You are on page 1of 17

The Journal of Neuroscience, November 22, 2023 • 43(47):7929–7945 • 7929

Development/Plasticity/Repair

Anatomical Diversity of the Adult Corticospinal Tract


Revealed by Single-Cell Transcriptional Profiling
Noa Golan,1,2 Daniel Ehrlich,1,4 James Bonanno,1,2 Rory F. O’Brien,2 Matias Murillo,1,2 Sierra D. Kauer,2
Neal Ravindra,5,6 David Van Dijk,5,6 and William B. Cafferty2,3
1
Interdepartmental Neuroscience Program, Yale University School, New Haven, Connecticut 06511, 2Department of Neurology, Yale University
School, New Haven, Connecticut 06511, 3Department of Neuroscience, Yale University School, New Haven, Connecticut 06511, 4Department of
Psychiatry, Yale University School, New Haven, Connecticut 06511, 5Department of Internal Medicine, Yale University School, New Haven,
Connecticut 06511, and 6Department of Computer Science, Yale University School, New Haven, Connecticut 06511

The corticospinal tract (CST) forms a central part of the voluntary motor apparatus in all mammals. Thus, injury, disease,
and subsequent degeneration within this pathway result in chronic irreversible functional deficits. Current strategies to repair
the damaged CST are suboptimal in part because of underexplored molecular heterogeneity within the adult tract. Here, we
combine spinal retrograde CST tracing with single-cell RNA sequencing (scRNAseq) in adult male and female mice to index
corticospinal neuron (CSN) subtypes that differentially innervate the forelimb and hindlimb. We exploit publicly available
datasets to confer anatomic specialization among CSNs and show that CSNs segregate not only along the forelimb and hind-
limb axis but also by supraspinal axon collateralization. These anatomically defined transcriptional data allow us to use
machine learning tools to build classifiers that discriminate between CSNs and cortical layer 2/3 and nonspinally terminating
layer 5 neurons in M1 and separately identify limb-specific CSNs. Using these tools, CSN subtypes can be differentially identi-
fied to study postnatal patterning of the CST in vivo, leveraged to screen for novel limb-specific axon growth survival and
growth activators in vitro, and ultimately exploited to repair the damaged CST after injury and disease.
Key words: Corticospinal tract; plasticity; regeneration; repair; single-cell RNA sequencing; spinal cord injury

Significance Statement
Therapeutic interventions designed to repair the damaged CST after spinal cord injury have remained functionally suboptimal in part
because of an incomplete understanding of the molecular heterogeneity among subclasses of CSNs. Here, we combine spinal retro-
grade labeling with scRNAseq and annotate a CSN index by the termination pattern of their primary axon in the cervical or lumbar
spinal cord and supraspinal collateral terminal fields. Using machine learning we have confirmed the veracity of our CSN gene lists to
train classifiers to identify CSNs among all classes of neurons in primary motor cortex to study the development, patterning, homeo-
stasis, and response to injury and disease, and ultimately target streamlined repair strategies to this critical motor pathway.

Introduction postnatally, with pioneer axons extending to spinal enlarge-


The corticospinal tract (CST) is the major descending motor ments, followed by periods of exuberant gray matter terminal
pathway responsible for driving fine coordinated movement in arborization and pruning via activity-dependent mechanisms
mammals (Lemon, 2008). Corticospinal neurons (CSNs) in layer during a protracted critical period that sculpts a mature motor
(L)5b (L5) of sensorimotor cortex project their axons through pathway (Gianino et al., 1999; Martin, 2005). Complex wiring of
the internal capsule, decussate in the medulla, and innervate the mature CST, together with its central role in voluntary and
fine motor control, means that damage leads to significant and
every spinal segment along the neuroaxis. The CST is wired
lasting functional impairments. Efforts to repair the damaged
CST have focused on either nullifying the effects of the axon
Received Apr. 26, 2022; revised July 28, 2023; accepted Aug. 1, 2023.
growth inhibitory environment of the mature CNS or recapitu-
Author contributions: N.G., D.E., and W.B.C. designed research; N.G., D.E., J.B., R.F.O., M.M., S.D.K., N.R.,
and W.B.C. performed research; N.G., D.E., M.M., N.R., D.V., and W.B.C. contributed unpublished reagents/
lating cell autonomous developmental mechanisms (Schwab and
analytic tools; N.G., D.E., J.B., R.F.O., M.M., S.D.K., and W.B.C. analyzed data; and N.G., D.E., and W.B.C. wrote Strittmatter, 2014; Gutilla and Steward, 2016; He and Jin, 2016;
the paper. Hilton and Bradke, 2017; Bradbury and Burnside, 2019; Fawcett,
This work was supported by National Institutes of Health–National Institute of Neurological Disorders and 2020). Although strides have been made in stimulating axotom-
Stroke Grants R01NS09593001 and R21NS108053.
ized CSNs to regenerate or intact CSNs to undergo plasticity af-
The authors declare no competing financial interests.
Correspondence should be addressed to William B. Cafferty at william.cafferty@yale.edu.
ter injury, recovery of significant motor function remains to be
https://doi.org/10.1523/JNEUROSCI.0811-22.2023 realized (Park et al., 2008; Fink et al., 2017; Kauer et al., 2022).
Copyright © 2023 the authors The inefficacy of current interventions could in part be because
7930 • J. Neurosci., November 22, 2023 • 43(47):7929–7945 Golan et al. · Transcriptional Index of the Adult Corticospinal Tract

of an incomplete understanding of the molecular heterogeneity Intraspinal retrograde AAV injections for scRNAseq. To complete
among CSNs. For instance, in vitro and in vivo screening approaches retrograde labeling of cervical-projecting CSNs, adult mice (2–3 months
designed to identify axon growth modulators may overlook mo- old, n ¼ 19, 10 males, 9 females) were anesthetized with ketamine
lecular heterogeneity within cortical neuron subclasses including (100 mg/kg; Covetrus) and xylazine (15 mg/kg; Covetrus) and placed
CSNs, consequently diluting the most potent candidates among in a stereotaxic frame (Stoelting). An incision was made over the cer-
differentially sensitive neuronal subtypes (Blackmore et al., 2010; vical enlargement and the C5–C8 vertebrae revealed by blunt dissec-
tion of overlying muscle. A bilateral laminectomy was performed to
Buchser et al., 2010; Sekine et al., 2018).
expose the underlying C5–C8 spinal cord, and a small incision was
Recent data from our laboratory supports differential sensitiv-
made in the dura mater. The tip of a pulled glass capillary tube
ity of CSN subdivisions. We showed that novel proplasticity attached to a 5 ml Hamilton syringe loaded into a Micro4 infusion de-
factors identified in intact CSNs undergoing functional plasticity vice (World Precision Instruments) was slowly inserted stereotaxi-
after unilateral pyramidotomy (PyX) stimulates growth of lesioned cally to a depth of 500 mm into the C6 level of the spinal cord and
and intact forelimb (FL) but not hindlimb (HL) CST axons (Fink ;600 mm lateral from the midline. Thirty seconds after the introduc-
et al., 2017; Kauer et al., 2022). Proaxon growth candidates Lppr1 tion of the capillary tube, 100 nl of retro-AAV-CAG-GFP (catalog
and Inpp5k were identified via retrograde labeling of sprouting #37825-AAVrg, Addgene) was infused into the spinal cord over
CSNs in the cervical spinal cord, suggesting that retrograde label- 2 min. The tip was left in situ for an additional 30 s before removal.
ing of plastic CSNs from the lumbar cord may identify a separate This procedure was completed seven additional times bilaterally at
set of factors. the same coordinates at evenly spaced injection sites between C6 and C7
resulting in a total infusion of 800 nl (100 nl over eight sites). Muscle
Single-cell RNA sequencing (scRNAseq) approaches offer the
layers were sutured with VICRYL (Ethicon) and skin with monofilament
sensitivity to identify transcriptional heterogeneity among CSNs. suture. All animals received postsurgical antibiotics (Ampicillin, 100 mg/
Indeed, previous studies have revealed the rich phenotypic diver- kg subcutaneously) and analgesia (Buprenorphine, 0.05 mg/kg subcuta-
sity among CNS cell types (Arlotta et al., 2005; Macosko et al., neously) for 2 d after lesion. All animals recovered uneventfully.
2015; Saunders et al., 2018; Tasic et al., 2018; Milich et al., 2021; To complete retrograde labeling of lumbar-projecting CSNs, adult
Yao et al., 2020; Muñoz-Castañeda et al., 2021). Central to the mice (2–3 months old, n ¼ 19, 10 males, 9 females) were anesthetized
utility of exploiting transcriptional indices is understanding the with ketamine (100 mg/kg) and xylazine (15 mg/kg) and placed in a cus-
functional or anatomic role of potential subclasses of cells. For tom-built spine stabilizer (Farrar et al., 2012). Using the last rib as a
instance, scRNAseq of corticocortical projection neurons showed landmark, an incision was made, and a bilateral laminectomy was per-
that although these cells form a single genetic cluster, their ana- formed to expose L4–L5 spinal cord. The tip of a pulled glass capillary
tube attached to a 5 ml Hamilton syringe loaded into a Micro4 infusion
tomically traced subdivisions have diverse gene expression (Kim
device was slowly inserted stereotaxically to a depth of 500 mm into the
et al., 2020). L4 level of the spinal cord and ;500 mm lateral from the midline. Thirty
To identify CSN transcriptional heterogeneity, we combined seconds after introduction of the capillary tube, 100 nl of retro-AAV-
retrograde adeno-associated virus (AAV) tracing from the cervi- CAG-GFP was infused into the spinal cord over 2 min. The tip was
cal and lumbar spinal cord with scRNAseq of adult CSNs. We left in situ for an additional 30 s before removal. This procedure was
used machine learning to develop classifiers that go beyond cell completed seven additional times bilaterally at the same coordinates
marker characterization and provide robust tools for unbiased at evenly spaced injection sites between L4 and L5, resulting in a total
classification of CSNs among other neurons in primary motor infusion of 800 nl (100 nl over eight sites). All animals received post-
cortex (M1) and among limb-specific CSNs. We exploited taxo- surgical antibiotics (Ampicillin, 100 mg/kg subcutaneously) and anal-
nomic definitions of projection neuron cell types in M1 to show gesia (Buprenorphine, 0.05 mg/kg subcutaneously) for 2 d postlesion.
that CSNs can be anatomically and molecularly defined based on All animals recovered uneventfully.
Injections for single molecule fluorescent in situ hybridization valida-
supraspinal terminals in addition to their spinal projections.
tion of scRNAseq. For confirmation studies, adult mice (2–3 months old,
Critically, we present evidence showing that intact adult CSNs n ¼ 3/probe) received bilateral injections of retro-AAV-CAG-GFP into
express a constellation of genes previously shown to be unique to the cervical cord and retro-AAV-CAG-tdTomato (catalog #59462-
nonspinally terminating projection neurons. Using intersectional AAVrg, Addgene) into the lumbar cord as described above for a
viral tracing, we confirm that forelimb and hindlimb CSNs total of 16 spinal injections (100 nl over eight sites in the cervical
differentially innervate supraspinal structures, further sup- enlargement, and 100 nl over eight sites in the lumbar enlargement).
porting emerging data that CSNs influence motor output All animals received postsurgical antibiotics (Ampicillin, 100 mg/kg
beyond directly modulating spinal motor circuitry (Nelson subcutaneously) and analgesia (Buprenorphine, 0.05 mg/kg subcu-
et al., 2021). We believe these sequencing data can be lever- taneously) for 2 d postlesion. All animals recovered uneventfully.
aged for in vitro and in vivo screening, targeting, and ex- Intersectional tracing of supraspinal CST terminals for BrainJ analysis.
To label the FL and HL corticospinal tract for projection analysis, two
ploitation strategies to enhance our understanding of the
groups of adult C57/blk6 mice received dual injections of pAAV-CAG-
development, patterning, housekeeping, and response to FLEX-tdTomato (catalog #28306-AAV1, Addgene) into either the cau-
injury within the CST. dal FL motor cortex (n ¼ 4; AP, between 0.0 mm and 0.5 mm rostral
to bregma; ML, between 1.0 mm and 1.3 mm lateral to bregma; four
Materials and Methods injections, 100 nl/site) or the HL motor cortex (n ¼ 4; AP, between 0.5
Mice mm and 1 mm caudal to bregma; ML, between 1.5 mm to 2.0 mm lat-
Three mouse lines were used for experiments—Rbp4 Cre mice (gift from eral to bregma; four injections, 100 nl/site), and pENN.AAV.hSyn.Cre.
David Berson, Brown University), Ai14 (The Jackson Laboratory), and WPRE.hGH (catalog #105553-AAVrg, Addgene) into either the cervical
C57BL/6J (The Jackson Laboratory). Rbp4 Cre mice were crossed with (C6/7) or lumbar spinal cord (L4/5), as described above. All animals
the cre-dependent tdTomato reporter line Ai14 to label deep-layer corti- received postsurgical antibiotics (Ampicillin, 100 mg/kg subcutaneously)
cal neurons and to confirm the veracity of our CSN classifier (CSN-C). and analgesia (Buprenorphine, 0.05 mg/kg subcutaneously) for 2 d post-
lesion. All animals recovered uneventfully.
Surgery
All procedures and postoperative care were performed in accordance Intact adult single-cell isolation for scRNAseq
with the guidelines of the Institutional Animal Use and Care Committee Adult intact cell isolation was done following our previously pub-
at Yale University. lished protocol (Golan and Cafferty, 2021). Briefly, mice that received
Golan et al. · Transcriptional Index of the Adult Corticospinal Tract J. Neurosci., November 22, 2023 • 43(47):7929–7945 • 7931

