counting algorithm was used to count HCR spots. The endogenous YFP protein was used to delineate regions used for the analysis. Plotted are mean±standard error. HCR spot counts are significantly different in the presence of probes than without probes (p<0.05 for both B1 and B2 amplifier sets, Welch's t-test; n=4 fields of view each). Scale bars: 50 μm.



FIG. 12A-12C: (12A) Volume rendering of Thy1-YFP (green) brain tissue acquired by lightsheet microscopy with HCR-ExFISH targeting YFP (red) and Gad1 (blue) mRNA. (12B) A maximum intensity projection (˜8 μm in Z) of a small subsection of the volume, showing the high resolution of imaging and single molecule localization of imaging expanded specimens with lightsheet imaging (scale bar: 10 μm, in pre-expansion units, expansion factor, 3×). (12C) Zoom in of the volume rendering in (12A) (scale bar: 20 μm, in pre-expansion units, 3×).



FIG. 13A-13G: (13A) Schematic showing two color amplification of the same target. A transcript of interest is targeted by probes against alternating parts of the sequence, and bearing two different HCR initiators, allowing for amplification in two colors. (13B) Confocal image showing FISH staining with HCR amplification against the Camk2a transcript in two colors (red and blue; YFP fluorescence shown in green). (13C) The result of an automated two-color spot co-localization analysis performed on the data set shown in (13B). Each purple spot represents a positive co-localization identified by the algorithm and overlaid on the confocal image of YFP. (13D, 13E) Zoom in of dendrites showing two color FISH staining with HCR amplification against Camk2a transcripts. (13F, 13G) As in (13D, 13E) but against D1g4 transcripts. Top row shows the raw two color staining data corresponding to the bottom row showing co-localized spots identified by the automated algorithm. Scale bars: (13B, 13C) 10 μm (3×); (13D-13G) 2 μm (3×). (13B-13G) are MIP of ˜1.6 μm thickness in unexpanded coordinates.



FIG. 14A-14B: (14A) Schematic for HCR amplification and reversal. HCR amplification is initiated with custom-made HCR hairpins bearing toe-holds for toe-hold mediated strand displacement. After amplification, the addition of a disassembling strand initiates the disassembly of the HCR polymers via strand displacement. (14B) ExFISH-treated Thy1-YFP brain slice (YFP in blue) is shown stained with YFP FISH probes bearing HCR initiators and amplified with custom made HCR hairpins bearing toe-holds for strand displacement (green dots). The different panels show the state of HCR reversal at different times after the addition of strands to initiate the disassembly of the HCR polymers. Scale bars: 20 μm (in post-expansion units).



FIG. 15: Dependence of RNA FISH spot intensity on degree of expansion and concentration of LabelX. HeLa cells, treated with LabelX diluted to different final concentrations of Label-It Amine concentration, were expanded and stained with a probe-set against GAPDH. After staining, the gelled samples were expanded in 1× PBS (˜2× expansion ratio) and water (˜4× expansion ratio) and the spot intensity for the different samples was quantified. Plotted are mean±standard error; N=6 cells.





DETAILED DESCRIPTION

The present invention provides for the anchoring of nucleic acids into the swellable gel of Expansion Microscopy (ExM), both for in site genomic and transcriptomic assessment, as well as to enable nucleic acid barcodes to be used to identify essentially arbitrary numbers of molecules. International patent application serial number PCT/US15/16788, which is incorporated herein by reference, teaches that the resolution of conventional microscopy can be increased by physically expanding specimens, a process termed ‘expansion microscopy’ (ExM). In short, biological specimens are embedded in a swellable gel material, subjected to a treatment to disrupt native biological networks, and then expanded. The advantages to ExM include tissue clearing, resolution improvement, and higher tolerance to sectioning error due to the specimen expansion in the z-axis.


In ExM, fluorophores were anchored directly to the polymer gel, so that proteins could be visualized; however, RNA molecules were not preserved in the gel and are instead lost during the expansion process. Thus, there was no way to probe the transcriptomic information of the sample.


In one embodiment, the invention provides methods that covalently anchor native nucleic acid molecules and antibody barcodes to the expandable gel matrix of expansion microscopy (ExM). Nucleic acids are modified using a small molecule tag, which lets them participate in free radical polymerization during gelling. During the gel formation step, any biomolecules bearing reactive groups are anchored into the gel and isotropically separated as the gel expands.


In one embodiment, the invention provides a nucleic acid reactive reagent that also carries a chemical group that can get incorporated into the gel. After treatment of samples with this reagent, nucleic adds, including DNA and RNA, are covalently labeled with this reagent. Afterwards, during gel formation, labeled nucleic acids are covalently incorporated into the gel. Using such anchored nucleic acids, the information in the nucleic acid can be used as a barcode, e.g. barcoded antibodies can be used for multiplexed in situ staining for ExM, enabling “arbitrary-color” imaging.


By covalently anchoring the nucleic acids, existing technologies for reading out RNA and DNA can be applied to the expanded context. These strategies include single molecule FISH (Imaging individual mRNA molecules using multiple singly labeled probes. Nature Methods, 2008 Oct; 5(10):877-9), oligo-paint (“Versatile design and synthesis platform for visualizing genomes with Oligopaint FISH probes.” PNAS 109.52 (2012): 21301-21306) and many other hybridization based readout strategies. Furthermore, the covalent anchoring allows for sequential hybridization, leading to various multiplexing strategies including serial, spectral, and temporal barcoding schemes. The present invention provides methods for labeling and staining with DNA-barcoded primary antibodies, allowing for an arbitrary number of protein tags to be utilized with ExM, This is a key step towards “infinite color” imaging, since previous the expansion microscopy method only enabled 3-color imaging.


In a further embodiment, the invention provides a method for performing sequential hybridizations against nucleic acids covalently incorporated into an ExM gel. Firstly, buffer condition for hybridizing complementary oligonucleotides bearing fluorophores to the nucleic acids in the ExM gel are provided. Second, the ExM gel is re-embedded in a polyacrylamide gel to minimize distortions resulting from changes in buffer. Third, chemical and enzymatic strategies for removing oligonucleotides hybridized to nucleic acids which are covalently anchored to the gel have been developed, which enables re-staining with the same or different oligonucleotides. Chemical strategies include using formamide and high temperatures to de-hybridize oligonucleotides forming duplexes with nucleic acids in the gel. Enzymatic strategies involve using endonucleases that specifically digest the oligonuetides which are hybridized to nucleic acids while leaving the nucleic acids anchored in the gel intact.


In a further embodiment the invention provides for the multiplexed imaging of proteins and transcripts using Expansion Microscopy. First, a strategy to barcode primary antibodies with oligonucleotides by both covalently and non-covalently associating oligonucleotides with their target antibodies has been developed. While covalent attachment schemes involve reacting to amines and sugar chains found on antibodies, non-covalent attachment schemes use secondary Fab fragments conjugated to oligonucleotide barcodes. Second, a set of conditions for performing immunostaining using these oligonucleotide barcoded primary antibodies has been developed. These conditions include unique buffer compositions for minimizing non-specific binding, as well as temperature ranges for obtaining adequate immunostaining. The oligonucleotides which are reacted to these antibodies possess a chemical group that can be incorporated into the ExM gel to gel formation. Therefore, during gel formation, these oligonucleotides are all anchored into the ExM gel while all proteins are degraded. In addition, a strategy for the multiplexed read out of the oligonucleotides and nucleic acids, including RNA and DNA, in the ExM gel using sequential hybridization has been developed. This approach consists of sequentially hybridizing complementary strands bearing fluorophores to each unique oligonucleotide or nucleic acid, one by one, serially. Finally, the set of capabilities offered by out technique enable exponential barcoding schemes demonstrated recently by a few groups. For instance, this approach allows for barcoding nucleic acids via temporal color barcodes or temporal binary barcodes.


One embodiment of a method for in situ genomic and transcriptomic assessment of target nucleic acids present in a biological sample comprises the steps of:

    • a) treating the biological sample with a small molecule linker capable of linking to at least one target nucleic acid and to a swellable material;
    • b) embedding the biological sample wherein the small molecule linker is bound to the at least one target nucleic acid in the biological sample and to the swellable material;
    • c) subjecting the biological sample to a physical disruption method;
    • d) swelling the swellable material to form an expanded biological sample;
    • e) providing at least one oligonucleotide complementary to the at least one target nucleic acid, wherein the at least one oligonucleotide hybridizes to the at least one target nucleic acid; and
    • f) genomically or transcriptomically assessing the expanded biological sample.


In this and other methods, the small molecule linkers are attached to target nucleic acids via a chemical reactive group capable of covalently binding the target nucleic acid. The small molecule linker may be labeled and or the at least one oligonucleotide may be labeled.


In another embodiment, embedding the biological sample in a swellable material may comprise permeating the biological sample with a composition comprising precursors of a swellable polymer and forming a swellable polymer in situ.


In another embodiment, the at least one target nucleic acid is anchored to the swellable material.


In another embodiment, the physical disruption method is an enzymatic digestion.


In another embodiment of the just described method, the target nucleic acids are DNA and/or RNA.


In another embodiment, the expanded biological sample expresses one or more labeled target nucleic acids.


In another embodiment, the expanded sample may be buffered prior to providing at least one oligonucleotide. After buffering, the expanded sample may be re-embedded in a non-swellable material prior to genomically or transcriptically assessing the expanded biological sample. Buffering enables removal of the at least one oligonucleotide through chemical or enzymatic means. For example, formamide and high temperature could be used to chemically remove the at least one oligonucleotide while endonucleases that specifically digest the at least one oligonucleotide could accomplish the same task enzymatically. After buffering, serial or sequential genomic and transcript assessments may be performed on the same expanded sample by repeating the steps of removing the at least one oligonucleotide and providing either the same or different at least one oligonucleotide.


Methods

a. ExM-FISH and ExM FISH-HCR


Secondary antibodies were conjugated to DNA oligo barcodes bearing 5′ acrydite and 3′ amine via the Solulink commercial kit. After primary and secondary antibody staining, samples were gelled, digested, and expanded following ExM procedure. Following expansion, the gelled samples were re-embeded in a 4% polyacrylamide gel by incubating the expanded gel with acrylamide, bis-acrylamide, and radical initiators. To perform in situ hybridization, gelled samples were incubated with fluorescently labeled oligos and excess oligos were subsequently washed out. To perform in situ hybridization with Hybridization Chain Reaction (HCR) signal amplification, gelled samples were incubated with oligo probes bearing a complementary region to the antibody conjugated oligo barcodes and a site for HCR initiation. After washing out excess probes, HCR hairpins were washed in to initiate the amplification.


b. Primary-Fab Antibody Conjugation and Staining


Fab Secondary antibodies were conjugated to DNA oligo barcodes bearing 5′ acrydite and 3′ amine via the Solulink commercial kit. To conjugate IgG primary antibodies with oligo tagged Fabs. Fabs were incubated with primary antibodies along with fluorescently labeled oligonucleotides complementary to the barcodes. Subsequently, excess fabs and oligos were removed using centrifugal spin filters.


