Skip to main content
eLife logoLink to eLife
. 2019 Jul 15;8:e46683. doi: 10.7554/eLife.46683

Critical roles of ARHGAP36 as a signal transduction mediator of Shh pathway in lateral motor columnar specification

Heejin Nam 1, Shin Jeon 1,2, Hyejin An 1, Jaeyoung Yoo 1, Hyo-Jong Lee 3, Soo-Kyung Lee 2,4, Seunghee Lee 1,
Editors: Marianne E Bronner5, Francois Guillemot6
PMCID: PMC6658197  PMID: 31305241

Abstract

During spinal cord development, Sonic hedgehog (Shh), secreted from the floor plate, plays an important role in the production of motor neurons by patterning the ventral neural tube, which establishes MN progenitor identity. It remains unknown, however, if Shh signaling plays a role in generating columnar diversity of MNs that connect distinct target muscles. Here, we report that Shh, expressed in MNs, is essential for the formation of lateral motor column (LMC) neurons in vertebrate spinal cord. This novel activity of Shh is mediated by its downstream effector ARHGAP36, whose expression is directly induced by the MN-specific transcription factor complex Isl1-Lhx3. Furthermore, we found that AKT stimulates the Shh activity to induce LMC MNs through the stabilization of ARHGAP36 proteins. Taken together, our data reveal that Shh, secreted from MNs, plays a crucial role in generating MN diversity via a regulatory axis of Shh-AKT-ARHGAP36 in the developing mouse spinal cord.

Research organism: Chicken, Mouse

Introduction

Spinal motor neurons (MNs) innervating the limb are contained within the lateral motor columns (LMCs), which are produced specifically at brachial and lumbar levels of the spinal cord. LMC neurons are subsequently divided into two populations, lateral LMC (LMCl) neurons that innervate the dorsal part of the limb and medial LMC (LMCm) neurons that innervate the ventral part of the limb (Landmesser, 1978; Hollyday, 1980a; Hollyday, 1980b; Tosney and Landmesser, 1985b; Tosney and Landmesser, 1985a; Sockanathan et al., 2003; Ji et al., 2006). The molecular mechanism of how morphogenetic signaling molecules cooperate with transcription factors to define MN subtype specification has been extensively studied (Shirasaki and Pfaff, 2002; Lee and Pfaff, 2001). Retinoic acid (RA) is essential for the diversification of MN subtype and MN columnar organization. At brachial level, within the LMCs, RA is synthesized in subpopulations of MNs expressing the RA-synthesizing enzyme, RALDH2, and specifies migrating MN precursor cells into the LMCl neurons (Vermot et al., 2005; Sockanathan et al., 2003). However, it remains poorly understood whether other signaling molecules also contribute to the specification and/or maintenance of LMC and other MN columns.

Sonic hedgehog (Shh) signaling is a highly conserved pathway that is essential in directing cell proliferation and patterning during early embryogenesis (Ingham et al., 2011; Briscoe and Thérond, 2013). In spinal neuron development, Shh, released from the notochord and floor plate, leads to the generation of distinct classes of progenitor domains including MN progenitor (pMN) within the ventral side of spinal cord (Lee and Pfaff, 2001). In Shh-responsive cells, protein kinase A (PKA) plays crucial roles in fate specification and proliferation by modulating the activity of Shh signaling (Kotani, 2012; Asaoka, 2012). In the absence of Shh, PKA phosphorylates Gli transcription factors, promoting the production of repressor forms of Gli and thus repressing the Shh target gene expression, while Shh antagonizes this action of PKA (Pan et al., 2009). As both decreased and increased activity of PKA results in abnormal cell proliferation and cell fate specification, the basal level of PKA activity should be precisely controlled spatiotemporally to ensure its proper action (Kotani, 2012). PKA activation occurs upon binding of cyclic AMP (cAMP) to its regulatory subunits (PKAR), causing the release of its catalytic subunits (PKAC) (Taylor et al., 1990). Shh is able to induce phosphoinositide 3-kinase (PI3-kinase)-dependent AKT phosphorylation in cell lines such as LIGHT cells and HUVEC cells (Kanda et al., 2003; Riobó et al., 2006). Interestingly, PI3-kinase-dependent AKT activation further potentiates Shh signaling in the neuronal fate specification (Riobó et al., 2006). It was suggested that AKT might target proteins that modulate PKA kinase activity or the interaction between PKA and Gli (Riobó et al., 2006), but the identity of such proteins remain unknown.

During MN development, two LIM-homeodomain (LIM-HD) transcription factors, Islet-1 (Isl1) and LIM homeobox-3 (Lhx3) act as essential players in MN fate specification (Pfaff et al., 1996; Sharma et al., 1998; Tanabe et al., 1998; Thaler et al., 2002; Lee et al., 2013) by forming a hexameric complex, named the Isl1-Lhx3 complex, which consists of two Isl1:Lhx3 dimers and a nuclear LIM interactor (NLI) dimer (Lee et al., 2008; Lee et al., 2013). Genome-wide analyses of the Isl1-Lhx3 complex binding sites from chromatin immunoprecipitation-sequencing (ChIP-seq) datasets (Lee et al., 2013; Mazzoni et al., 2013) combined with MN transcriptome analyses (Lee et al., 2012; Mazzoni et al., 2013) identified many novel signaling pathways and regulators that are directly regulated by the Isl1-Lhx3 complex. Studies of individual target genes of the Isl1-Lhx3 complex in MN specification uncovered critical effector genes in MN specification such as genes involved in cholinergic neuronal identity determination (Cho et al., 2014), miR-218 as a downstream effector of Isl1-Lhx3 complex (Thiebes et al., 2015) and STAM1 as an endosomal sorting machinery necessary for ventral motor axon projection (Nam and Lee, 2016).

ARHGAP36, a putative Rho GTPase-activating protein, was identified from a genome-scale cDNA overexpression screen as a positive regulator of the Shh pathway (Rack et al., 2014). Overexpression of ARHGAP36 recapitulates Shh signaling transduction and these effects are independent of Smo and require kinesin family member 3a (Kif3a) and intraflagellar transport protein 88 (Ift88) (Rack et al., 2014). Recently, ARHGAP36 has emerged as a potent antagonist of PKA signaling. ARHGAP36 interacts with PKAC and inhibits PKAC catalytic activity as a PKA pseudosubstrate inhibitor (Eccles et al., 2016). It also targets PKAC for ubiquitin-dependent proteolysis by the endosomal sorting complex required for transport (ESCRT) pathway (Eccles et al., 2016). Moreover, ARHGAP36 interaction with Patched1 leads to the removal of centrosomal ARHGAP36 with ciliary Patched1 and the accumulation of centrosomal PKA that phosphorylates Inversin and this Patched1-ARHGAP36-PKA-Inversin axis determines the ciliary translocation of Smoothened and consequent hedgehog pathway activation (Zhang et al., 2019). Although recent studies in cultured cells suggest that ARHGAP36 regulates Shh activity through inhibiting PKA kinase activity, it remains unexplored whether ARHGAP36 functions as a key modulator of Shh signaling pathway in vivo.

Here we report that Shh expression is induced in postmitotic MNs at brachial and lumbar levels but not at thoracic level at later stages of development when motor columnar identities are established. Shh is required for proper LMC formation as the knock-down of Shh in the developing chick spinal cord and the deletion of Shh in the developing MNs of mouse embryos result in reduction of FoxP1+ LMC. We further show that ARHGAP36 is a critical MN-enriched Shh transduction component and Arhgap36 is a direct target gene of Isl1-Lhx3 complex during MN generation. The action of ARHGAP36 is to promote Gli-dependent transactivation partly through inhibition of PKA activity. Moreover, AKT, known as an inhibitor of PKA activity, interacts with ARHGAP36 and stabilizes ARHGAP36 protein, which further enforces suppression of PKA activity by ARHGAP36. Consistently, blocking AKT activity reduces the protein level of ARHGAP36 and hinders the efficiency of in vitro MN differentiation from mouse ESCs. Consistently, deletion of Arhgap36 gene in mouse results in defects in LMC formation at brachial level, which might be caused by dysregulation of Shh signaling pathway. Our results define a new regulatory axis of Shh-AKT-ARHGAP36-PKA in LMC specification, in which ARHGAP36 functions as an essential effector of Shh and AKT in repressing PKA activity.

Results

Shh is expressed in LMC neurons in developing mouse and chick spinal cord

While searching for extrinsic signaling molecules for LMC specification other than RA, we found that Shh shows an interesting expression pattern in ventro-lateral region of spinal cord where LMC neurons are located. At earlier stages, Shh is mainly detected in the notochord (NC) and floor plate (FP) of the mouse and chick embryos (Bitgood and McMahon, 1995; Oppenheim et al., 1999; Martí and Bovolenta, 2002) (Figure 1A). When MNs begin to be segregated into distinct motor columns, Shh is also detected in the LMC region at brachial and lumbar level but not at the thoracic level in chick (Bitgood and McMahon, 1995; Oppenheim et al., 1999; Martí and Bovolenta, 2002) and mouse embryos (Figure 1A and B). To provide the detailed expression of Shh, we performed in situ hybridization (ISH) for chick Shh and immunohistochemistry (IHC) for well-defined markers for motor columns in chick embryos (HH St.29). Our analyses showed that Shh is expressed in LMCm (Isl1+/FoxP1+) but not LMCl (Isl1-/FoxP1+) at brachial level, and interestingly, it is expressed in LMCl (Isl1-/FoxP1+) at lumbar level. Shh expression is clearly excluded from MMC (Lhx3+ MNs) at all axial levels, as demonstrated by converging analyses of Lhx3 and Shh (Figure 1B and C). The expression of Shh in motor neurons of mouse embryo is much lower than that of in chick embryo and it is rather restricted to LMCl region at different developmental stages examined so far (Figure 1A). As we examined the comparable developmental stages in mouse and chick embryos, the sensitivity differences in ISH outcomes may reflect species differences. Nonetheless, our analyses in both mouse and chick embryos demonstrate the expression of Shh in postmitotic motor neurons.