retrograde injections into either the lumbar or cervical cord were form nanoliter-scale gel beads-in-emulsions (GEMs). Each gel bead has
anesthetized with isoflurane and transcardially perfused with artificial primers containing (1) an Illumina R1 sequence [read 1 sequencing
CSF (aCSF) 10 d after retro-AAV injection. The aCSF contained the primer (R1)], (2) a 16 nt 10x barcode, (3) a 12 nt unique molecular iden-
following (in mM): 0.5 CaCl2, 25 glucose, 96 HCl, 20 HEPES, 10 MgSO4, tifier (UMI), and (4) a poly-dT primer sequence (30 nt). After dissolu-
1.25 NaH2PO4, 3 myo-inositol, 12 N-acetylcysteine, 96 NMDG, 2.5 KCl, tion of the Gel Beads in a GEM, the primers were released and mixed
25 NaHCO3, 5 sodium L-ascorbate, 3 sodium pyruvate, 0.01 taurine, 2 with cell lysate and Master Mix. Incubation of the GEMs then produced
thiourea, 13.2 trehalose, and was bubbled with carbogen gas (95% O2 barcoded, full-length cDNA from polyadenylated mRNA.
and 5% CO2; Tasic et al., 2018). The brain was dissected and submerged Post-GEM-RT cleanup, cDNA amplification, and library construction.
in ice-cold carbogenated aCSF for 3 min before being transferred to a Silane magnetic beads were used to remove leftover biochemical
brain matrix (Braintree Scientific) where four 500 mm sections through reagents and primers from the post-GEM reaction mixture. Full-length,
M1 were sliced and transferred to a Petri dish containing ice-cold carbo- barcoded cDNA was then amplified by PCR to generate sufficient mass
genated aCSF. Regions of M1 (11.0 to 0.5 mm relative to bregma) for library construction. Enzymatic fragmentation and size selection
containing labeled CSNs were dissected under a fluorescent microscope were used to optimize the cDNA amplicon size before library construc-
and transferred to a 5 ml Eppendorf tube with dissociation buffer with tion. R1 was added to the molecules during GEM incubation. Primer 5
papain. (P5), P7, a sample index, and R2 (read 2 primer sequence) were added
The dissociation buffer contained the following (in mM): 82 sodium during library construction via End Repair, A-tailing, Adaptor Ligation,
sulfate, 30 potassium sulfate, 10 HEPES, 10 glucose, and 5 magnesium and PCR. The final libraries contain the P5 and P7 primers that were
chloride (Saunders et al., 2018). Thirty minutes before dissociation, the used in Illumina bridge amplification.
dissociation buffer was warmed to 34°C, and 5 ml was added to a vial of Sequencing libraries. The Single Cell 39 protocol produces Illumina-
lyophilized papain (catalog #LK003178, Worthington Biochemical). The ready sequencing libraries. A Single Cell 39 library contains standard
solution was diluted 1:2 with additional dissociation buffer before sam- Illumina paired-end constructs that begin and end with P5 and P7. The
ple dissociation. Single Cell 39 16 bp 10x barcode and 12 bp UMI are encoded in Read 1,
The sample in dissociation buffer with papain was incubated whereas Read 2 is used to sequence the cDNA fragment (91 bp).
at 34°C for 70 min on a shaker at medium speed. After 70 min, the Minimum sequencing depth is 20,000 read pairs per cell. The RNAseq
dissociation solution with papain was replaced with a Stop buffer data is deposited in the National Center for Biotechnology Information
containing 5 ml of dissociation buffer, 5 mg ovomucoid protease in- Gene Expression Omnibus (GEO) and is accessible through the GEO
hibitor (catalog #LK003182, Worthington Biochemical) and 10 mg series.
bovine serum albumin (catalog #AB01088, AmericanBio) for 5 min
on ice. The Stop solution was then replaced with 800 ml of dissocia- Single-cell RNAseq analysis
tion buffer and triturated with Pasteur pipettes with decreasing diam- Quality control and normalization. Single-cell analyses were per-
eters (600, 300, and 150 mm) and placed on ice. To differentiate formed with Python version 3.8.5 software and the SciPy (version 1.5.2)
between intact neurons and nuclei, a cell-permeable dye was added at and NumPy (version 1.19.2) modules. After extracting the single-cell
a concentration of 1:1000 for 5 min (catalog #L34974, Invitrogen). gene expression matrix, we used the scprep (version 1.0.3) package in
The sample was then centrifuged at 300  g for 10 min at 4°C and Python to eliminate non-neurons and normalize the data (https://github.
resuspended in 500 ml of dissociation buffer. com/KrishnaswamyLab/scprep/releases). Briefly, we filtered out cells that
had a library size of ,2500 UMIs as well as cells that had mitochondrial
Fluorescence activated cell sorting gene expression in the top 10th percentile. To exclude doublets, we used
In total 44 mice were used for the scRNAseq studies, and processing of two methods. (1) We used the Python package Scrublet and removed all
tissue was completed in seven batches on different days for the following: cells that were detected as doublets (Wolock et al., 2019), and (2) we
three FL-traced cohorts (FL1, n ¼ 8, 4 female/4 male; FL2, n ¼ 6, 3 removed cells that expressed both the male-specific genes (Eif2s3y or
female/3 male; FL3, n ¼ 5, 2 female/3 male), three HL-traced cohorts Ddx3y) and female-specific genes (Xist or Tsix). After a library size nor-
(HL1, n ¼ 6, 3 female/3 male; HL2, n ¼ 7, 3 female/4 male; HL3, n ¼ 6, malization, we used the Python package MAGIC (version 3.0.0) to impute
3 female/3 male), and one naive Rbp4 cre:Ai14 (n ¼ 6 rbp4 cre:Ai14, 3 missing data (van Dijk et al., 2018). Using the MAGIC output gene
female/3 male). Fluorescent single cells were isolated from rbp4 cre:Ai14 expression matrix, we filtered out contaminating cell types (endothelial
mice and C57BL/6J mice injected with retro-AAV-CAG-GFP. Sorting cells, microglia, astrocytes, and oligodendrocytes) as well as inhibitory
was performed on the Sony SH800 Cell Sorter with a 130 mm nozzle, interneurons (via negative expression of vglut2), resulting in an approxi-
ensuring a sheath pressure and sample pressure of ,9 psi. To ensure py- mate 1% reduction from the starting neuronal population. We then
ramidal neurons were sorted, size reference beads were used to set a square-root transformed the data for further downstream analysis.
minimum diameter of 10 mm [polystyrene particles (PPS)-5 and PPS-6, Batch correction. To batch correct collected data, we used the ingest
Spherotech] for all collected cells. To exclude nonintact cells, the cells function from the Python package Scanpy (version 1.8.2). Once the data-
were sorted for the presence of the cell-permeable dye. A sample of non- sets had been integrated, a graph was built in the integrated space, and
fluorescent tissue from adjacent regions of the cortex was used as nega- MAGIC was used to impute gene expression based on the integrated
tive control to set the gate for the presence of fluorescence (GFP or graph.
tdTomato). The sample was then sorted into Eppendorf tubes containing To identify supraspinal connectivity of neurons sequenced in this
4°C dissociation buffer and immediately sequenced at the Yale Center study, we took a similar approach. Briefly, the dataset collected in Yao et
for Genome Analysis. al. (2021) was used as the reference dataset. We then used the ingest
In total 44 animals were used (4 rbp4 cre:Ai14; 16 C57BL/6J or Ai14 function in Scanpy and projected the dataset collected in this study into
injected with retro-AAV-CAG-GFP into C6/7; 24 C57BL/6J or Ai14 the manifold calculated from Yao et al. (2021). Last, we used the label-
injected with retro-AAV-CAG-GFP into L4/5). Dissection of L5 of M1 mapping output from Scanpy to identify intratelencephalic (IT), extrate-
was guided by fluorescence and was consistent across all animals. lencephalic (ET), near-project (NP), and corticothalamic (CT) cells.
Removing putatively mislabeled neurons from downstream analyses.
10x Single-cell RNAseq To conduct accurate differential expression analyses, we first needed to
Construction of 10x genomic single-cell/nuclei 39 RNA-seq libraries, identify putatively mislabeled neurons. Because of inherent inefficiencies
CITE-seq libraries, and sequencing. The single-cell protocol enables in viral labeling and fluorescence-activated cell sorting (FACS), we used
short read sequencing to deliver a scalable microfluidic platform for digi- a computational approach. We approximated the distribution for each
tal gene expression of 50010,000 individual cells per sample. of our cell populations (L5 nonspinal, L2/3, and CSNs) using a Gaussian
Gel beads-in-emulsions generation and barcoding. Single-cell suspen- kernel density estimate (KDE) using the Python package SciPy. To iden-
sion in Real Time (RT) Master Mix was loaded on the Single Cell A tify putative nonspinal nonfluorescent neurons, we calculated the likeli-
Chip and partitioned with a pool of ;750,000 barcoded gel beads to hood that each non-CSN (L5 and L2/3) came from the CSN distribution.
7932 • J. Neurosci., November 22, 2023 • 43(47):7929–7945 Golan et al. · Transcriptional Index of the Adult Corticospinal Tract