Cultured HeLa cells were fixed with 4% formaldehyde. Subsequently, staining antibody mixture was prepared by mixing appropriate purified primary-fab conjugated in a blocking buffer containing dextran sulfate, normal donkey serum, and rabbit gamma globulin. Finally, fixed cells were incubated with the antibody mixture overnight and any excess was washed off


ExFISH: Design and Validation of RNA Anchoring Chemistry

Because of the nature of the reactions occurring during ExM, covalently linking RNAs directly to the ExM gel is necessary. Although transcripts are crosslinked to proteins during fixation, the strong proteolysis of ExM precludes a reliance on proteins for RNA retention (FIGS. 4A, 4B). Thus, covalently securing RNA molecules directly to the ExM gel via a small molecule linker enables the interrogation of these molecules post-expansion. A reagent was synthesized from two building blocks: a molecule containing both an amine as well as an alkylating group that primarily reacts to the N7 of guanine, and a molecule that contains an amine-reactive succinamide ester and a polymerizable acrylamide moiety. Commercially available reagents exist that satisfy each of these two profiles, such as Label-It Amine (MirusBio) and 6-((Acryloyl)amino)hexanoic acid (Acryloyl-X SE, here abbreviated AcX, Life Technologies; all reagents are listed in Table 1). FIG. 1A depicts this molecule, which enables RNA to be covalently functionalized with a free radical polymerizable group, and which will be referred to as LabelX. As shown in FIG. 5E, LabelX does not impede smFISH readout. The original ExM protocol and the use of LabelX allows a procedure wherein a sample could be treated with LabelX to make its RNAs gel-anchorable, followed by gel formation, proteolysis, and osmotic swelling as performed in the original ExM protocol. Once a sample was thus expanded, the RNAs could then be interrogated through FISH (FIG. 1B).


To quantify RNA transcript anchoring yield after expansion, smFISH probes were used, targeting mRNAs of varying copy number (7 targets, with copy number ranging from ˜10 to ˜10,000 per cell, n=59 cells across all 7 targets). smFISH images, taken with probes delivered before (FIG. 1C) and after (FIG. 1D) expansion, to the same cells, showed no loss of transcript detectability with expansion for both low- and high-copy number transcripts (FIG. 1E). The ratio of transcripts detected was near unity at low transcript counts (e.g., in the 10's), however, more transcripts were detected after expansion for highly expressed mRNAs (e.g., in the 1,000's) (FIGS. 9A, 9B, Table 2). This difference arises from the high density of smFISH spots for these targets in the un-expanded state, with the expansion process de-crowding spots that previously were indistinguishable. For example, for smFISH against ACTB, we were able to resolve individual ACTB mRNA puncta post-expansion even within transcriptional foci in the nucleus (FIG. 1C, versus FIG. 1D), which can be dense with mRNA due to transcriptional bursting. Thus, ExFISH is capable of supporting single molecule RNA readout in the expanded state. Since Label-It also reacts to DNA, the ExFISH process enables uniform expansion of the nucleus (FIGS. 7A-C). The isotropy of ExFISH (FIG. 8) was numerically similar to that observed when protein targets were labeled and expanded in the original ExM protocol'. In recent ExM protocols in which proteins are anchored to the same hydrogel as used in ExFISH, with a similar linker“, the distortion is small (a few percent distortion, in cells and tissues). These earlier results, since they were obtained with similar polymer chemistry, serve to bound the ExFISH distortion. The expansion factor is slightly lower than in our original ExM paper (i.e., ˜3.3× versus ˜4×, expansion factors can be found in Figure Legends of this manuscript) due to the salt required to support hybridization of probes.


Nanoscale Imaging of lncRNA with ExFISH


Long non-coding RNAs (lncRNAs) known to serve structural roles in cell biology were imaged. The IncRNA XIST was imaged. Its role in inactivating the X chromosome may depend on initial association with specific chromatin subregions through a process which is still being revealed11. The pre-expansion image (FIG. 1F) shows two bright globular fluorescent regions, presumably corresponding to the X chromosomes of HEK cells undergoing inactivation11-13, but post-expansion, individual puncta were apparent both within the globular regions as well as nearby (FIG. 1G). ExFISH was used additionally to examine the previously described14 ring-shaped morphology of ensembles of NEAT1 lncRNAs (FIG. 1H), which has been hypothesized to play an important role in gene expression regulation and nuclear mRNA retention15. Before expansion, NEAT1 presents in the form of bright, diffraction-limited puncta (FIG. 1H, FIG. 1I), but after expansion, the ring-shaped morphology becomes clear (FIG. 1H, FIG. 1I). Given the complex 3-D structure of the genome16, mapping lncRNAs may be useful in defining key chromatin regulatory complexes and their spatial configurations.


Super-Resolved, Multiplexed Imaging of RNA with ExFISH


The combination of covalent RNA anchoring to the ExM gel, and the de-crowding of the local environment that results from expansion, could facilitate strategies that have been proposed for multiplexed RNA readout17-19 based upon sequential hybridization with multiple probe sets. In order to facilitate multiple cycles of FISH, we re-embedded expanded specimens in charge-neutral polyacrylamide. This process allowed expanded gels to be immobilized for multi-round imaging, and additionally stabilized the expanded specimen throughout salt concentration changes in the protocol. Such re-embedded samples exhibited similar expansion factors as non-re-embedded samples (i.e., ˜3×), and were robust to multiple wash-stain cycles as assessed by repeated application of the same probe set (FIG. 2A, FIG. 9A, showing 5 rounds of smFISH staining against GAPDH on cultured cells). This stability was observed even under stringent wash conditions designed to minimize cycle-to-cycle crosstalk (e.g., 100% formamide). Across the 5 rounds, there was no distortion of the locations of individual RNA spots from round to round (FIG. 2B), nor variance in detection efficiency or signal-to-noise ratio (FIGS. 2C, 2D). Having validated the cycle-to-cycle consistency, we next demonstrated the capability of multiplexed ExFISH by applying probes for GAPDH, UBC, NEAT1, USF2, ACTB, and EEF2 in series, enabling 6 individual RNAs to be identified and localized in the same cell (FIG. 2E, FIG. 9B). Thus, serial FISH is applicable to samples expanded after securing RNA to the swellable polymer as here described, making it straightforward to apply probe sets computationally designed to yield more information per FISH cycle, e.g. MERFISH18-20.


3D Nanoscale Imaging of RNA in Mouse Brain Tissue

ExM allows for facile super-resolution imaging of thick 3-D specimens such as brain tissue on conventional microscopy hardware1. ExFISH was applied to samples of Thy1-YFP mouse brain tissue21, using the YFP protein to delineate neural morphology (FIGS. 3A, 3B). Endogenous YFP protein was anchored to the polyacrylate gel via AcX using the proExM protocol9, and RNA anchored via LabelX. Since smFISH yields signals too dim to visualize in intact tissues using confocal imaging, the previously described technique of hybridization chain reaction (HCR)5 was applied, in particular the next-generation DNA HCR amplifier architecture6 (schematic in FIG. 10). In samples containing mouse cortical and hippocampal regions, mRNAs for YFP (FIG. 3C) and glutamic acid decarboxylase 1 Gad1 (FIG. 3D) were easily visualized using a widefield microscope, with YFP mRNA well localized to YFP-fluorescing cells (FIG. 3E), and Gad1 mRNA localized to a population of cells with characteristic arrangement throughout specific layers of the cortex and hippocampus22. Examining brain specimens at high magnification using a confocal spinning disk microscope revealed that individual transcripts could be distinguished due to the physical magnification of ExM (FIG. 3F, with YFP and Gad1 mRNA highlighted), with even highly overexpressed transcripts (e.g., YFP) cleanly resolved into individual puncta (FIG. 3F). When FISH probes were omitted, minimal background HCR amplification was observed (FIGS. 11A-C). Given that ExM enables super-resolution imaging on diffraction limited microscopes, which can be scaled to very fast imaging speeds23, we used a commercially available lightsheet microscope on a Thy1-YFP brain slice to enable visualization of multiple transcripts, with single molecule precision, throughout a volume of ˜575 μm×575 μm×160 μm thick in just 3 hours (˜6×1010 voxels in 3 colors; FIGS. 12A-C).


HCR amplifies a target binding event into a bright fluorescent signal (FIG. 10). A stringent method for assessing detection accuracy is to label individual RNAs with different probe sets bearing different colors24,25, which shows that 50-80% of mRNAs thus targeted will be doubly labeled, when assessed in cell culture; a 50% co-localization is interpreted as √{square root over (0.5)}˜70% detection efficiency (assuming probe independence); this is a lower bound as it excludes false positives. In order to assess the false positive and negative rates for single molecule visualization in expanded tissues, pairs of probe sets targeting the same transcript with different initiators were delivered. This scheme results in amplified fluorescent signals of two different colors from the same target (FIGS. 13A-B), giving a measure of the hybridization efficiency. Delivering probe sets against a nonexistent transcript also gives a measure of false positive rate. A probe set was delivered against a missense probe (D1g4 reversed, FIG. 3G(i-ii)) as well as a nonexistent transcript (mCherry, Table 3), using Thy1-YFP mouse brain samples, and found a low but nonzero spatial density of dim, yet amplified, puncta (1 per 61 μm3 in unexpanded coordinates, D1g4 reversed; 1 per 48 μm3, mCherry). Essentially zero of these puncta exhibited co-localization (0/1,209 spots, D1g4 reversed; 4/1,540 spots mCherry). In contrast, when a transcript was present (Actb), a large fraction of the puncta exhibited co-localization (an average of 58% of probes in one color co-localized with other color, 15,866/27,504 spots, FIG. 3H(i-ii), Table 3), indicative of a 75% detection efficiency, comparable to the non-amplified single molecule studies described above.


