Science Abp9262
Science Abp9262
C
resolution (Fig. 1, D and E, and table S1). We
omparing brains between animals has neurogenesis throughout life; however, after performed immunofluorescence stainings and
been a means to analyze the evolution- brain injury neurogenesis is almost absent in situ RNA hybridization chain reaction (HCR)
ary origin and diversification of this (10, 11). The molecular relationship between to localize major cell types in the tissue accord-
structure. Comprehensive single-cell RNA neurogenesis seen in salamanders and mam- ing to marker expression (Fig. 1F). Glial fibrillary
sequencing (scRNA-seq) and single- mals has not been explored. Furthermore, acidic protein+ (GFAP+) ependymoglia line
nucleus RNA sequencing (snRNA-seq) have similarities and differences between homeo- the ventricle of every region, whereas Mex3a+
increased the resolution of cell identity and static and regenerative neurogenesis in the neuroblasts are sparsely distributed along the
development of the vertebrate brain. Cell types salamander brain are unclear. ventricle. GABAergic neurons were sparsely
from the dorsal region of the telencephalon— To understand the organizational features distributed along the medial, dorsal, and lateral
which in mammals includes the hippocampus, of the axolotl telencephalon, we used single- region and clustered together densely in the
amygdala, claustrum, olfactory (piriform) cortex, nuclei genomic methods and spatial profiling. striatum, whereas glutamatergic neurons were
and elaborate neocortex—have been resolved We analyzed cell types present in different re- located in the medial, dorsal, lateral, and ven-
and compared between amniotes, including gions and defined their similarities to amniote tral pallium. Small populations of myelin basic
turtle, lizards, birds, mouse, and human (1–4). telencephalic cell types. To delineate the cellu- protein+ (MBP+) oligodendrocytes and ion-
These studies have revealed evolutionary rela- lar and molecular dynamics of homeostatic ized calcium binding adaptor molecule 1+
tionships in cell types and brain regions among neurogenesis in the axolotl and its relation to (IBA1+) microglia cells were dispersed through-
amniotes; however, little is known about their adult neurogenesis in mice, we used clonal out all regions.
conservation in other vertebrates. tracing, trajectory analysis, and multiomic We next analyzed the abundance of each
The axolotl (Ambystoma mexicanum), as an sequencing. Analyzing regenerative neuro- cell cluster in the sampled pallial regions (Fig.
amphibian, represents one of the closest living genesis, we determined the similarities and 1G and fig. S1F). Oligodendrocytes, microglia,
relatives to amniotes and is therefore suited differences to homeostatic neurogenesis and and endothelial cell clusters originated from
for comparative studies of brain cell types, neu- found that regenerated neurons reestablish each region in similar proportions. By contrast,
ronal connectivity, and function. The axolotl is neuronal input from other regions of the ependymoglia, neuroblast, glutamatergic, and
also able to regenerate the telencephalon after telencephalon. Together, our comprehensive GABAergic neuron clusters showed variable
removal of the dorsal region by activating analyses of the axolotl telencephalon yielded region contributions, with some cell clusters
neurogenesis (5), which is also present during insights into the organization, evolution, and being completely region-restricted, whereas
post-embryonic life (6). Neurogenesis can be regeneration of a tetrapod nervous system. others could be found throughout the sampled
found in all metazoans with a nervous system pallium regions. These data provide an overview
(7–9), and in the mouse, cells of the adult sub- snRNA-seq of the axolotl telencephalon of cell populations in the axolotl telencephalon
ventricular zone (SVZ) undergo continuous We used snRNA-seq to generate a comprehen- and suggest substantial regional specificity in
sive dataset of the cell types and states from the neurogenic programs.
1
Research Institute of Molecular Pathology, Vienna Biocenter
axolotl telencephalon. We microdissected the
telencephalon into medial (containing medial Regional conservation of axolotl
(VBC), Campus Vienna Biocenter, 1030 Vienna, Austria.
2
Department of Biosystems Science and Engineering, ETH pallium and septum), dorsal (containing dorsal glutamatergic neurons
Zürich, 4058 Basel, Switzerland. 3Roche Institute for
pallium), and lateral (containing lateral pallium, Glutamatergic neurons of the amniote telen-
Translational Bioengineering (ITB), Roche Pharma Research and
Early Development, Roche Innovation Center Basel, Basel, ventral pallium, and dorsal striatum) regions. cephalon show a high degree of transcrip-
Switzerland. 4University of Basel, 4001 Basel, Switzerland. Additionally, we profiled all of these regions as a tomic diversification (1, 2). Our data revealed
*Corresponding author. Email: [email protected] (K.L.); whole with single-nucleus multiome sequenc- 29 glutamatergic neuron clusters differentially
[email protected] (E.M.T.); [email protected]
(B.T.) ing [snRNA-seq and single-nucleus assay for distributed in medial, dorsal, and lateral re-
†These authors contributed equally to this work. transposase-accessible chromatin with high- gions (Fig. 2, A to C, fig. S2A, and table S1).