Figure 1. Shh is expressed in LMC neurons in developing mouse and chick spinal cord.

Figure 1.

(A) ISH analysis of mouse spinal cord (dotted outline) showed the expression of Shh in FP and LMC region (arrowhead) at later stages when motor columns are specified. (B) ISH analysis of chick Shh combining with IHC of Isl1/FoxP1 and Lhx3 in chick spinal cord. At HH St.29, Shh is mainly detected in LMCm (Isl1+/FoxP1+) neurons at brachial levels and LMCl (Isl1-/FoxP1+) neurons at lumbar levels but not in motor neurons at thoracic levels of chick spinal cord (dotted outline). Scale bars: 100 μm. (C) Schematic drawing shows the LMCm, LMCl, and MMC motor columns in the ventral spinal cord with representative markers.

Shh, expressed in LMC neurons, is necessary for LMC specification in developing chick spinal cord

To test whether Shh contributes to motor columnar fate determination, we generated a short-hairpin RNA (shRNA) construct that targets chick Shh. We misexpressed shRNA-Shh or shRNA-vector control in neural progenitors of the developing spinal cord by using in ovo electroporation and harvested embryos 4 days post electroporation when motor columns are established. In ovo electroporation of shRNA vector resulted in the expression of GFP throughout the electroporated spinal cord (Figure 2A). First, we confirmed the specific reduction of Shh expression in LMC region but not in the floor plate of the spinal cord (Figure 2A and C). In our electroporation condition, the electroporation efficiency of the floor plate cells is generally very low. Thus, we expected that Shh knock-down would be mostly effective in motor neurons but not in the floor plate, allowing us to focus on analyzing the effect of Shh loss-of-function (LOF) on motor columnar specification without significant changes in progenitor proliferation or early patterning of the neural tube. Furthermore, we analyzed the chick embryos that do not express GFP in the floor plate, which was further confirmed by ISH analyses of Shh expression (Figure 2A and C). Our analyses including careful quantification indeed revealed that there’s no significant change in proliferation (BrdU+ cells), survival (cCasp3+ cells) of neural progenitors or ventral neural patterning (Olig2+ and Nkx2.2+ cells) (Figure 2A and C). These rigorous analyses exclude the possibility that any observed phenotypes are caused by the effect of Shh deletion in the progenitors. Newborn motor neurons express Isl1, Lhx3 and Hb9. As the motor neurons are segregated into different motor columns, MMC neurons keep on expressing Isl1, Lhx3 and Hb9 while LMC neurons lose Lhx3 expression and gain FoxP1 expression. Thus, FoxP1 marks LMC neurons including both LMCm (Isl1+/FoxP1+) and LMCl (Hb9+/FoxP1+) neurons, whereas Isl1 and Hb9 label both MMC and hypaxial motor column (HMC) neurons (Figures 1C and 5E). The number of LMCl (Hb9+/FoxP1+) neurons of the sh-Shh injected embryos showed approximately 26% reduction compared to the uninjected control side (Figure 2B and C). However there was no effect on other motor columns, and consequently the number of total MNs was reduced by knock-down of Shh (Figure 2B and C), suggesting that Shh plays an essential role in specifying the FoxP1+ LMC neuronal identity.

Figure 2. Shh signaling is required for LMC specification in chick spinal cord.

Figure 2.

(A) IHC analyses in chick spinal cords electroporated with sh-Shh and sh-vector construct. ISH analysis showed the reduced expression of chick Shh in sh-Shh electroporated chick embryo but not in control sh-vector injected chick embryo. Knock-down of chick Shh did not affect the proliferation (BrdU) or survival (cCasp3) of neural progenitor cells and ventral neural patterning (Olig2 and Nkx2.2). (B) Knock-down of chick Shh reduced the number of LMCl (Hb9+/FoxP1+) neurons but had no effect on other motor columns such as LMCm, MMC, and HMC and consequently reduced the number of total MNs compared to the uninjected control side. +, electroporated side; -, non-electroporated control side. (C) Quantification of the relative intensity of Shh ISH signal in motor neurons, % marker+ (BrdU, Nkx2.2, Olig2, and cCasp3) cells relative to uninjected side and % motor columns relative to uninjected side of the spinal cord. Each set of chick electroporation experiments in this figure was repeated independently at least three times with 6 to 10 embryos. Embryos were harvested 4 days post electroporation (dpe). Data are mean ± s.d. **p<0.001, ***p<0.0001; ns, non-significant (Student’s t-test). n = 6 ~ 15 independent images per each sample. Scale bars: 100 μm.

Figure 2—source data 1. Source data for Figure 2C.
DOI: 10.7554/eLife.46683.004

Shh is also required for LMC formation in developing mouse spinal cord

To further support the function of Shh in LMC formation, we tried to delete Shh gene in mouse MNs by crossing Shhf/f mice with MN specific Cre recombinase expressing mice. Hb9-Cre turned out to be problematic for our experiments, because Hb9 is expressed (therefore Hb9-Cre is active) in the notochord, which secretes Shh required for the neural tube development (Harrison et al., 1999). Isl1-Cre, whose Cre expression occurs as motor neurons emerge from progenitors, might lead to severe defects in the limb development as Isl1-Cre inactivates Shh in the developing limb (Harfe et al., 2004; Yang et al., 2006; Itou et al., 2012), which can complicate our analyses of LMC motor neuron development. Finally, Olig2-Cre mice in which the Cre recombinase is active in MN progenitors, but not in the floor plate cells (Dessaud et al., 2007; Sagner et al., 2018), was used to inactivate Shh in postmitotic MNs. Consistent with the results of reduced LMCs in chick spinal cord by knock-down of Shh, there was ~30% reduction of LMCm (Isl1+/FoxP1+) neurons and LMCl (Hb9+/FoxP1+ or Lhx1+/FoxP1+) neurons in Shh conditional knock-out (Shh-cKO) embryos compared to that of WT control littermate embryos at embryonic day (E) 12.5 (Figure 3A and C). But neither MMC (Hb9+/Lhx3+) neurons nor HMC (Hb9+/Isl1+) neurons were affected (Figure 3A and C), which resulted in reduction of total number of MNs in Shh-cKO compared to control littermates. These results suggest that Shh plays an essential role in specifying the FoxP1+ LMC neuronal identity but not MMC (Hb9+/Lhx3+) or HMC (Hb9+/Isl1+) neuronal identity in developing mouse spinal cord.

Figure 3. Shh is required for LMC formation in developing mouse spinal cord.

(A) IHC analyses of E12.5 Shh-cKO (Shhf/f;Olig2-Cre) mutant embryos (n = 4) (lower panel) and control littermates (n = 4) (upper panel). The cervical level of ventral spinal cord is shown. LMCm (Isl1+/FoxP1+) neurons and LMCl (Hb9+/FoxP1+ or Lhx1+/FoxP1+) neurons (yellow bracket) in Shh conditional knock-out (Shh-cKO) were significantly reduced. On the other hand, the number of MMC (Hb9+/Lhx3+) and HMC (Hb9+/Isl1+) neurons did not change (white bracket). (B) IHC analyses of Olig2, Sox2, and Ki67 in E12.5 Shh-cKO mutant embryo and control littermates (cervical level). No significant difference in the expression of Sox2, Olig2 and Ki67 within the spinal cord. Scale bars: 100 μm. (C) Quantification of the number of LMCm (Isl1+/FoxP1+), LMCl (Hb9+/FoxP1+ or Lhx1+/FoxP1+), MMC (Hb9+/Lhx3+) and HMC (Hb9+/Isl1+) neurons, Olig2+, Sox2+, Ki67+ cells and total MNs at cervical level in E12.5 mouse embryonic spinal cord. Data are mean ± s.d. ***p<0.0001, ****p<0.00001; ns, non-significant (Student’s t-test). n = 5 ~ 28 independent images per each sample.

Figure 3—source data 1. Source data for Figure 3C.
DOI: 10.7554/eLife.46683.008

Figure 3.

Figure 3—figure supplement 1. Reduced expression of ARHGAP3 does not affect progenitor cell proliferation in Shh-cKO mouse spinal cord.

Figure 3—figure supplement 1.

(A) IHC analysis for ARHGAP36 in E12.5 Shh-cKO (n = 4) and littermate control (n = 4) embryos showing the reduced expression of ARHGAP36 in ventro-lateral region of the spinal cord at cervical level. (B) IHC analysis for BrdU incorporation in E11.5 Shh-cKO (n = 4) and littermate control (n = 5) embryos showing no defect in proliferation of progenitor cells within the spinal cord. Scale bars: 100 μm. Data are mean ± s.d. ****p<0.00001; ns, non-significant (Student's t-test). n = 14 ~ 32 independent images per each sample.
Figure 3—figure supplement 1—source data 1. Source data for Figure 3—figure supplement 1.
DOI: 10.7554/eLife.46683.007

To test whether the reduced number of FoxP1+ cell was resulted from the defects in the proliferation of the neural stem cells, in particular the MN progenitors, we examined Ki67+ cells, BrdU incorporation, and expression patterns of Sox2 that labels the proliferating progenitor cells in the ventricular zone and Olig2, a marker of MN progenitors (pMN) (Figure 3B and C and Figure 3—figure supplement 1B). There was no significant difference in the expression of Ki67, BrdU, Sox2 and Olig2 within the spinal cord of Shh-cKO mutants compared with control embryos (Figure 3C), suggesting that the specific deletion of Shh in MNs does not perturb the proliferation of neural stem cells and the overall dorsal-ventral patterning of the spinal cord.