Two-color HCR ExFISH was used against mRNAs to image their position within cellular compartments such as dendritic spines, which require nanoscale resolution for accurate identification or segmentation. The D1g4 mRNA was probed, which encodes the prominent postsynaptic scaffolding protein PSD-95, and which is known to be dendritically enriched7. A degree of co-localization (53%, 5,174/9,795 spots) was obtained, suggesting a high detection efficiency, 73% (FIG. 3I). The mRNA was also probed for Camk2a, finding a detection efficiency of 78% (co-localization, 61%, 8,799/14,440 spots, FIGS. 13D-E). Puncta which were co-localized were focused on, thus suppressing false positive errors, and giving a lower-bound on transcript detection (FIG. 13). Focusing on individual dendrites in these expanded samples revealed that individual D1g4 (FIG. 3J(i-ii)) and Camk2a (FIG. 3K(i-ii)) mRNAs could indeed be detected in a sparse subset of dendritic spines as well as fine dendritic processes. To facilitate multiplexed HCR readout, we developed modified HCR hairpins that can be disassembled using toe-hold mediated strand displacement26 (FIGS. 14A-B). These modified HCR amplifiers enable multiple cycles of HCR by disassembling the HCR polymer between subsequent cycles. Given that neurons can have tens of thousands of synapses, and mRNAs can be low copy number, the ability to map mRNAs at synapses throughout neuronal arbors may be useful for a diversity of questions in neuroscience ranging from plasticity to development to degeneration.


Discussion

A novel reagent, easily synthesized from commercial precursors, that enables RNA to be covalently anchored for expansion microscopy is presented. The resulting procedure, ExFISH, enables RNAs to be probed through single-molecule FISH labeling as well as hybridization chain reaction (HCR) amplification. RNA retention before versus after expansion was validated, finding excellent yield, and de-crowding of RNAs for more accurate RNA counts and localization. This enabled visualization, with nanoscale precision and single molecule resolution, RNA structures such as XIST and NEAT1, long non-coding RNAs whose emergent structure has direct implications for their biological roles. The anchoring was robust enough to support serial smFISH, including repeated washing and probe hybridization steps, and multiplexed readout of RNA identity and location, implying that using probes designed according to specific coding strategies17-19 would support combinatorial multiplexing, in which each additional cycle yields exponentially more transcript information. The covalent anchoring of RNA to the ExM gel may also support enzymatic reactions to be performed in expanded samples—such as reverse transcription, rolling circle amplification (RCA), fluorescent in situ sequencing (FISSEQ)27, and other strategies for transcriptomic readout or SNP detection28, within intact samples.


ExM, being a physical form of magnification, enables nanoscale resolution even on conventional diffraction limited microscopes. Expanding samples makes them transparent and homogeneous in index of refraction, in part because of the volumetric dilution, and in part because of washout of non-anchored components1. Thus, strategies combining ExM with fast diffraction limited methods like lightsheet microscopy23 may result in “best of both worlds” performance metrics: the voxel sizes of classical super-resolution methods, but the voxel acquisition rates of increasingly fast diffraction limited microscopes1. The de-crowding of RNAs enables another key advantage: reducing the effective size of the self-assembled amplification product of HCR, which were applied here, following the protocols of refs.5,6, to enable nanoscale resolution visualization of RNA in intact tissues (a paper conducted in parallel has also recently performed single molecule HCR FISH29). An HCR amplicon of size 500 nm in the post-expanded sample would, because of the greater distance between RNAs, have an effective size of 500/3.5=˜150 nm. The lower packing density of amplicons facilitates the imaging of more transcripts per experiment19 with nanoscale precision. Other methods of achieving brighter signals may be possible. For example, brighter fluorophores such as quantum dots30 or bottlebrush fluorophores31 could obviate the need for signal amplification, in principle. The expanded state may enable better delivery of these and other bulky fluorophores into samples. Other amplification strategies may be possible as well, including enzymatic (e.g., RCA28, tyramide amplification22, HRP amplification) as well as nonenzymatic (e.g., branched DNA) methods, although reaction efficiency and diffusion of reagents into the sample must be considered.


ExFISH may find many uses in neuroscience and other biological fields. In the brain, for example, RNA is known to be trafficked to specific synapses as a function of local synaptic activity32 and intron content33, and locally translated7,34,35, and the presence and translation of axonal RNAs remains under investigation36. It is anticipated that, coupled to straightforward multiplexed coding schemes, this method can be used for transcriptomic profiling of neuronal cell-types in situ, as well as for the super-resolved characterization of neuronal connectivity and synaptic organization in intact brain circuits, key for an integrative understanding of the mechanisms underlying neural circuit function and dysfunction. More broadly, visualizing RNAs within cells, and their relationship with RNA processing and trafficking machinery, may reveal new insights throughout biology and medicine.


Method Information









TABLE 1







List of reagents and suppliers










Chemical


Part


Supplies
Chemical Name
Supplier
Number





ExM Gel or
Sodium Acrylate
Sigma
408220


Preparation
(purity note:*)





Acrylamide
Sigma
A9099



N,N′-
Sigma
M7279



Methylenebisacrylamide





Ammonium Persulfate
Sigma
A3678



N,N,N′,N′-
Sigma
T7024



Tetramethylethylenediamine





VA-044
Wako
27776-





21-2



4-Hydroxy-TEMPO
Sigma
176141


Hybrid-
Dextran Sulfate
Sigma
D8906-50g


ization
SSC
Thermo
AM9765


Buffer

Fisher




Formamide
Thermo
AM9342




Fisher



Fixation and
Paraformaldehyde
Electron
15710


Permeabil-

Microscopy



ization
Tissue-prep Buffered
Electron
15742-10



10% Formalin
Microscopy




Triton X-100
Sigma
93426



Ethyl Alcohol
Sigma
E7023



Glycine
Sigma
50046



10x PBS
Thermo
AM9624




Fisher



Protein
Proteinase K
New
P8107S


Digestion

England






text missing or illegible when filed





Ethylenediaminetetraacetic
Sigma
EDS



acid





Sodium Chloride
Sigma
S9888



Tris-HCl
Life
AM9855


HCR
Amplification Buffer
Molecular
N/A


Ampli-


text missing or illegible when filed




fication
Tween 20
Sigma
P1379


LabelX
LABEL-IT ® Amine
Mirus Bio
MIR 3900



text missing or illegible when filed

Acryloyl-X, SE
Thermo
A20770




Fisher



LabelX
MOPS
Sigma
M9381-25G


Treatment





Reembeded
DNAse I
Sigma
4716728001


Gels





Bind-silane
Bind-Silane
Sigma
GE17-1330-






text missing or illegible when filed






*check for yellow color upon resuspension: that indicates poor quality; solution should be clear (see http://expansionmicroscopy.org)



text missing or illegible when filed indicates data missing or illegible when filed
















TABLE 2








Mean (Ratio of #




spots detected in












Decades
individual cells after





(Transcript
ExM, to # spots
Standard
Sample
p-


Abundance)
detected before ExM)
Deviation
size (n)
Value















10
s
1.082
0.177
14
0.107


100
s
1.105
0.138
29
3.24 × 10text missing or illegible when filed


1000
s
1.596
0.562
16
7.09 × 10text missing or illegible when filed






text missing or illegible when filed indicates data missing or illegible when filed




















TABLE 3











Density



Total Spot




(Co-



Count



Volume
localized



(Averaged


Hybrid-
analyzed
Punctatext missing or illegible when filed


Tar
Across
Cotext missing or illegible when filed
Cotext missing or illegible when filed
ization
(μmtext missing or illegible when filed in
per





















ActB

27504
z
15866
0.577





0.76

236749
0.067



Dlg4

9795

5174
0.528





0.727

236749
0.022



Camk2a

14440

8799
0.609






text missing or illegible when filed indicates data missing or illegible when filed
















TABLE 4







Accession
Probe Sequence
Initiat or Type





YFP B1 1
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATctcgcccttgct
B1



caccat



YFP B1 2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATcaccaccccggt
B1



gaacag



YFP B1 3
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATtccagctcgacc
B1



aggatg



YFP B1 4
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATtgtggccgttta
B1



cgtcgc



YFP B1 5
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATctcgccggacac
B1



gctgaa



YFP B1 6
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATtaggtggcatcg
B1



ccctcg



YFP B1 7
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATacttcagggtca
B1



gcttgc



YFP B1 8
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATcttgccggtggt
B1



gcagat



YFP B1 9
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATgtgggccagggc
B1



acgggc



YFP B1 10
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATagccgaaggtgg
B1



tcacga



YFP B1 11
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATggcgaagcactg
B1



caggcc



YFP B1 12
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATttcatgtggtcg
B1



gggtag



YFP B1 13
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATacttgaagaagt
B1



cgtgct



YFP B1 14
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATgtagccttcggg
B1



catggc



YFP B1 15
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATaagatggtgcgc
B1



tcctgg



YFP B1 16
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATagttgccgtcgt
B1



ccttga



YFP B1 17
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATcacctcggcgcg
B1



ggtctt



YFP B1 18
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATagggtgtcgccc
B1



tcgaac



YFP B1 19
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATtcagctcgatgc
B1



ggttca



YFP B1 20
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATctccttgaagtc
B1



gatgcc



YFP B1 21
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATtgccccaggatg
B1



ttgccg



YFP B1 22
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATtgtagttgtact
B1



ccagct



YFP B1 23
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATgatatagacgtt
B1



gtggct



YFP B1 24
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATttcttctgcttg
B1



tcggcc



YFP B1 25
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATtgaagttcacct
B1



tgatgc



YFP B1 26
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATctcgatgttgtg
B1



gcggat



YFP B1 27
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATgcgagctgcacg
B1



ctgccg



YFP B1 28
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATtgttctgctggt
B1



agtggt



YFP B1 29
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATggggccgtcgcc
B1



gatggg



YFP B1 30
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATtggttgtcgggc
B1



agcagc



YFP B1 31
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATcggactggtagc
B1



tcaggt



YFP B1 32
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATgttggggtcttt
B1



gctcag



YFP B1 33
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATaccatgtgatcg
B1



cgcttc



YFP B1 34
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATcggtcacgaact
B1



ccagca



YFP B1 35
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATgccgagagtgat
B1



cccggc



YFP B1 36
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTAATtacttgtacagc
B1



tcgtcc






Gad1 21
TTGAAAAATCGAGGGTGACCTGAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



Gad1 22
CCAATGATATCCAAACCAGTAGAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



Gad1 23
GATGTCAGCCATTCACCAGCTAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



Gad1 24
TCATATGTGAACATATTGGTATAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



Gad1 25
ATGAGAACAAACACGGGTGCAAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



Gad1 26
TCTCTCATCTTCTTAAGAGTAAAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



Gad1 27
TCTTTATTTGACCATCCAACGAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