A snRNA-seq analysis of E Marker gene sets for 95 molecularly distinct cell types G EPEN12
EPEN10
the axolotl telencephalon Dorsal EPEN2
EPEN6
Ependymoglia
EPEN14
EPEN1
EPEN7
EPEN0
EPEN3
Neuroblast
95 clusters NB2
NB7
NB9
NB4
1
Expression
NB8
NB12
NB0
NB14
NB10
UMAP 2
NB6
0
GLUT14
GLUT7
UMAP 1 GLUT4
All Clusters GLUT18
GLUT19
GABA Endo F GLUT0
GLUT15
Glut MGs GFAP Mex3a GLUT6
GLUT17
Glutamatergic
GLUT20
GLUT16
C Region
GLUT23
GLUT12
GLUT24
GLUT3
GLUT8
GLUT1
GLUT21
GLUT27
GLUT25
GLUT26
GLUT2
GLUT28
M D L M D L GLUT9
GLUT22
Gad2 Slc17a7 GLUT10
GABA24
GABA8
Medial GABA5
GABA0
Dorsal GABA21
Lateral GABA20
GABA16
Undetermined GABA6
GABA2
GABA23
D Gli2 G2M Score Mex3a
GABA27
GABA7
GABA18
GABAergic
GABA17
GABA12
GABA4
GABA29
GABA14
GABA10
M D L M D L GABA28
GABA22
MBP IBA1 GABA3
GABA9
GABA11
Snap25 Gad2 Slc17a6/7 GABA19
GABA15
GABA13
GABA26
GABA1
GABA25
OLIG10
OLIG15
Others
MG8
ENDO11
ENDO12
ENDO14
Expression
1 Max 100 % per region 0
Fig. 1. Cellular diversity of the axolotl telencephalon. (A) Schematic ependymoglia), Mex3a (neuroblasts), Snap25 (neurons), Gad2 (GABAergic
highlighting the regions of the axolotl telencephalon used for snRNA-seq. neurons), and Slc17a6/7 (glutamatergic neurons). (E) Heatmap illustrates the
(B) UMAP of all cell types across all regions colored by cell-type class. Subtypes expression of marker genes (table S1) for the 95 distinct cell types. (F) Antibody
are shown in different shades. GABA, GABAergic neuron; Glut, glutamatergic stainings and HCR for main cell types: GFAP (ependymoglia), Mex3a (neuroblasts),
neuron; NB, neuroblast; Epen, ependymoglia cell; Endo, endothelial cell; MGs, Gad2 (GABAergic neurons), Slc17a7 (glutamatergic neurons), MBP (oligodendro-
microglia; and Oligo, oligodendrocyte. (C) UMAP plot of regional distribution of cytes), and IBA1 (microglia cells). Scale bars, 100 mm. (G) Stacked barplots
all cell types. Shades indicate true region identity versus predicted regional illustrating the regional distribution of the populations of cells. GABA, GABAergic
identity. (D) UMAP plot of the expression of markers for ependymoglia, neuron; Glut, Glutamatergic neuron; NB, neuroblast; Epen, ependymoglia cell; Endo,
neuroblast, and neuronal cell types: Gli2 (ependymoglia), G2M score (active endothelial cell; MGs, microglia; Oligo, oligodendrocyte.
Max
27
axolotl glutamatergic neuron 28
Expression
Ndst4
Reln
Rbfox3
Nrxn3
Sema5a
Elmo1
Znf385b
Cdh11
Pik3cb
C1ql3
Sorcs1
Vwa5b1
Dnah3
Fstl4
Inpp5a
Spock1
Satb1
Arpp21
Crhr1
Rasa2
Nptx2
Pdzd2
Nfatc3
C1ql1
Tnfaip8l3
Rtn4rl1
Kcng1
Nxph1
Asic4
Scn5a
Grid1
Gria4
Htr5a
Tacr1
Adcy8
Cdh12
Cacng3
Elfn1
Cox1
Sim1
Shisa6
Tmem132c
Prkg2
Khdrbs2
Adcy2
Gpr22
Mchr2
Trpc6
Sox6
Syt6
Cntn5
Lhx6
Trpc7
Ptprt
Pex5l
Arhgap27
Nmbr
Cntfr
Bdnf
Medial
types and turtle glutamatergic Dorsal
Lateral
neuron types [data are from Undetermined
0
(2)]. Vertical lines indicate
highest correlated cluster to
D CA DG pDC aDC DVR PT LC
Medial
Dorsal
Lateral
E
Undeter. Cluster 11
Dorsal
turtle, and horizontal lines 22
|
* 25
|
indicate highest correlated
Elmo1 9
|
cluster to axolotl. (Right) 20
Cluster Score
10
0.10
|
*
Stacked barplots illustrating 24
|
0.05
| | |
11 Cluster 1
|
cells from each cluster. 1 | | |
|
|
|
Asterisks indicate clusters 2
|
21
|
with agreeing assignments | |
8
|
* | | | | | |
Cluster Score
between all genes correlation 6
|
0.20
* |
Satb1 13
|
and integration analysis. CA, 28
| |
0.05
| | | |
cornu ammonis; DG, dentate 27 | Cluster 6
26
|
* | |
gyrus; pDC and aDC, posterior 16
|
* |
3
|
* | | |
DVR, dorsal ventricular ridge; Region * 5
Cluster Score
0.16
| |
12
|
aDVR
|
lateral cortex. (E) Spatial
0.04
18
|
|
Rbfox3 pDVR 4 Cluster 7
|
*
mapping of glutamatergic 7
| |
|
CA1 * |
neuron clusters 11, 1, 6, and 7. 14
|
CA3 |
15
|
*
(F) Heatmap illustrating the DG
e34
e33
e38
e37
e36
e35
e32
e31
e30
e29
e28
e27
e26
e24
e16
e15
e14
e13
e08
e07
e25
e23
e22
e21
e20
e19
e18
e02
e01
e06
e05
e04
e03
e17
e12
e11
e10
e09
0
100
% per
Cluster Score
pDC
expression of Satb1, Rorb,
3
region
aLC
Reln, Grik1, and Tbr1. pLC
Spearman’s rho Turtle Glutamatergic Clusters Highest Corr. | -log10 adj. p-value
1
|
(G) HCR for Satb1 and Rorb. Expression PT -0.13 0.00 0.17 Axolotl Turtle 4 8 12 16
1 Max
(H) Projection mapping of
input into glutamatergic F 1
G H Neurobiotin
tracing
1. Main OB 2. Acc. OB 3. Injection site 4. Caudal T. 5. Thalamus
3
cluster 1 by using Neurobiotin- 5
1
2
mediated anterograde and 7
9 3
retrograde tracing. Dots indi- 11
4
13 Satb1
cate locations of cell bodies, 15 5
Max
17
and lines indicate locations of 19
Expression
21
fibers. Main OB, main olfactory 24 Rorb
26 D
bulb; Acc. OB, accessory 28
Satb1
M L
0
DAPI
Satb1
Grik1
Rorb
Tbr1
Reln
Potential homologies of axolotl glutamatergic cordant similarities, using both gene sets as (12, 13). We found that the majority of medial
neurons to telencephalon neurons were probed indicators of conservation. In parallel, cluster glutamatergic clusters (Glut4, -5, -7, -15, and
with two separate comparisons, by using either similarities were also assessed through inte- -18) and two dorsal clusters (Glut12 and -16)
species-shared differentially expressed genes or gration of single-cell and single-nuclei datasets showed highest correlation to either turtle
species-shared differentially expressed tran- from axolotl, turtle pallium (2), and mouse cornu amonis (CA) or dentate gyrus (DG)
scription factors (TFs) to distinguish between telencephalon (3). clusters (Fig. 2D and fig. S2B). Correlations to
convergent evolution versus homology (Fig. 2D; Anatomical and functional evidence sug- the mouse dataset also revealed that medial
fig. S2, B to F; and tables S2 and S3). We focused gests that the amphibian medial pallium is glutamatergic clusters (Glut5, -7, -14, -17, -24, and