Arhgap36 is identified as a direct target gene of the Isl1-Lhx3 complex

Given this novel action of Shh in MNs is distinct from the established role of Shh pathway in neural progenitors for patterning the ventral neural tube, we considered the possibility that MN-specific downstream effector of Shh mediates the Shh activity in driving LMC formation. To identify the candidate effector genes, we searched for target genes of the Isl1-Lhx3 by analyzing the Isl1-Lhx3-bound genomic loci mapped by ChIP-seq analyses (Mazzoni et al., 2013; Lee et al., 2013). Among several putative target genes of the Isl1-Lhx3 complex from bioinformatics analysis of these ChIP-seq datasets, we identified only Arhgap36, rather than a cluster of HH-signaling components, whose function has been implicated in Shh signaling pathway (Rack et al., 2014). We identified the binding peak in the promoter region of Arhgap36 (Figure 4A). Within the binding site, we discovered a motif similar to the previously defined consensus HxRE (for hexamer response element) (Figure 4A and B), which is the binding site for the Isl1-Lhx3 complex (Lee et al., 2013; Lee et al., 2008). To test whether the Isl1-Lhx3 complex is recruited to the HxRE of the Arhgap36 gene in vivo, we performed ChIP assay with antibodies against Isl1 and Lhx3 using E12.5 mouse embryonic spinal cord extracts. Both Isl1 and Lhx3 strongly bound to the genomic region of the Arhgap36 gene containing the ChIP-seq peak while they showed much weaker binding to a negative control genomic region Untr6 (Mali et al., 2008) (Figure 4C). These results indicate that the endogenous Isl1-Lhx3 complex is recruited to the Arhgap36 gene in the developing spinal cord.

Figure 4. ChIP-seq peaks for the Isl1-Lhx3 complex in Arhgap36 and their in vivo recruitment of the Isl1-Lhx3 complex.

Figure 4.

(A) Isl1-Lhx3 complex binding sites in Arhgap36. The peak has HxRE motif. (B) A schematic representation of reporter constructs linked to two copies of Arhgap36-enhancer genomic DNA fragment. (C) Both Isl1 and Lhx3 were recruited to Isl1-Lhx3-bound ChIP-seq peak in Arhgap36 gene. ChIP was performed with anti-IgG antibody (control), anti-Isl1 and anti-Lhx3 antibodies using E12.5 mouse embryonic spinal cord extracts. Quantitative PCR amplification of the binding region of Arhgap36 and negative control region, Untr6. ChIP experiments were repeated independently twice. Data are presented as the mean of duplicate values and error bars represent standard deviation. (D) Luciferase assay for a reporter directed by two copies of Arhgap36-enhancer. Transfections were repeated independently at least three times. Data are presented as the mean of triplicate values and error bars represent standard deviation. (E) In ovo electroporation of LacZ (to measure electroporation efficiency) and a GFP reporter directed by two copies of Arhgap36-enhancer without or with co-expression of Isl1 and Lhx3. TATA-GFP vector with no HxRE was used as a negative control and this reporter was not activated even when Isl1 +Lhx3 expression induces ectopic MNs in dorsal spinal cord. Each set of DNA was injected and electroporated in chick neural tube and embryos (n = 5 ~ 10) were harvested 3 days post electroporation (three dpe). Hb9 staining labels endogenous and ectopically induced motor neurons in the spinal cord. +, electroporated side, –, non-electroporated side. White dotted lines indicate the outline of the spinal cord. Experiments were repeated independently at least three times. Scale bars: 100 μm. (F) Quantification of the number of Hb9+ cells relative to uninjected side of the spinal cord. Data are mean ± s.d. **p<0.001 (Student’s t-test). n = 5 ~ 8 independent images per each sample.

Figure 4—source data 1. Source data for Figure 4C.
DOI: 10.7554/eLife.46683.010
Figure 4—source data 2. Source data for Figure 4D.
DOI: 10.7554/eLife.46683.011
Figure 4—source data 3. Source data for Figure 4F.
DOI: 10.7554/eLife.46683.012

Arhgap36 HxRE is activated by the Isl1-Lhx3 complex

To determine whether ARHGAP36 expression is induced directly by Isl1-Lhx3 complex via the HxRE within the ChIP-seq peak, we constructed a luciferase reporter and a GFP reporter linked to two copies of the genomic fragment containing the HxRE in the peak (herein named Arhgap36-enh) (Figure 4B). In mouse embryonic P19 cells, co-expression of Isl1 and Lhx3, which form the Isl1-Lhx3 complex with endogenous NLI, strongly activated the luciferase reporter, whereas Isl1 or Lhx3 alone showed only marginal to no activations (Figure 4D). To test whether Isl1-Lhx3 complex can activate the Arhgap36-enh in vivo, we electroporated the chick neural tube with a GFP reporter linked to two copies of the Arhgap36-enh at a time when MNs are being specified, and found that GFP is specifically expressed in MNs (Figure 4E, upper panels). When we co-electroporated Isl1 and Lhx3 expression vectors with the GFP reporter, Arhgap36-enh was ectopically activated in the dorsal spinal cord (Figure 4E, lower panels), coincident with the occurrence of Hb9+ ectopic MNs (Figure 4E and F). As a negative control experiment, TATA-GFP construct containing no HxRE was electroporated into the chick neural tube and this GFP reporter was not activated even when ectopic Hb9 is induced by the expression of Isl1 and Lhx3 in the dorsal spinal cord (Figure 4E). Together, these results indicate that the Isl1-Lhx3 complex directly triggers the expression of ARHGAP36 via the HxRE motif in the Arhgap36 gene during MN differentiation.

ARHGAP36 is expressed in developing spinal MNs

The binding of Isl1-Lhx3 complex to the Arhgap36 gene raises the possibility that the expression of ARHGAP36 is induced as MNs become specified in the developing spinal cord. In support of this idea, the expression of ARHGAP36 was induced when MNs were derived from mouse embryonic stem cells (mESCs) under MN differentiation condition (Lee et al., 2012; Wichterle et al., 2002) (Figure 5A). To further test this possibility, we performed ISH and IHC on mouse embryonic spinal cord. Consistent with the finding that Isl1-Lhx3 triggers the expression of ARHGAP36, ARHGAP36 began to be expressed in newborn MNs around E9.5 (Figure 5B) and its expression was strongly induced in MNs at E10.5-E11.5 (Figure 5B) along the rostrocaudal axis of the spinal cord, which is soon after Isl1+/Lhx3+ MNs are born. From E12.5, the expression of ARHGAP36 is most highly enriched in LMCl (Isl1-/FoxP1+) region, some in MMC-rhomboideus (Hb9+/Lhx3low) and a very little in the most medial part of MMC but not in LMCm (Isl1+/FoxP1+) at cervical level. At thoracic level, ARHGAP36 is also expressed in preganglionic motor column (PGC) (FoxP1+/Isl1+) and HMC (Isl1+/Hb9+) neurons but with relatively lower expression compared to the cervical level. At lumbar level, ARHGAP36 is relatively highly enriched in LMCl (Isl1-/FoxP1+) and show very low level in the most medial part of MMC (Figure 5C and E). To examine the co-localization of ARHGAP36 with Shh, we performed ISH of Shh and IHC of ARHGAP36 in mouse E12.5 spinal cord at cervical level. Shh is co-localized with ARHGAP36 mostly in LMCl region in mouse spinal cord (Figure 5D). FoxP1 is expressed high in LMC region at brachial and lumbar levels as well as in PGC region at thoracic level, which is co-expressed in all ARHGAP36+ cells. Also ARHGAP36 protein was mainly localized in the cytoplasm (Figure 5B and C), suggesting that ARHGAP36 protein might function as a modulator of a cytoplasmic signaling cascade within MNs. We also examined the expression of Arhgap36 in chick embryo and found that it is ubiquitously expressed within the spinal cord but not in other tissues (Figure 7—figure supplement 3A).

Figure 5. Expression of ARHGAP36 in the developing spinal MNs.

Figure 5.

(A) Induction of ARHGAP36 expression in MNs differentiated from mouse ESCs was determined by qRT-PCR. Relative expression levels are shown as the mean of duplicate values obtained from representative experiments. Error bars represent standard deviation. (B,C) ARHGAP36 was specifically expressed in MNs of mouse embryos at E9.5, E10.5, E11.5 and E12.5 stages, as shown by ISH with a probe detecting ARHGAP36 and IHC for ARHGAP36, Isl1/FoxP1, Isl1/Hb9, Lhx3/Hb9 and FoxP1. From E12.5 and onward, ARHGAP36 expression was highly enriched in LMCl (Isl1-/FoxP1+) region, some in MMC-rhomboideus (Rb) (Hb9+/Lhx3low) and a very little in the most medial part of MMC but not in LMCm (Isl1+/FoxP1+) at cervical level. ARHGAP36 is also expressed in PGC (FoxP1+/Isl1+) and HMC (Isl1+/Hb9+) neurons at thoracic level but with relatively lower expression compared to the cervical level. At lumbar level, ARHGAP36 is enriched in LMCl (Isl1-/FoxP1+) of the spinal cord. Scale bars: 100 μm. (D) Co-localization of ARHGAP36 with Shh shown by ISH of Shh and IHC of ARHGAP36 in mouse E12.5 spinal cord at cervical level. Shh is co-localized with ARHGAP36 mostly in LMCl region in mouse spinal cord. Scale bars: 100 μm. (E) Schematic drawing shows the LMCm, LMCl, HMC, MMC and MMC-rhomboideus (Rb) motor columns in the ventral spinal cord with representative markers.