We removed putative CSNs (KDE . 50th percentile of CSN distribution) (VT1000 S, Leica), mounted on Superfrost Plus slides and processed for
from the non-CSN samples. Additionally, we used a similar approach to single-molecule fluorescent in situ hybridization (smFISH) according to
exclude a portion of L5 nonspinal neurons as they were putative L2/3. the ACD RNAScope fluorescent protocol. Briefly, sections were either im-
Differential gene expression analysis. Once we identified a pure mediately stored at 80°C until they were ready for processing, or sec-
nonspinally projecting layer 5 neuron population, we used the Python tions were postfixed in 4% PFA and stained with a single probe, Wnt7b
package diffxpy (https://github.com/theislab/diffxpy) to run a Wilcoxon (catalog #401131, Advanced Cell Diagnostics), Cacng7 (catalog #556171-
rank-sum test with a Benjamini–Hochberg correction for multiple com- C2, Advanced Cell Diagnostics), and Slc16a2 (catalog #545291, Advanced
parisons to identify differentially expressed (DE) genes between GFP1 Cell Diagnostics). Each experiment included a positive and negative con-
CSNs and nonspinal L5 cells. Using the significantly differentially trol probe to ensure validity. Before coverslipping, immunofluorescence
expressed genes (q , 0.05), we used recursive feature elimination using using primary antibodies directed against GFP (1:2000; catalog #ab13970,
sklearn.feature_selection.RFE (Pedregosa et al., 2011) to identify the top Abcam), and mCherry (1:2000; catalog #ab167453, Abcam) and detected
10 genes that best differentiate between CSNs and non-CSNs. Differentially with secondary antibodies Alexa Fluor 488 and 568, (1:500, 1:500; catalog
expressed genes were then used for QIAGEN Ingenuity Pathway Analysis #A11011, Life Technologies) was used to visualize traced CSNs.
(IPA; Krämer et al., 2014; https://www.qiagen.com/us/products/discovery-
and-translational-research/next-generation-sequencing/informatics-and- smFISH of adult cortical tissue analysis
data/interpretation-content-databases/ingenuity-pathway-analysis) for All analyses were performed by an experimenter blinded to probe iden-
identification of commonly regulated pathways and for building support tity. For quantification of smFISH results, images were taken at 40 on
machine vectors. a Leica SP8 confocal microscope. Three images were taken per hemi-
Linear support vector machine classification. To classify cells as ei- sphere, three sections were taken per animal, and three animals were
ther CSNs or non-CSNs, we trained a support vector machine (SVM) used for the analysis of each probe. For analysis, each image was sepa-
classifier with a linear kernel to predict cell origin based on the top 50 rated into individual channels (probe, forelimb and hindlimb CSN, and
enriched genes in CSNs and the top 50 enriched genes in non-CSNs, DAPI). The images of the traced neurons were then exported to Adobe
reserving half of our cells for cross-validation (Pedregosa et al., 2011). Photoshop, where the quick selection tool was used to select cell body
The SVM was balanced for class weight as there were an uneven number regions of interest (ROIs) and create masks. This was done manually as
of CSNs and non-CSNs. Probability outcomes were predicted by setting CSN axons can be very thick, and automatic ROI selection often cannot
probability to True during training. SVM performance was evaluated distinguish between CSN axons and cell bodies. Images of the probe and
using the area under the receiver operating characteristics curve (AUC the ROI masks are then uploaded to CellProfiler software, and puncta
ROC). Classifier performance was statistically compared using a random- per cell are automatically calculated according to the CellProfiler manual
ized permutation test. (McQuin et al., 2018).
The limb-specific classifier was built in a similar way. Briefly, the top Immunohistochemistry protocol. Ten days after retro-AAV injec-
50 enriched forelimb genes and the top 50 enriched hindlimb genes were tions, mice were killed with isoflurane and transcardially perfused with
used to build a linear SVM, with half of the cells reserved for cross-vali- 0.9% NaCl with 10 units/ml heparin (1000 unit/ml; Covetrus) followed
dation. SVM performance was evaluated using AUC ROC. Classifier by 4% paraformaldehyde in PBS. Brains and spinal cords were dissected
performances were statistically compared using a randomized permuta- and postfixed in 4% PFA overnight at 4°C, and 40-mm-thick sections of
tion test. brain were then sectioned on a vibratome (Leica Microsystems). The sec-
Classification of Rbp41 neurons. Rbp41 neurons were run through tions were then processed for mCherry with tyramide signal amplification
the same QC quality control pipeline outlined above. They were batch (catalog #NEL700A001KT, PerkinElmer). Immunofluorescence used pri-
corrected with the sequenced cells in this study using the ingest function mary antibodies directed against mCherry (1:500,000; catalog #ab167453,
in Scanpy. Their expression was batch normalized with MAGIC based Abcam), Synaptophysin (1:10,000; catalog #MAB329, EMD Millipore),
on the combined graph from the batch correction. Rbp41 neurons with and NeuN (1:200; catalog #24307, Cell Signaling Technology) and detected
batch corrected expression values were then run through the CSN-C, with secondary antibodies Alexa Fluor 488, 568, and 647 (1:500; catalog
and neurons with a probability of being a CSN . 0.75 were classified as #A11039, #A11011, #A21235, Life Technologies).
CSNs. BrainJ processing. Ten days after AAV injections, mice were killed
CSN visualization. For low-dimensional visualization of all scRNAseq with isoflurane and transcardially perfused with 0.9% NaCl with 10
data, we used the Python package PHATE (potential of heat diffusion units/ml heparin (1000 unit/ml; Covetrus), followed by 4% PFA in PBS.
for affinity-based transition embedding; version 1.0.7) in three dimen- Brains were dissected and postfixed in 4% PFA overnight at 4°C. The
sions with the default settings (Moon et al., 2019). All PHATE projec- next day, 75-mm-thick sections were cut using a vibratome (Leica
tions are displayed in 2D; examples of 3D animated versions are in VT1000 S). Sections underwent an immunohistochemistry protocol to
Movies 1, 2, 3 (see Figs. 3C, 6D, 7A). Animated versions of all PHATE pro- enhance tdTomato expression for microscopy. Briefly, sections received
jections are available on request. 3 washes of PBS-T, followed by a 10 min incubation in 0.3% H2O2 at
MELD analysis of intermediate FL-HL CSN classification. To investi- room temperature, three more washes of PBS-T, and blocking in 10%
gate the distribution of cervical- and lumbar-labeled CSNs, and investi- normal donkey serum for 1 h at room temperature. Next, sections were
gate the possibility of an intermediate population, we used the Python incubated overnight in Rb-anti-mCherry (1:1000). The next day, sec-
package MELD (version 1.0.0) to identify the degree to which individual tions were washed 3 in PBS-T and then incubated for 2 h in 546 G-
CSNs separated into the two groups (cervical or lumbar) defined by anti-Rb (1:1000). After 3 final PBS-T washes, every other section was
labeling, which was expressed as a sample-associated relative likelihood mounted on Superfrost Plus slides with DAPI mounting media (1 mg/
(Burkhardt et al., 2021). This analysis was performed on unimputed ml; 1:10,000). Sections were imaged at 4 using an Olympus VS200
square-root-transformed data to avoid the possibility that imputing over Slide Scanner for projection analysis. BrainJ software was used, as previ-
the whole dataset could have softened the distinction between the cervi- ously described (Botta et al., 2020), to perform analysis of supraspinal
cal and lumbar groups. forelimb and hindlimb ipsilateral and contralateral projections. Briefly,
sections were ordered and registered to the Allen Mouse Brain Common
Immunohistochemistry and single-molecule fluorescent in situ hybridiza- Coordinate Framework (CCF) (http://atlas.brain-map.org/) template
tion validation using Elastix software, version 5.0.1. Next, ilastix, version 1.3.3 was used
Single-molecule fluorescent in situ hybridization of adult cortical tissue. to identify brainwide soma and projections in the ;80 mounted sections
Ten days after retro-AAV injections, mice were killed with isoflurane and using pixel-based machine learning. BrainJ output absolute density
transcardially perfused with 0.9% NaCl with 10 units/ml heparin (1000 measurements for 1429 subregions of the brain, as defined by the Kim
unit/ml; Covetrus) followed by 4% paraformaldehyde (PFA) in PBS. enhanced and unified mouse brain atlas (Chon et al., 2019). We then
Brains and spinal cords were dissected and postfixed in 4% PFA overnight classified each subregion as ET, IT, NP, or CT using the Allen Institute
at 4°C. The next day, 15-mm-thick sections were cut using a vibratome for Brain Science framework. A mixed-design ANOVA was performed
Golan et al. · Transcriptional Index of the Adult Corticospinal Tract J. Neurosci., November 22, 2023 • 43(47):7929–7945 • 7933

Figure 1. Single-cell RNAseq pipeline for adult CSNs. A, Schematic of spinal tracing via injecting retro-AAV-CAG-GFP at spinal level C6/7 and retro-AAV-CAG-tdTomato at spinal level L4/5.
B–D’, Photomicrographs from M1 (;0.0 mm AP bregma, medial cortex) showing FL-CSNs (B), HL-CSNs (C), and an overlay with both single and dual-projecting CSNs (D, D’) 14 d after spinal
infusion. Schematic overview of the dissection-sequencing procedure. Ei–iv, Mice received injections of retro-AAV-CAG-GFP into either C6/7 or L4/5 (E). After a 2 week incubation, three 500
mm sections through M1 were collected (Ei), layers 2/5 and 5 were macrodissected (Eii) and dissociated, and traced CSNs (green), L2/3 cells (yellow), and L5 nonfluorescent cells (blue) were
then sorted using FACS (Eiii) and sequenced using 10x Chromium (Eiv). To exclude putative CSNs from the unlabeled layer 5 population of sequenced neurons, we used KDE analysis to quantify
the transcriptional overlap between populations in low-dimensional space. F, Right, untraced layer 5 neurons (blue) with .50% probability (blue, white outline) of originating from the CSN
distribution (green) were excluded. To exclude putative layer 5 neurons from the layer 2/3 population of sequenced neurons, we used KDE to quantify the transcriptional overlap between these
populations in low-dimensional space. G, Right, Layer 2/3 neurons (orange) with .50% probability (orange, white outline) of originating from the layer 5 distribution (blue) were excluded.
Scale bars: B–D, 500 mm; D’, 100 mm.

in R software for each projection subtype to analyze differences between HL CSNs, we microinfused retro-AAV-CAG-GFP into the gray
forelimb–hindlimb and ipsilateral–contralateral projections. An alpha matter of either C6/7 or L4/5 spinal cord in adult mice (Fig. 1E).
level of 0.05 was set a priori, and Bonferroni-adjusted post hoc tests were After 10 d we macrodissected layers 2/3 and 5 of M1 (11.0 to
performed when main effects were found. 0.5 mm relative to bregma), enzymatically and physically disso-
ciated the tissue to create a single-cell suspension. Dissociated
Data availability CSNs were then incubated in a cytoplasmic dye to aid whole-cell
All analyses were performed using in-house-developed code and imple-
detection for subsequent FACS (Golan and Cafferty, 2021).
mented in Python. The CSN and limb-specific CSN gene lists along with
example classifiers and appropriate sample data and code can be found Intact CSNs were then collected for scRNAseq using the 10
on GitHub at https://github.com/cafferty-lab/CSN_classification. Chromium system. As not every CSN was fluorescently labeled
because of inherent inefficiencies with viral labeling, KDE analy-
sis was used to identify putative CSNs from the nonfluorescent
Results layer 5 samples based on gene expression. Control layer 5 cells
A novel pipeline for scRNAseq of retrogradely labeled CSNs with a .50% probability of originating from the CSN distribu-
CSNs extend axons from L5b of M1 to innervate every spinal tion were classified as putative CSNs and excluded from down-
segment, synapsing principally on spinal interneurons to control stream analyses (Fig. 1F). To overcome inexact microdissection
FL and HL motor output. Spinal retrograde tracing from C6/7 of layer 2/3 neurons from layer 5 neurons, KDE analysis was
and L4/5 show that CSNs that innervate the FL are spatially and used to exclude putative layer 5 neurons from the layer 2/3 sam-
anatomically distinct from those that innervate the HL, mirror- ple for downstream analysis (Fig. 1G).
ing functional dexterity differences and hinting that FL and Sequenced neurons had a mean library size of 19,432 UMIs
HL CSNs are also molecularly heterogeneous (Fig. 1A–D’). To (median, 18,474 UMIs; Fig. 2A) and 5357 genes per cell (median,
explore the molecular heterogeneity among CSN populations, 5537). Previous studies have detailed molecular differences
we completed scRNAseq on retrogradely labeled FL and HL between and among excitatory and inhibitory neurons spanning
CSNs and compared gene expression between these anatomically the entire cortical depth (Arlotta et al., 2005; Lein et al., 2007;
discrete populations with layer 2/3 and nonspinal projecting L5 Morris et al., 2010; Molyneaux et al., 2015; Tasic et al., 2016;
neurons in M1 of adult mice. As relatively large pyramidal neu- Saunders et al., 2018; Tasic et al., 2018). Here, we focused specifi-
rons, CSNs have a lower nucleus to cytoplasm ratio, hence a large cally on molecular differences between layer 2/3 neurons and L5
portion of transcripts are extranuclear (Bakken et al., 2018). neurons in M1, as well as differences among L5 neurons, and
Therefore, it was critical to maintain plasma membrane integrity comprehensively characterize transcriptional specificity between
to achieve maximal quality for whole-cell sequencing. To that CSNs. To achieve this, we compared gene expression between
end, we created a pipeline for labeling, extracting, dissociating, retrogradely traced CSNs (n ¼ 1776 cells), nonfluorescent non-
and sequencing adult neurons with long-distance axonal projec- spinal L5 neurons (n ¼ 3000 cells), and layer 2/3 neurons (n ¼
tions (Golan and Cafferty, 2021). To independently label FL and 719) in M1, sequencing a total of 5495 adult neurons.
7934 • J. Neurosci., November 22, 2023 • 43(47):7929–7945 Golan et al. · Transcriptional Index of the Adult Corticospinal Tract

Figure 2. Gene expression confirms sequenced cell identity. A, Sequenced CSNs, L2/3 neurons, and nontraced L5 cells have overlapping distributions of library size; mean library size of
19,432 UMIs; median, 18,474 UMIs. B, Differential gene expression analysis revealed 32 genes were enriched in neurons from male mice and 213 genes enriched in neurons from female mice
(Wilcoxon rank sum test with a Benjamini–Hochberg correction, p adjusted , 0.05). C, IPA was used to gain biological insight between the sexes; only three pathways were significantly
enriched—the synaptogenesis, protein ubiquitination, and eNOS signaling (right-tailed Fisher’s exact test with a Benjamini–Hochberg correction, p , 0.05). D–H, Heat maps representing nor-
malized imputed expression of sex-specific genes (D), intact cell (IC) genes (E), cell-type genes (F), neurotransmitter (NT) genes (G), and cortical layer genes (H), confirming that sequenced cells
were sex balanced, intact, neurons, glutamatergic, and from expected layers, respectively.