then on glutamatergic clusters that have con- homologous to the mammalian hippocampus -27) correlated most to hippocampus-related
clusters (fig. S2, D to F). Data integration into the region containing Glut1 (Fig. 2H). level of effector genes, suggesting evolution-
additionally showed co-clustering of a large Labeling of cell bodies in the main and ac- ary divergence of these cells. Together, these
number of medial glutamatergic clusters to cessory olfactory bulb, caudal pallium (lateral data show that the axolotl telencephalon con-
either turtle or mouse hippocampus clusters amygdala) (19), and the thalamus indicate tains putative LGE-derived, CGE-derived, MGE-
(fig S3, A to E). To reveal the locations of that neurons in these regions project into the derived, and septal GABAergic neurons and
hippocampus-correlated glutamatergic clusters Glut1-containing domain. These data show a that LGE-like striatum and olfactory bulb
and unravel a potential subdivision into CA strong correspondence between presence of classes have strong transcriptional similarities
or DG, we performed spatial transcriptomics neurons with transcriptional similarity to between axolotl, turtle, and mouse (tables S2
using Visium (10x Genomics), resulting in spa- amniote olfactory cortex neurons and presence and S3).
tial resolution of approximately 1 to 30 cells of projections consistent with a role in olfac- In mammals and turtles, MGE-derived and
(Fig. 2E and figs. S4A and S5) (14). We found tory processing. CGE-derived interneurons are differentially dis-
that all aforementioned clusters except Glut18 tributed across cortical layers (2, 21). In axolotl,
showed exclusive localization in the medial Amniote-conserved signatures of axolotl we found MGE-like neurons [Somatostatin+
pallium; however, clear subdivisions were not GABAergic neurons (Sst+) and Satb1+] equally distributed in the
detectable, which was further confirmed by We identified 30 clusters (n = 15,665 cells) of medial pallium and enriched in the outer re-
means of HCR for ETS translocation variant 1 GABAergic (Gad1+/Gad2+) neurons (Fig. 3, A to gions of the dorsal and lateral pallium (Fig. 3E
(Etv1) (CA), Prospero homeobox 1 (Prox1), and B, and table S1) in the axolotl. In many ver- and fig. S8A). CGE-like neurons [Zinc finger
LIM domain only 3 (Lmo3) (DG) (fig. S4B). tebrates, GABAergic interneurons are born and BTB domain containing 16+ (Zbtb16+)]
These data show that neurons of the axolotl in the lateral, caudal, and medial ganglionic were equally distributed in the medial pallium
medial pallium have transcriptional similarities eminences (LGE, CGE, and MGE, respectively) but closer to the ventricle in the dorsal pallium.
to amniote hippocampal neurons, but a clear and migrate to the pallium during develop- LGE-like striatal GABAergic neurons [Forkhead
distinction into CA1, CA3, and DG cannot be ment (20). Comparative scRNA-seq analyses box P1+ (Foxp1+)] were found exclusively in the
| |
n=15,665
neuron types. (A) UMAP 2 26 * 29
11 * 3
| | | | |
plots of 30 GABAergic 19 * 10
20 18 15 13 * 1 |
14
| | | |
* 8 |
GABAergic neurons types, 16 7 27 * 5
1 * 24
as well as two marker 12 25 * 9
| |
UMAP 2
genes, Lhx6 and Meis2. 21 * 11 | |
10 29 4
|
(B) Heatmap illustrating 3 * 13
|
UMAP 1 8 * 19
|
the expression of differen- 5 28 * 26
|
0 * 15
| |
tial expressed markers for 24 Expression
* 14
* 2 | | |
|
each GABAergic neuron 1 Max
* 17 |
| |
Region Lhx6 Meis2 * 6 | |
cluster. (C) Heatmap illus- * 20 | |
|
* 7 |
|
trating the expression of 23
|
* 16 | | |
|
TFs known to define 18
|
GABAergic subtypes (LGE-, 21
|
* 27
|
CGE-, and MGE-derived) * 28
i16 |
Foxp1
Tshz1
Pbx3
Prox1
Nr2e1
Satb1
Bcl11a
Lmo3
Etv1
Foxp2
Meis2
Nr2f2
Id2
Z eb 2
Sp8
Lhx6
Sox6
Zbtb16
A rx
100
i04
i05
i06
i14
i15
i17
i18
i07
i08
i09
i10
i11
i12
i13
i02
i03
Lateral
0
Dorsal
i01
Medial Undetermined
for each axolotl GABAergic % per
B region
neuron type. (Right) Nxph1 Highest Match
Turtle GABAergic Clusters
St6galnac5 Expression Spearman’s rho | -log10 adj. p-value
Stacked barplots illustrat- 0 Max Medial Lateral
|
Arhgap6 10 20 Dorsal Undetermined
Kcnab1 0.4 0 -0.2 Axolotl Turtle
ing the regional distribution
Gad2 DAPI
Unc13b
analysis between expres- Synpr
Adarb2 2
sion profiles of axolotl Ncam2
Arpp21
GABAergic neuron types Sulf2 1
Scube2
and turtle GABAergic neu- Shisa6
Adamts6 0
ron types [data are from Pnoc
Ptprd
(2)]. Vertical lines indicate Cntfr 4
DAPI
Baiap3
highest correlated cluster Kl
Dach1
to turtle, and horizontal Dach2 2
Sst
Cpne6
lines indicate highest Syt6
Prkd1
correlated cluster to turtle. Foxp1 0
Bcl11b
Asterisks indicate Snca 4
Ebf1
clusters with agreeing
DAPI
Nova1 3
Gpr149
assignments between all Tac1
Asic4 2
genes correlation and
Satb1
Oprk1
Cdh13 1
integration analysis. Sep., Sorcs1
Tacr3
Septum. (E) HCR and Cntn5 0
Npy
snRNA-seq quantifications Tafa1
Slc17a6 3
Zbtb16 DAPI
Col19a1
Nmur1
Crtac1 2
Rxfp1
Kctd8
Shisa9 1
Cdh23
Brinp1
0
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
0
Expression
Axolotl GABAergic Clusters 0 Max
known to be expressed in the developing me- enriched for expression of Epidermal growth tains three main ependymoglia types (quiescent,
dial pallium in human, mouse, and chicken factor receptor kinase substrate 8-like protein active, and pro-neuro) that divide into regionally
(23). HCR confirmed expression of Wnt2b, 2 (Eps8l2). Because Wnt and Sfrp genes have distinct subtypes and, barring pro-neuro epen-
Wnt3a, and b-catenin target gene Axin2 in a been implicated in patterning of the develop- dymoglia, continue to express pallial patterning
restricted domain of the medial pallium at ing pallium, we performed HCR in embryonic genes in the postembryonic brain.