Gad1 28
GCTCCCCCAGGAGAAAATATCCAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



Gad1 29
ATGATGCTGTACATATTGGATAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



Gad1 30
ACTTCTGGGAAGTACTTGTAACAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



Gad1 31
ACAGCCGCCATGCCTTTTGTCTAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



Gad1 32
TGTTCTGAGGTGAAGAGGACCAAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



Gad1 33
GCTTTCTTTATGGAATAGTGACAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



Gad1 34
TTGTCGGTTCCAAAGCCAAGCGAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



Gad1 35
TCATTGCACTTTATCAAAATCAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



Gad1 36
TCTAAATCAGCCGGAATTATCTAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



Gad1 37
TGTTTGGCATCAAGAATTTTTGAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



Gad1 38
GCATTGACATAAAGGGGAACATAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



Gad1 39
CCGTAAACAGTCGTGCCTGCGGAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



Gad1 40
TCCGCAATTTCCTGGATTGGATAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



Gad1 41
CAAAGGTTGTATTTCTCACATAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



Gad1 42
CCACCACCCCAGGCAGCATCCAAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



Gad1 43
CGGTGCTTCCGGGACATGAGCAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



Gad1 44
TTGGCCCTTTCTATGCCGCTGAAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



Gad1 45
TTGTGAGGGTTCCAGGTGACTGAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



Gad1 46
GCAGAGCACTGGAGCAGCACGCAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



Gad1 47
ATACCCTTTTCCTTGACCAGAAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



Gad1 48
CCTGCACACATCTGGTTGCATCAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



ActB B2 2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAggaatacagcccgg
B2



ggagcatc



ActB B2 4
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAcacccacataggag
B2



tccttctg



ActB B2 6
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAcaatggggtacttc
B2



agggtcag



ActB B2 8
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAggtgccagatcttc
B2



tccatgtc



ActB B2 10
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAtcatcttttcacgg
B2



ttggcctt



ActB B2 12
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAtggctacgtacatg
B2



gctggggt



ActB B2 14
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAcaatgcctgtggta
B2



cgaccaga



ActB B2 16
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAcctcgtagatgggc
B2



acagtgtg



ActB B2 18
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAatcttcatgaggta
B2



gtctgtca



ActB B2 20
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAatttccctctcagc
B2



tgtggtgg



ActB B2 22
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAtcgaagtctagagc
B2



aacatagc



ActB B2 24
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAtagctcttctccag
B2



ggaggaag



ActB B2 26
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAcggaaccgctcgtt
B2



gccaatag



ActB B2 28
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAcaggattccatacc
B2



caagaagg



ActB B2 30
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAtcaacgtcacactt
B2



catgatgg



ActB B2 32
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAgtggtaccaccaga
B2



cagcactg



ActB B2 34
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAagagcagtaatctc
B2



cttctgca



ActB B2 36
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAttgcgctcaggagg
B2



agcaatga



ActB B2 38
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAaaggtggacagtga
B2



ggccagga



ActB B2 40
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAgaggggccggactc
B2



atcgtact



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTgcgcagcgatatcg
B1


HCR 1
tcatccat



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTccattcccaccatc
B1


HCR 3
acaccctg



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTtacctctcttgctc
B1


HCR 5
tgggcctc



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTcccagttggtaaca
B1


HCR 7
atgccatg



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTcacgcagctcattg
B1


HCR 9
tagaaggt



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTtgaaggtctcaaac
B1


HCR 11
atgatctg



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTcatacagggacagc
B1


HCR 13
acagcctg



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTtgaccccgtctccg
B1


HCR 15
gagtccat



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTggatggcgtgaggg
B1


HCR 17
agagcata



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTaagctgtagccacg
B1


HCR 19
ctcggtca



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTagcttctctttgat
B1


HCR 21
gtcacgca



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTgatgcggcagtggc
B1


HCR 23
catctcct



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTatgacctggccgtc
B1


HCR 25
aggcagct



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTggctggaaaagagc
B1


HCR 27
ctcagggc



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTttgaatgtagtttc
B1


HCR 29
atggatgc



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTttggcatagaggtc
B1


HCR 31
tttacgga



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTctgtcagcaatgcc
B1


HCR 33
tgggtaca



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTttgatcttcatggt
B1


HCR 35
gctaggag



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTgagccaccgatcca
B1


HCR 37
cacagagt



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTtgcttgctgatcca
B1


HCR 39
catctgct



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTtagaagcacttgcg
B1


HCR 41
gtgcacga



Act Short
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgTTtagaagcacttgcg
B1


HCR 41
gtgcacga



DLG4 B1 2
GGGCTGTGTTCCAGAGGGGGCGAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



DLG4 B1 4
GTGTCCGTGTTGACAATCACAGAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



DLG4 B1 6
TCCTCATACTCCATCTCCCCCTAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



DLG4 B1 8
GTGCCACCTGCGATGCTGAAGCAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



DLG4 B1 10
GGAATGATCTTGGTGATAAAGAAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



DLG4 B1 12
AACAGGATGCTGTCGTTGACCCAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



DLG4 B1 14
AGGGCCTCCACTGCAGCTGAATAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



DLG4 B1 16
GCTGGGGGTTTCCGGCGCATGAAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



DLG4 B1 18
CTGAAGCCAAGTCCTTTAGGCCAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



DLG4 B1 20
ACGTAGATGCTATTATCTCCAGAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



DLG4 B1 22
CCGATCTGCAACCTGCCATCCTAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



DLG4 B1 24
TCCTCATGCATGACATCCTCTAAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



DLG4 B1 26
TTGGCCACCTTTAGGTACACAAAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



DLG4 B1 28
GAGGTTGTGATGTCTGGGGGAGAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



DLG4 B1 30
TCGGTGCCCAAGTAGCTGCTATAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1



gggAAgAg



DLG4 B2 1
TCTTCATCTTGGTAGCGGTATTAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



DLG4 B2 3
GGAGAATTGGCCTGGTTGGGGAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



DLG4 B2 5
GTTCCGTTCACATATCCTGGGGAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



DLG4 B2 7
AGACCTGAGTTACCCCTTTCCAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



DLG4 B2 9
GATGGGTCGTCACCGATGTGTGAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



DLG4 B2 11
AGGCGGCCATCCTGGGCTGCAGAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



DLG4 B2 13
GTCACCTCCCGGACATCCACTTAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



DLG4 B2 15
TAGAGGCGAACGATGGAACCCGAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



DLG4 B2 17
TTGATAAGCTTGATCTCTATGAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



DLG4 B2 19
ATGTGCTGGTTCCCAACGCCCCAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



DLG4 B2 21
TGGGCAGCGCCTCCTTCGATGAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



DLG4 B2 23
CCCACACTGTTGACCGCCAGGAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



DLG4 B2 25
TCATATGTGTTCTTCAGGGCTGAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



DLG4 B2 27
TAGCTGTCACTCAGGTAGGCATAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



DLG4 B2 29
CTGATCTCATTGTCCAGGTGCTAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2



CAATCATC



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAACGGGTGCAGGTGAT
B1


1
GGTAGCCA



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAATCCTCAAAGAGCTG
B2


2
GTACTCTT



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAAACAGAGAAGGCTCC
B1


3
CTTTCCCA



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAACCAGCCAGCACCTT
B2


4
CACACACC



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAAATAATCTTGGCAGC
B1


5
ATACTCCT



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAATGATCTCTGGCTGA
B2


6
AAGCTTCT



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAACGGGCCTCACGCTC
B1


7
CAGCTTCT



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAATATTGGGGTGCTT
B2


8
CAACAAGC



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAAGAGATGCTGTCATG
B1


9
GAGTCGGA



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAATCGAAGATAAGGTA
B2


10
GTGGTGCC



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAAAACAGTTCCCCACC
B1


11
AGTAACCA



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAACTGTAATACTCCCG
B2


12
GGCCACAA



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAAATACAGTGGCTGGC
B1


13
ATCAGCTT



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAACAGTGTAGCACAGC
B2


14
CTCCAAGA



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAACGATGCACCACCCC
B1


15
CATCTGGT



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAGCCAGCAACAGATT
B2


16
CTCAGGCT



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAAACAGCAGCGCCCTT
B1


17
GAGCTTCG



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAATCTATGGCCAGGCC
B2


18
AAAGTCTG



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAACATGCCTGCTGCTC
B1


19
CCCCTCCA



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAAGGTATCCAGGTGT
B2


20
CCCTGCGA



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAATCCTTCCTCAGCAC
B1


21
TTCTGGGG



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAGCCCACAGGTCCAC
B2


22
GGGCTTCC



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAAAAGATATACAGGAT
B1


23
GACGCCAC



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAATCATCCCAGAACGG
B2


24
GGGATACC



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAATGCTGGTACAGGCG
B1


25
ATGCTGGT



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAGATGGGAAATCATA
B2


26
GGCACCAG



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAAGGGGTGACGGTGTC
B1


27
CCATTCTG



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAAGCATCTTATTGAT
B2


28
CAGATCCT



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAAATGCGTTTGGACGG
B1


29
GTTGATGG



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAACATGGGTGCTTGAG
B2


30
AGCCTCAG



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAAGCCACGGTGGAGCG
B1


31
GTGCGAGA



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAATCCACGGTCTCCTG
B2


32
TCTGTGCA



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAACTGGCATTGAACTT
B1


33
CTTCAGGC



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAGTGGTGAGGATGGC
B2


34
TCCCTTCA



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAAGAGAAGTTCCTGGT
B1


35
GGCCAGCA



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAATTCTTCTTGTTTCC
B2


36
TCCGCTCT



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAATCAGAAGATTCCTT
B1


37
CACACCAT



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAATCTTCGTCCTCAAT
B2


38
GGTGGTGT



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAAATTTCCTGTTTGCG
B1


39
CACTTTGG



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAGCTTCGATCAGCTG
B2


40
CTCTGTCA



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAAGACTCAAAGTCTCC
B1


41
ATTGCTTA



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAGTCATTCCAGGGTC
B2


42
GCACATCT



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAACCCAGGGCCTCTGG
B1


43
TTCAAAGG



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAACGATGAAAGTCCAG
B2


44
GCCCTCCA



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAAGACCACAGGTTTTC
B1


45
AAAATAGA



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAATGGTGGTGTGCAC
B2


46
GGGCTTGC



Camk2a iso2
gAggAgggCAgCAAACgggAAgAgTCTTCCTTTACgAAATCAGGTGGATGTG
B1


47
AGGGTTCA



Camk2a iso2
CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCAAATATAGGCGATGCA
B2