Differential gene expression analysis of neurons from male (nonspinal L5 neurons and layer 2/3 neurons), revealing 197
and female mice revealed 32 DE genes in male mice and 213 DE CSN upregulated genes and 639 downregulated genes; (2) traced
genes in female mice (Fig. 2B). QIAGEN IPA revealed that the CSNs versus nonspinal L5 neurons, revealing 215 CSN upregu-
synaptogenesis, protein ubiquitination, and eNOS (endothelial lated genes and 634 downregulated genes; and (3) traced CSNs
nitric oxide synthase) signaling pathways were differentially versus layer 2/3 neurons, revealing 467 CSN upregulated genes
enriched between male and female mice (Fig. 2C). To ensure and 1533 downregulated genes (Fig. 3A; Extended Data Fig. 3-1
there was no sex bias in this study, we assessed the expression contains the entire DE lists). To explore the biological import
of female-specific genes Xist and Tsix as well as the male-spe- of the transcriptional differences among CSNs and other gluta-
cific genes Ddx3y and Eif3s3y (Fig. 2D), showing female and matergic neurons in M1, we completed QIAGEN IPA (Krämer
male representation in all assessed populations. Next, expres- et al., 2014). Consistently among the top enriched pathways
sion of Ywhaz, a gene whose mRNA is enriched in the cyto- across all comparisons were those associated with axon guid-
plasm (Yao et al., 2021), and Neurod2, a nuclear gene, confirms ance, signal transduction, and synaptic transmission (Fig. 3B).
that all sequenced cells contained both a nucleus and cytoplasm These pathways are consistent with previous work showing that
(Fig. 2E). To broadly confirm the identity of sequenced cells, axon guidance cues including Epha4, Efnb3, Ryk, and Wnt3a
we assessed their expression of previously identified cell-type- have previously been shown to be critical in wiring the CST
specific genes. All sequenced cells expressed the neuron-specific (Canty and Murphy, 2008).
gene Snap25 and showed a dearth in expression for other cell- To explore differences among these populations, we used
type-specific genes including the endothelial-specific gene PHATE for dimensionality reduction and visualization, as unlike
Cldn5, the astrocyte-specific gene Gfap, the microglial-specific the visualization techniques PCA, t-SNE, and UMAP, PHATE
gene Cx3cr1, and the oligodendrocyte-specific gene Olig2 (Fig. maintains both local and global structure and was designed for
2F). Neurotransmitter gene expression confirmed that all scRNAseq visualization (van Dijk et al., 2018; Moon et al., 2019).
sequenced neurons were enriched in the glutamate trans- A PHATE projection colored by cell origin (Fig. 3C; Movie 1)
porter Slc17a7 compared with the GABAergic gene Gad1, illustrates the transcriptional overlap between these populations.
the cholinergic gene Chat, or the serotonergic gene Sert To quantify, KDE analysis of each population was plotted, and a
(Fig. 2G). Layer 2/3 neurons showed an elevated expression similarity index was calculated based on the overlapping regions
for the previously identified layer 2/3-enriched gene Cux2 of the KDE (Fig. 3D). These analyses demonstrate that CSNs are
(Tasic et al., 2016). CSNs and nonspinal L5 neurons showed a most similar to nonspinal L5 neurons (similarity index ¼ 0.5),
moderate enrichment for established L5 genes Ctip2, Fezf2, and whereas they are least similar to layer 2/3 neurons (similarity
Crym, relative to other cortical layer genes Reln (L1) and Ctgf index ¼ 0.23). Nonspinal L5 neurons show an intermediate level
(L6; Fig. 2H), confirming L5 identity. Although CSNs expressed of similarity with layer 2/3 neurons in comparison with CSNs
previously identified L5 genes, none were specific to any L5 (similarity index ¼ 0.41).
population, suggesting that more targeted analyses are neces- Based on enrichment of guidance cues in CSNs, we selected
sary to identify unique molecular characteristics of CSNs. Wnt7b as a primary candidate to validate our scRNAseq data as
it is significantly enriched in all traced CSNs (Fig. 3E). To con-
Identifying CSN-enriched genes in M1 firm this finding in vivo, we infused retro-AAV-CAG-GFP
To identify CSN-enriched genes within M1, we performed the into the gray matter of C6/7 spinal cord and retro-AAV-CAG-
following DE analyses: (1) traced CSNs versus non-CSNs tdTomato into the gray matter of L4/5 spinal cord in adult
Golan et al. · Transcriptional Index of the Adult Corticospinal Tract J. Neurosci., November 22, 2023 • 43(47):7929–7945 • 7935

Figure 3. CSNs are unique among neurons in M1 and are enriched in Wnt7b expression. A, Differential gene expression analysis between traced CSNs and a combination of layer 2/3 and L5 nonfluor-
escent cells shows that 197 genes were upregulated and 639 downregulated in CSNs, 217 genes were upregulated and 634 downregulated in CSNs compared with layer 5 nontraced neurons, and 467
genes were upregulated and 1533 downregulated in CSNs compared with layer 2/3 neurons (Wilcoxon rank sum test with a Benjamini–Hochberg correction, p adjusted , 0.05). B, QIAGEN IPA was
used to identify enriched pathways in each of these analyses, (right-tailed Fisher’s exact test with a Benjamini–Hochberg correction, p adjusted , 0.05). C, D, PHATE projection (C, Movie 1) and kernel
density estimate plots with hierarchical clustering and a similarity index (D) colored by cell identity illustrate differences between CSNs, L5 nontraced, and L2/3 neurons. E, Wnt7b is significantly enriched
in CSNs compared with layer 5 nontraced and L2/3 neurons (Wilcoxon rank sum test with a Benjamini–Hochberg correction; ****p adjusted , 0.001). F, Schematic showing labeling of cervical CSNs
with retro-AAV-CAG-GFP (rAAV-GFP, cyan) and lumbar CSNs with retro-AAV-CAG-tdTomato (rAAV-tdT, magenta). G, H, smFISH of retrogradely labeled CSNs shows an enrichment of Wnt7b in FL-CSNs
and HL-CSNs compared with nonfluorescent DAPI1 cells in layer 5b (data shown are average number of puncta per cell 6 SEM, one-way ANOVA with a Bonferroni correction, ****p , 0.0001). Scale
bars: 10 mm. Data tables from the scRNAseq containing the results of the DE analysis comparing CSNs to nontraced L5 and L2/3 neurons are provided in Extended Data Figure 3-1.

wild-type mice, differentially labeling FL and HL CSNs and


completing smFISH (Fig. 3F). Photomicrographs of L5b from
M1 prepared 2 weeks after spinal infusion show GFP1 FL
CSNs and tdTomato1 HL CSNs are significantly enriched in
Wnt7b relative to DAPI1 control cells (Fig. 3G,H).

Classifier analysis to predict CSN identity


Based on the high similarity index between CSNs and nonspinal
L5 neurons, we reasoned it would be unlikely that any single
gene could uniquely identify CSNs from other L5 neurons. To
that end, we first explored 12 genes that have previously been
used to describe L5 pyramidal neurons (Fig. 4A; Arlotta et al.,
2005; Fink et al., 2015; Tasic et al., 2016; Economo et al., 2018;
Tasic et al., 2018; Poplawski et al., 2020). Of these genes, many
are enriched in CSNs compared with other populations includ-
ing Ctip2, Cd55, Cdh13, Crym, and Fezf2. However, all are also
expressed at moderate levels in nontraced L5 neurons and are
only detected in a subset of CSNs. Accordingly, PHATE projec- Movie 1. 3D PHATE visualization of CSNs, L2/3, and nontraced L5 neurons in M1. 3D
tions for Ctip2 and Rbp4 show an incomplete and nonspecific PHATE projection of 5495 intact adult neurons from M1 colored by cell identity (traced CSNs,
expression in CSNs, and although Wnt7b shows greater specificity, green; nontraced L5, blue; L2/3 neurons, yellow). [View online]
7936 • J. Neurosci., November 22, 2023 • 43(47):7929–7945 Golan et al. · Transcriptional Index of the Adult Corticospinal Tract

Figure 4. Using linear SVM classifiers to identify CSNs among cells in M1. A, Dot plot of previously characterized L5-enriched genes illustrates enrichment and expression of each gene in
CSNs, nontraced L5, and L2/3 neurons. The size of the dot denotes the proportion of each population that expresses a given gene, and the color denotes the normalized mean expression within
that subtype. B, PHATE projections colored by normalized gene expression of previously characterized L5 genes including Ctip2 and Rbp4, along with newly identified wnt7b, illustrate expres-
sion in CSNs, nontraced L5 neurons, and L2/3 neurons. Top right, Boxed PHATE projection illustrates neuronal cell types in their transcriptional space (CSNs, green; nontraced L5 neurons, blue;
L2/3 neurons, orange). C, An ROC curve showing the performance of an SVM) trained to classify CSNs based on L5-C genes; the top 100, top 10, or random 10 novel genes enriched in CSN-C,
or 10 random genes. AUC is calculated for each classifier, showing that CSN-C outperforms L5-C, and top 10 and random 10 CSN genes outperform the L5-C (randomized permutation test,
p , 0.001). D, PHATE projections showing CSN classification performance with either the CSN-C (top left, forest green) or L5-C (bottom left, purple), Top right, CSNs missed by L5-C (black), and a
quantification of the proportion of CSNs that were classified correctly. CSN-C correctly classified 92% of CSNs. L5-C correctly classified 76% of CSNs. E, Rbp4 cre:Ai14 mice received infusions of
retro-AAV-CAG-GFP into either the cervical, C6/7, or the lumbar, L4/5, spinal cord. F, F’, Low-power (F) and high-power (F’) photomicrographs show GFP (cyan) expression in a section through
M1 from an Rbp4 cre:Ai14 (magenta) mouse. Cyan cells represent traced CSNs. Magenta cells are Rbp41 neurons that do not project to the spinal cord, white cells are GFP1/Rbp41 neurons
(F’). G, Quantification of the proportion of Rbp41 neurons that express GFP in coronal sections through M1 (1.7 to 1.2, relative to Bregma; mean, 7.7%). CSN-C prediction of CSN identity of
sequenced Rbp41 neurons. H, CSN-C predicts 7% of Rbp4 neurons are CSNs with a 75% probability cutoff. Scale bars, F, 500 mm; F’, 100 mm.

coverage remains incomplete (Fig. 4B). To improve on these genes most differentially expressed between CSNs and non-CSNs (CSN-
as a means for CSN identification, we sought to distinguish a CSN C). For comparison, we trained additional CSN classifiers based
transcriptional signature. To that end, we trained a linear classifier on the top 10 genes differentially expressed and a random 10 genes
to predict cell identity based on expression of 100 genes that are from the top 100 that were differentially expressed (Fig. 4C). We
Golan et al. · Transcriptional Index of the Adult Corticospinal Tract J. Neurosci., November 22, 2023 • 43(47):7929–7945 • 7937