the border to the septum. Lateral and ventral axolotl brains (stage 44). We detected Wnt3a
ependymoglia showed strong expression of in medial ventricular cells and Sfrp1 lateral Identification of glutamatergic and
Secreted frizzled-related protein (Sfrp1), a and ventral ventricular cells; however, Eps8l2 GABAergic neuroblasts
gene known to be expressed in the antihem was not expressed (Fig. 4E and fig. S9F). These In addition to ependymoglia, we identified
in amniotes (23). Dorsal ependymoglia were data show that the axolotl telencephalon con- cells that we termed neuroblasts because of
Wnt3a
n=3,590 3 Wnt7b
Active 5 Irs1
Etv1 DAPI
4 Cdk14
14 Ephb2
Gli2
Sfrp1 Wnt2b
9 Wnt5b
7 Nox4
13 Meis2 8
Chk1
1 10 6 Fgfr3
0 Ski
Pro-Neuro Shisa6
St6galnac5
Grin1
UMAP 2
Eps8l2
Gria3
12 2 Grin2b
Quiescent Kif1a
UMAP 1 Fgf14
Hip1
B Region
Axin2 DAPI
Notch2 Edn3 Grm3
Eps8l2
Grm8
Lrrtm4
Edn3
Wnt8b
Wnt2b
Wnt3a
Rspo2 Wnt3a D Sfrp1
Medial Lateral Expression Fgfrl1 E Stage 44
D
DAPI DAPI
Dorsal Undetermined 1 Max Ar M M
Sfrp1 P
OT
HB L L
Notch1
OT
0
0
100
4
3
6
5
11
10
12
2
0
9
8
13
1
7
14
F
Expression
0 Max Ependymoglia clusters
Neuroblasts
9
3 1
n=1,501 Slc17a6/7 I Ependymal B cells A cells C Mitosis
2 * 2
|
Late NB * 12
|
11 VGLUT+ * 13
|
13 14 3
*
| | | |
4 * 0
0
Neuroblasts
* 14
7
Axolotl neuroblaset and ependymoglia clusters
Gad2 * 1
* 9
|
Early 12 G * 7
|
1.00
|
8 10 * 6
||
NBs 5
G2M Score
* 5
* 10
|
|
6 GABA+ * 11
|||
0.25
UMAP 2
* 8 | | | | | |
* 4
* 1 |
|| |
UMAP 1 12
E.a E.q E.pn NB.e NB *
|
* 2 | |
H * 6 | | |
Ependymoglia
10
|
11 | | |
Medial
| | | | |
9
5
* 0
Slc17a7 8
Slc17a7 Mex3a
4 |
|
*
Mex3a Mex3a 13
|
DAPI 7
|
Dorsal
2
23
32
34
5
14
13
22
6
1
4
0
15
12
10
8
16
17
30
Fig. 4. Axolotl telencephalic ependymoglia and neuroblasts. (A) (Left) telencephalon. Scale bars, 50 mm. (F) UMAP plots of the regional distribution
Morphology of an axolotl ependymoglia cell and (right) UMAP plot of 15 neuroblast clusters. Black boxes outline the four main cell types: early
15 ependymoglia clusters. Boxes outline the three main cell types: quiescent neuroblasts, VGLUT+ and GABA+ neuroblasts, as well as late neuroblasts.
ependymoglia, active ependymoglia, and pro-neuro ependymoglia. (B) UMAP plot The red box outlines the early neuroblast population. (G) Boxplot of G2M score
of the regional distribution of ependymoglia and UMAP plots colored by for quiescent, active, and pro-neuro ependymoglia as well as neuroblasts.
expression for Notch2 (quiescent and active ependymoglia), Edn3 (quiescent (H) HCR for Slc17a7, Gad2, and Mex3a. Arrows indicate coexpressing cells. Scale
ependymoglia), G2M score (active ependymoglia), Notch1 (active ependymoglia), bars, 10 mm. (I) Correlation analysis between expression profiles of axolotl
and Grin1 (pro-neuro ependymoglia). (C) Heatmap illustrating the expression neuroblasts, ependymoglia, and adult mouse SVZ cell types [scRNA-seq data are
of differentially expressed cell-to-cell communication–related genes in quiescent, from (25)]. Vertical lines indicate highest correlated cluster to mouse, and
active, and pro-neuro ependymoglia. (D) HCR and snRNA-seq quantifications horizontal lines indicate highest correlated cluster to axolotl. Asterisks indicate
for Wnt2b, Wnt3a, Eps8l2, Sfrp1, and Axin2 in medial, dorsal, and lateral regions. clusters with agreeing assignments between all genes correlation and integration
Scale bars, 25 mm. (E) HCR for Wnt3a and Sfrp1 in the stage 44 developing analysis. C, C cells.