48
GGCTGACT






mCherry 2C
cttcttcaccttttgaaaccatAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


1
gggAAgAg



mCherry 2C
ccatatgaactttaaatctcatAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


3
gggAAgAg



mCherry 2C
cttcaccttcaccttcaatttcAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


5
gggAAgAg



mCherry 2C
cacctttagtaactttcaatttAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


7
gggAAgAg



mCherry 2C
catacataaattgtggtgacaaAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


9
gggAAgAg



mCherry 2C
ttaaataatctggaatatcagcAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


11
gggAAgAg



mCherry 2C
tcaaaattcataactctttcccAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


13
gggAAgAg



mCherry 2C
ctctcaatttaactttataaatAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


15
gggAAgAg



mCherry 2C
ccatagtttttttttgcataacAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


17
gggAAgAg



mCherry 2C
tcaatctttgtttaatttcaccAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


19
gggAAgAg



mCherry 2C
taatattaacattataagcaccAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


21
gggAAgAg



mCherry 2C
tttcatattgttcaacaatagtAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


23
gggAAgAg



mCherry 2C
attctttaataatagccatattAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


2
CAATCATC



mCherry 2C
attcatgaccattaactgaaccAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


4
CAATCATC



mCherry 2C
cagtttgagtaccttcatatggAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


6
CAATCATC



mCherry 2C
tatcccaagcaaatggtaatggAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


8
CAATCATC



mCherry 2C
gatgtttaacataagcttttgaAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


10
CAATCATC



mCherry 2C
ttaaaaccttctggaaatgacaAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


12
CAATCATC



mCherry 2C
gagtaacagtaacaacaccaccAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


14
CAATCATC



mCherry 2C
gaccatctgatggaaaattagtAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


16
CAATCATC



mCherry 2C
ttctttctgatgaagcttcccaAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


18
CAATCATC



mCherry 2C
gtaattgaactggttttttagcAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


20
CAATCATC



mCherry 2C
tcattatgtgaagtaatatccaAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


22
CAATCATC



mCherry 2C
atttatataattcatccataccAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


24
CAATCATC






DLG4
AATACCGCTACCAAGATGAAGAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


ShHCR mis 1
CAATCATC



DLG4
TCCCCAACCAGGCCAATTCTCCAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


ShHCR mis 3
CAATCATC



DLG4
CCCCAGGATATGTGAACGGAACAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


ShHCR mis 5
CAATCATC



DLG4
TGGAAAGGGGTAACTCAGGTCTAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


ShHCR mis 7
CAATCATC



DLG4
CACACATCGGTGACGACCCATCAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


ShHCR mis 9
CAATCATC



DLG4
CTGCAGCCCAGGATGGCCGCCTAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


ShHCR mis 11
CAATCATC



DLG4
AAGTGGATGTCCGGGAGGTGACAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


ShHCR mis 13
CAATCATC



DLG4
CGGGTTCCATCGTTCGCCTCTAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


ShHCR mis 15
CAATCATC



DLG4
TCATAGAGATCAAGCTTATCAAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


ShHCR mis 17
CAATCATC



DLG4
GGGGCGTTGGGAACCAGCACATAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


ShHCR mis 19
CAATCATC



DLG4
TCATCGAAGGAGGCGCTGCCCAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


ShHCR mis 21
CAATCATC



DLG4
TCCTGGCGGTCAACAGTGTGGGAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


ShHCR mis 23
CAATCATC



DLG4
CAGCCCTGAAGAACACATATGAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


ShHCR mis 25
CAATCATC



DLG4
ATGCCTACCTGAGTGACAGCTAAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


ShHCR mis 27
CAATCATC



DLG4
AGCACCTGGACAATGAGATCAGAAAgCTCAgTCCATCCTCgTAAATCCTCAT
B2


ShHCR mis 29
CAATCATC



DLG4
CGCCCCCTCTGGAACACAGCCCAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


ShHCR mis 2
gggAAgAg



DLG4
CTGTGATTGTCAACACGGACACAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


ShHCR mis 4
gggAAgAg



DLG4
AGGGGGAGATGGAGTATGAGGAAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


ShHCR mis 6
gggAAgAg



DLG4
GCTTCAGCATCGCAGGTGGCACAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


ShHCR mis 8
gggAAgAg



DLG4
TCTTTATCACCAAGATCATTCCAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


ShHCR mis 10
gggAAgAg



DLG4
GGGTCAACGACAGCATCCTGTTAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


ShHCR mis 12
gggAAgAg



DLG4
ATTCAGCTGCAGTGGAGGCCCTAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


ShHCR mis 14
gggAAgAg



DLG4
TCATGCGCCGGAAACCCCCAGCAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


ShHCR mis 16
gggAAgAg



DLG4
GGCCTAAAGGACTTGGCTTCAGAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


ShHCR mis 18
gggAAgAg



DLG4
CTGGAGATAATAGCATCTACGTAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


ShHCR mis 20
gggAAgAg



DLG4
AGGATGGCAGGTTGCAGATCGGAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


ShHCR mis 22
gggAAgAg



DLG4
TAGAGGATGTCATGCATGAGGAAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


ShHCR mis 24
gggAAgAg



DLG4
TTGTGTACCTAAAGGTGGCCAAAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


ShHCR mis 26
gggAAgAg



DLG4
CTCCCCCAGACATCACAACCTCAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