compared the performance of these classifiers to one that was feature elimination was used to determine the top 10 FL- and
trained on sole expression of all 12 previously characterized L5 HL-specific genes that most robustly distinguished these two
genes, termed L5 classifier (L5-C), and another classifier trained populations. The top genes that define the FL population include
on a random set of 10 genes. The performance of each classi- calcium-related genes Cacng7 and Efcab12, as well as transport-
fier was evaluated using the ROC AUC, and the classifiers ers Slc27a1 and Slc4a2 (Fig. 5F). The top genes that define the
were compared using a permutation test. This revealed that HL population include ion transporters such as Slc16a2, tran-
CSN-C (AUC ¼ 0.96), top 10 genes (AUC ¼ 0.95), random 10 scription-related genes such as Zfp281, and receptors such as
CSN genes (AUC ¼ 0.91) significantly outperform L5-C Ptger1 (Fig. 5F).
(AUC ¼ 0.87; Fig. 4C). To visually compare the differences PHATE projections show Cacng7 expression is significantly
between these classifiers, we plotted CSN-C CSNs and L5-C enriched in FL traced neurons (Fig. 5G) and Slc16a2 expression
CSNs in PHATE. CSN-C correctly classified 92% of CSNs, is significantly enriched in HL traced neurons (Fig. 5H). To vali-
whereas L5-C classified 78% of CSNs (Fig. 4D). date these results in vivo, mice received injections of retro-AAV-
CAG-GFP into spinal gray matter at C6/7 and retro-AAV-CAG-
Classifier analysis to predict CSN identity of Rbp41 neurons tdTomato into spinal gray matter at L4/5. Visualization of
To validate the use of the CSN-C as a tool for predicting CSN Cacng7 using smFISH confirmed differential expression in
identity, we sequenced neurons from Rbp4 cre:Ai14 mice, a GFP1 FL–projecting CSNs compared with tdTomato1 HL–
line previously used to comprehensively target and label L5 projecting CSNs (Fig. 5I,J). A similar analysis was then repeated
pyramidal neurons (Alcamo et al., 2008; Tasic et al., 2018). To with the HL enriched gene Slc16a2. In vivo smFISH confirmed
quantify the proportion of Rbp41 neurons that project to the HL-enrichment of Slc16a2 in tdTomato1 HL–projecting CSNs
spinal cord (Rbp41 CSNs), we delivered retro-AAV-CAG- relative to GFP1 FL–projecting neurons (Fig. 5K,L).
GFP into the cervical and lumbar cord of Rbp4 cre:Ai14 mice
(Fig. 4E). Photomicrographs show that a minority of Rbp41 Classifier analysis to predict FL and HL identity
neurons project to the spinal cord (Figs. 4F,F’). Quantification Although DE analysis revealed FL- and HL-enriched genes,
of the proportion of Rbp41 neurons that expressed GFP, and because of the high similarity index between FL and HL CSN
therefore have terminals in either the cervical or lumbar spinal populations, none of these genes can independently predict FL
cord, revealed that on average, 7.7% of Rbp41 neurons are or HL identity. To explore, we first examined limb specificity in
also CSNs (Fig. 4G). Classification of sequenced Rbp41 neu- the previously identified L5-enriched genes (Fig. 5M). Crym is
rons using CSN-C predicted that 7% of Rbp41 neurons were modestly enriched in FL-CSNs (Fink et al., 2015), and Crim1 was
putative CSNs using a 75% probability cutoff (Fig. 4H). These modestly enriched in HL-CSNs (Sahni et al., 2021a,b). As the
findings confirm the validity of our CSN gene list to predict majority of these genes showed no limb specificity, we trained a
CSN identity among L5 neurons in M1. linear classifier to predict limb specificity based on expression of
100 genes that are most differentially expressed between FL and
Molecular identification of limb-specific CSN subtypes HL CSNs, termed limb-specific CSN-C (LS-CSN-C). We com-
We next wanted to explore potential transcriptional heterogene- pared the performance of this classifier with L5-C. We evaluated
ity between anatomically discrete FL- and HL-projecting CSNs. the performance of each classifier with ROC AUC, and the two
PHATE visualization of traced neurons colored by anatomic ori- classifiers were compared using a permutation test. This revealed
gin illustrates there is a transcriptional separation between FL that LS-CSN-C (AUC ¼ 0.94) significantly outperforms L5-C
and HL traced cells (Figs. 5A). To quantify, a KDE of each popu- (AUC ¼ 0.76; Fig. 5N).
lation was plotted, and a similarity index was calculated based on
the overlapping regions of the KDE. FL and HL CSNs are some- CSN subtypes defined by supraspinal connectivity
what similar to each other (similarity index ¼ 0.74). Because of Recent studies from the Brain Initiative Cell Consensus Network
the elevated similarity index relative to the more definitive sepa- (BICCN; Callaway et al., 2021) performing scRNAseq on M1
ration between CSNs versus L5 controls and CSNs versus layer and anterolateral motor cortex (ALM) have classified glutama-
2/3 (Fig. 3D), we reasoned that a population of CSNs may have a tergic projection neurons into the following four broad subtypes:
projection to both the cervical and lumbar spinal cord. Indeed, a IT neurons that have many projection targets including the stria-
small percentage of traced CSNs were double positive for GFP tum and cerebral cortex (Lin et al., 2018; Zhang et al., 2021); ET
and tdTomato when tracers were injected into the cervical and neurons that project from M1 to the brainstem and medulla
hindlimb respectively (Fig. 1A–D’). To explore whether these (Economo et al., 2018; Tasic et al., 2018; Yao et al., 2021); NP
cells represented a true population of FL/HL CSNs or whether en neurons that have projections in local areas including M1, M2,
passant labeling of HL axons occurred when infusing tracer into and S1 (Tasic et al., 2018); and CT) neurons (Yao et al., 2021;
the cervical cord, we used MELD to identify CSNs whose tran- Fig. 6A). The 10-sequenced deep layer glutamatergic neurons
scriptomic characteristics were intermediate between the FL and collected in Yao et al. (2021) separate into four discrete popula-
HL populations as previously identified by cervical and lumbar tions representing the IT, ET, NP, and CT subtypes when exam-
tracing (Fig. 5B,C). We found 188 such CSNs, 60 of which had ined in PHATE (Fig. 6B). CSNs collected in this study, when
been previously identified as FL and 128 as HL (Fig. 5D). These projected into the same PHATE graph, populate all four sub-
data suggest that as much as 10% of CSNs may have a projection types, and therefore we classified them as CSN-IT, CSN-ET,
into both the cervical and lumbar spinal cord. CSN-NP, and CSN-CT (Fig. 6C,D; Movie 2). Differential gene
DE analysis revealed that there were 492 FL-enriched genes expression analysis among these four putative subdivisions of
and 788 HL-enriched genes (Fig. 5E; Exended Data Fig. 5-1, CSNs revealed an enrichment for previously identified projection
entire DE lists). Identifying a set of enriched FL and HL CSN subtype genes, Fam84b for CSN-ET cells, Slc30a3 in CSN-IT
genes is central to understanding the potential independent cells, Foxp2 in CSN-CT, and Sla2 in CSN-NP cells (Tasic et al.,
homeostatic biology and injury-induced changes within these 2018; Yao et al., 2021; Fig. 6E), and an additional set of subcate-
discrete populations. To prioritize candidate genes, recursive gory-specific genes (Fig. 6E,F). We completed QIAGEN IPA on
7938 • J. Neurosci., November 22, 2023 • 43(47):7929–7945 Golan et al. · Transcriptional Index of the Adult Corticospinal Tract

Figure 5. A, Forelimb and hindlimb CSN specialization. PHATE projection colored by cell identity of retrogradely labeled GFP1 cervical CSNs (cyan) and tdTomato1 lumbar CSNs (magenta). B,
Histogram of MELD prediction confidence of lumbar identity in all CSNs, showing predicted intermediate cells in gray. C, PHATE projection showing MELD prediction confidence of lumbar identity. D,
Comparison of counts for tracing-identified lumbar and cervical CSN populations (top) with MELD-predicted counts for lumbar, cervical, and intermediate CSN populations (bottom). E, DE analysis identified
492 upregulated FL genes and 788 upregulated HL genes (Wilcoxon rank sum test, Benjamini–Hochberg correction, p adjusted , 0.05). F, Violin plots of the top 10 most differentially expressed genes
in FL CSNs (cyan) and HL CSNs (magenta; Wilcoxon rank sum test, Benjamini–Hochberg correction, p adjusted , 0.05). G, PHATE projection colored by Cacng7 expression illustrates FL enrichment. H,
PHATE projection colored by Slc16a2 expression illustrates HL enrichment. I, J, smFISH of retrogradely labeled GFP1 FL-CSNs and tdTomato1 HL-CSNs shows an enrichment of Cacng7 in FL-CSNs (data
shown are average number of puncta per traced CSN 6 SEM; unpaired t test, ***p , 0.0001). K, L, smFISH of retrogradely labeled GFP1 FL-CSNs and tdTomato1 HL-CSNs shows an enrichment of
Slc16a2 in HL-CSNs (data shown are average number of puncta per traced CSN 6 SEM; unpaired t test, ***p , 0.001). M, Dot plot of previously characterized L5-enriched genes illustrates enrichment
and expression of each gene in FL and HL CSNs. The size of the dot denotes the proportion of each population that expresses a given gene, and the color denotes the normalized mean expression within
that subtype. These L5 genes are not uniquely nor differentially enriched in FL or HL CSNs. N, An ROC curve showing the performance of a linear SVM trained to classify limb-specific CSNs based on previ-
ously characterized L5 genes (L5-C) or our novel limb-specific gene list (LS-CSN-C). AUC is calculated for each classifier, showing that LS-CSN-C outperforms C-L5. Scale bars; K, bottom left 30 mm; K, bot-
tom right, 15 mm. Data tables from the scRNAseq containing the results of the DE analysis comparing FL versus HL CSNs are provided in Extended Data Figure 5-1.

genes significantly differentially expressed among these CSN associated with synaptic signaling and neurovascular coupling
subcategories (Figs. 6G–J). When comparing the CSN-ET (Fig. 6G). Comparing CSN-IT cells with the other three catego-
population with the other three categories, we found there ries, we found there were 539 DE genes showing an enrichment
were 972 DE genes showing an enrichment in pathways in pathways associated with axon guidance and CREB signaling
Golan et al. · Transcriptional Index of the Adult Corticospinal Tract J. Neurosci., November 22, 2023 • 43(47):7929–7945 • 7939

Figure 6. Transcriptionally distinct CSN subtypes can be defined by supraspinal terminal fields. A, Schematic based on the BICCN M1 taxonomy showing that projection neurons can be classi-
fied into four categories based on their primary terminal field, including ET (areas shaded in orange in the schematic), IT (areas shaded green), CT (areas shaded purple), and NP (areas shaded
red). B, PHATE projection showing 10x V3-sequenced M1 neurons from Yao et al. (2021) colored by ET (orange), IT (green), CT (purple), and NP (red) projection subtype. C, PHATE projection of
7940 • J. Neurosci., November 22, 2023 • 43(47):7929–7945 Golan et al. · Transcriptional Index of the Adult Corticospinal Tract

As CSNs are represented in all four projection subtype cate-


gories, we infer that transcriptional subdivisions among CSNs
are therefore defined by the spinal termination of their primary
axon in the spinal cord and by the location of their supraspinal
terminal fields (Fig. 6K–N).
To explore CSN supraspinal connectivity in vivo, we injected
retro-AAV-CAG-tdTomato unilaterally into the gray matter of
C6/7 and L4/5 spinal cord and surveyed for the presence of
tdTomato1ve CSN terminals in the BICCN identified supraspi-
nal structures (Fig. 6A,K–N). We observed td-Tomato1ve CSN
terminals in the brainstem (Fig. 6K,K’), the caudoputamen of the
striatum (Fig. 6L,L’), the motor thalamus (Fig. 6M,M’), and
regions near M1 including S1 and M2 (Fig. 6N,N’), confirming
that CSNs project to both the spinal cord and to these supraspinal
structures. To further explore whether these collaterals rep-
resented putative active synapses, we completed immunohis-
tochemistry, showing that traced CSN terminals express
the presynaptic protein synaptophysin and colocalize with
NeuN-positive neurons.