the expression of Mex3a, an RNA-binding pro- these two clusters showed strongest correla- intermediates. We identified many genes with
tein expressed in proliferating neuroblasts in tion to A cell clusters 6 and 15, which were trajectory-specific varying expression along
the Xenopus laevis central nervous system (24). defined previously as dividing neuroblasts and pseudotime, but also some genes with pseudo-
Additionally, neuroblasts were characterized early A cells, as well as to IP cell clusters in the temporal expression consistent across trajecto-
by the absence of ependymoglia markers such developmental dataset (Fig. 4I and fig. S10, A ries (Fig. 5D).
as Gli2, Aquaporin-4 (Aqp4), and Potassium and B). Together, these results show that the To resolve the gene regulatory relationships
inwardly rectifying channel subfamily J axolotl telencephalon contains neuroblast pop- underlying glutamatergic neurogenesis, we
member 10 (Kcnj10). We detected 15 neuro- ulations that already express neurotransmitter leveraged our single-nucleus multiome se-
blast clusters, which formed two distinct groups signatures of downstream neurons. Moreover, quencing of the axolotl whole pallium. We
that expressed either Slc17a6/7 (VGLUT+) or neuroblasts are most similar to mouse progen- identified proximal and distal candidate
Gad1/2 (GABA+) (Fig. 4F; fig. S9, G and H; itor cells and neuroblasts, whereas ependymoglia regulatory regions differentially accessible in
and table S1). In contrast to intermediate pro- harbor transcriptional similarities to mouse each of the terminal glutamatergic neuron
genitors (IPs) found in other vertebrate brains, ependymal cells as well as neural stem cells. clusters and assessed at which stage in the
Mex3a+ neuroblasts were largely nonprolifer- respective trajectory they become accessible
ative (Fig. 4G and fig. S9I). Two neuroblast Transcriptional dynamics of postembryonic (Fig. 5, E and F; fig. S13, A and B; and table
clusters (7 and 8) showed an increased G2M glutamatergic neurogenesis S4). Most elements identified as specific for a
score when compared with the rest and were We labeled ependymoglia using Cre-loxP– given terminal glutamatergic cluster already
therefore termed early neuroblasts. VGLUT+ mediated tracing to investigate their self- obtained accessibility in the corresponding
neuroblasts (except cluster 4) were enriched in renewing properties and determine the clonal neuroblast clusters earlier in the trajectory.
medial and dorsal datasets; GABA+ neuroblasts patterns they generate during post-embryonic We inferred a gene regulatory network (GRN)
(except cluster 5) were mostly found in the neurogenesis (Fig. 5A). This uncovered dis- using Pando (31) by combining gene expres-
lateral dataset. HCR for Scl17a6 or Gad2 in tinct neurogenesis patterns in medial, dorsal, sion, chromatin accessibility, and TF binding
proportions
Cell state
post-embryonic neuro-
genesis. (A) Schematic
D Glut 0
Pseudotime Pseudotime Pseudotime
describing the out- Glut 1 Glut 6
vs 1
comes of Cre-loxP fate
Exp.
mapping performed to
assess clonal dynamics 0
Pseudotime Pseudotime Pseudotime
and potential clone Elmo1
Etv1
Gria3
Msi2
Nfix
Sez6l
Elmo1
Foxo3
Mef2c Rorb
Nfix Sema5a
Foxp1 Msi2 Sez6
Htr5a Nfix Slc2a13
shapes during homeo- Glut 7 Glut 11 Glut 8
static neurogenesis of B Hippocampal Lateral cortex Dorso-medial 1
Exp.
trajectory trajectory trajectory
the axolotl telencephalon Glut Glut 0 Glut 11 Glut 6
adjacent to measured NB 13 0
NBs
NBs 4, 7 Pseudotime Pseudotime Pseudotime
Epen
clonal patterns in medial, Etv1 Meis2 Sez6l Bcl11a Mex3a Rbfox3 Foxo3 Msi2 Slc2a13
Glra2 Nfix Shisa6 Mef2c Nfix Rorb Htr5a Nfix Sox6
NBs 3,9
dorsal, and lateral pal- Glut 7
E
Normalized accessibility
Normalized accessibility
lium. (B) Glutamatergic NB 2 Glut 0 Glut 0
NBs 7,11
trajectories reflecting Glut 7 Glut 7
(range 0 − 1.9)
(range 0 − 2.1)
NB 1
postembryonic neuro- Glut 1
Glut 1 Glut 1
Glut 8
genesis of axolotl neurons Glut 11 Glut 11
of neuron clusters. 7
(C) Pseudotemporal cell 0
type progression from
11
ependymoglia to gluta-
matergic neurons during 1
Ndst4
Trarg1
Shisal1
Reln
Grik3
Klhdc8b
Mdga1
Plpp4
Satb1
Nmb
Grik3
Tmem88b
Syndig1
Rapgef4
Mmel1
Opn3
Slitrk1
Mras
Elmo1
Megf11
Shank3
Slit3
Daam1
Epha4
Grin2a
Atp2b1
Eml4
Nrp1
Wnt4
Tp53i11
Trpm4
Sorbs1
Btbd3
Tmcc1
Rims4
Negr1
Unc13b
Garem1
Rtn4rl1
C1ql1
Brs3
Grik1
Fam131a
Oprm1
C1ql3
Pde1b
Fibcd1
Garem1
Fam131b
Tiam1
Rtn4rl1
C1ql1
Kank2
Atp2b2
Cpne2
Msi2
Plxdc2
Brinp2
Meis2
Pdzd2
Rhbdf2
Flrt2
Nr3c2
C1ql2
Kank2
Npffr2
Slc35g2
Unc13c
Unc13c
Shisa6
Sstr5
St6galnac5
Sstr5
Kcnq5
Kcnq5
Kcnh7
Cabp7
Cpne7
neurogenesis in the three
Ptprt
Penk
Nsmf
trajectory groups.