ShHCR mis 28
gggAAgAg



DLG4
ATAGCAGCTACTTGGGCACCGAAAgCATTCTTTCTTgAggAgggCAgCAAAC
B1


ShHCR mis 30
gggAAgAg










smFISH









Probe Name
Oligonucleotide Sequence
Sequence Name





UBC
atggtcttaccagtcagagt
hUBC_1



gacattctcgatggtgtcac
hUBC_2



gggatgccttccttatcttg
hUBC_3



atcttccagctgttttccag
hUBC_4



cagtgagtgtcttcacgaag
hUBC_5



tcctggatctttgctttgac
hUBC_6



cagggtagactctttctgga
hUBC_7



cttcacgaagatctgcatcc
hUBC_8



tcttggatctttgccttgac
hUBC_9



cagtgagtgtcttcacgaag
hUBC_10



tgacgttctcgatagtgtca
hUBC_11



tccttgtcttggatctttgc
hUBC_12



cagggtagactctttctgga
hUBC_13



cttcacgaagatctgcatcc
hUBC_14



agagtgatggtcttaccagt
hUBC_15



tcttggatctttgccttgac
hUBC_16



cttcacgaagatctgcatcc
hUBC_17



agagtgatggtcttaccagt
hUBC_18



tcttggatctttgccttgac
hUBC_19



tgtttcccagcaaagatcaa
hUBC_20



cttcacgaagatctgcatcc
hUBC_21



agagtgatggtcttaccagt
hUBC_22



tcttggatctttgccttgac
hUBC_23



tgtttcccagcaaagatcaa
hUBC_24



cttcacgaagatctgcatcc
hUBC_25



agagtgatggtcttaccagt
hUBC_26



tcttggatctttgccttgac
hUBC_27



tgtttcccagcaaagatcaa
hUBC_28



gacattctcgatggtgtcac
hUBC_29



gggatgccttccttatcttg
hUBC_30



tgtttcccagcaaagatcaa
hUBC_31



agagtggactctttctggat
hUBC_32





EEF2
atctggtctaccgtgaagtt
hEEF2_1



ttggccttcttgtccatgat
hEEF2_2



gtatcagtgaagcgtgtctc
hEEF2_3



ttgacttgatggtgatgcaa
hEEF2_4



ctcgtagaagagggagatgg
hEEF2_5



tccttgctctgcttgatgaa
hEEF2_6



gggagtcaatgaggttgatg
hEEF2_7



cggtccatcttgttcatcat
hEEF2_8



gtggagatgatgacgttcac
hEEF2_9



gtaccgaggacaggatcgat
hEEF2_10



caaactgcttcagggtgaag
hEEF2_11



aacttggccacatacatctc
hEEF2_12



atgtcctctactttcttggc
hEEF2_13



ttcatgatcgcatcaaacac
hEEF2_14



gtccagtttgatgtccagtt
hEEF2_15



gatggtgatcatctgcaaca
hEEF2_16



tttggggtcacagcttttaa
hEEF2_17



gtagaaccgacctttgtcgg
hEEF2_18



ccatgatcctgaccttcagg
hEEF2_19



ttcttcccaggggtatagtt
hEEF2_20



tctggattggcttcaggtag
hEEF2_21



ggcccatcatcaagattgtt
hEEF2_22



gtcttcaccaggaactggtc
hEEF2_23



ctgacgctgaacttcatcac
hEEF2_24



atgatatgctctcccgactc
hEEF2_25



gactcttcactgaccgtctc
hEEF2_26



cttcatgtacagccggttgt
hEEF2_27



tcgcctttatcgatgtcctc
hEEF2_28



tgatgtcggtgaggatgttg
hEEF2_29



cactgtccttgatctcgttg
hEEF2_30



gtcagcacactggcatagag
hEEF2_31



atctccacaaggtagatggg
hEEF2_32





USF2
ggatccagacccgggtccag
usf2_withUT R_1



tactggatgttgtggtcgcc
usf2_withUT R_2



catttgtctctgtgcggaac
usf2_withUT R_3



attttggatcacagcctgtc
usf2_withUT R_4



gactgccaccattgctgaag
usf2_withUT R_5



ctgggaaataggcaaatcgt
usf2_withUT R_6



gacacagccgtagtatctcc
usf2_withUT R_7



gtctgaagcacatcctgggg
usf2_withUT R_8



ggcgatcgtcctctgtgttc
usf2_withUT R_9



tggttccatcaatttttgga
usf2_withUT R_10



ttctcctctcatctcggggt
usf2_withUT R_11



ctccacttcgttgtgctggg
usf2_withUT R_12



cagttgttgatcttgtccct
usf2_withUT R_13



gattttcgaaagctggacga
usf2_withUT R_14



gttgtctgcgttacagtctg
usf2_withUT R_15



ggccttggacaggatccctc
usf2_withUT R_16



cgcaactcccggatgtaatc
usf2_withUT R_17



ctgcatgcgctggttggtct
usf2_withUT R_18



gctcggcctctttgaaggtc
usf2_withUT R_19



agctcgttgtccatctgcag
usf2_withUT R_20



caccatctccaggttgtgct
usf2_withUT R_21



tgtatccacagaaatgcatt
usf2_withUT R_22



ggaggataccgtttccaagt
usf2_withUT R_23



gtgagaccactagaagtgcc
usf2_withUT R_24



cataggtccaggccccgggt
usf2_withUT R_25



cagggacccagaaacaagag
usf2_withUT R_26



gggccagtttattgcagtta
usf2_withUT R_27





TOP2A
ctgggcggagcaaaatatgt
hTOP2A_CD S_1



tcttcatcgtaaacccacat
hTOP2A_CD S_2



ccggatcaattgtgactcta
hTOP2A_CD S_3



ccttttccattattccatat
hTOP2A_CD S_4



agaagttaggagctgtccaa
hTOP2A_CD S_5



ccagcaatatcatatgctct
hTOP2A_CD S_6



ttactggcagtttatttcca
hTOP2A_CD S_7



tgttgatccaaagctcttgg
hTOP2A_CD S_8



aactggacttgggccttaaa
hTOP2A_CD S_9



atcattggcatcatcgagtt
hTOP2A_CD S_10



gtcaggataagcgtacactc
hTOP2A_CD S_11



ggaaaaccccatatttgtct
hTOP2A_CD S_12



tttcttgtactgaagaccca
hTOP2A_CD S_13



ttggtcctgatctgtcataa
hTOP2A_CD S_14



ctccagaaaacgatgtcgca
hTOP2A_CD S_15



gttaaccattcctttcgatc
hTOP2A_CD S_16



agctaattgggcaaccttta
hTOP2A_CD S_17



atgtatcgtggactagcaga
hTOP2A_CD S_18



acgctggttgtcatcatata
hTOP2A_CD S_19



ttcttctccatccatcaaac
hTOP2A_CD S_20



cccttgaagttcttgtaact
hTOP2A_CD S_21



tatgagaggaggtgtcttct
hTOP2A_CD S_22



tgtatggtattccctatagt
hTOP2A_CD S_23



tcagtttagcagattcagca
hTOP2A_CD S_24



cttcacaggatccgaatcat
hTOP2A_CD S_25



gtggaatgactctttgacca
hTOP2A_CD S_26



tgctcctatctgattctgaa
hTOP2A_CD S_27



agtggaggtggaagactgac
hTOP2A_CD S_28



aattcaaagctggatccctt
hTOP2A_CD S_29



caggatcaggcttttgagag
hTOP2A_CD S_30



cttggatttcttgcttgtga
hTOP2A_CD S_31



tatggaagtcatcactctcc
hTOP2A_CD S_32





NEAT1
gacctagtctccttgccaag
NEAT1_1



ggatattttccatgcagcct
NEAT1_2



acaagttgaagattagccct
NEAT1_3



ccttggtctggaaaaaaagg
NEAT1_4



cgagctaagttcagttccac
NEAT1_5



ggccgagcgaaaattacata
NEAT1_6



cctgtcaaacatgctaggtg
NEAT1_7



actgccacctggaaaataaa
NEAT1_8



gtgagctcacaagaagagtt
NEAT1_9



accagatgaccaggtaatgt
NEAT1_10



cggtccatgaagcatttttg
NEAT1_11



tcgccatgaggaacactata
NEAT1_12



aatctgcaggcatcaattga
NEAT1_13



cctggaaacagaacattgga
NEAT1_14



gcatctgctgtggacttttt
NEAT1_15



ggctctggaacaagcattta
NEAT1_16



tgcagcatctgaaaaccttt
NEAT1_17



accggaggctcaatttagaa
NEAT1_18



caaggttccaagcacaaaac
NEAT1_19



acagcttagggatcttcttg
NEAT1_20



tggcatcaacgttaaaatgt
NEAT1_21



tctacaaggcatcaatctgc
NEAT1_22



aagaacttctccgagaaacg
NEAT1_23



gccccaagttatttcatcag
NEAT1_24



gcgtttagcacaacacaatg
NEAT1_25



ggaatgaccaacttgtaccc
NEAT1_26



caatgcccaaactagacctg
NEAT1_27



tcctagtaatctgcaatgca
NEAT1_28



agcaagaacaaaagagcact
NEAT1_29



ggtcctcttactagaatgcc
NEAT1_30



ctgtgtcacctgttttcagt
NEAT1_31



cctttggttctcggaaaact
NEAT1_32



agctggtaaagacatttccc
NEAT1_33



ctctgaaacaggctgtcttg
NEAT1_34



gcccatctttcaagtgacta
NEAT1_35



aaccacctaagttgctaagg
NEAT1_36



tcgtcttaagtggtccctta
NEAT1_37



atccagaagagcccatctaa
NEAT1_38



acctgtgacaaatgaggaac
NEAT1_39



agatgtgtttctaaggcacg
NEAT1_40



acagtgaccacaaaaggtta
NEAT1_41



agcaaaggtacatggattct
NEAT1_42



cagggttttcagatcacaca
NEAT1_43



ccccaagtcattggttaaga
NEAT1_44



tcccaacgacagtaattgtt
NEAT1_45



cccatacatgcgtgactaat
NEAT1_46



caacagcatacccgagacta
NEAT1_47



acagagcaacataccagtac
NEAT1_48









Cell Culture and Fixation

HeLa (ATCC CCL-2) cells and HEK293-FT cells (Invitrogen) were cultured on Nunc Lab-Tek II Chambered Coverglass (Thermo Scientific) in D10 medium (Cellgro) supplemented with 10% FBS (Invitrogen), 1% penicillin/streptomycin (Cellgro), and 1% sodium pyruvate (BioWhittaker). Cells were authenticated by the manufacturer and tested for mycoplasma contamination to their standard levels of stringency, and were here used because they are common cell lines for testing new tools. Cultured cells were washed once with DPBS (Cellgro), fixed with 10% formalin for 10 mins, and washed twice with 1× PBS. Fixed cells were then stored in 70% Ethanol at 4° C. until use.


Preparation of LabelX

Acryloyl-X, SE (6-((acryloyl)amino)hexanoic acid, succinimidyl ester, here abbreviated AcX; Thermo-Fisher) was resuspended in anhydrous DMSO at a concentration of 10 mg/mL, aliquoted and stored frozen in a desiccated environment. LABEL-IT® Amine Modifying Reagent (Mirus Bio, LLC) was resuspended in the provided Mirus Reconstitution Solution at 1 mg/ml and stored frozen in a desiccated environment. To prepare LabelX, 10 μL, of AcX (10 mg/mL) was reacted with 100 μL of LABEL-IT® Amine Modifying Reagent (1 mg/mL) overnight at room temperature with shaking. LabelX was subsequently stored frozen (−20° C.) in a desiccated environment until use.


Mouse perfusion


All methods for animal care and use were approved by the Massachusetts Institute of Technology Committee on Animal Care and were in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals. All solutions below were made up in 1× phosphate buffered saline (PBS) prepared from nuclease free reagents. Mice were anesthetized with isoflurane and perfused transcardially with ice cold 4% paraformaldehyde. Brains were dissected out, left in 4% paraformaldehyde at 4° C. for one day, before moving to PBS containing 100 mM glycine. Slices (50 μm and 200 μm) were sliced on a vibratome (Leica VT1000S) and stored at 4° C. in PBS until use. The mouse used in FIG. 3 and related analyses was a Thy1-YFP (Tg(Thy1-YFP)16 Jrs) male mouse in the age range 6-8 weeks. No sample size estimate was performed, since the goal was to demonstrate a technology. No exclusion, randomization or blinding of samples was performed.


LabelX Treatment of Cultured Cells and Brain Slices

Fixed cells were washed twice with 1× PBS, once with 20 mM MOPS pH 7.7, and incubated with LabelX diluted to a desired final concentration in MOPS buffer (20 mM MOPS pH 7.7) at 37° C. overnight followed by two washes with 1× PBS. For cells, ranges of LabelX were used that resulted in a LABEL-IT® Amine concentration of 0.006-0.02 mg/mL; higher concentrations resulted in somewhat dimmer smFISH staining (FIG. 15), but otherwise no difference in staining quality was observed with LABEL-IT® Amine concentrations in this range. For FIG. 1e, FIG. 4, FIG. 5, and FIG. 6 fixed cells were incubated with LabelX diluted to a final LABEL-IT® Amine concentration of 0.02 mg/mL. For all other experiments in cells, fixed cells were treated with LabelX diluted to a final LABEL-IT® Amine concentration of 0.006 mg/mL.


Brain slices, as prepared above, were incubated with 20 mM MOPS pH 7.7 for 30 mins and subsequently incubated with LabelX diluted to a final LABEL-IT® Amine concentration of 0.1 mg/mL (due to their increased thickness and increased fragmentation from formaldehyde post-fixation) in MOPS buffer (20 mM MOPS pH 7.7) at 37° C. overnight. For YFP retention, slices were treated with 0.05 mg/mL AcX in PBS for >6 hours @ RT.


smFISH in Fixed Cultured Cells Before Expansion


Fixed cells were briefly washed once with wash buffer (10% formamide, 2× SSC) and hybridized with RNA FISH probes in hybridization buffer (10% formamide, 10% dextran sulfate, 2× SSC) overnight at 37° C. Following hybridization, samples were washed twice with wash buffer, 30 mins per wash, and washed once with 1× PBS. Imaging was performed in 1× PBS.


smFISH probe sets targeting the human transcripts for TFRC, ACTB, GAPDH, XIST, and 5′ portion of NEAT1 were ordered from Stellaris with Quasar 570 dye. Probe sets against UBC, EEF2, USF2, TOP2A and full length NEAT1 were synthesized, conjugated to fluorophores, and subsequently purified by HPLC as described previously37. Oligonucleotide sequences for probe sets and accession numbers can be found in Table 4.