Limb-specific specialization among supraspinal CSN


Movie 2. 3D PHATE visualization of CSNs and deep layer glutamatergic projection neurons subtypes
sequenced from Yao et al. (2021) in the Yao et al. (2021) manifold. PHATE projection of We found that traced CSNs maintained a supraspinal cluster sep-
CSNs and M1 projection neurons from Yao et al. (2021), colored by BICCN taxonomy, ET (or- aration when plotted in their native PHATE space (Fig. 7A;
ange), IT (green), CT (purple), and NP (red) projection subtype. [View online] Movie 3), and FL and HL CSNs were represented in each supra-
spinal subcategory (Fig. 7B). However, the relative distribution
(Fig. 6H). Comparing CSN-CT cells with the other categories, we of CSN subcategory distribution was different between FL and
found there were 656 DE genes showing an enrichment in axon HL. For FL CSNs 19.6% were ET, 36.5% IT, 40% CT, and 3.8%
guidance and RhoGDI signaling (Fig. 6I). Comparing CSN-NP NP. HL CSNs were 37.6% ET, 31.5% IT, 26.8% CT, and 4% NP
cells with the other categories we found there were 1735 DE (Fig. 7C,D). These data suggest that FL and HL CSNs may differ-
genes showing an enrichment in axon guidance and cAMP sig- entially innervate supraspinal structures. To explore we used
naling and GPCR signaling (Fig. 6J). The entire DE lists are avail- intersectional viral tracing to independently label all CSN termi-
able in Extended Data Figure 6-1. nals whose axons innervate either cervical or lumbar spinal cord.
We injected retro-AAV-CAG-Cre into either C5/6 or L4/5 spinal
cord and AAV-CAG-FLEX-tdTomato into FL M1 or HL M1,
/
respectively. Using the Java plug-in BrainJ (Botta et al., 2020), we
CSNs collected in this study projected onto the same PHATE space as in B from Yao et al. analyzed the density of tdTomato1ve CST projections across the
(2021) colored by projection subtype show that CSNs are represented in all four projection entire CST. We found that the projection density was higher on
classes. D, PHATE projections of CSNs and M1 projection neurons from Yao et al. (2021; the ipsilateral side compared with the contralateral side (mixed-
Movie 2). E, Violin plot showing gene expression in each projection subtype. F, PHATE projec- model ANOVA, p , 0.001). We also found that the FL-CST had
tions showing enrichment of established ET projection gene Fam84b and novel ET enriched
a higher density of projections compared with the HL-CST
gene Gprc5, enrichment of established IT gene Slc30a3 and novel IT enriched gene Frzb,
enrichment of established CT gene Foxp2 and novel CT enriched gene Syt6, and enrichment
(mixed-model ANOVA p ¼ 0.008), consistent with prior reports
in established NP gene Sla2 and novel NP enriched gene Lypd1. G, DE analysis identified 791 that FL-CSNs are more numerous than HL-CSNs (Wang et al.,
upregulated and 181 downregulated genes in CSN-ET cells versus CSN-IT, CSN-CT, and CSN- 2018). Next, we assessed FL and HL-CSN supraspinal projection
NP cells, listed with the top six enriched pathways from QIAGEN IPA (Wilcoxon rank sum density. We found an enrichment of FL-CSN-ET projections
test, Benjamini–Hochberg correction, p adjusted , 0.05). H, DE analysis identified 275 up- compared with HL-CSN-ET projections (Fig. 7E,F; mixed-model
regulated and 263 downregulated genes in CSN-IT cells versus CSN-ET, CSN-CT, and CSN-NP ANOVA p ¼ 0.003), and FL-CSN-NP projections compared
cells, listed with the top six enriched pathways from QIAGEN IPA (Wilcoxon rank sum test, with HL-CSN-NP projections (Fig. 7Q,R; mixed-model ANOVA
Benjamini–Hochberg correction, p adjusted , 0.05). I, DE analysis identified 215 upregu- p , 0.001). In contrast, we found no significant differences
lated and 441 downregulated genes in CSN-CT cells versus CSN-ET, CSN-IT, and CSN-NP cells,
between FL and HL for IT (Fig. 7I,J; p ¼ 0.38) and CT (Fig. 7M,
listed with the top six enriched pathways from QIAGEN IPA (Wilcoxon rank sum test,
Benjamini–Hochberg correction, p adjusted , 0.05). J, DE analysis identified 675 upregu-
N; p ¼ 0.085) projections. Overall, these data demonstrate the
lated and 1060 downregulated genes in CSN-NT cells versus CSN-ET, CSN-IT, and CSN-CT cells, supraspinal anatomic diversity of FL-CSNs and HL-CSNs and
listed with the top six enriched pathways from QIAGEN IPA (Wilcoxon rank sum test, suggest that the transcriptional differences among these popula-
Benjamini–Hochberg correction, p adjusted , 0.05). To validate CSN innervation of ET, IT, tion support unique circuit wiring.
CT, and NP structures, retro-AAV-CAG-tdTomato was injected in the cervical and lumbar cord.
K–N’, Terminals in supraspinal brain regions were examined including the brainstem (ET, or- CSN ET specialization
ange, K), the striatum (IT, green, L), motor thalamus (CT, purple, M), and primary motor cor-
These data reveal that supraspinal structures are differentially in-
tex (NP, red, N). Photomicrographs of CSN terminals in the brainstem (ET, K’), striatum (IT,
L’), motor thalamus (CT, M’), and primary motor cortex (NP, N’) stained with antibodies
nervated by FL-CSNs and HL-CSNs. To explore the transcrip-
against synaptophysin (green) and NeuN (white) show presumptive synaptic connections within tional differences among these populations that may drive and/
these supraspinal terminal fields. Scale bars; K’-N’, 100 mm; insets in each, 30 mm. Data tables or support the unique supraspinal terminal distribution within
from the scRNAseq containing the results of the DE analysis comparing each CSN supraspinal sub- these subdivisions we completed a differential gene expression
type versus all the other subtypes are provided in Extended Data Figure 6-1. analysis among FL and HL ET, IT, CT, and NP categories.
Golan et al. · Transcriptional Index of the Adult Corticospinal Tract J. Neurosci., November 22, 2023 • 43(47):7929–7945 • 7941

Figure 7. Forelimb- and hindlimb-specific supraspinal CSN innervation and specialization. A, B, CSNs in native PHATE space colored by supraspinal categories (A; Movie 3), and FL and HL
identity (B). C, D, Divergent numbers of FL and HL CSNs transcriptionally defined by BICCN supraspinal structures. E, Photomicrographs show representative images of FL (top, cyan outline) and
HL (bottom, magenta outline) td-tomato1ve CST projections after intersectional tracing in the brainstem (ET population). F, Projection density of FL-CSN-ET was significantly higher than in
HL-CSN-ET (n ¼ 4 FL traced mice, n ¼ 3 HL traced mice, mixed-design ANOVA, **p ¼ 0.003). G, Schematic illustrates FL axons (thick cyan lines) more densely innervating ET structures
7942 • J. Neurosci., November 22, 2023 • 43(47):7929–7945 Golan et al. · Transcriptional Index of the Adult Corticospinal Tract

dysregulated. Between FL-ET and FL-CT, 465 genes were upreg-


ulated, and 1065 genes were downregulated, with GPCR signal-
ing and RHOGDI signaling among the top enriched pathways.
Between FL-ET and FL-NP, 630 genes were upregulated, and
1808 genes were downregulated, with CREB signaling Gai sig-
naling among the dysregulated pathways (Fig. 7H).
For the HL-CSN-ET population, 807 genes were upregulated
and 588 genes downregulated in HL-CSN-ET cells compared
with HL-CSN-IT, HL-CSN-CT, and HL-CSN-NP combined
(Fig. 7H’). Comparisons among the subcategories revealed that
between HL-ET and HL-IT, 538 genes were upregulated, and
821 genes were downregulated, with axon guidance signaling
and CREB signaling among the top pathways dysregulated.
Between HL-ET and HL-CT, 735 genes were upregulated, and
1065 genes were downregulated, with Gai signaling and RHOGDI
signaling among the top enriched pathways. Between HL-ET and
HL-NP, 1234 genes were upregulated, and 2193 genes were down-
regulated, with spliceosomal cycle signaling and EIF2 signaling
among the top pathways (Fig. 7H’).
Movie 3: 3D PHATE visualization of CSNs traced from the cervical and lumbar enlarge-
ments colored by BICCN taxonomy in their native manifold. 3D PHATE visualization of CSNs CSN IT specialization
in their native manifold colored by BICCN taxonomy, CSN-ET (orange), CSN-IT (green), CSN- We found that 282 genes were upregulated and 228 genes down-
CT (purple), and CSN-NP (red) projection subtype. [View online] regulated in FL-CSN-IT cells compared with FL-CSN-ET, FL-
CSN-CT, and FL-CSN-NP combined (Fig. 7L). A more specific
comparison among subcategories revealed that between FL-IT
There were 964 genes upregulated and 484 genes downregu- and FL-ET, 1089 genes were upregulated, and 477 genes were
lated in FL-CSN-ET cells compared with FL-CSN-IT, FL-CSN- downregulated, with valine degradation and isoleucine degrada-
CT, and FL-CSN-NP combined (Fig. 7H). A more granular com- tion among the top dysregulated pathways. Between FL-IT and
parison among subcategories revealed that between FL-ET and FL-CT, 234 genes were upregulated, and 359 genes were downre-
FL-IT, 477 genes were upregulated and 1089 downregulated, and gulated, with Rho family signaling and SEMA signaling among
QIAGEN IPA revealed that GPCR (G-protein-coupled receptor) the top dysregulated pathways. Between FL-IT and FL-NP, 820
signaling and CREB signaling were among the top pathways genes were upregulated, and 1155 genes were downregulated,
with glutamate receptor signaling and putrescine degradation
among dysregulated pathways (Fig. 7L).
/ For the HL-CSN-IT population, 388 genes were upregulated,
compared with HL axons (thin magenta lines). H, Number of DE genes comparing FL-CSN-ET and 488 genes downregulated in HL-CSN-IT cells compared
with FL-CSN-IT, FL-CSN-CT, and NP combined (orange to black), and FL-CSN-ET versus IT (or- with HL-CSN-ET, HL-CSN-CT, and HL-CSN-NP combined
ange, upregulated, to green, downregulated), FL-CSN-ET versus CT (orange to purple) FL- (Fig. 7L’). Comparisons among the subcategories revealed that
CSN-NP combined (orange to black). H’, These comparisons are repeated for the HL-CSN-ET between HL-IT and HL-ET, 821 genes were upregulated,
population. I, Photomicrographs show FL and HL td-tomato1ve CST projections after inter- and 538 genes were downregulated, with axon guidance sig-
sectional tracing in the striatum (IT population). J, There was no significant difference in the
naling and valine degradation among the top dysregulated
projection density between ipsilateral and contralateral FL-CSN-IT and HL-CSN-IT projections
(mixed-design ANOVA, p ¼ 0.38). K, Schematic illustrates FL axons and HL axons innervating
pathways. Between HL-IT and HL-CT, 523 genes were up-
IT structures (green). L, Number of DE genes comparing FL-CSN-IT with FL-CSN-ET, FL-CSN- regulated, and 749 genes were downregulated, with SEMA
CT, and FL-CSN-NP combined (green to black), and FL-CSN-IT versus ET (orange to green), signaling and RHOGDI signaling among the top enriched
FL-CSN-IT versus CT (purple to green), and FL-CSN-IT vs NP (green to red). L’, These compari- pathways. Between HL-IT and HL-NP, 1377 genes were up-
sons are repeated for the HL-CSN-IT population. M, Photomicrographs show images of FL regulated, and 1758 genes were downregulated, with zymos-
and HL td-tomato1ve CST projections after intersectional tracing in the thalamus (CT popu- terol biosynthesis and putrescine degradation among the
lation). VPL, Ventral posterolateral nucleus of thalamus. N, There was no significant differ- top pathways (Fig. 7L’).
ence in the projection density between FL-CSN-CT and HL-CSN-CT projections (mixed-design
ANOVA, p ¼ 0.085). O, Schematic illustrates FL axons and HL axons innervating the thala-
mus. P, Number of DE genes comparing FL-CSN-CT to FL-CSN-ET, FL-CSN-IT, and NP com- CSN CT specialization
bined (purple to black), and FL-CSN-CT versus ET (purple to orange), FL-CSN-CT vs FL-CSN-IT We found that 201 genes upregulated, and 386 genes
(purple to green), and FL-CSN-CT versus FL-CSN-NP (purple to red). P’, These comparisons downregulated in FL-CSN-CT cells compared with FL-
are repeated for the HL-CSN-ET population. Q, Photomicrographs show images of FL and HL CSN-ET, FL-CSN-IT, and FL-CSN-NP combined (Fig. 7P).
td-tomato1ve CST projections after intersectional tracing in M2 (NP population). R, A more specific comparison among subcategories revealed
Projection density of FL-CSN-NP was significantly higher than in HL-CSN-ET (n ¼ 4 FL traced that between FL-CT and FL-ET, 1065 genes were upregu-
mice, n ¼ 3 HL traced mice, mixed-design ANOVA, ***p , 0.001). S, Schematic illustrates lated, and 465 genes were downregulated, with circadian
FL axons more densely innervating NP structures compared with HL axons. T, Number of DE
rhythm signaling and synaptogenesis signaling among the
genes comparing FL-CSN-NP to FL-CSN-ET, FL-CSN-IT, and FL-CSN-CT combined (red to
black), and FL-CSN-NP versus TT (red to orange), FL-CSN-IT versus NP (green to red), and FL-
top pathways dysregulated. Between FL-CT and FL-IT, 359 genes
CSN-NP vs CT (purple to red). T’, These comparisons are repeated for the HL-CSN-ET popula- were upregulated, and 234 genes were downregulated, with axon
tion. Scale bars: E, 500 mm; I, M, Q, 100 mm. Data tables from the scRNAseq containing the guidance signaling and CREB signaling in neurons among the
results of the DE analysis comparing FL CSN supraspinal subtypes and HL within each CSN top dysregulated pathways. Between FL-CT and FL-NP, 757
supraspinal subtypes are provided in Extended Data Figure 7-1. genes were upregulated, and 1068 genes were downregulated,
Golan et al. · Transcriptional Index of the Adult Corticospinal Tract J. Neurosci., November 22, 2023 • 43(47):7929–7945 • 7943