Figure 5—source data 1. Source data for Figure 5A.
DOI: 10.7554/eLife.46683.014

Shh pathway is activated by ARHGAP36 expression in spinal cord

To test whether ARHGAP36 is able to mediate Shh activity within the developing spinal cord, we have ectopically expressed ARHGAP36 in the neural tube using in ovo electroporation and examined the expression pattern of MN genes as well as genes in spinal progenitor domain and Shh pathway by IHC and ISH. ARHGAP36 misexpression resulted in a strong ventralization of the dorsal spinal cord (Figure 6—figure supplement 1A), mimicking Shh activity. Apparently, the size of the spinal cord was increased with ARHGAP36 expression and MN genes such as Hb9, Isl1/2 and Slc18a3 were highly upregulated in the electroporated side by ARHGAP36 (Figure 6—figure supplement 1A). Nkx2.2, a marker for p3 domain (V3 interneuron progenitors), and Olig2, a marker for pMN, were also ectopically expressed in the electroporated side (Figure 6—figure supplement 1A). As the effect of ARHGAP36 expression highly resembled that of activated Shh pathway, we examined two downstream target genes of Shh pathway, Ptch1 and Gli1. Both genes were also upregulated in the dorsal spinal cord, demonstrating activation of Gli-dependent transcription (Figure 6—figure supplement 1A). Given the requirement of Shh in LMC and the specific expression of ARHGAP36 in LMC neurons among mature MNs, we considered the possibility that ARHGAP36 has a role in LMC formation. When ARHGAP36 is misexpressed preferentially in ventral spinal cord and harvested at 4 days post electroporation (dpe), the number of FoxP1+ LMC neurons increased drastically, but there was no change in MMC (Hb9+/Lhx3+) neurons (Figure 6A and B). To avoid the defects in proliferation and neural patterning possibly caused by the aberrant activation of Shh in the neural progenitors, we adopted Gal4/UAS system to drive the motor neuron specific expression of ARHGAP36. Under this condition, ARHGAP36 was expressed specifically in postmitotic MNs and resulted in significant increase of FoxP1+ LMC neurons but had no effect on MMC (Hb9+/Lhx3+) neurons (Figure 6A and B). These data suggest that ARHGAP36 is sufficient to direct LMC fate-determination.

Figure 6. Expression of ARHGAP36 promotes LMC specification in developing chick spinal cord.

(A) ARHGAP36 constructs were injected and electroporated in chick neural tube and embryos (n = 8 ~ 15) were harvested 4 days post electroporation (four dpe). Ectopic expression of ARHGAP36 driven by CMV promoter in most injected cells induced robust expression of FoxP1+ LMC neurons (orange bracket) in ventral spinal cord but had no effect on MMC (Hb9+/Lhx3+) neurons (white bracket). Targeting the expression of ARHGAP36 specifically in motor neurons using Hb9-Gal4/UAS-ARHGAP36 system also lead to the robust induction of FoxP1+ LMC neurons (orange bracket) but had no effect on MMC (Hb9+/Lhx3+) neurons (white bracket). +, electroporated side; -, non-electroporated control side. Experiments were repeated independently at least three times. Scale bars: 100 μm. (B) Quantification of the number of FoxP1+ neurons and MMC (Hb9+/Lhx3+) neurons on the electroporated (+) and non-electroporated (-) sides of the spinal cord. Data are mean ± s.d. **p<0.001, ****p<0.00001; ns, non-significant (Student’s t-test). n = 6 ~ 20 independent images per each sample.

Figure 6—source data 1. Source data for Figure 6B.
DOI: 10.7554/eLife.46683.019

Figure 6.

Figure 6—figure supplement 1. Activation of Shh pathway by ARHGAP36 expression in spinal cord.

Figure 6—figure supplement 1.

(A) In ovo electroporation of ARHGAP36 construct. ARHGAP36 construct was injected and electroporated in chick neural tube and embryos (n = 8 ~ 12) were harvested 3 days post electroporation (three dpe). Ectopic expression of ARHGAP36 induced expression of ventral spinal neuronal genes including motor neuron genes (Hb9, Isl1/2, Slc18a3), ventral progenitor genes (Nkx2.2 and Olig2) and downstream target genes of Shh pathway (Ptch1 and Gli1) as shown by ISH and IHC. +, electroporated side; -, non-electroporated control side. Experiments were repeated independently at least three times. Scale bars: 100 μm. (B) Western blot analysis showed that ARHGAP36 inhibits PKA activity. PKA expression resulted in producing Gli3R, repressor form of Gli3 (lane 2) in HEK293T cells. Co-expression of ARHGAP36 blocks the formation of Gli3R by PKA (lane 4). β-tubulin was used as a loading control. (C) ARHGAP36 inhibited the level of phosphorylation of putative PKA substrates shown by western blot with anti-p-SER antibody and the level of phospho-CREB (p-CREB), a known PKA target in NIH3T3 cells. W indicates PKA wild type and M indicates PKA kinase dead mutant (K73H). β-tubulin was used as a loading control. (D) Luciferase reporter assay in HEK293T cells. The activation of CRE-luc is directed by PKA-phosphorylated CREB and this activity was blunted by co-expression of ARHGAP36, indicating that ARHGAP36 inhibits the kinase activity of PKA. Data are mean ± s.d.
Figure 6—figure supplement 1—source data 1. Source data for Figure 6—figure supplement 1D.
DOI: 10.7554/eLife.46683.017
Figure 6—figure supplement 2. ARHGAP36 is not sufficient to induce MN differentiation in mESCs.

Figure 6—figure supplement 2.

(A) Schematic representation of inducible ARHGAP36-ESCs before and after treatment with Dox (Doxycycline). TRE, tetracycline response element; rtTA, reverse tetracycline transactivator. (B) Illustration of experimental design to differentiate ESCs to MNs. RA and SAG or RA and Dox were treated at day two and EBs were harvested at day six for further analyses. EB, embryoid body; RA, retinoic acid; SAG, Smoothened agonist. (C) IHC analyses in ESC-derived MNs cultured with RA +SAG or RA +Dox at differentiation day 6. Hb9 labels MNs and it was induced only by RA +SAG. Treatment of RA +Dox induced ARHGAP36 and pan-neuronal marker TuJ1, but not MN marker Hb9.

PKA activity is inhibited by ARHGAP36

To further test whether ARHGAP36 functions through Shh, we examined the effects of ARHGAP36 on Gli3 processing in cells, which is modulated by PKA in response to Shh (Pan et al., 2009). In support of the idea that the expression of ARHGAP36 in chick spinal cord induces Gli-dependent transcription through blocking PKA activity, ARHGAP36 blunted the activity of PKA from producing Gli3R, the repressor form of Gli3 (Figure 6—figure supplement 1B, lane 4). We then examined whether the kinase activity of PKA was inhibited by ARHGAP36 by measuring the level of phosphorylated CREB (Nichols et al., 1992), a direct target of PKA as well as phospho serine (pSER) levels of putative PKA substrates in the whole cell lysate. WT PKA, but not a kinase dead mutant form of PKA (K73H) (Iyer et al., 2005; Zhong et al., 1997), showed a robust increase in pSER and p-CREB levels (Figure 6—figure supplement 1C, lanes 2, 3). The pSER and p-CREB levels in the presence of WT PKA were drastically reduced by ARHGAP36 overexpression (Figure 6—figure supplement 1C, lane 6). PKA phosphorylates CREB, and p-CREB in turn binds and activates CRE-luciferase reporter (Nichols et al., 1992; Grewal et al., 2000). Indeed, the CRE-Luc reporter was strongly activated by PKA, but this activation was blunted by co-expression of ARHGAP36 in HEK293T cells (Figure 6—figure supplement 1D). Taken together, our data indicate that ARHGAP36 inhibits PKA and de-represses Gli activity.

ARHGAP36 alone is not sufficient to induce MNs from mouse embryonic stem cells

As ARHGAP36 has a potent activity in Shh signaling stimulation and MN induction in chick spinal cord, we tested whether ARHGAP36 alone is sufficient in inducing MNs from mouse embryonic stem cells (mESCs). We generated a mouse ESC line, in which doxycycline (Dox) induces the expression of ARHGAP36 (iARHGAP36-ESCs) and tested whether ARHGAP36 can replace the activity of Shh (Figure 6—figure supplement 2). The iARHGAP36-ESCs enabled us to control the exact timing of ARHGAP36 expression by treating the cells with Dox (Figure 6—figure supplement 2A). We used conventional MN differentiation method with retinoic acid (RA) and Shh agonist (Smoothened agonist, SAG) to compare the efficiency of MN generation (Figure 6—figure supplement 2B). iARHGAP36-ESCs treated with RA and SAG exhibited effective MN differentiation, as determined by the induction of MN markers such as Hb9. iARHGAP36-ESCs treated with RA and Dox without SAG differentiated into neurons as marked by TuJ1 expression, but failed to induce the MN gene, Hb9 (Figure 6—figure supplement 2C), suggesting that ARHGAP36 alone is not sufficient to activate Shh downstream pathway to promote the initial ventralization and MN induction in mESCs. These results suggest that Shh ligand is likely needed for ARHGAP36 to function properly in vivo.

ARHGAP36 mediates the positive effect of AKT in Shh signaling

To fully understand the nature of ARHGAP36 function, we tried to identify signaling pathways that regulate the activity of ARHGAP36 through post-translational modifications, including phosphorylation. We adopted GPS 3.0 website for predicted sites based on protein sequences (Xue et al., 2005). We found several predicted phosphorylation sites in ARHGAP36 proteins, and AKT kinase was one of the high ranked kinase (data not shown). Phosphoinositide 3-kinase (PI3-kinase)-dependent AKT activation plays an essential role in Shh signaling by antagonizing PKA-mediated Gli inactivation in the specification of neuronal fates in chicken neural explants (Riobó et al., 2006). To test whether ARHGAP36 functions as a downstream effector of AKT, we transfected ARHGAP36 with AKT constructs in HEK293T cells and measured the protein levels of ARHGAP36 in the presence of AKT. As ARHGAP36 is not expressed endogenously in HEK293T cells, ARHGAP36 proteins are expressed only from the ARHGAP36-encoding plasmid, in which CMV promoter drives the transcription of ARHGAP36. Interestingly, wild type AKT (WT) and constitutively active myristoylated form of AKT (CA), but not a nonphosphorylatable dominant negative form of mutant AKT (DN), stabilized ARHGAP36 proteins robustly (Figure 7A), suggesting that AKT increases ARHGAP36 proteins likely by stabilizing ARHGAP36 protein, rather than activating the ARHGAP36 promoter transcriptionally. We also found that the half-life of ARHGAP36 protein, treated with cycloheximide that blocks the protein translation, was prolonged in the presence of AKT (Figure 7—figure supplement 1). This stabilization of ARHGAP36 protein by AKT WT was reversed by AKT inhibitor, but the CA form of AKT was not affected by AKT inhibitor (Figure 7B). Also AKT and ARHGAP36 associated with each other in co-immunoprecipitation assays and this association was decreased by treatment of AKT inhibitor (Figure 7C). The CA form of AKT interacted with ARHGAP36 more robustly than WT AKT (Figure 7D). These results show that activated AKT interacts with ARHGAP36 and stabilizes ARHGAP36 proteins (Figure 7E). It needs to be further confirmed whether ARHGAP36 interacts with AKT directly and is a genuine substrate of AKT.