Gelation, Digestion and Expansion

Monomer solution (1× PBS, 2 M NaCl, 8.625% (w/w) sodium acrylate, 2.5% (w/w) acrylamide, 0.15% (w/w) N,N′-methylenebisacrylamide) was mixed, frozen in aliquots, and thawed before use. Monomer solution was cooled to 4° C. before use. For gelling cultured cells treated with LabelX, a concentrated stock of VA-044 (25% w/w, chosen instead of the Ammonium persulfate (APS)/Tetramethylethylenediamine (TEMED) of the original ExM protocol1 because APS/TEMED resulted in autofluorescence that was small in magnitude but appreciable in the context of smFISH) was added to the monomer solution to a final concentration of 0.5% (w/w) and degassed in 200 μl aliquots for 15 mins. Cells were briefly incubated with the monomer solution plus VA-044 and transferred to a humidified chamber. Subsequently, the humidified chamber was purged with nitrogen gas. To initiate gelation, the humidified chamber was transferred to a 60° C. incubator for two hours. For gelling brain slices treated with LabelX, gelation was performed as in the original ExM protocol (since, with HCR amplification, the slight autofluorescence of APS/TEMED was negligible). Gelled cultured cells and brain slices were digested with Proteinase K (New England Biolabs) diluted 1:100 to 8 units/mL in digestion buffer (50 mM Tris (pH 8), 1 mM EDTA, 0.5% Triton X-100, 500 mM NaCl) and digestion was carried out overnight at 37° C. The gels expand slightly in the high osmolarity digestion buffer (˜1.5×). After digestion, gels were stored in 1× PBS until use and expansion was carried out as previously described.


smFISH Staining After Expansion


Expanded gels were incubated with wash buffer (10% formamide, 2× SSC) for 30 mins at room temperature and hybridized with RNA FISH probes in hybridization buffer (10% formamide, 10% dextran sulfate, 2× SSC) overnight at 37° C. Following hybridization, samples were washed twice with wash buffer, 30 minutes per wash, and washed once with1× PBS for another 30 mins. Imaging was performed in 1× PBS.


Image Processing and Analysis of smFISH performed on Cultured Cells


Widefield images of smFISH staining performed before or after expansion were first processed using a rolling-ball background subtraction algorithm (FIJI)38 with a 200 pixel radius. Subsequently, maximum intensity Z-projections of these images were generated. Spots were then localized and counted using a code developed by the Raj lab and available online (http://rajlab.seas.upenn.edu/StarSearch/launch.html). This image analysis was performed for FIGS. 1C-E, FIGS. 2A-C, FIG. FIGS. 5A-E, FIGS. 6A-G, FIGS. 7A-E, FIGS. 9A-B, 11A-C.


Analysis of Expansion Isotropy

smFISH images before and after expansion of TOP2A was rigidly aligned via two control points using the FIJI plugin Turboreg39. Spots were localized and counted via a custom spot counting Matlab code developed by the Raj lab (complete source code and instructions can be found at https://bitbucket.org/arjunrajlaboratory/rajlabimagetools/wiki/Home). Length measurements were performed among all pairs of points before expansion and the corresponding pairs of points after expansion via a custom Matlab script. Measurement error was defined as the absolute difference between the before and after expansion length measurements (FIG. 8C).


Re-embedding of Expanded Gels in Acrylamide Matrix

For serial staining in cells, expanded gels were re-embeded in acrylamide to stabilize the gels in the expanded state. Briefly: gels were expanded in water and cut manually to ˜1 mm thickness with a stainless steel blade. Cut gels were incubated in 3% acrylamide, 0.15% N,N′-Methylenebisacrylamide with 0.05% APS, 0.05% TEMED and 5 mM Tris ph 10.5 for 20 minutes on a shaker. There is a ˜30% reduction in gel size during this step. Excess solution is removed from the gels and the gels are dried with light wicking from a laboratory wipe. Gels are placed on top of a bind-silane treated (see below) coverslip or glass bottom plate with a coverslip placed on top of the gels before moving into a container and purged with nitrogen. The container is moved to a 37° C. incubator for gelation for 1.5 hours.


Staining of Re-Embedded Gels

Re-embeded staining of gels were performed with exact conditions as described above for expanded gels, except post-hybridization washes were changed to twice with wash buffer (10% formamide), 60 minutes per wash.


Probes were removed for multiple rounds of hybridization via treatment with DNAse I or 100% formamide. For DNAse I, samples were treated with DNAse I at 0.5 U/μL for 6 hours at RT. For formamide stripping, samples were treated with 100% formamide at 6 hours at 37C.


Bind-silane Treatment of Coverslips

Coverslips and glass bottom 24 well plates were treated with Bind-Silane, a silanization reagent which incorporates acryloyl groups onto the surface of glass to perform in free radical polymerization. Briefly, 5 μL of Bind-Silane reagent was diluted into 8 mL of ethanol, 1.8 mL of ddH2O and 200 μL of acetic acid. Coverslips and glass bottom 24 well plates were washed with ddH2O followed by 100% ethanol, followed by the diluted Bind-Silane reagent. After a brief wash with the diluted Bind-Silane reagent, the cover-slip was dried, then washed with 100% ethanol, and then dried again. Coverslips were prepared immediately before use.


Probe Design for HCR-FISH

Probe sequences and accession numbers for mRNA targets can be found in Table 4. Probes were designed for HCR-FISH by tiling the CDS of mRNA targets with 22-mer oligos spaced by 3-7 bases. HCR initiators were appended to tiled sequences via a 2 base spacer (AA). For 2 color probe-sets, even and odd tiled probes were assigned different HCR-initiators to allow for amplification in different color channel.


RNA FISH with Hybridization Chain Reaction (HCR) Amplification


Gelled samples were incubated with wash buffer (20% formamide, 2× SSC) for 30 mins at room temperature and hybridized with HCR initiator tagged FISH probes in hybridization buffer (20% formamide, 10% dextran sulfate, 2× SSC) overnight at 37° C. Following hybridization, samples were washed twice with wash buffer, 30 mins per wash, and incubated with 1× PBS for 2 hrs at 37° C. Subsequently, samples were incubated with 1× PBS for at least 6hrs at room temperature. Before HCR amplification, hybridized samples were pre-incubated with amplification buffer (10% dextran sulfate, 5× SSC, 0.1% Tween 20) for 30 mins. To initiate amplification, HCR hairpin stocks (Alexa 456 and Alexa 647 fluorophores) at 3 μM were snap-cooled by heating to 95° C. for 90 seconds, and leaving to cool at room temperature for 30 mins. Gelled samples were then incubated with HCR hairpins diluted to 60 nM in amplification buffer for 3 hrs at room temperature. After amplification, gels were washed with 5× SSCT (5× SSC, 0.1% Tween 20) twice with one hour per wash.


Imaging of Cultured Cells using ExFISH


Both cultured cells as well as LabelX treated and expanded cultured cells were imaged on a Nikon Ti-E epifluorescence microscope with a SPECTRA X light engine (Lumencor), and a 5.5 Zyla sCMOS camera (Andor), controlled by NIS-Elements AR software. For FIGS. 1C, 1D, and FIGS. 6A-G, FIGS. 7A-E, and FIGS. 8-D a 40×1.15 NA water immersion objective was used. For all other experiments with cultured cells, a 60×1.4 NA oil immersion objective was used.


For imaging smFISH probes labeled with fluorophores, the following filter cubes (Semrock, Rochester, N.Y.) were used: Alexa 488, GFP-1828A-NTE-ZERO; Quasar 570, LF561-B-000; Alexa 594, FITC/TXRED-2X-B-NTE; Atto 647N, Cy5-4040C-000.


Imaging of Expanded Brain Slices

For epifluorescence imaging of brain sections before and after expansion (FIGS. 3A-E) and to quantify expansion factors of tissue slices specimens were imaged on a Nikon Ti-E epifluorescence microscope with a 4×0.2 NA air objective, a SPECTRA X light engine (Lumencor), and a 5.5 Zyla sCMOS camera (Andor), controlled by NIS-Elements AR software.


Post-expansion confocal imaging of expanded brain tissue was performed on an Andor spinning disk (CSU-X1 Yokogawa) confocal system with a 40×1.15 NA water objective (FIGS. 3F-K, FIGS. 13A-G) on a Nikon TI-E microscope body. GFP was excited with a 488 nm laser, with 525/40 emission filter. Alexa 546 HCR amplicons were excited with a 561 nm laser with 607/36 emission filter. Alexa 647 amplicons were excited with a 640 nm laser with 685/40 emission filter.


Gels were expanded in with 3 washes, 15 minutes each of 0.05× SSC. The expansion factor can be controlled with the salt concentration. It was found that 0.05× SSC gives 3× expansion, while still giving enough salt for hybridization stability. To stabilize the gels against drift during imaging following expansion, gels were placed in glass bottom 6 well plates with all excess liquid removed. If needed, liquid low melt agarose (2% w/w) was pipetted around the gel and allowed to solidify, to encase the gels before imaging.


Lightsheet imaging was performed on a Zeiss Z.1 lightsheet microscope. Briefly, the sample was fixed on a custom-made plastic holder using super glue and mounted on the freely rotating stage of the Z.1 lightsheet. Lightsheets were generated by two illumination objectives (5×, NA 0.1), and the fluorescence signal detected by a 20× water immersion objective (NA 1.0). Both lightsheets were used for data collection. The image volume dimensions of a single tile were 1400×1400×1057 pixels, with a voxel size of 227 nm laterally and 469 nm axially. The laserlines used for excitation were 488 nm, 561 nm and 638 nm. The individual laser transmissions were set to 5%, with the maximum output of 50 mW (488 nm and 561 nm) and 75 mW (638 nm). Optical filters used to separate and clean the fluorescence response included a Chroma T5601 pxr as a dichroic, and a Chroma 59001 m for GFP and 59007 m for Alexa 546 and Alexa 647. Two PCO.Edge 5.5 m sCMOS cameras were used to capture two fluorescence channels simultaneously. Tiled datasets were taken with the Zeiss ZEN Software, and subsequently merged and processed with FIJI, Arivis Vision4D and Bitplane Imaris.


Two Color Analysis in Slices

A sliding window averaging (or minimization) scheme in Z (3 optical sections) was used to suppress movement artifacts before spot detection processing. RNA puncta were detected via a custom 3D spot counting Matlab code developed by the Raj lab; complete source code and instructions can be found at https://bitbucket. org/arjunrajlaboratory/rajlabimagetools/wiki/Home.


Spot centroids were extracted from both color channels, and spots were determined to be co-localized if their centroids were within a 3 pixel radius in the x,y dimensions and a 2 pixel radius in the z dimension.


HCR Reversal via Toe-Hold Mediated Strand Displacement

HCR amplification commences upon the addition of two HCR metastable amplifier hairpins. We designed a pair of HCR amplifiers, B2H1T and B2H2 (see below for sequence), where B2H1T bears a 6 bp toe-hold for strand displacement. To initiate HCR amplification, aliquots of these amplifiers at 3 μM were snap-cooled by heating to 95° C. for 90 seconds, and leaving to cool at room temperature for 30 mins. Gelled samples were then incubated with HCR hairpins diluted to 60 nM in amplification buffer for 3hrs at room temperature. After amplification, gels were washed with 5× SSCT (5× SSC, 0.1% Tween 20) twice with one hour per wash. Subsequently, HCR reversal was initiated by the addition of a displacement strand (see below for sequence) at 200 nM in 5× SSCT.









