Tissue Procurement
All human tissue procurement procedures were approved by the Institutional Review Boards at the University of Texas at Dallas. Human DRGs were procured from organ donors through a collaboration with the Southwest Transplant Alliance. DRGs and other nervous tissues (sympathetic chain ganglia) were recovered using a ventral approach as previously described 34,62. Upon removal from the body, DRGs were prepared three different ways. One, DRGs used for histology and sequencing work were frozen in pulverized dry ice, transferred into prechilled epitubes, and stored in a -80°C freezer. Two, DRGs used for neuronal dissociation were placed into freshly prepared artificial cerebral spinal fluid (aCSF) over ice. Three, DRGs used for transmission electron microscopy were placed into 4% paraformaldehyde for 48 hours at 4°C and then transferred to 1X Phosphate Buffered Saline (PBS) and shipped on ice to the University of Adelaide in Australia. A detailed protocol of the procurement process, including recipes for aCSF, can be found on protocols.io 34.
Donor medical history was provided by the Southwest Transplant Alliance and Anabios and includes medical details from the donor’s family members and hospital records. Donor demographics, medical history, and DRG level details (majority are lumbar DRGs) are provided in Supplementary Table 1. The frozen DRGs were gradually embedded in OCT in a cryomold by adding small volumes of OCT over dry ice to avoid thawing. Tissues were sectioned on a cryostat and utilized for histology and spatial transcriptomics. After sectioning, the remaining tissue blocks were wrapped in tin foil, and then returned to the -80°C freezer for future use.
Hematoxylin and Eosin staining, imaging, and analysis
A single DRG from 90 organ donors was sectioned at 20μm onto SuperFrost Plus charged slides (Fisher Scientific; Cat 12-550-15). The donors were randomly selected from our tissue bank as part of routine tissue morphology checking as part of our quality control process. Lumbar DRGs were preferentially selected when available in our tissue bank, but in some cases lower thoracic DRGs were used. The DRG levels that were assessed are indicated in Supplementary Table 1. Sections were only briefly thawed in order to adhere to the slide but were immediately returned to the -20°C cryostat chamber until completion of sectioning. The slides were removed from the cryostat and immediately immersed in 10% formalin (Fisher Scientific; Cat 23-245684) for 15 minutes. The tissues were then sequentially dehydrated in 50% ethanol (5 min; Fisher Scientific; Cat 04-355-223), 70% ethanol (5 min), and two times in 100% ethanol (5 min) at room temperature. The slides were air dried briefly, and then each section was covered with isopropanol (Sigma; L9516) and incubated for 1 minute at room temperature. The excess isopropanol was removed, and the slides were allowed to air dry again briefly (< 5 minutes). Hematoxylin (Sigma; MHS16) was applied to each tissue section until covered and incubated for 7 minutes at room temperature. The excess Hematoxylin was removed by tapping, and the slides were immersed 30 times in ultrapure water (dipping into the water). Bluing Buffer (Agilent; CS70230-2) was applied to each tissue section until covered and incubated for 2 minutes at room temperature. The excess Bluing Buffer was removed by tapping, and the slides were immersed 5X in ultrapure water (dipping into the water). Eosin mix (1:10 of 0.45M Tris Acetic Acid Buffer to Eosin (Sigma; HT110216)) was applied to each tissue section until covered and incubated for 1 minute at room temperature. The excess Eosin mix was removed by tapping, and the slides were immersed 15X in ultrapure water (dipping into the water). The excess water was removed from the slide using a tapping motion and Kim wipe. The slides were allowed to completely air dry before being coverslipped with Prolong Gold Antifade (Fisher Scientific; Cat P36930).
DRG sections were mosaically imaged at 10X using default brightfield settings on an Olympus vs120 slide scanner. The raw images were opened in CellSens (Olympus; v1.18) and qualitatively scored for the presence of Nageotte nodules throughout the entire DRG section. A qualitative scoring system (ranked from high to very low) was developed by comparing sections to one another and noting the extremes: DRG sections that had an abundance of Nageotte nodules (high) versus those with very little-to-none (very low). The person analyzing was blinded to the donor’s demographics and medical history. Once each section was scored, the medical information from each donor was probed and grouped into categories based on diagnoses of diabetes or other pain conditions. The diabetes diagnosis is known for each donor as insulin is monitored while the donors are on life support. There were 5 categories of donors: diabetics with indices for peripheral neuropathy (medical history statements of having peripheral neuropathy and/or amputation), diabetics taking analgesics (analgesic(s) usage indicated in medical history with no obvious signs of drug abuse/addiction), diabetics not taking analgesics (no history of taking analgesics, drug abuse/addiction included), non-diabetics (no DRG-affiliated pain condition, and no diabetes), non-diabetics with other pain conditions (fibromyalgia, arthritis, neuropathy, back pain as indicated in medical history). The donors included in each category are indicated in Supplemental Table 1.