(D) Pseudotemporal gene Glut NBs Epen acc. scale
G H I 100
−2.5 5.0
expression changes dur- Nr3c2 Etv1
% specific
Global Gene Zfpm2 Aff3 2.0 0.4
ing neurogenesis for each Hippocampal trajectory
exp.
Mitf
Zeb2
Neuronal % exp. 0.3
Regulatory Network St18 Hivep2 0.25 0.5
terminal branch from the -log10(padj) Neurod4 0.50 50 0.2
Lhx9 0.75
5 Nfix
trajectories of each 10 # of outgoing Nfib Nfic Cux2
0
Number of Connected Genes
15 connections Znf469
group. (E) Representa-
Nfib
Nfia
Nfatc3
Stat5b
Etv1
Mef2a
Tcf4
Eomes
Rorb
Foxo3
Gli3
Foxp1
Nr3c2
Pou2f2
Meis2
Nfic
Pknox2
Tcf7l2
Gli2
% specific Bach2
Mef2c
Sox6
Nfix
Ahr
Mitf
20 0
100
tive example peaks Interactions Hippocampal
Positive 200
associated with (left) Negative 300 Tox2 1.6 40 Lateral cortex trajectory 0.2
exp.
Nfib
Rorb
Nfia
Gli3
Rreb1
Eomes
Tcf7l1
Foxo3
Tcf4
Zeb1
Etv1
Tead4
Rfx4
Nfatc3
Pbx3
Meis2
Pknox2
Gli2
Tcf7l2
Nr3c2
Mef2c
Nfic
Nfix
Mef2d
Tead4
Znf536
Egr4 Foxp4 % exp. Dorso-medial trajectory 0.3
Gli2 Npas3 Prdm16 Fosb Nr4a1 Meis1 Pbx3 0.25 0.4 40
8, 6, 7, 0, 11, and 1. Tcf7l2 Creb5 Sox6
Sox2
Foxm1
Pbx1 0.50 0.1
Dach1 0.75
Zeb1
(G) UMAP embedding of Znf516
Bnc2 E2f1
E2f2 0
the inferred gene mod- Dach2 Nsd2 Sall3
Gli3
Rreb1
Rfx4
Tcf7l1
Nfib
Nfia
Tead4
Mef2a
Eomes
Etv1
Nfatc3
Foxo3
Tcf4
Lef1
Rorb
Foxp1
Gli2
Tcf7l2
Nr3c2
Pknox2
Meis2
Foxp2
Nfic
Sox6
Nfix
Hivep1 Rxra
ules based on coex- Dorso-medial
pression and inferred Transcription Factors
interaction strength
between TFs. Size represents the number of connections for each TF. Color scale indicates expression, and size indicates the percent of cells
(H) Trimmed GRN UMAP embedding of the inferred gene modules based expressing. (I) Barplot of the top 25 TFs ranked by number of connections for
on coexpression and inferred interaction strength between TFs for (top) each TF and colored by fraction of trajectory-specific peaks out of total
hippocampal, (middle) lateral cortex, and (bottom) dorsal-medial trajectories. number of peaks in the global module.
at 1, 2, 4, 6, 8, and 12 weeks post injury (Fig. 6A). erating brains (Fig. 6B and fig. S14, A and B). At 2 weeks post injury, the injury site was
To visualize the location of EdU+ cells during At 1 week post injury, the injury site was still starting to close from the accumulation of
the regeneration process, we fluorescently open, and EdU+ ependymoglia were present EdU+ cells. Throughout the following time
labeled EdU using click chemistry on regen- in medial and lateral wound-adjacent regions. points, EdU+ cells remained accumulated at
EdU DAPI
D0 D5 1 wpi D12 2 wpi D19 D26 4 wpi 6 wpi 8 wpi 12 wpi
proportion
Epen MG 0.4
6 wpi 8 wpi 12 wpi
0.3
EdU DAPI
Div-seq NB 0.2
n=19300 0.1
0.0
1 2 4 6 8 12
time (wpi)
UMAP 2
Glut
Epen a Epen q Glut Oligo MG
Fractional abundance of ependymoglia clusters per time point
Epen n NB GABA Endo
UMAP 1 GABA
F
Active Quiescent Pro-neuro
2e-04 4e-04
norm. % occurr.
Ependymoglia 1 wpi
Steady-State 3 Clusters 2 wpi
n=3590 5 4 wpi
4 6 wpi
8 wpi
Div-Seq 8 10 19 12 wpi
0
n=3753 13 SS
12
21
G
11 16 1 172213 4 14 6 15 9 1211 0 2 8 20 2118 7 19 5 1016 3
20 22 % Exp.
18 Plat 20
17 Anxa2 40
2 0
1 wpi 8 wpi 7 14 Kazald1 60
UMAP 2
Anxa1
Fn1
Col7a1
Col3a1
Runx1
Akt1
Cbfb
Ncaph
Smc4
Bub1
Nrxxn1
Syp
Smc2
Col1a2
Anxa2
Plod2
S100a10
Slc6a12
Slc1a2
Ccdc80
Tnc
Dlgap5
Snap25
Plat
Div-seq 0
projected
to SS
9
5
L GlutGlut
1 1
M Neurogenesis
NB 7,13
8 Glut 1 Glut 11
Correlation (Div-seq)
2
10
0.5
0.4
11 Glut 11
12
NB 4, 2
UMAP 2
4
0.0
0. 0
24
13
1 wpi 8 wpi 6
UMAP 2
−0.5
UMAP 1 −0.4
2 wpi 12 wpi 15
4 wpi SS −0.5 0.0 0.5 −0.5 0.0 0.5
0
600
0
600
0
125
0
150
0
500
0
100
6 wpi Pseudotime
UMAP 1 0 1 Correlation (Steady-state)
N 1 Main OB 2 Acc. OB NB 1 Main OB 2 Acc. OB
NB
8 weeks post injury
3
3
Non-injured
1
1
4
2 3 Injec. site 4 Caudal pal. 4 3 Injec. site 4 Caudal pal.