with glutamate receptor signaling and GABA receptor signaling train a limb-specific CSN linear classifier based on the top 100
among the dysregulated pathways (Fig. 7P). DE genes between FL and HL (Fig. 5N). These gene lists can be
For the HL-CSN-CT population, 425 genes were upregulated used in combination to identify FL and HL CSNs from a mixed
and 805 genes downregulated in HL-CSN-IT cells compared population of neurons in any single-cell sequencing dataset from
with HL-CSN-ET, HL-CSN-CT, and HL-CSN-NP combined the intact adult mouse brain. Additionally, we found that adult
(Fig. 7P’). Comparisons among the subcategories revealed that CNSs do not fit exclusively into the BICCN-defined extratelence-
between HL-CT and HL-ET, 1370 genes were upregulated, and phalic category (also known as pyramidal tract; Callaway et al.,
735 genes were downregulated, with synaptogenesis signaling 2021). Rather, they express a profile aligning with all nonspinally
and serotonin receptor signaling among the top pathways dysre- projecting categories IT, CT, and NP (Fig. 6A–D). Furthermore,
gulated. Between HL-CT and HL-IT, 749 genes were upregu- these new CSN subcategories CSN-ET, CSN-IT, CSN-CT, and
lated, and 523 genes were downregulated, with circadian rhythm CSN-NP each display a unique gene expression profile (Fig. 6E–J).
signaling and the opioid signaling pathway among the top Finally, we confirmed that CSNs innervate supraspinal structures
enriched pathways. Between HL-CT and HL-NP, 1109 genes consistent with projection categories ET, IT, CT, and NP, high-
were upregulated, and 1500 genes were downregulated, with va- lighting the capacity of the CST to influence supraspinal motor
line degradation and glutamate receptor signaling among the top processing.
dysregulated pathways (Fig. 7P’).
Molecular specialization among forelimb and hindlimb
CSN NP specialization CSNs
We found that 709 genes upregulated and 1163 genes downregu- CSN identity has been explored during development by combin-
lated in FL-CSN-NP cells compared with FL-CSN-ET, FL-CSN- ing retrograde tracing from the pons and upper cervical spinal
IT, and FL-CSN-CT combined (Fig. 7T). A more specific com- cord postnatally with temporal bulk microarray (Arlotta et al.,
parison among subcategories revealed that between FL-NP and 2005) and RNA sequencing (Molyneaux et al., 2015) of FACS-
FL-ET, 1808 genes were upregulated, and 630 genes were down- purified traced neurons. Authors identified Ctip2, Crym, and
regulated, with GADD45 signaling and senescence signaling Crim1 as CSN-specific genes up to P14, leaving the CSN tran-
among the top pathways dysregulated. Between FL-NP and FL- scriptome temporally and spatially incomplete. Building on these
IT, 1155 genes were upregulated, and 630 genes were downre- transformative studies, we combined spinal retrograde labeling
gulated, with cAMP signaling and GPCR signaling among the with scRNAseq to comprehensively profile FL and HL CSNs. We
top dysregulated pathways. Between FL-NP and FL-CT, 1068 were able to identify and anatomically confirm robust CSN-specific
genes were upregulated, and 757 genes were downregulated, and limb-specific genetic signatures that expand beyond previously
with cAMP signaling and GPCR signaling among the top dysre- identified single genes. Our findings revealed a novel constellation
gulated pathways. (Fig. 7T). of genes that can be used to assign CSN identity with greater fidel-
For the HL-CSN-NP, 1096 genes were upregulated, and ity/confidence over previously identified CSN markers.
1706 genes were downregulated in HL-CSN-NP cells compared Indeed, the power of CSN subtype identity was previously
with HL-CSN-ET, HL-CSN-IT, and HL-CSN-CT combined highlighted in a study that sought to explore transcriptional
(Fig. 7T’). Comparisons among the subcategories revealed that divergence among rostral- (FL) and caudal (HL)-projecting
between HL-NP and HL-ET 2193 genes were upregulated, and CSNs during postnatal patterning. Authors identified Klhl14
1234 genes were downregulated, with molecular mechanisms in and Crim1 as crucial for segment-specific axon targeting by
cancer signaling and axon guidance signaling among the top completing bulk microarray sequencing of neurons in the
pathways dysregulated. Between HL-NP and HL-IT 1758 genes rostral forelimb area (RFA) and comparing them with neu-
were upregulated, and 1377 genes were downregulated, with rons in caudal forelimb area and hindlimb M1 regions (Sahni
axon guidance signaling and the serotonin receptor signaling et al., 2021a,b). To elaborate on the findings, we explored
pathway among the top enriched pathways. Between HL-NP limb-specific differences in the adult. Neither Klhl14 or
and HL-CT 1500 genes were upregulated, and 1109 genes were Crim1 are significantly enriched in FL or HL CSNs in adult-
downregulated, with GPCR signaling and CREB signaling in hood, likely reflecting downregulation of patterning machin-
neurons among the top dysregulated pathways (Fig. 7T’). The ery in the adult. However, here, CSNs were harvested for
entire DE lists are available in Extended Data Figure 7-1. scRNAseq from 11.0 to 0.5 mm relative to bregma, poten-
tially undersampling CSNs from the RFA.

Discussion Functional specialization among forelimb and hindlimb


In this study we used a combination of viral retrograde labeling CSNs
from the cervical and lumbar spinal cord, FACS, and scRNAseq Functional subtypes among M1 CSNs were previously described
to annotate a comprehensive transcriptional index of adult using slice electrophysiology and morphologic analyses (Oswald
CSNs. Using a refined extraction, dissociation, and FACS pipe- et al., 2013). Unbiased clustering found that among cortical neu-
line, we were able to maintain cytoplasmic integrity of large-di- rons, FL- and HL-projecting CSNs fired action potentials at
ameter pyramidal neurons for whole-cell sequencing (Fig. 1; the fastest rate and with the most regular pattern, and that burst
Golan and Cafferty, 2021) of intact adult CSNs (Figs. 2, 3). Using firing was exclusive to FL CSNs. This is consistent with FL-
this approach, we identified 849 genes that were significantly dif- enriched expression of Cacng7, a type II transmembrane AMPA
ferentially expressed between CSNs and nonspinally projecting receptor regulatory protein that enhances glutamate-evoked cur-
L5 neurons. We built a linear classifier to confirm the veracity of rents from glutamate receptors (Kato et al., 2007).
this constellation of genes to identify CSNs with improved fidel- There is also evidence that FL and HL CSNs play separate
ity over a classifier trained on a set of established L5 genes (Fig. roles in sensorimotor function. A previous study showed that the
4C). Furthermore, we found that 1280 genes were differentially lumbar CST functions exclusively to gate sensory input via primary
expressed between FL and HL CSNs. These data allowed us to afferent depolarization, and CST-mediated motor commands to the
7944 • J. Neurosci., November 22, 2023 • 43(47):7929–7945 Golan et al. · Transcriptional Index of the Adult Corticospinal Tract

lumbar spinal cord are not direct but rather use polysynaptic cir- that CSNs terminate in multiple supraspinal locations. By aligning
cuits in the upper cervical cord (Moreno-Lopez et al., 2021). Our sequenced CSNs with previously published M1 sequencing data-
data support this finding as mice deficient in the HL-enriched gene sets, we found that CSNs are not all characterized solely as ET cells
Slc16a2 do not display gross motor abnormalities but do exhibit but rather span all four designated subtypes.
shorter latency paw withdrawal during a hot plate test, suggestive of There is also evidence that CSN supraspinal connectivity
hyperalgesia (Wirth et al., 2009; Moreno-Lopez et al., 2021). reflects functional differences among CSNs. Studies exploring FL-
This spinal enlargement level transcriptional resolution re- CSNs with supraspinal collaterals in the striatum found that even
solves broad FL and HL-level functional differences; however, among this one CSN subclass, there was anatomic and functional
there could also be within and between spinal segment specific- heterogeneity. Using intersectional polysynaptic tracing, authors
ity. Indeed, it was recently shown that different phases of FL found that CSNs synapse onto both D1 and D2 neurons in the
movement are coordinated by CSNs projecting to discrete spinal striatum and GAD2 and calbindin D28K interneurons in the spi-
segments (Ueno et al., 2018). nal cord. By combining two-photon imaging of FL-CSNs la-
beled via retro-AAV-GCaMP6f infusion into the cervical cord
Plasticity of forelimb and hindlimb CSNs with a lever-pulling task, authors found that striatal CSNs dis-
Data from our laboratory support the notion that FL and HL play three unique activity patterns during pulling onset, pulling
CSNs require independent interventions to stimulate functional offset, and throughout the task (Nelson et al., 2021). This study
axon growth. Previously, we completed bulk RNAseq of retro- provides robust evidence that supraspinal CST connectivity is
gradely traced intact CSNs after contralateral PyX-induced func- critical to refining motor function, further necessitating a need
tional sprouting into the denervated side. DE analyses revealed for a comprehensive evaluation of the functional phenotypes of
that members of the LPAR1-interacting pathway and the 3-phos- these molecularly divergent CSN subtypes.
phoinositide degradation pathway were enriched in sprouting In summary, from these data we conclude that CSNs in M1
CSNs. Viral overexpression of candidate genes within these path- are molecularly unique among nonprojection layer 2/3 and
ways in intact CSNs after PyX resulted in significant sprouting of layer 5 neurons and can be further subdivided based on the
intact CST axons into the denervated side of the spinal cord location of their spinal rostrocaudal and supraspinal terminal
(Fink et al., 2017; Kauer et al., 2022). Assessment of fine motor arbors. Leveraging this molecular heterogeneity provides a
skills showed that the lesioned FL performed better than the powerful opportunity to explore the transcriptional machinery
lesioned HL, suggesting that our treatments were having a that drives development, maintains homeostasis, and actuates
greater impact in FL CSNs. These data support limb-specific reactive changes after trauma and disease within this critical
therapeutic interventions as the progrowth factors we identi- motor pathway. Comprehensive transcriptional insight in
fied in our original in vivo screen were identified via retro- these physiological and maladaptive processes will enable
grade tracing from the cervical spinal cord. We hypothesize refined therapeutic interventions to repair the damaged
that tracing from the lumbar cord after contralateral PyX may CST with enhanced precision.
reveal a unique set of proaxon growth modulators.

CSNs segregate by supraspinal collateralization


References
Alcamo EA, Chirivella L, Dautzenberg M, Dobreva G, Fariñas I, Grosschedl
In addition to FL and HL specialization, CSNs can also be R, McConnell SK (2008) Satb2 regulates callosal projection neuron iden-
classified based on their supraspinal projections. We found tity in the developing cerebral cortex. Neuron 57:364–377.
that FL-CSNs innervate ET and NP structures more densely Arlotta P, Molyneaux BJ, Chen J, Inoue J, Kominami R, Macklis JD (2005)
than HL-CSNs. Furthermore, each of these subdivisions can Neuronal subtype-specific genes that control corticospinal motor neuron
development in vivo. Neuron 45:207–221.
also be defined transcriptionally, suggesting they each influ-
Bakken TE, et al. (2018) Single-nucleus and single-cell transcriptomes com-
ence fine motor processing in a previously underappreciated pared in matched cortical cell types. PLoS One 13:e0209648.
fashion. Their expression of a range of axon guidance ligands Blackmore MG, Moore DL, Smith RP, Goldberg JL, Bixby JL, Lemmon VP
and receptors, suite of G-protein coupled receptors and syn- (2010) High content screening of cortical neurons identifies novel regula-
aptogenesis signaling components suggest they may each sup- tors of axon growth. Mol Cell Neurosci 44:43–54.
port a unique response to injury, disease, and aging. In alignment Botta P, Fushiki A, Vicente AM, Hammond LA, Mosberger AC, Gerfen CR,
with these findings, previous work using scRNAseq of adult motor Peterka D, Costa RM (2020) An amygdala circuit mediates experience-
dependent momentary arrests during exploration. Cell 183:605–619.e22.
regions established a taxonomy of neuronal cell types (Tasic et al., Bradbury EJ, Burnside ER (2019) Moving beyond the glial scar for spinal
2016, 2018; Callaway et al., 2021; Yao et al., 2021). To describe the cord repair. Nat Commun 10:3879.
diversity observed among neuronal cell types spanning the cortical Buchser WJ, Slepak TI, Gutierrez-Arenas O, Bixby JL, Lemmon VP (2010)
depth in M1, cell-type-specific genes describing IT, ET, NP, and Kinase/phosphatase overexpression reveals pathways regulating hippo-
CT neurons were identified (Yao et al., 2021). The ET designation campal neuron morphology. Mol Syst Biol 6:391.
was confirmed in part by retrograde tracing from the pons and Burkhardt DB, Stanley JS 3rd, Tong A, Perdigoto AL, Gigante SA,
Herold KC, Wolf G, Giraldez AJ, van Dijk D, Krishnaswamy S (2021)
medulla and scRNAseq of ALM neurons (Economo et al., 2018;
Quantifying the effect of experimental perturbations at single-cell re-
Tasic et al., 2018). However, these studies did not include tracing solution. Nat Biotechnol 39:619–629.
from the spinal cord, thus rendering a full ET atlas incomplete. Callaway EM, et al. (2021) A multimodal cell census and atlas of the mamma-
Previous studies have combined scRNAseq with single-cell recon- lian primary motor cortex. Nature 598:86–102.
structions from sparsely labeled L5 neurons. These studies identi- Canty AJ, Murphy M (2008) Molecular mechanisms of axon guidance in the
fied a subtype of L5 ET neurons with projections in the spinal developing corticospinal tract. Prog Neurobiol 85:214–235.
cord, in addition to many supraspinal regions (Callaway et al., Chon U, Vanselow DJ, Cheng KC, Kim Y (2019) Enhanced and unified ana-
tomical labeling for a common mouse brain atlas. Nat Commun 10:5067.
2021; Peng et al., 2021). Furthermore, axonal barcoding of the Economo MN, Viswanathan S, Tasic B, Bas E, Winnubst J, Menon V,
CST revealed that many CSNs innervate a range of supraspinal Graybuck LT, Nguyen TN, Smith KA, Yao Z, Wang L, Gerfen CR,
structures in addition to their terminal fields in the spinal cord Chandrashekar J, Zeng H, Looger LL, Svoboda K (2018) Distinct descending
(Hausmann et al., 2022). These data are consistent with our findings motor cortex pathways and their roles in movement. Nature 563:79–84.
Golan et al. · Transcriptional Index of the Adult Corticospinal Tract J. Neurosci., November 22, 2023 • 43(47):7929–7945 • 7945