Pre-mounted section Immunofluorescence staining, imaging, and analysis
3-4 20μm tissue sections (technical replicates) were acquired from each DRG (sample sizes indicated in figure captions) and placed onto SuperFrost Plus charged slides (Fisher Scientific; Cat 12-550-15). Slides were removed from the cryostat and immediately transferred to cold 10% formalin (pH 7.4) for 15 minutes. The tissues were then dehydrated in 50% ethanol (5 min), 70% ethanol (5 min), 100% ethanol (5 min), 100% ethanol (5 min) at room temperature. The slides were air dried briefly and then boundaries were drawn around each section using a hydrophobic pen (ImmEdge PAP pen, Vector Labs). When hydrophobic boundaries had dried, the slides were submerged in blocking buffer (10% Normal Goat Serum, 0.3% Triton-X 100 in 1X PBS) for 1 hour at room temperature. Slides were then rinsed in 1X PBS, placed in a light-protected humidity-controlled tray and incubated in primary antibody diluted in blocking buffer overnight at 4°C. A list of all primary and secondary antibodies is shown in Supplementary Table 5. The next day, slides were washed in 1X PBS and then incubated in their respective secondary antibody (1:2000) with DAPI (1:5000; Cayman Chemical; Cat # 14285) diluted in blocking buffer for 1 hour at room temperature. The sections were washed in 1X PBS and then covered with True Black (diluted at 1:20 in 70% Ethanol; Biotium; 23014), a blocker of lipofuscin, for 1 minute. Sections were then rinsed vigorously with ultrapure water, and then washed in 1X PBS. The slides were then air dried and coverslipped with Prolong Gold Antifade reagent. A negative control consisting of 1 section from each DRG was processed in every immunohistochemistry experiment and was exposed to all of the same reagents except for primary antibody.
DRG sections were imaged on a vs120 slide scanner (Evident Scientific) or an FV3000 or FV4000 confocal microscope (Evident Scientific) at 10X, 20X, 40X, 60X, or 100X magnification as indicated in the figure captions. The acquisition parameters were set based on guidelines for the vs120, FV3000, and FV4000 provided by Evident Scientific. The raw image files were brightened and contrasted in Olympus CellSens software (v1.18) for display purposes. For quantification and DPN vs non-diabetic comparisons experiments, all acquisition and brightness/contrast adjustment parameters were kept the same in order to make direct comparisons between samples.
For the Nageotte nodule quantification experiment (GFAP + Peripherin IHC), all neurons with a visible cytoplasm (peripherin+) were counted and all of the Nageotte nodules were counted (cluster of DAPI+ nuclei and GFAP+ signal) in Olympus CellSens (v1.18). The percentage of neurons with a Nageotte nodule morphology was calculated by dividing the Nageotte nodule counts by the total neuronal population (sum of Nageotte nodules and neurons) and multiplying by 100. Three 20X mosaic sections (vs120) were analyzed per donor, and then the final percentages from each section were averaged for each donor.
For peripherin fiber density analysis, a single 10X confocal image (FV3000) was acquired for each section of DRG (three sections total from each DRG, 3 images/donor). A single negative control DRG section from each donor (exposed to all reagents except for primary antibody) was imaged with the same settings. The neuron-rich area of the DRG was manually outlined in Olympus CellSens (v1.18) and its area was provided by the software. The peripherin signal within the neuron-rich area was autodetected using the Count and Measure feature in Olympus CellSens (v1.18). The peripherin signal within the soma of the neurons was then manually removed in the software. The remaining area of peripherin signal (axonal only) was provided by the CellSens software (v1.18) and divided by the neuron-rich area for each section and then averaged across all sections for each DRG.