2
D D
A P A P
V V
Fig. 6. Axolotl telencephalon regeneration after injury. (A) Schematic cluster occurrence per time point. (G) Dotplot of selected ependymoglia
describing the Div-Seq protocol used during axolotl telencephalon regener- cluster 21 differentially expressed genes. (H) Gene average expression
ation. (B) EdU stainings of all regeneration time points. Scale bars, 50 mm. change per time point, grouped by GO terms. (I) HCR for Kazald1 and Runx1 in
(C) UMAP plot of all EdU+ cells across all regeneration time points, colored by steady state, 2 days post injury (dpi) and 1 week post injury (wpi). Scale
cell-type class. Predicted cell clusters are shown in different shades. GABA, bars, 50 mm. (J) (Left) Correlation projection of all Div-Seq glutamatergic
GABAergic neuron; Glut, Glutamatergic neuron; NB, neuroblast; Epen, neurons (pink) to steady-state glutamatergic neurons (gray). (Right) Barplots
ependymoglia cell; Endo, endothelial cell; and MGs, microglia. (D) Change of largest glutamatergic neuron clusters recovered throughout regeneration
in cell type relative abundance along regeneration time points. (E) (Left) time points. (K) HCR for Satb1 and Rorb throughout regeneration. Scale
UMAP plot of all Div-Seq ependymoglia (shades of pink indicate different time bars, 50 mm. (L) Trajectories reflecting regenerative neurogenesis of
points) and noninjured brain steady-state (SS) ependymoglia (gray). (Right) glutamatergic neuron clusters 1 and 11. UMAPs are colored by (left) cell type
UMAP plot of clustering of all ependymoglia. (F) Heatmap of normalized and (right) pseudotime. (M) Correlations of lineage driver genes with the
assignment probability for (top) Glut1 and (bottom) Glut11 trajectories, in domain. (Insets) Labeled cell bodies in the olfactory bulb, accessory olfactory
(y axis) regenerative and (x axis) steady-state neurogenesis. (N) Whole- bulb, injection site, and caudal telencephalon. Scale bars, (overviews)
mount cleared Neurobiotin stainings on (left) noninjured brains and (right) 100 mm; (zooms) 50 mm. Main OB, main olfactory bulb; Acc. OB, accessory
brains 8 weeks post injury. Neurobiotin was injected in the Satb1+, Rorb+ olfactory bulb; and caudal T., caudal telencephalon.
the regeneration site until tissue architecture state (Fig. 6F). Cluster 21 cells differentially reestablish afferent and efferent projections.
was largely reestablished. expressed genes such as Kazal type serine We injected Neurobiotin into the Satb1+, Rorb+
Next, we investigated the transcriptomes of peptidase inhibitor domain 1 (Kazald1) and domain in noninjured as well as regenerated
EdU+ cells during the regeneration time course. RUNX family transcription factor 1 (Runx1) brains at 8 weeks post injury and performed
We used our steady-state data as a reference and were enriched in Gene Ontology (GO) whole-mount immunohistochemistry to iden-
and identified all major cell types, including terms relating to wound healing and cell tify cell bodies and projections. Similarly to
ependymoglia, neuroblasts, glutamatergic and adhesion, indicating early response programs noninjured brains, stained cell bodies were
GABAergic neurons, as well as endothelial to injury (Fig. 6, F to H). Staining for Kazald1 located in the olfactory bulb, accessory olfactory
cells and microglia (Fig. 6C). Each cell type and Runx1 confirmed absence of expression bulb, and the caudal telencephalon (amygdala),
was represented in different proportions in the uninjured telencephalon and strong indicating that the input from these regions is
throughout regeneration (Fig. 6D and fig. S14C), expression in a subpopulation of cells at reestablished in the regenerated telencephalon
reflecting a wave of neurogenesis induced by 1 week post injury, with an induction of ex- at 8 weeks post injury (Fig. 6N).
the injury. Whereas at 1 week post injury, active pression as early as 2 days post injury (Fig. 6I).
ependymoglia constituted the majority of EdU+ Cluster 8 was enriched for Lc27 and Inositol Discussion
cells, neuroblasts were the most abundant cell polyphosphate-5-phosphatase (Inpp5d), whereas Using snRNA-seq, multiomic sequencing, and
type at 2 and 4 weeks post injury. Starting from cluster 21 was enriched for cilia-related genes, Div-Seq along with spatial transcriptomics,
the putative ganglionic eminences. In axolotl, facilities, and each animal is kept individually. Brain regeneration with Div-Seq
GABAergic neurogenesis likely continues in All handling and surgical procedures were Axolotls were injected intraperitoneally with
the postembryonic brain because we detected carried out in accordance with the local ethics EdU. After the desired pulse-chase period,
GABA+ neuroblasts and found one cluster (NB 5) committee guidelines. Animal experiments were nuclei were isolated, EdU staining was per-
that contains cells from medial and dorsal performed as approved by the Magistrate formed immediately by using Click-iT EdU
pallium, which suggests local pallial GABAergic of Vienna (Genetically Modified Organism Flow Cytometry assay Kit (Thermo Fisher
neurogenesis, a phenomenon observed in the Office and MA58, City of Vienna, Austria, Scientific, #C10424), and EdU+ nuclei were
developing primate brain (37). license GZ51072/2019/16 and license GZ665226/ sorted by means of fluorescence-activated cell
We found that axolotl ependymoglia show 2019/21). sorting (FACS). Nuclei from the Div-Seq dataset
transcriptomic signatures of both mouse SVZ were classified into cell types, clusters, and
ependymal cells and B cells and function as snRNA-seq library preparation and sequencing
brain regions by using the steady-state data
stem cells during homeostatic neurogenesis. We used snRNA-seq (10x Genomics) to profile as a reference. Subsequently, ependymoglia
Postembryonic ependymoglia in different pal- medial, dorsal, and lateral regions of the were subset from the steady-state and Div-Seq
lial regions maintained expression of develop- telencephalon. We additionally profiled all datasets and integrated by using harmony,
mental patterning genes thought to regulate these regions as a whole with single-nucleus with the library chemistry as a covariate.