Figure 7. AKT potentiates Shh signaling through stabilization of ARHGAP36 proteins and AKT-ARHGAP36 axis is required for LMC specification.

(A) ARHGAP36 was stabilized dramatically by AKT WT and CA, but not by DN in HEK293T cells. ARHGAP36 was transiently transfected with AKT constructs in HEK293T cells and the protein levels were monitored by western blotting. β-tubulin was used as a loading control. (B) 10 μM of AKT inhibitor (iAKT1/2) was treated for 20 hr and the protein level of ARHGAP36 was monitored. AKT inhibitor reversed the effect of AKT WT in stabilizing ARHGAP36 protein but had no effect on constitutively active form of AKT. (C) Co-immunoprecipitation assay with HEK293T cells transiently transfected with the expression vectors for HA-tagged AKT and ARHGAP36 showed that AKT WT co-purified ARHGAP36, and this interaction was decreased by iAKT1/2, the AKT inhibitor. (D) The CA form of AKT interacted with ARHGAP36 more robustly than AKT WT. ARHGAP36 with either HA-tagged AKT WT or AKT CA was transfected into HEK293T cells and immunoprecipitated with anti-HA antibody that pull-downs AKT. Anti-IgG antibody was used as a negative control. (E) Illustration of the modulatory pathway showing that activated AKT stabilizes ARHGAP36 proteins, which in turn blocks the kinase activity of PKA, which results in Gli-dependent transcriptional activation via dephosphorylation of Gli. (F) IHC analyses in the chick neural tube electroporated with AKT WT, CA and DN. Embryos (n = 8–10) were harvested 4dpe. AKT WT or CA increased the number of FoxP1+ cells by almost two fold in the electroporated side (+) compared to the non-electroporated control side (-). Experiments were repeated independently at least three times. Scale bars: 100 μm. (G) The analysis of ectopic FoxP1+ neuron formation by ARHGAP36 in the presence of either AKT DN or LacZ in the chick neural tube. Embryos (n = 8–10) were harvested 4dpe. +, electroporated side; -, non-electroporated control side. AKT DN completely blocked the effect of ARHGAP36 in inducing ectopic FoxP1 expression in the electroporated cells. Experiments were repeated independently at least three times. Scale bars: 100 μm. (H,I) Quantification of the number of FoxP1+ neurons on the electroporated (+) and non-electroporated (-) sides of the spinal cord. Data are mean ± s.d. *p<0.01, **p<0.001, ****p<0.00001 (Student’s t-test). n = 6 ~ 27 independent images per each sample.

Figure 7—source data 1. Source data for Figure 7H and 7I.
DOI: 10.7554/eLife.46683.025

Figure 7.

Figure 7—figure supplement 1. AKT stabilizes protein level of ARHGAP36.

Figure 7—figure supplement 1.

Measurement of half-life of ARHGAP36 in HEK293T cells transfected with empty vector or vector encoding AKT WT, following treatment with 200 ng/ml of the protein synthesis inhibitor cycloheximide (CHX). The half-life of ARHGAP36 was roughly estimated to ~1 hr for vector alone, while it was not degraded within 24 hr for AKT WT. These experiments were repeated multiple times, and we obtained similar results.
Figure 7—figure supplement 1—source data 1. Source data for Figure 7—figure supplement 1.
DOI: 10.7554/eLife.46683.022
Figure 7—figure supplement 2. AKT inhibition blocks MN differentiation from mouse ESCs.

Figure 7—figure supplement 2.

(A) Illustration of experimental design to differentiate ESCs to MNs. RA and SAG were treated at day two and AKT inhibitor was treated from day four and EBs were harvested at day six for further analyses. EB, embryoid body; RA, retinoic acid; SAG, Smoothened agonist. (B) IHC analyses in ESC-derived MNs cultured with RA +SAG with or without iAKT at differentiation day 6. Hb9 labels MNs. ARHGAP36 was induced in MN differentiation condition. Treatment of iAKT reduced expression of ARHGAP36, FoxP1 and Hb9 but not pan-neuronal marker TuJ. (C) Western blotting analysis showed reduced expression of ARHGAP36, FoxP1 and Isl1/2 but not pan-neuronal marker TuJ by AKT inhibitor treatment. (D) RT-PCR analysis revealed that ARHGAP36 mRNA level was not affected by AKT inhibition. Cyclophilin A was used as a loading control.
Figure 7—figure supplement 3. Expression of Arhgap36 in chick spinal cord and AKTs and PKAs in developing mouse spinal cord.

Figure 7—figure supplement 3.

(A) ISH for Arghap36 in HH25 and HH29 chick embryos showing its broad expression in the spinal cord at brachial level. (B) ISH for AKT1, AKT2 and AKT3 in E13.5 mouse embryos showing their expression in ventro-lateral region of the spinal cord at cervical level. (C) ISH for the PKA catalytic subunits CA and CB and the regulatory subunits RIa, RIb, RIIa and RIIb in E12.5 mouse embryos showing their expression in ventro-lateral region of the spinal cord at cervical level. Scale bars: 100 μm.

AKT is required for MN differentiation in mouse ESCs

AKT is a key player in activation of MN survival pathways after spinal cord injury (Yu et al., 2005) and it is downregulated in amyotrophic lateral sclerosis (ALS) (Peviani et al., 2014), suggesting that AKT may also play a role in embryonic MN development. Given that ARHGAP36 was induced when mESCs differentiated into MNs (Figure 5A), we hypothesized that AKT regulates the protein levels of ARHGAP36 affecting the efficiency of MN differentiation from mESCs. We used the MN differentiation condition with RA and SAG, a Smoothened agonist that stimulates Shh pathway, followed by treatment with AKT inhibitor for 2 days (Figure 7—figure supplement 2A) and harvested differentiated EBs for immunostaining (Figure 7—figure supplement 2B) and immunoblotting (Figure 7—figure supplement 2C) to monitor MN differentiation. Treatment of AKT inhibitor decreased ARHGAP36 protein levels as well as MN markers such as Isl1/2, FoxP1 and Hb9 but not pan-neuronal marker TuJ1 (Figure 7—figure supplement 2B and C). AKT inhibitor did not affect the mRNA level of ARHGAP36 (Figure 7—figure supplement 2D). These results suggest that AKT activity plays an important role in MN differentiation likely through modulating the level of ARHGAP36 proteins.

AKT-ARHGAP36 axis modulates Shh signaling in LMC specification

To further investigate the roles of AKT in modulating Shh signaling in LMC specification, we examined the expression pattern of AKTs using ISH. AKT1, AKT2, and AKT3 showed relatively low expression in the spinal cord but they were specifically enriched in the lateral region of the spinal cord (Figure 7—figure supplement 3B). We also examined the expression patterns of PKA catalytic isoforms and regulatory isoforms using ISH. Most of them were expressed in the lateral region of the spinal cord, while PKA CA, CB, RIb and RIIa were more enriched in the LMC region (Figure 7—figure supplement 3C). Given the relatively high expression of AKT and PKA in ventro-lateral region of the spinal cord and the role of Shh in inducing the activation of AKT in cell lines such as LIGHT cells and HUVEC cells (Kanda et al., 2003; Riobó et al., 2006), we proposed that Shh expressed in the motor neurons triggers AKT activation, which in turn stabilizes the protein level of ARHGAP36 in LMC neurons. Indeed, we detected reduced expression of ARHGAP36 in Shh-cKO (Figure 3—figure supplement 1A) suggesting that the protein level of ARHGAP36 can be modulated through AKT activation by Shh in LMC neurons of developing mouse spinal cord. To test the activity of AKT in inducing FoxP1+ LMC MNs, we injected WT, CA and DN form of AKT in chick spinal neural tube and monitored the expression of FoxP1. Interestingly, AKT WT and CA increased the number of cells expressing FoxP1 by almost two fold in the electroporated side of the spinal cord compared to the non-electroporated side (Figure 7F and H), while AKT DN resulted in further reduction of endogenous FoxP1 in LMC region (Figure 7F and H). Furthermore, this AKT DN actively blocked the effect of ARHGAP36 in inducing ectopic FoxP1 in the electroporated cells (Figure 7G and I), suggesting that AKT is required for the ARHGAP36 to function as a modulator of Shh signaling in LMC specification. Taken together, our results demonstrate that AKT-directed maintenance of physiological levels of ARHGAP36 is likely critical for effectively activating Shh signaling through inhibition of PKA in LMC MN formation.