For the p-eIF4E experiment analysis, a mosaically tiled 20X image (Evident Scientific, vs120) was acquired for each section of DRG (three sections total from each DRG, 3 images/donor). A single negative control DRG section from each donor (exposed to all reagents except for primary antibody) was imaged with the same settings. The cell body of all neurons within the field of view were manually outlined using the Closed Polygon tool in Olympus CellSens (v1.18) and the software output the mean fluorescence intensity of the p-eIF4E signal within the ROI. This was performed on both the experimental and negative control sections. For each donor, the final p-eIF4E mean fluorescence intensity value was corrected by subtracting the mean fluorescence intensity value of the negative control.
Filament Tracing and Free-floating Immunofluorescence staining
A DPN donor with the highest Nageotte score (Donor #6) was selected for filament tracing in order to grant the highest likelihood of tracing Nageotte nodule fibers to their point of origin. For free-floating immunofluorescence staining, 50 μm DRG sections were acquired on a cryostat and then immediately submerged in 10% formalin (pH 7.4) in a 24-well plate. The sections were fixed for 15 minutes, and then washed in an adjacent well containing 1X PBS. The sections were then transferred to a well containing blocking solution (10% Normal Goat Serum, 0.3% Triton-X 100 in 1X PBS) for 1 hour at room temperature before being transferred to primary antibody (peripherin, Supplementary Table 5) diluted in blocking solution over night at 4°C.
The next day, the sections were washed in 1X PBS, and then placed into a well containing secondary antibody (1:2000, Supplementary Table 5) with DAPI (1:5000) diluted in blocking buffer for 1 hour at room temperature while being shielded from light. The sections were then washed in 1X PBS, mounted onto slides, and treated with True Black (diluted at 1:20 in 70% Ethanol; Biotium; 23014) for 1 minute. Sections were then rinsed vigorously with ultrapure water, and then washed in 1X PBS. The slides were then air dried and coverslipped with Prolong Gold Antifade reagent.
60X z-stack images with optimal z-slices (0.3 μm) of the entire z plane were acquired on an FV4000 confocal microscope (Evident Scientific). The images were loaded into Imaris (v10), converted to IMS files, and then the peripherin signal was traced using the semi-automatic filament tracing tool. Starting points were manually selected at fibers within the Nageotte nodules, and at a neighboring neuron’s soma. The fibers were traced using the semi-automatic filament path finding tool which path finds continuous peripherin signal through the z-plane. Only fibers that originated from the Nageotte nodule arborizations or the neuronal soma and had paths that were continuously autodetected through the z-plane were traced. Other axons in the image that were not traced were either discontinuous or did not originate from the designated starting points.
RNAscope in situ hybridization staining, imaging, and analysis
DRGs were sectioned at 20μm onto SuperFrost Plus charged slides (Fisher Scientific; Cat 12-550-15). Sections were only briefly thawed in order to adhere to the slide but were immediately returned to the -20°C cryostat chamber until completion of sectioning. The slides were removed from the cryostat and immediately immersed in cold (4°C) 10% formalin (Fisher Scientific; Cat 23-245684) for 15 minutes. The tissues were then sequentially dehydrated in 50% ethanol (5 min; Fisher Scientific; Cat 04-355-223), 70% ethanol (5 min), and two times in 100% ethanol (5 min) at room temperature. The slides were air dried briefly and then boundaries were drawn around each section using a hydrophobic pen (ImmEdge PAP pen; Vector Labs). Once the hydrophobic boundaries had dried, the slides were immediately processed for RNAscope in situ hybridization.
RNAscope in situ hybridization multiplex version 2 (Advanced Cell Diagnostics; Cat 323100) was conducted on human DRGs using the fresh frozen protocol as described by ACD (acdbio; manual # 323100-USM with rev date: 02272019). Hydrogen Peroxide (ACD; Cat 322381) was applied to each section until fully covered and incubated for 10 minutes at room temperature. The slides were then washed in distilled water and then were incubated one at a time in Protease III reagent (ACD; Cat 322381) for 10 seconds at room temperature. The protease incubation time was optimized as recommended by ACD for the tissue and specific lot of Protease reagent. Slides were washed briefly in 1X phosphate buffered saline (PBS, pH 7.4) at room temperature. Each slide was then placed in a prewarmed humidity control tray (ACD; Cat 321710) containing dampened filter paper (ThermoFisher Scientific; Cat 84784) and a mixture of Channel 1, Channel 2, and Channel 3 probes (50:1:1 dilution as directed by ACD due to stock concentrations) was pipetted onto each section until fully covered. This was performed one slide at a time to avoid liquid evaporation and section drying. The humidity control tray containing the slides was placed in a HybEZ oven (ACD; Cat 321710) for 2 hours at 40°C. A table of all probes used is shown in Supplementary Table 5. Following probe incubation, the slides were washed two times in 1X RNAscope wash buffer (ACD; Cat 310091) and then placed in 5X SSC buffer (Sigma; Cat S6639) over night at room temperature.