B2H1T:



ggCggTTTACTggATgATTgATgAggATTTACgAggAgCTCAgTCCATCC






TCgTAAATCCTCATCAATCATCAAATAG.





B2H2:


/5′-Alexa546-C12/


CCTCgTAAATCCTCATCAATCATCCAgTAAACCgCCgATgATTgATgAgg





ATTTACgAggATggACTgAgCT.





Displacement Strand:


CTATTTGATGATTGATGAGGATTTAcGAGGATGGAcTGAGcT.






REFERENCES:



  • 1. Chen, F., Tillberg, P. W. & Boyden, E. S. Expansion microscopy. Science (80-.). 347, 543-548 (2015).

  • 2. Femino, A. M., Fay, F., Fogarty, K. & Singer, R. Visualization of Single RNA Transcripts in Situ. Science (80,). 280, 585-590 (1998).

  • 3. Levsky, J. M. & Singer, R. H. Fluorescence in situ hybridization: past, present and future. J. Cell Sci. 116, 2833-2838 (2003).

  • 4. Raj, A., van den Bogaard, P., Rifkin, S. A., van Oudenaarden, A. & Tyagi, S. Imaging individual mRNA molecules using multiple singly labeled probes. Nat. Methods 5, 877-9 (2008).

  • 5. Choi, H. M. T. et al. Programmable in situ amplification for multiplexed imaging of mRNA expression. Nat. Biotechnol. 28, 1208-12 (2010).

  • 6. Choi, H. M. T., Beck, V. A. & Pierce, N. A. Next-Generation in Situ Hybridization Chain Reaction: Higher Gain, Lower Cost, Greater Durability. ACS Nano 8, 4284-4294 (2014).

  • 7 Cajigas, I. J. et al. The local transcriptome in the synaptic neuropil revealed by deep sequencing and high-resolution imaging. Neuron 74, 453-66 (2012).

  • 8. Wang, F. et al. RNAscope: A novel in situ RNA analysis platform for formalin-fixed, paraffin-embedded tissues. J. Mol. Diagnostics 14, 22-29 (2012).

  • 9. Tillberg, P. W. et al. Expansion Microscopy of Biological Specimens with Protein Retention. Nat. Biotechnol.

  • 10. Chozinski, T. J. et al. Expansion microscopy with conventional antibodies and fluorescent proteins. Nat. Methods (2016). doi:10.1038/nmeth.3833

  • 11. Engreitz, J. M. et al. The Xist 1ncRNA exploits three-dimensional genome architecture to spread across the X chromosome. Science 341, 1237973 (2013).

  • 12. Panning, B., Dausman, J. & Jaenisch, R. X chromosome inactivation is mediated by Xist RNA stabilization. Cell 90, 907-16 (1997).

  • 13. Plath, K., Mlynarczyk-Evans, S., Nusinow, D. A. & Panning, B. Xist RNA and the mechanism of X chromosome inactivation. Annu. Rev. Genet. 36, 233-78 (2002).

  • 14. Mito, M., Kawaguchi, T., Hirose, T. & Nakagawa, S. Simultaneous multicolor detection of RNA and proteins using super-resolution microscopy. Methods (2015). doi:10.1016/j.ymeth.2015.11.007

  • 15. Clemson, C. M. et al. An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles. Mol. Cell 33, 717-26 (2009).

  • 16. Lieberman-Aiden, E. et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 326, 289-93 (2009).

  • 17. Lubeck, E. & Cai, L. Single-cell systems biology by super-resolution imaging and combinatorial labeling. Nat. Methods 9, 743-8 (2012).

  • 18. Lubeck, E., Coskun, A. F., Zhiyentayev, T., Ahmad, M. & Cai, L. Single-cell in situ RNA profiling by sequential hybridization. Nat. Methods 11, 360-1 (2014).

  • 19. Chen, K. H., Boettiger, A. N., Moffitt, J. R., Wang, S. & Zhuang, X. Spatially resolved, highly multiplexed RNA profiling in single cells. Science (80-.). 348, aaa6090-aaa6090 (2015).

  • 20. Beliveau, B. J. et al. Versatile design and synthesis platform for visualizing genomes with Oligopaint FISH probes. Proc. Natl. Acad. Sci. U. S. A. 109, 21301-6 (2012).

  • 21. Feng, G. et al. Imaging neuronal subsets in transgenic mice expressing multiple spectral variants of GFP. Neuron 28, 41-51 (2000).

  • 22. Lein, E. S. et al. Genome-wide atlas of gene expression in the adult mouse brain. Nature 445, 168-76 (2007).

  • 23. Huisken, J., Swoger, J., Bene, F. Del, Wittbrodt, J. & Stelzer, E. H. K. Optical Sectioning Deep Inside Live Embryos by Selective Plane Illumination Microscopy. Science 305, 1007-1009 (2004).

  • 24. Batish, M., van den Bogaard, P., Kramer, F. R. & Tyagi, S. Neuronal mRNAs travel singly into dendrites. Proc. Natl. Acad. Sci. 109, 4645-4650 (2012).

  • 25. Cabili, M. N. et al. Localization and abundance analysis of human lncRNAs at single-cell and single-molecule resolution. Genome Biol. 16, 20 (2015).

  • 26. Zhang, D. Y. & Seelig, G. Dynamic DNA nanotechnology using strand-displacement reactions. Nat. Chem. 3, 103-113 (2011).

  • 27. Lee, J. H. et al. Highly Multiplexed Subcellular RNA Sequencing in Situ. Science (80-.). 343, 1360-1363 (2014).

  • 28. Ke, R. et al. In situ sequencing for RNA analysis in preserved tissue and cells. Nat. Methods 10, 857-60 (2013).

  • 29. Shah, S. et al. Single-molecule RNA detection at depth via hybridization chain reaction and tissue hydrogel embedding and clearing. Development In Review, (2016).

  • 30. Bruchez, M. et al. Semiconductor nanocrystals as fluorescent biological labels. Science 281, 2013-6 (1998).

  • 31. Fouz, M. F. et al. Bright Fluorescent Nanotags from Bottlebrush Polymers with DNA-Tipped Bristles. ACS Cent. Sci. 1, 431-438 (2015).

  • 32. Steward, O., Wallace, C. S., Lyford, G. L. & Worley, P. F. Synaptic activation causes the mRNA for the leg Arc to localize selectively near activated postsynaptic sites on dendrites. Neuron 21, 741-751 (1998).

  • 33. Buckley, P. T. et al. Cytoplasmic Intron Sequence-Retaining Transcripts Can Be Dendritically Targeted via ID Element Retrotransposons. Neuron 69, 877-884 (2011).

  • 34. Steward, 0. & Schuman, E. M. Compartmentalized synthesis and degradation of proteins in neurons. Neuron 40, 347-359 (2003).

  • 35. Buxbaum, A. R., Wu, B. & Singer, R. H. Single -Actin mRNA Detection in Neurons Reveals a Mechanism for Regulating Its Translatability. Science (80-.). 343, 419-422 (2014).

  • 36. Jung, H., Yoon, B. C. & Holt, C. E. Axonal mRNA localization and local protein synthesis in nervous system assembly, maintenance and repair. Nat. Rev. Neurosci. 13, 308-24 (2012).

  • 37. Raj, A. & Tyagi, S. Detection of individual endogenous RNA transcripts in situ using multiple singly labeled probes. Methods in enzymology 472, (Elsevier Inc., 2010).

  • 38. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676-82 (2012).

  • 39. Thévenaz, P., Ruttimann, U. E. & Unser, M. A pyramid approach to subpixel registration based on intensity. IEEE Trans. Image Process. 7,27-41 (1998).



While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims
  • 1. A method for in situ genomic and transcriptomic assessment of target nucleic acids present in a biological sample comprising the steps of: a) treating the biological sample with a small molecule linker capable of linking to at least one target nucleic acid and to a swellable material;b) embedding the biological sample wherein the small molecule linker is bound to the at least one target nucleic acid in the biological sample and to the swellable material;c) subjecting the biological sample to a physical disruption method;d) swelling the swellable material to form an expanded biological sample;e) providing at least one oligonucleotide complementary to the at least one target nucleic acid, wherein the at least one oligonucleotide hybridizes to the at least one target nucleic acid; andf) genomically or transcriptomically assessing the expanded biological sample.
  • 2. The method according to claim 1, wherein the small molecule linkers are attached to target nucleic acids via a chemical reactive group capable of covalently binding the target nucleic acid.
  • 3. The method according to claim 1, wherein the small molecule linker is labeled.
  • 4. The method according to claim 1, wherein the at least one oligonucleotide is labeled.
  • 5. The method according to claim 1, wherein embedding the biological sample in a swellable material comprises permeating the biological sample with a composition comprising precursors of a swellable polymer and forming a swellable polymer in situ.
  • 6. The method according to claim 1, wherein the at least one target nucleic acid is anchored to the swellable material.
  • 7. The method according to claim 1, wherein the physical disruption method is an enzymatic digestion.
  • 8. The method according to claim 1, wherein the target nucleic acids are DNA and/or RNA.
  • 9. The method according to claim 3, wherein the expanded biological sample expresses one or more labeled target nucleic acids.
  • 10. The method according to claim 1, further comprising the additional step of buffering the expanded sample.
  • 11. The method according to claim 10, further comprising the additional step of re-embedding the buffered expanded biological sample in a non-swellable material.
  • 12. The method according to claim 11, further comprising the step of removing the at least one oligonucleotide complementary to the at least one target nucleic acid.
  • 13. The method according to claim 12, wherein the steps of providing at least one oligonucleotide, genomically or transcriptomically assessing the expanded biological sample and removing the at least one oligonucleotide are repeated so as to allow serial or sequential genomic or transcriptomic assessments of the expanded biological sample.
  • 14. The method of claim 12, wherein removing the at least one oligonucleotide which is hybridized to the at least one target nucleic acid comprises formamide and high temperatures.
  • 15. The method of claim 12, wherein removing the at least one oligonucleotide which is hybridized to the at least one target nucleic acid comprises endonucleases that specifically digest the at least one oligonucleotide.
RELATED APPLICATION

This application claims the benefit of U.S. Provisional Application Ser. No. 62/202,421, filed Aug. 7, 2015, the contents of which are incorporated herein by reference in its entirety.

GOVERNMENT SUPPORT

This invention was made with government support under 5-DPI-NS087724 awarded by NIH, Hertz Foundation, ODGE Lemelson & Viterbi, 5-DPI-N S087724 awarded by NIH and NSF. The government has certain rights in the invention.

Provisional Applications (1)
Number Date Country
62202421 Aug 2015 US