Requirement of ARHGAP36 for LMC formation in mice

To further define the roles of ARHGAP36 in developing mouse embryos, we generated Arhgap36 deficient mice using Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and CRISPR-associated proteins (Cas) system (Figure 8—figure supplement 1A and B) (Yang et al., 2014; Ran et al., 2013; Jinek et al., 2012). By IHC with anti-ARHGAP36 antibody, we confirmed that ARHGAP36 proteins were not expressed in Arhgap36-/- and Arhgap36-/y mutant spinal cord at different time points (Figure 8A and Figure 8—figure supplement 2A). We then examined the specification of MNs, especially formation of LMC MNs at cervical level where ARHGAP36 is highly expressed (Figure 5B and C). At earlier stages of mouse embryo from E9.5 to 11.5, there was no obvious defect in proliferation of neural progenitor cells, ventral neural patterning and overall MN generation (Figure 8—figure supplement 1C). At E12.5, the overall number of MNs in Arhgap36-/- mutant spinal cord was similar to that of control littermates and the total number of FoxP1+ MNs at cervical level was not affected either (Figure 8—figure supplement 1D). However, we found that LMCm (Isl1+/FoxP1+) neurons are increased, whereas LMCl (Lhx1+/FoxP1+) neurons are decreased in Arhgap36-/- mutant spinal cord (Figure 8—figure supplement 1D). Concomitantly, there was an increase in cleaved Caspase3 positive apoptotic cells in the absence of ARHGAP36 (Figure 8—figure supplement 1D). LMCl neurons are born later than LMCm neurons, and these LMCl neurons have to migrate through earlier born LMCs. These results suggest that later born presumptive LMCl neurons may fail to fully differentiate into LMCl neurons and, instead, either adopt LMCm characteristics or undergo cell death in the absence of ARHGAP36. Intriguingly, from E13.0, the numbers of LMCm (Isl1+/FoxP1+) and LMCl (Lhx1+/FoxP1+) neurons were reduced in Arhgap36-/- spinal cord (Figure 8A and C), which may be caused by the increased cell death (Figure 8—figure supplement 1D). These results suggest that ARHGAP36 is required for proper generation or maintenance of LMC MNs. In contrast, there was no significant difference in MMC (Hb9+/Lhx3+), HMC (Hb9+/Isl1+) MNs and V2-INs (Chx10+/Lhx3+) (Figure 8A and C). At thoracic levels, nNOS+ PGC, HMC (Hb9+/Isl1+) and MMC (Hb9+/Lhx3+) MNs in Arhgap36 deficient spinal cord were expressed comparably to those in control littermate (Figure 8B). Arhgap36 gene is located in the X chromosome and we examined whether there is phenotypic difference between male and female mutant embryos. While there was no significant difference in MMC (Hb9+/Lhx3+) and nNOS+ PGC MNs between sexes, the number of FoxP1+ LMC neurons at cervical level was reduced in female mutant embryo (Figure 8) but not in male mutant embryo at E13.5 (Figure 8—figure supplement 2). The molecular basis underlying this sexual dimorphism needs to be further investigated in the future. Taken together, these data support the notion that specific expression of ARHGAP36 in a subpopulation of MNs (i.e., LMC neurons) at later developmental stages directs the formation or maintenance of LMC MNs.

Figure 8. ARHGAP36 is required for LMC formation in mice.

(A) IHC analyses of E13.5 Arhgap36-/- mutant embryo (n = 4) (lower panel) and their littermate controls (n = 5) (upper panel). Ventrolateral quadrants of the cervical level of spinal cord are shown in all panels. IHC with anti-ARHGAP36 antibody confirms the absence of ARHGAP36 expression in Arhgap36-null embryos. LMCm (Isl1+/FoxP1+) and LMCl (Lhx1+/FoxP1+) neurons were significantly reduced in Arhgap36-/-. On the other hand, the numbers of MMC (Hb9+/Lhx3+), HMC (Isl1+/Hb9+) neurons and V2-INs (Lhx3+/Chx10+) did not change. Scale bars: 100 μm. (B) At thoracic level, there was no difference in PGC (nNOS+ or Isl1+/FoxP1+), HMC (Hb9+/Isl1+), and MMC (Hb9+/Lhx3+) neurons compared to control littermates. Scale bars: 100 μm. (C) Quantification of the number of LMCm (Isl1+/FoxP1+), LMCl (Lhx1+/FoxP1+), MMC (Hb9+/Lhx3+), HMC (Isl1+/Hb9+) and V2-INs (Lhx3+/Chx10+) at cervical level in E13.5 mouse embryonic spinal cord. Data are mean ± s.d. *p<0.01, **p<0.001, ****p<0.00001; ns, non-significant; n = 6 ~ 12 independent images per each sample. (D) Proposed model. In Shh-cKO or Arhgap36-/- mutant embryos, LMCm and LMCl neurons are reduced with no expansion of other motor columns, and thus this results in the reduction of total MNs compared to WT control. AKT, activated in response to Shh, stabilizes ARHGAP36 protein, which in turn inhibits the kinase activity of PKA. This results in Gli-dependent transcriptional activation and LMC formation in MNs at cervical level of the spinal cord.

Figure 8—source data 1. Source data for Figure 8C.
DOI: 10.7554/eLife.46683.031

Figure 8.

Figure 8—figure supplement 1. ARHGAP36 is required for proper LMC formation in mice.

Figure 8—figure supplement 1.

(A) Schematic showing the CRISPR/Cas9-mediated ARHGAP36 knockout mouse generation. Sequences for target exon2 of WT allele (upper) and ARHGAP36 KO allele (lower). The target sequence of sgRNA is marked in red, and the PAM sequence is in blue. (B) Genotyping PCR results show one DNA band for KO alleles from KO mouse genomic DNA and two DNA bands of WT and KO alleles from heterozygous. (C) IHC analyses of E11.5 Arhgap36-/- mutant embryos (lower panels, n = 3) and control littermates (upper panels, n = 5). There is no defect in cell proliferation (BrdU), ventral patterning (Nkx2.2and Olig2) and motor neuron generation (Hb9) at early developmental stage. Data are mean ± s.d. ns, non-significant; n = 7 ~ 14 independent images per each sample. (D) IHC analyses of E12.5 Arhgap36-/- mutant embryos (lower panels, n = 4) and control littermates (upper panels, n = 4). At cervical level, LMCm neurons are increased and LMCl neurons are decreased in Arhgap36-/- mutant embryo. At the same time, there is an increase in apoptotic marker (cCasp3) in Arhgap36-/- mutant spinal cord (arrowheads). Scale bars: 100 μm.
Figure 8—figure supplement 1—source data 1. Source data for Figure 8—figure supplement 1C.
DOI: 10.7554/eLife.46683.028
Figure 8—figure supplement 2. ARHGAP36 is not required for LMC formation in male mouse embryos.

Figure 8—figure supplement 2.

(A) IHC analyses of E13.5 Arhgap36-/y mutant male embryos (n = 4) (lower panel) and their littermate controls (n = 5) (upper panel). At cervical level, the numbers of FoxP1+ LMC MNs and Hb9+/Lhx3+ MMC neurons did not change in Arhgap36-/y. (B) At thoracic level, there was no difference in nNOS+ PGC neurons, Hb9+/Isl1+ HMC neurons, and Hb9+/Lhx3+ MMC neurons compared to control littermates. Scale bars: 100 μm. (C) Quantification of the number of FoxP1+ LMC and Hb9+/Lhx3+ MMC at cervical level and FoxP1+/Isl1+PGC and Hb9+/Lhx3+ MMC at thoracic level in E13.5 male mouse embryonic spinal cord. Data are mean ±s.d. ns, non-significant; n = 6 ~ 12 independent images per each sample.
Figure 8—figure supplement 2—source data 1. Source data for Figure 8—figure supplement 2C.
DOI: 10.7554/eLife.46683.030

Discussion

While the role of Shh signaling pathway to induce ventral progenitor domains in the spinal cord has been relatively well characterized (Persson et al., 2002; Jessell, 2000), its role in LMC specification has not been studied. In this report, we discovered a novel role of Shh in inducing LMC specification, which involves coordination of multiple signaling pathways by ARHGAP36, a key modulator of Shh signaling pathway. First, we discovered that Shh is expressed in MNs at brachial and lumbar levels of the spinal cord where LMC neurons are specified and is required for proper LMC formation in developing chick and mouse spinal cord (Figures 13). Second, we identified ARHGAP36, along with Shh, as a protein highly expressed in the LMC regions of differentiating MNs (Figure 5C). Third, ARHGAP36 modulates the activity of PKA, an inhibitor of Shh pathway, thereby enhancing the activity of Gli-dependent transcription in the spinal cord (Figure 6—figure supplement 1). Fourth, ARHGAP36 levels seem to be tightly regulated by AKT during MN generation, as shown by the increase of ARHGAP36 protein levels by WT AKT and a constitutively active form of AKT and a decrease in ARHGAP36 levels by a dominant negative form of AKT and AKT inhibitor (Figure 7 and Figure 7—figure supplements 12). Fifth, deletion of Arhgap36 in mouse results in specific reduction of FoxP1+ LMC MNs in the developing mouse embryonic spinal cord (Figure 8), which is similar to what was observed in Shh knock-down chick spinal cord and Shh-cKO mouse spinal cord (Figures 2 and 3). Taken together, our results reveal a regulatory axis consisting of Shh-AKT-ARHGAP36-PKA, which plays crucial roles in modulating the activity of Shh signaling in a spatiotemporal manner for LMC specification.