The following morning, the slides were washed two times in 1X RNAscope wash buffer (ACD; Cat 310091) and placed in the 40°C oven for 30 minutes after submersion in AMP-1 reagent. Washes and amplification were repeated using AMP-2 and AMP-3 reagents with a 30-minute and 15-minute incubation period, respectively. HRP-C1 reagent was applied to all sections and then incubated in the oven at 40°C for 15 minutes. The slides were then washed in 1X RNAscope wash buffer (ACD; Cat 310091). TSA Plus Akoya Dyes in Fluorescin, Cyanine-3, and Cyanine-5 (Akoya; NEL741001KT, NEL744001KT, NEL745001KT) were prepared at 1:1000 in TSA buffer (ACD; Cat 322809). The Akoya dye assigned to Channel 1 probe was applied to each section until fully covered and incubated for 30 minutes in the 40°C oven. The slides were washed and then covered in HRP blocker (ACD; Cat 323110) for 15 minutes at 40°C. The slides were washed again and then the same steps were repeated using HRP-C2 and HRP-C3 reagents with their assigned Akoya dye. DAPI (ACD; Cat 323110) was applied to each section for 1 minute at room temperature and then washed in 1X PBS (pH 7.4) before being washed, air dried, and cover-slipped (Globe Scientific; Cat 1415-15) with Prolong Gold Antifade mounting medium (Fisher Scientific; Cat P36930).
A positive and negative control was run on a single section from each DRG for every RNAscope experiment. The positive control probe cocktail (Supplementary Table 5) contains probes for high, medium and low-expressing mRNAs that are present in all cells (ubiquitin C > Peptidyl-prolyl cis-trans isomerase B > DNA-directed RNA polymerase II subunit RPB1) and allows us to gauge tissue quality and experiment conditions. All tissues showed robust signal for all 3 positive control probes. A negative control probe against the bacterial DapB gene (Supplementary Table 5) was used to check for lipofuscin and background label.
DRG sections were imaged on an FV3000 or FV4000 confocal microscope (Evident Scientific) at 10X, 20X, 40X or 60X magnification as indicated in the figure captions. The acquisition parameters were set based on guidelines for the FV3000 and FV4000 provided by Evident Scientific. The raw image files were analyzed in CellSens (Olympus; v1.18). The True black lipofuscin quencher (used in Immunofluorescence) is not compatible with RNAscope. Large globular structures and/or signal that auto fluoresced in all channels (particularly brightest in 488 and 555 wavelengths) was considered to be background lipofuscin and was not analyzed. Aside from adjusting brightness/contrast, we performed no digital image processing to subtract background.
Transmission electron microscopy tissue preparation, staining, and imaging
Upon receipt from UTDallas, the DRGs were cut into smaller pieces of about 1 mm3 in size, fixed in 2.5% glutaraldehyde 2.5 % in phosphate buffer, pH 7.4 at 4°C for 24 hours, washed in PBS, and transferred into 2% aqueous osmium tetroxide solution for 1 hour. The samples were then dehydrated in a graded series of ethanol and embedded in TAAB epon araldite embedding medium at 60°C for 48 hours. Ultrathin sections of 70-90 nm thickness were cut using a ultramicrotome (Leica), stained with 4% uranyl acetate and Reynolds lead citrate for 8 minutes and examined using an electron microscope (FEI Tecnai 120kV Spirit). Images were captured using an AMT Camera with AMT_V7.0.1 software.