Farrar MJ, Bernstein IM, Schlafer DH, Cleland TA, Fetcho JR, Schaffer CB Morris JA, et al. (2010) Divergent and nonuniform gene expression patterns
(2012) Chronic in vivo imaging in the mouse spinal cord using an in mouse brain. Proc Natl Acad Sci U S A 107:19049–19054.
implanted chamber. Nat Methods 9:297–302. Muñoz-Castañeda R, et al. (2021) Cellular anatomy of the mouse primary
Fawcett JW (2020) The struggle to make CNS axons regenerate: why has it motor cortex. Nature 598:159–166.
been so difficult? Neurochem Res 45:144–158. Nelson A, Abdelmesih B, Costa RM (2021) Corticospinal populations broad-
Fink KL, Strittmatter SM, Cafferty WB (2015) Comprehensive corticospinal cast complex motor signals to coordinated spinal and striatal circuits. Nat
labeling with mu-crystallin transgene reveals axon regeneration after spi- Neurosci 24:1721–1732.
nal cord trauma in ngr1-/- Mice. J Neurosci 35:15403–15418. Oswald MJ, Tantirigama ML, Sonntag I, Hughes SM, Empson RM (2013)
Fink KL, López-Giráldez F, Kim IJ, Strittmatter SM, Cafferty WBJ (2017) Diversity of layer 5 projection neurons in the mouse motor cortex. Front
Identification of intrinsic axon growth modulators for intact CNS neu- Cell Neurosci 7:174.
rons after injury. Cell Rep 18:2687–2701. Park KK, Liu K, Hu Y, Smith PD, Wang C, Cai B, Xu B, Connolly L, Kramvis
Gianino S, Stein SA, Li H, Lu X, Biesiada E, Ulas J, Xu XM (1999) Postnatal I, Sahin M, He Z (2008) Promoting axon regeneration in the adult CNS
growth of corticospinal axons in the spinal cord of developing mice. by modulation of the PTEN/mTOR pathway. Science 322:963–966.
Brain Res Dev Brain Res 112:189–204. Pedregosa F, Varoquaux G, Gramfort A, Michel V, Thirion B, Grisel O,
Golan N, Cafferty WB (2021) Dissociation of intact adult mouse cortical pro- Blondel M, Prettenhofer P, Weiss R, Dubourg V, Vanderplas J, Passos A,
jection neurons for single-cell RNA-seq. STAR Protoc 2:100941. Cournapeau D, Brucher M, Perrot M, Duchesnay E (2011) Scikit-learn:
Gutilla EA, Steward O (2016) Selective neuronal PTEN deletion: can we take the machine learning in Python. J Mach Learn Res 12:2825–2830.
brakes off of growth without losing control? Neural Regen Res 11:1201– Peng H, et al. (2021) Morphological diversity of single neurons in molecu-
1203. larly defined cell types. Nature 598:174–181.
Hausmann FS, Barrett JM, Martin ME, Zhan H, Shepherd GMG (2022) Poplawski GHD, Kawaguchi R, Van Niekerk E, Lu P, Mehta N, Canete P, Lie
Axonal barcode analysis of pyramidal tract projections from mouse fore- R, Dragatsis I, Meves JM, Zheng B, Coppola G, Tuszynski MH (2020)
limb M1 and M2. J Neurosci 42:7733–7743. Injured adult neurons regress to an embryonic transcriptional growth
He Z, Jin Y (2016) Intrinsic control of axon regeneration. Neuron 90:437–451. state. Nature 581:77–82.
Hilton BJ, Bradke F (2017) Can injured adult CNS axons regenerate by reca- Sahni V, Itoh Y, Shnider SJ, Macklis JD (2021a) Crim1 and Kelch-like 14 exert
pitulating development? Development 144:3417–3429. complementary dual-directional developmental control over segmentally
Kato AS, Zhou W, Milstein AD, Knierman MD, Siuda ER, Dotzlaf JE, Yu H, specific corticospinal axon projection targeting. Cell Rep 37:109842.
Hale JE, Nisenbaum ES, Nicoll RA, Bredt DS (2007) New transmembrane Sahni V, Shnider SJ, Jabaudon D, Song JHT, Itoh Y, Greig LC, Macklis JD
AMPA receptor regulatory protein isoform, gamma-7, differentially regu- (2021b) Corticospinal neuron subpopulation-specific developmental
lates AMPA receptors. J Neurosci 27:4969–4977. genes prospectively indicate mature segmentally specific axon projection
targeting. Cell Rep 37:109843.
Kauer SD, Fink KL, Li EHF, Evans BP, Golan N, Cafferty WB (2022) Inositol
Saunders A, Macosko EZ, Wysoker A, Goldman M, Krienen FM, de Rivera H,
polyphosphate-5-phosphatase K (Inpp5k) enhances sprouting of cortico-
Bien E, Baum M, Bortolin L, Wang S, Goeva A, Nemesh J, Kamitaki N,
spinal tract axons after CNS trauma. J Neurosci 42:2190–2204.
Brumbaugh S, Kulp D, McCarroll SA (2018) Molecular diversity and special-
Kim EJ, Zhang Z, Huang L, Ito-Cole T, Jacobs MW, Juavinett AL, Senturk G,
izations among the cells of the adult mouse brain. Cell 174:1015–1030.e16.
Hu M, Ku M, Ecker JR, Callaway EM (2020) Extraction of distinct neuro-
Schwab ME, Strittmatter SM (2014) Nogo limits neural plasticity and recov-
nal cell types from within a genetically continuous population. Neuron
ery from injury. Curr Opin Neurobiol 27:53–60.
107:274–282.e6.
Sekine Y, Lin-Moore A, Chenette DM, Wang XX, Jiang ZX, Cafferty WB,
Krämer A, Green J, Pollard J Jr, Tugendreich S (2014) Causal analysis approaches
Hammarlund M, Strittmatter SM (2018) Functional genome-wide screen
in Ingenuity Pathway Analysis. Bioinformatics 30:523–530.
identifies pathways restricting central nervous system axonal regenera-
Lein ES, et al. (2007) Genome-wide atlas of gene expression in the adult
tion. Cell Rep 24:269–269.
mouse brain. Nature 445:168–176.
Tasic B, et al. (2016) Adult mouse cortical cell taxonomy revealed by single
Lemon RN (2008) Descending pathways in motor control. Annu Rev cell transcriptomics. Nat Neurosci 19:335–346.
Neurosci 31:195–218. Tasic B, et al. (2018) Shared and distinct transcriptomic cell types across neo-
Lin HM, Kuang JX, Sun P, Li N, Lv X, Zhang YH (2018) Reconstruction of cortical areas. Nature 563:72–78.
intratelencephalic neurons in the mouse secondary motor cortex reveals Ueno M, Nakamura Y, Li J, Gu Z, Niehaus J, Maezawa M, Crone SA,
the diverse projection patterns of single neurons. Front Neuroanat 12:86. Goulding M, Baccei ML, Yoshida Y (2018) Corticospinal circuits from
Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, Tirosh I, the sensory and motor cortices differentially regulate skilled movements
Bialas AR, Kamitaki N, Martersteck EM, Trombetta JJ, Weitz DA, Sanes through distinct spinal interneurons. Cell Rep 23:1286–1300.e7.
JR, Shalek AK, Regev A, McCarroll SA (2015) Highly parallel genome- van Dijk D, Sharma R, Nainys J, Yim K, Kathail P, Carr AJ, Burdziak C,
wide expression profiling of individual cells using nanoliter droplets. Cell Moon KR, Chaffer CL, Pattabiraman D, Bierie B, Mazutis L, Wolf G,
161:1202–1214. Krishnaswamy S, Pe’er D (2018) Recovering gene interactions from sin-
Martin JH (2005) The corticospinal system: from development to motor con- gle-cell data using data diffusion. Cell 174:716–729.e27.
trol. Neuroscientist 11:161–173. Wang Z, Maunze B, Wang Y, Tsoulfas P, Blackmore MG (2018) Global con-
McQuin C, Goodman A, Chernyshev V, Kamentsky L, Cimini BA, Karhohs nectivity and function of descending spinal input revealed by 3D micros-
KW, Doan M, Ding L, Rafelski SM, Thirstrup D, Wiegraebe W, Singh S, copy and retrograde transduction. J Neurosci 38:10566–10581.
Becker T, Caicedo JC, Carpenter AE (2018) CellProfiler 3.0: next-genera- Wirth EK, Roth S, Blechschmidt C, Hölter SM, Becker L, Racz I, Zimmer A,
tion image processing for biology. PLoS Biol 16:e2005970. Klopstock T, Gailus-Durner V, Fuchs H, Wurst W, Naumann T, Bräuer
Milich LM, Choi J, Ryan C, Yahn SL, Tsoulfas P, Lee JK (2021) Single cell A, de Angelis MH, Köhrle J, Grüters A, Schweizer U (2009) Neuronal
analysis of the cellular heterogeneity and interactions in the injured 3’,3,5-triiodothyronine (T3) uptake and behavioral phenotype of mice de-
mouse spinal cord. J Exp Med 218:e20210040. ficient in Mct8, the neuronal T3 transporter mutated in Allan-Herndon-
Molyneaux BJ, Goff LA, Brettler AC, Chen HH, Hrvatin S, Rinn JL, Arlotta P Dudley syndrome. J Neurosci 29:9439–9449.
(2015) DeCoN: genome-wide analysis of in vivo transcriptional dynamics Wolock SL, Lopez R, Klein AM (2019) Scrublet: computational identification
during pyramidal neuron fate selection in neocortex. Neuron 85:275–288. of cell doublets in single-cell transcriptomic data. Cell Syst 8:281–291.e9.
Moon KR, van Dijk D, Wang Z, Gigante S, Burkhardt DB, Chen WS, Yim K, Yao Z et al. (2020) An integrated transcriptomic and epigenomic atlas of
Elzen AVD, Hirn MJ, Coifman RR, Ivanova NB, Wolf G, Krishnaswamy mouse primary motor cortex cell types. Nature 598:103–110.
S (2019) Visualizing structure and transitions in high-dimensional bio- Yao Z, et al. (2021) A transcriptomic and epigenomic cell atlas of the mouse
logical data. Nat Biotechnol 37:1482–1492. primary motor cortex. Nature 598:137– 143.
Moreno-Lopez Y, Bichara C, Isope P, Cordero-Erausquin M (2021) Sensory Zhang M, Eichhorn SW, Zingg B, Yao Z, Cotter K, Zeng H, Dong H, Zhuang
gating precedes motor command in evolution of the corticospinal tract. X (2021) Spatially resolved cell atlas of the mouse primary motor cortex
eLife 10:e65304. by MERFISH. Nature 598:137–143.

You might also like