Once MN progenitors, produced in the pMN progenitor domain of the ventral neural tube in response to the morphogen Shh, give birth to MNs (Jessell, 2000), MNs are further organized into distinct motor columns that are responsible for innervating each target muscle along the rostral-caudal neural tube (Dasen and Jessell, 2009; Stifani, 2014). MMC neurons innervate dorsal epaxial muscles, whereas HMC neurons project to the ventral hypaxial muscles. The LMC neurons innervate limb muscles and PGC neurons innervate sympathetic ganglia (Stifani, 2014; Dasen and Jessell, 2009). Motor column specific transcription factors and morphogenetic signaling molecules collaborate to define MN subtype specification (Shirasaki and Pfaff, 2002; Lee and Pfaff, 2001). RA is essential for the diversification of MN subtype and MN columnar organization. Also Hox genes, which encode a family of transcription factors, determine MN subtypes and there is a clear relationship between Hox protein expression and motor columnar specification. Moreover, FoxP1 has been shown to function as a critical Hox cofactor in regulating MN subtype diversity especially for specification of both the LMC and PGC neurons (Rousso et al., 2008; Pfaff, 2008; Dasen et al., 2008; Arber, 2008). It has been shown that abnormal expression of Hox proteins within postmitotic MNs result in subtype switching (Jung et al., 2010; Wu et al., 2008; Dasen et al., 2005; Lin and Carpenter, 2003). The spatiotemporal expression of these HD factors together with extrinsic signaling suggest that MN subtype identity remains plastic even after they exit the cell cycle and it should be tightly regulated to generate proper MN columnar subtypes. It is intriguing that two prominent extrinsic cues, RA and Shh, participate in LMC specification. Future studies should be directed at elucidating whether Shh pathway involving ARHGAP36 integrates with RA and Hox genes in LMC specification.

Because LMC and PGC neurons do not express Lhx3, it is not clear whether ARHGAP36 induced by the Isl1-Lhx3 complex at earlier stages of MN development (Figure 5) persists in LMC and PGC neurons or another mechanism independently induces the expression of ARHGAP36 in these specific motor columns at later stages. Because Shh stabilizes ARHGAP36 through AKT activation (Figure 7) and Shh is only expressed in LMC neurons at later developmental stages (Figure 1), Shh may be responsible for the continuous existence of ARHGAP36 at least in LMC neurons via its ability to stabilize ARHGAP36 protein. It is also possible that Hox and its cofactor Pbx may additionally upregulate the expression of ARHGAP36 at later stages of MN columnar specification (Hanley et al., 2016).

We found that there was no obvious defect in the general MN formation in Arhgap36 knockout mouse embryonic spinal cord at early stages, suggesting that its function is not necessary for the initial MN differentiation. This conclusion is supported by the lack of Shh expression in early born MNs and with our notion that Shh agonist is likely needed for the activity of ARHGAP36 (Figure 6—figure supplement 2) as well as our finding that overexpression of AKT shows specific effect only on FoxP1+ LMC neurons (Figure 7). Because ARHGAP36 is also expressed in early born MNs, however, ARHGAP36 may also have a critical role in initial MN generation. This action of ARHGAP36 may have a redundant functional homologue, given no deficits were observed in early MN generation in Arhgap36 knockout mouse embryonic spinal cord. Furthermore, there was no obvious defect even in the LMC formation in male mutant spinal cord at later stages. Based on the amino acid sequence of ARHGAP36, it is predicted to be a Rho GAP family member, but it lacks the ‘arginine finger’ motif for Rho GTPase activity (Rittinger et al., 1997), suggesting that the catalytic GAP domain is not required for Gli activation. Arhgap6 is the closest homolog of Arhgap36, which does not affect Shh activation (Rack et al., 2014). It will be interesting to further investigate whether another functional homologue to ARHGAP36 controls initial MN generation as well as LMC formation particularly in male embryos. At E12.5, we observed that LMCm (Isl1+/FoxP1+) neurons are increased, whereas LMCl (Lhx1+/FoxP1+) neurons are decreased in Arhgap36 mutant spinal cord (Figure 8—figure supplement 1). But later on, the number of FoxP1+ LMC neurons decreased significantly in Arhgap36 deficient mouse embryonic spinal cord. These results suggest that the increased early born LMCm neurons in the Arhgap36 mutant spinal cord somehow adopt other cell fates or degenerate. These possibilities need to be further investigated in the future. Later born LMCl neurons have to migrate through earlier born LMCs (Sockanathan et al., 2003; Maden, 2006) but it is not known whether early born LMCm neurons can change their fate into LMCl neurons by receiving signals such as RA and Shh from the neighboring cells. It will be interesting to further investigate whether early born LMCm neurons affect LMCl neuron specification through Shh signaling pathway and its modulator ARHGAP36.

In the developing central nervous system, a number of differentiated cells degenerate during development (Buss et al., 2006; Oppenheim, 1991). Initially overproduced MNs undergo cell death by competing for a limited amount of neurotropic factors when they arrive at the target muscles (Yamamoto and Henderson, 1999). LMC neuronal axons navigate through a very long distance to their final destinations by integrating intrinsic factors and extrinsic pro- and anti-apoptotic signals. Based on the well-known function of AKT in cell survival, we speculate that ARHGAP36 proteins stabilized by AKT may also function to maintain the proper number of differentiated LMC neurons by blocking their apoptosis. Indeed, we observed an increase in cell death with a reduction of FoxP1+ LMC neurons in Arhgap36 KO mouse (Figure 8—figure supplement 1D), suggesting that ARHGAP36 may function as an essential downstream effector of AKT-directed cell survival.

It remains to be further investigated how ARHGAP36 affects the FoxP1+ LMC cell fate specification. It has been shown that FoxP1 expression is regulated by PI3K/AKT/p70S6K signaling cascade in breast cancer (Halacli and Dogan, 2015). These results further support our proposal that the expression of FoxP1 in LMC neurons is likely regulated by activated AKT in collaboration with ARHGAP36. Furthermore, ARHGAP36 is up-regulated in medulloblastoma where Shh pathway is aberrantly activated (Rack et al., 2014), suggesting that similar mechanisms may be applied to tumorigenesis. Therefore, it will be interesting to further study the function of ARHGAP36 in Shh-dependent tumorigenesis, for instance using the cerebellar granule neuron progenitors (GNPs) from Arhgap36-/- mice.

Materials and methods

Key resources table.

Reagent type
(species) or
resource
Designation Source or
reference
Identifiers Additional
information
Genetic reagent (M. musculus) Shhf/f Jackson Laboratories RRID:MGI:2165468
Genetic reagent (M. musculus) Olig2-Cre PMID: 18046410 Dr. Bennett G. Novitch (University of California, Los Angeles)
Genetic reagent (M. musculus) Arhgap36-/- This paper Exon2 targetting sgRNA made by ToolGEN, injected by KRIBB
Transfected construct (M. musculus) Arhgap36-(enhancer)2:LUC This paper Arhgap36 enhancer containing HxRE sequence cloned into TK-LUC vector
Transfected construct (M. musculus) Arhgap36-(enhancer)2:GFP This paper Arhgap36 enhancer containing HxRE sequence cloned into TATA-GFP vector
Transfected construct (R. norvegicus) Isl1 PMID: 22343290
Transfected construct (M. musculus) Lhx3 PMID: 22343290
Transfected construct (E. coli) β-galactosidase PMID: 22343290
Transfected construct (M. musculus) Arhgap36 Open Biosystems Accession: BC145645
Transfected construct (M. musculus) PKA WT PMID: 23644383
Transfected construct (M. musculus) PKA K73H PMID: 23644383
Transfected construct (M. musculus) AKT2 CA Addgene RRID:Addgene_9016 myristoylated form
Transfected construct (M. musculus) AKT2 DN Addgene RRID:Addgene_60128
Sequence-based reagent Shh shRNA_sense strand This paper PCR primers GAT CCA AGC TCT TCT ACG TCA TCG TTC AAG AGA CGA TGA CGT AGA AGA GCT TTT TTT A
Sequence-based reagent Shh shRNA_antisense strand This paper PCR primers AGC TTA AAA AAA GCT CTT CTA CGT CAT CGT CTC TTG AAC GAT GAC GTA GAA GAG CTT G
Sequence-based reagent Arhgap36 enhancer_F This paper PCR primers ACTGCCTATTCGCATCGGCCTTTGA, for cloning
Sequence-based reagent Arhgap36 enhancer_R This paper PCR primers TTCTGCGGAGCCATTAGTGCGATTG, for cloning
Sequence-based reagent mouse Arhgap36_F This paper PCR primers TGG GAT CCA AGA GGA AGA TG, for RT-PCR
Sequence-based reagent mouse Arhgap36_R This paper PCR primers CAG CCA CAT CAT GGA CAT TC, for RT-PCR
Sequence-based reagent mouse Cyclophilin A_F This paper PCR primers GTC TCC TTC GAG CTG TTT GC, for RT-PCR
Sequence-based reagent mouse Cyclophilin A_R This paper PCR primers GAT GCC AGG ACC TGT ATG CT, for RT-PCR
Sequence-based reagent mouse Arhgap36 enhancer_F This paper PCR primers ACC TTG TAG CAG GAC TGG GGT, for ChIP
Sequence-based reagent mouse Arhgap36 enhancer_R This paper PCR primers AGC CAT TAG TGC GAT TGC TCT, for ChIP
Sequence-based reagent Untr6_F PMID: 18854042 PCR primers TCA GGC ATG AAC CAC CAT AC, for ChIP
Sequence-based reagent Untr6_R PMID: 18854042 PCR primers AAC ATC CAC ACG TCC AGT GA, for ChIP
Antibody anti-Hb9/MNR2 (Mouse) DSHB DSHB Cat# 81.5C10, RRID:AB_2145209 IHC, 1:500
Antibody anti-Isl1
(Rabbit monoclonal)
Abcam Abcam Cat# ab109517, RRID:AB_10866454 IHC, 1:2000
Antibody anti-FoxP1
(Rabbit polyclonal)
Abcam Abcam Cat# ab16645, RRID:AB_732428 IHC, 1:1000
Antibody anti-Nkx2.2
(Mouse monoclonal)
DSHB DSHB Cat# 74.5A5, RRID:AB_531794 IHC, 1:100
Antibody anti-Pax6
(Mouse monoclonal)
DSHB DSHB Cat# pax6, RRID:AB_528427 IHC, 1:500
Antibody anti-Olig2
(Rabbit polyclonal)
Abcam Millipore Cat# AB15328, RRID:AB_2299035 IHC, 1:1000
Antibody anti-β-gal
(Chicken polyclonal)
Abcam Abcam Cat# ab9361, RRID:AB_307210 IHC, 1:5000
Antibody anti-Lhx3
(Rabbit polyclonal)
Abcam Abcam Cat# ab14555, RRID:AB_301332 IHC, 1:500
Antibody anti-nNOS
(Rabbit polyclonal)
Immunostar ImmunoStar Cat# 24287, RRID:AB_572256 IHC, 1:1000
Antibody anti-Chx10 (Guinea pig polyclonal) PMID: 18539116 IHC, 1:1000
Antibody anti-GFP
(Rabbit polyclonal)
Life Technologies Thermo Fisher Scientific Cat# A-11122, RRID:AB_221569 IHC, 1:1000
Antibody anti-Hb9
(Guinea pig polyclonal)
PMID: 30177510 IHC, 1:1000
rat Hb9 C-terminus (234–403 aa
Antibody anti-ARHGAP36
(Rabbit polyclonal)
This paper IHC, 1:2000
mouse ARHGAP36(201–590 aa)
Antibody anti-HA (Mouse monoclonal) Covance Covance Research Products Inc Cat# MMS-101R-500, RRID:AB_10063630 IP, IB, 1:5000
Antibody anti-Gli3
(Goat polyclonal)
R and D Systems R and D Systems Cat# AF3690, RRID:AB_2232499 IB, 1:250
Antibody anti-ARHGAP36 (Rabbit polyclonal) Sigma-Aldrich Sigma-Aldrich Cat# HPA002064, RRID:AB_1078891 IB, 1:2000
Antibody anti-β-tubulin
(Rabbit polyclonal)
Santa Cruz Santa Cruz Biotechnology Cat# sc-9104, RRID:AB_2241191 IB, 1:2000
Antibody anti-pSER Cell Signaling Cat. #9651 IB, 1:5000
Antibody anti-TuJ1 (Mouse monoclonal) Covance Covance Research Products Inc Cat# MMS-435P, RRID:AB_2313773 IB, 1:5000
IHC, 1:5000
Antibody anti-FoxP1
(Rabbit polyclonal)
abcam Abcam Cat# ab16645, RRID:AB_732428 IB, 1:1000
Antibody anti-pCREB
(Rabbit monoclonal)
Cell Signaling Cell Signaling Technology Cat# 9198, RRID:AB_2561044 IB, 1:1000
Cell line (M. musculus) P19 ATCC ATCC Cat# CRL-1825, RRID:CVCL_2153 embryonic carcinoma cells
Cell line (Homo-sapiens) HEK293T ATCC ATCC Cat# CRL-3216, RRID:CVCL_0063
Cell line (M. musculus) A172L ESC PMID: 22343290, 22039605
Chemical compound, drug iAKT1/2 Sigma Aldrich A6730 10 μM
Chemical compound, drug SAG Calbiochem MER-566660 0.25 μM
Chemical compound, drug Lipofectamine 2000 Invitrogen Cat. 52887
Chemical compound, drug Superfect Qiagen Cat. 301307
Chemical compound, drug SuperScript III First-Strand Synthesis System Invitrogen Cat. 18080085
Chemical compound, drug SYBR-Green kit Enzynomics RT501S
Software, algorithm GPS 3.0 PMID: 15980451 http://gps.biocuckoo.cn/