Dissociated neuronal cultures and Immunocytochemistry
After procurement, DRGs (donors 91 and 92, Supplemental Table 1) in aCSF were transported to the lab over ice (~30 minutes). The DRGs were trimmed of excess connective tissue, fat, and nerve roots to expose the DRG bulb. The DRG bulb was then cut into 3mm sections and placed in 5mL of pre-warmed digestion enzyme containing 1 mg/mL of Stemzyme I (Worthington Biochemical, LS004106), 0.1 mg/mL of DNAse I (Worthington Biochemical, LS002139), and 10ng/mL of recombinant human β-NGF (R&D Systems, 256-GF) in HBSS without calcium and magnesium (Thermo Scientific, 14170-112). The tubes were placed in a 37° C shaking water bath until the DRG sections dissociated (4-10 hours). The solution was then filtered through a 100µm mesh strainer. The resultant cell suspension was then gently added to a 15mL tube containing 3mL of 10% Bovine Serum Albumin (Biopharm, 71-040) in HBSS. The tubes were then centrifuged at 900g for 5 min at room temperature. The supernatant was aspirated, and the pellet was resuspended in prewarmed BrainPhys® media (Stemcell technologies, 05790) containing 1% penicillin/streptomycin (Thermo Fisher Scientific, 15070063), 1% Glutamax (Thermo Scientific, 35050061), 2% NeuroCult SM1 (Stemcell technologies, 05711), 2% HyClone™ Fetal Bovine Serum (Thermo Fisher Scientific, SH3008803IR), 1% N-2 (Thermo Scientific, 17502048), 0.1% 5-Fluoro-2′-deoxyuridine (FRDU, Sigma-Aldrich, F0503), and 10ng/ml of β-NGF. Cells were plated in a 24 well plate containing 12mm coverslips which were pre-coated with 0.1mg/mL of poly-D-lysine at a seeding density of 100 neurons per well. Cells were incubated at 37°C and 5% CO2 for 3 hours to allow for adherence. Following neuron adherence, wells were flooded with 1mL of prewarmed media, and half media changes were performed every other day.
On DIV 3 (donor 91) and DIV 5 (donor 92), cells were washed with 1X PBS and fixed with 10% formalin for 10 min at room temperature. Cells were then washed 3 times with 1X PBS and blocked with 10% normal goat serum in PBS for 1 hour at room temperature. Cells were then permeabilized with 10% Normal Goat Serum and 0.3% Triton X in PBS for 30 min at room temperature. To label neurons, cells were incubated with peripherin (1:1000, Supplemental Table 5) diluted in blocking buffer overnight at 4°C. The next day cells were washed 3 times with 1X PBS and incubated with a goat anti-chicken 488 secondary antibody (1:2000) and DAPI (1:5000, Cayman Chemical, 14285) diluted in blocking buffer for 1 hour at room temperature. Cells were washed with 1X PBS and then covered with True Black (diluted at 1:20 in 70% Ethanol; Biotium; 23014), a blocker of lipofuscin, for 1 minute. The cells were washed again in 1X PBS. Coverslips were lifted out of the 24 well plate and mounted onto glass slides with Prolong Gold Antifade reagent (Fisher Scientific, P36930). A negative control coverslip was processed similarly in each experiment but was not exposed to the primary antibody. All images were taken on an Olympus FV3000 confocal microscope at the University of Texas at Dallas.
Graphing and Statistical Analyses
Graphs were generated using GraphPad Prism version 8.4.3 (GraphPad Software, Inc. San Diego, CA USA). Statistical analyses (either t-test or one-way ANOVA) were run in GraphPad Prism as indicated in the figure captions. Sample size is also indicated on the graphs and/or figure captions. Graphical figures were made in Biorender. Box-and-whisker plots were generated with the R ggplot2 package to visualize expression of genes in Nageotte nodule barcodes. Dot/bubble plots were generated with the DotPlot() function in Seurat (v5.0.3) to visualize expression of genes in the VISIUM data.
Spatial transcriptomics
VISIUM tissue optimization and spatial gene expression protocols were followed exactly as described by 10x Genomics (https://www.10xgenomics.com/) using Haematoxylin and Eosin as the counterstain. Optimal permeabilization time was obtained at 12 min incubation with permeabilization enzyme 41. Imaging was conducted on an Olympus vs120 slide scanner. mRNA library preparation and sequencing (Illumina Novaseq, NextSeq 500, Nextseq 2000) were done at the Genome Center in the University of Texas at Dallas Research Core Facilities. DRGs from 6 DPN donors were used and processed in three separate VISIUM experiments (donors 1-4, 6, 14). The donor demographic information is provided in Supplementary Table 1.