dorsal and ventral pallial subdomain size multiome sequencing (10x Genomics). Libra- Neurogenesis trajectories in regeneration were
during development in amniotes (23). It is ries were sequenced and then aligned to the inferred similarly to steady-state trajectories.
possible that expression of these factors main- axolotl genome. Control and regenerated brains were injected
tains pallial domain proportions during con- with neuronal tracer Neurobiotin, whole-mount
tinuous neurogenesis. The mammalian SVZ Data integration and clustering
stained, and cleared to determine projection
also contains IPs and migratory neuroblasts Datasets from each chemistry were integrated patterns.
that express GABAergic or glutamatergic fate by using harmony integration. Identification
15. A. Joven et al., Cellular basis of brain maturation and dynamical modeling. Nat. Biotechnol. 38, 1408–1414 (2020). associated with regeneration initiation in planarians. Genes
acquisition of complex behaviors in salamanders. doi: 10.1038/s41587-020-0591-3; pmid: 32747759 Dev. 26, 988–1002 (2012). doi: 10.1101/gad.187377.112;
Development 145, dev.160051 (2018). doi: 10.1242/dev.160051; 29. M. Lange et al., CellRank for directed single-cell fate mapping. pmid: 22549959
pmid: 29217751 Nat. Methods 19, 159–170 (2020). 44. A. Maynard, tomasgomes/pallium_evo: Post-revision 1 release.
16. P. S. Ulinski, W. T. Rainey, Intrinsic organization of snake 30. J. A. Farrell et al., Single-cell reconstruction of developmental Zenodo (2022); doi: 10.5281/zenodo.6627781
lateral cortex. J. Morphol. 165, 85–116 (1980). doi: 10.1002/ trajectories during zebrafish embryogenesis. Science 360, 45. K. Lust et al., Single-cell analyses of axolotl forebrain
jmor.1051650108; pmid: 30180381 eaar3131 (2018). doi: 10.1126/science.aar3131; organization, neurogenesis, and regeneration. Zenodo (2022);
17. A. Diodato et al., Molecular signatures of neural connectivity in pmid: 29700225 doi: 10.5281/zenodo.6390083
the olfactory cortex. Nat. Commun. 7, 12238 (2016). 31. J. S. Fleck et al., Inferring and perturbing cell fate regulomes in
doi: 10.1038/ncomms12238; pmid: 27426965 human cerebral organoids. bioRxiv 457460 [Preprint] (2021). AC KNOWLED GME NTS
18. D. M. G. Johnson, K. R. Illig, M. Behan, L. B. Haberly, New doi: 10.1101/2021.08.24.457460 We thank the Treutlein, Camp, and Tanaka laboratories for their
features of connectivity in piriform cortex visualized by 32. N. Habib et al., Div-Seq: Single-nucleus RNA-seq reveals discussions, input, and support. FACS support was provided by the
intracellular injection of pyramidal cells suggest that “primary” dynamics of rare adult newborn neurons. Science 353, single-cell facility at D-BSSE, ETH Zurich. Illumina sequencing
olfactory cortex functions like “association” cortex in other 925–928 (2016). doi: 10.1126/science.aad7038; was done in the Genomics Facility at D-BSSE, ETH Zurich. We
sensory systems. J. Neurosci. 20, 6974–6982 (2000). pmid: 27471252 thank the IMP Bio-Optics facility and the animal caretaker team
doi: 10.1523/JNEUROSCI.20-18-06974.2000; pmid: 10995842 33. A. Rodrigo Albors et al., Planar cell polarity-mediated induction (Vienna Biocenter) for outstanding service. Funding: This work
19. N. Moreno, A. González, Evolution of the amygdaloid of neural stem cell expansion during axolotl spinal cord was supported by a Long-Term Fellowship from the Human
complex in vertebrates, with special reference to the anamnio- regeneration. eLife 4, e10230 (2015). doi: 10.7554/ Frontier Science Program LT000605/2018-L (K.L.); EMBO Long-
amniotic transition. J. Anat. 211, 151–163 (2007). eLife.10230; pmid: 26568310 Term Fellowship ALTF 738-2019 (T.G.); European Research Council
doi: 10.1111/j.1469-7580.2007.00780.x; pmid: 17634058 34. F. Sugahara et al., Evidence from cyclostomes for RegGeneMems 742046 (E.M.T.); Special research program of the
20. O. Marín, J. L. Rubenstein, A long, remarkable journey: complex regionalization of the ancestral vertebrate brain. Austrian Science Fund project F78 (E.M.T.); European Research
Tangential migration in the telencephalon. Nat. Rev. Neurosci. Nature 531, 97–100 (2016). doi: 10.1038/nature16518; Council Organomics 758877 (A.M. and B.T.); European Research
2, 780–790 (2001). doi: 10.1038/35097509; pmid: 11715055 pmid: 26878236 Council Braintime 874606 (B.T.); and Swiss National Science
21. Z. Almási, C. Dávid, M. Witte, J. F. Staiger, Distribution patterns 35. I. Carrera, S. Ferreiro-Galve, C. Sueiro, R. Anadón, Foundation Project Grant 310030_192604 (B.T.). Author
of three molecularly defined classes of GABAergic neurons I. Rodríguez-Moldes, Tangentially migrating GABAergic cells of contributions: Designed study: K.L., A.M., T.G., E.M.T., and B.T.
across columnar compartments in mouse barrel cortex. Front. subpallial origin invade massively the pallium in developing Prepared and collected the samples and performed lineage tracing
Neuroanat. 13, 45 (2019). doi: 10.3389/fnana.2019.00045; sharks. Brain Res. Bull. 75, 405–409 (2008). doi: 10.1016/ and staining: K.L. Performed single-nuclei experiments: A.M.
pmid: 31114486 j.brainresbull.2007.10.013; pmid: 18331906 Analyzed the sequencing data: T.G., A.M., and J.S.F. Prepared the