Raw sequencing files were processed with the 10X Genomics SpaceRanger pipeline (versions 1.1.0, 1.3.0, and 2.0.0) to generate count matrices of gene expression per VISIUM barcode for 16 DRG sections from 6 donors. VISIUM sections were examined on Loupe Browser (10X Genomics) to select for barcodes that overlapped Nageotte nodules and nearby neurons (one barcode away from Nageotte nodule). Any Nageotte nodule barcodes that also overlapped with a neuronal soma were excluded. 1094 Nageotte nodule barcodes and 1087 nearby neuronal barcodes were selected. Spatial sequencing metrics can be found in Supplementary Table 6.
Spatial RNA-seq analysis
Gene expression analysis of the barcodes was done with R (version 4.3.3) and consisted of quantification of gene expression in Nageotte Nodules, Enrichr analysis of top genes, spatial deconvolution, clustering of Nageotte nodule barcodes, and interactome analysis.
Analysis of Gene Expression and Top Genes in Nageotte Nodules
The raw counts from all nodule barcodes were pseudo bulked and normalized to library size, to generate counts per million (CPM) values for each gene in the Nageotte nodule barcodes. As the 10X VISIUM assay library preparation does not have a gene length bias, the CPM values were deemed sufficient to allow comparisons between genes. A recently published dataset from our group of single-nuclei RNA-sequencing from human DRGs 63 was used with Seurat’s FindAllMarkers() to determine a list of neuronally enriched genes using the following criteria: 1) a difference of at least 0.1 when subtracting the percentage of non-neuronal cells that express the gene from the percentage of neurons that express the gene, 2) at least 2-fold increase between the number of non-neurons that express the gene and the number of neurons that express the gene, 3) adj. p-value <0.05 in neurons and >0.05 in every other cell type. This list was intersected with the filtered Nageotte nodule gene expression table to generate a table with 609 neuronally enriched genes, and a table with 15651 non-enriched genes.
Enrichr Analysis
The complete gene expression table was filtered to include only protein-coding genes and exclude genes from the mitochondrial chromosome and those that code for ribosomal proteins. After filtering, the gene expression values were re-normalized to sum one million. The top 300 genes were analyzed with Enrichr to determine ontology terms enriched in the gene set in the following databases: 1) MGI Mammalian Phenotype Level 4 2021, 2) GO Molecular Function 2023, 3) GO Biological Process 2023, 4) GO Cellular Compartment 2023.
Spatial Deconvolution
All barcodes from the 16 sections with greater than 200 unique genes were processed with SONAR 64 to predict cell type proportions using a signature matrix of marker genes per cell type. The signature matrix was generated by subsetting the raw count matrix of a single-nuclei dataset of human DRG cells to include only those genes that were highly enriched in a cell type (log-fold change > 1.0, expression in > 50% of cells, and adj. p-value < 10-20). Deconvolution results were used to estimate the contribution of different cell types to the transcriptomes of barcodes with Nageotte nodules, adjacent neurons, and all other barcodes (barcodes that overlapped both nodules and neurons were excluded from all 3 categories). They were also used to predict cell type composition of the different clusters of Nageotte nodules.
Clustering of Nageotte nodule barcodes
The SCT pipeline from Seurat (version 5.0.3), followed by Harmony integration and FindNeighbors()/FindClusters(), was used to perform unsupervised clustering of the Nageotte nodule barcodes. A UMAP plot of the clustering with cells labeled by donor was used to confirm that the Nageotte barcodes did not exhibit donor-specific groupings. The cells in each cluster were pseudo bulked to obtain expression in CPM for all genes per cluster and FindMarkers() was used to determine enriched genes per cluster.
Interactome Analysis
To explore potential signaling mechanisms between Nageotte nodules and neighboring neurons, reads from neuronal barcodes adjacent to the nodules were pseudo bulked and normalized to CPM. A curated ligand-receptor database and interactome platform (https://sensoryomics.shinyapps.io/Interactome/; 65) were used to hypothesize interactions with ligands from each Nageotte nodule cluster and receptors on the neurons adjacent to nodules. In the first interactome analysis, to determine the top potential interactions per cluster, the interactions were ranked based on the sum of the ligand and receptor CPM expression values. In the second analysis, to determine interactions that may be more prominent for each cluster, only ligand genes that were differentially expressed (adj. p-value <0.05) in each cluster were used. Lastly, we also observed potential interactions with ligands from the surrounding neurons and receptors on the nodules (using only those receptor genes that were enriched in a cluster with adj. p-value <0.05). Ligand and receptor genes were also labeled with the protein class of their gene product using the PANTHER database 66.