DNA constructs

Arhgap36-(enhancer)2:LUC and Arhgap36-(enhancer)2:GFP reporters were constructed with two copies of enhancer genomic fragments (268 bp) into synthetic TK-LUC or TATA-GFP vectors; primers used for enhancer genomic PCR are forward: 5’-ACT GCC TAT TCG CAT CGG CCT TTG A-3’ and reverse: 5’-TTC TGC GGA GCC ATT AGT GCG ATT G-3’. Rat Isl1 and mouse Lhx3 and LacZ genes were cloned in pCS2 containing a HA, Flag or myc-epitope tag for expression in mammalian cells and chick embryos, as previously described (Lee et al., 2012; Lee et al., 2004; Lee and Pfaff, 2003; Thaler et al., 2002). Mouse Arhgap36 gene was purchased from Open Biosystems and cloned into HA or Flag-tagged pCS2 vectors and UAS enhancer-CMVmini promoter containing vector. Hb9 promoter region (1212 bp) was cloned to pCS2 vector containing Gal4 transcriptional activator gene. PKA WT and K73H mutant were cloned into HA-pCS2 vector. HA-tagged AKT2 WT, dominant negative form (Addgene #60128) and constitutively active form AKT2 (Addgene #9016) were cloned into pcDNA3 or pCS2 vector. Short hairpin RNA (shRNA) of chick Shh was cloned into EFU6-300 vector; sense strand: 5’-GAT CCA AGC TCT TCT ACG TCA TCG TTC AAG AGA CGA TGA CGT AGA AGA GCT TTT TTT A-3’, antisense strand: 5’-AGC TTA AAA AAA GCT CTT CTA CGT CAT CGT CTC TTG AAC GAT GAC GTA GAA GAG CTT G-3’.

Chick in ovo electroporation, Immunohistochemistry and in situ hybridization assays

DNAs were injected into the lumen of the neural tube of HH stage 13 chick embryos, which were then electroporated (Thaler et al., 2002). The embryos were harvested 3 or 4 days post-electroporation and fixed in 4% paraformaldehyde, embedded in OCT and cryosectioned in 12 μm thickness for IHC assays or 18 μm thickness for ISH with digoxigenin-labeled probes. Each set of chick electroporation experiments was repeated independently at least three times. Representative sets of images from reproducible results were presented.

For IHC assays, the following antibodies were used; mouse anti-Hb9/MNR2 (DSHB, 5C10, 1:500), rabbit anti-Isl1 (abcam, ab109517, 1:2000), rabbit anti-FoxP1 (abcam, ab16645, 1:1000), mouse anti-Nkx2.2 (DSHB, 5A5, 1:100), mouse anti-Pax6 (DSHB, 1:500), rabbit anti-Olig2 (abcam, ab15328, 1:1000), chicken anti-β-gal (Abcam, ab9361, 1:5000), rabbit anti-Lhx3 (abcam, ab14555, 1:500), rabbit anti-nNOS (Immunostar, 1:1000), guinea pig anti-Chx10 (1:1000) (Lee et al., 2008; Thaler et al., 1999) and rabbit anti-GFP (Life Technologies, A11122, 1:1000). Guinea pig anti-Hb9 antibody was raised from guinea pig using rat Hb9 C-terminus (234–403 aa) as an antigen and rabbit anti-ARHGAP36 antibody was raised from rabbit with mouse ARHGAP36 (201–590 aa) as an antigen.

For ISH analyses, mouse Arhgap36, Akt1, Akt2, Akt3, Prkaca, Prkacb, Prkar1a, Prkar1b, Prkar2a, Prkar2b chick Ptch1, Gli1, and Shh were cloned to pBluescript vector and used to generate digoxigenin-labeled riboprobes.

Mice

All mouse works were performed under an approved protocol (SNU-150123-1-2) by the Institutional Animal Care and Use Committee (IACUC) at Seoul National University. Arhgap36 mice were generated using CRISPR/Cas9 system. We injected Cas9 mRNA and single guide RNA (sgRNA) directly into mouse embryos to target exon 2 of Arhgap36 gene and obtained a mouse line with seven nucleotides deleted (Figure 8—figure supplement 1). This resulted in a premature stop codon producing a truncated ARHGAP36 protein of 84 amino acids. Targeting sgRNA is 5’-GAC TGC TTA CCA CGA ACT CGT GG-3’, (ToolGEN). Cas9 mRNA and sgRNA mixtures were injected into one-cell embryos (Korea Research Institute of Bioscience and Biotechnology). Arhgap36-/y male mice were crossed with Arhgap36+/- female mice to get KO mutant embryos for the analyses. Shhf/f and Olig2-Cre mouse lines were described previously (Lewis et al., 2001; Dessaud et al., 2007). Shhf/f mice were obtained from the Jackon Laboratories (Bar Harbor, ME). Mouse embryos were collected at indicated developmental stages and processed similarly to chick embryos as described above.

Luciferase reporter assays

P19 embryonic carcinoma cells (ATCC, #CRL-1825) were cultured in MEM supplemented with 10% fetal bovine serum (FBS). For luciferase assays, P19 cells were plated in 48-well plate and incubated for 24 hr, followed by transient transfections using Lipofectamine 2000 (Invitrogen). An actin-β-galactosidase plasmid was co-transfected for normalization of transfection efficiency and empty vectors were used to equalize the total amount of DNA. Cells were harvested 36–40 hr after transfection. Cell extracts were assayed for luciferase activity and the values were normalized with β-galactosidase activity. All transfections were repeated independently at least three times. Data are presented as the mean of triplicate values obtained from representative experiments. Error bars represent standard deviation.

Co-immunoprecipitation and immunoblotting assays

HEK293T cells (ATCC, #CRL-3216) were cultured in DMEM media supplemented with 10% FBS. For co-immunoprecipitation, HEK293T cells were seeded on 10 cm tissue culture dishes, cultured in DMEM media supplemented with 10% FBS, and transfected with the expression vectors tagged with Flag, and HA using Superfect (Qiagen) or calcium phosphate. Cells were treated with AKT1/2 kinase inhibitor, iAKT1/2 (Sigma Aldrich, A6730) at 10 μM for 20 hr and then cells were harvested and lysed in IP buffer (20 mM Tris-HCl, pH 8.0, 0.5 % NP-40, 1 mM EDTA, 150 mM NaCl, 2 mM PMSF, 10% Glycerol, 4 mM Na3VO4, 200 mM NaF, 20 mM Na-pyroPO4, and protease inhibitor cocktail). In these studies, immunoprecipitations were performed with mouse anti-HA antibody (Covance). Immunoblotting assays were performed using goat anti-Gli3 (R and D Systems, AF3690, 1:250), rabbit anti-ARHGAP36 (Sigma, HPA-002064, 1:2000), mouse anti-HA (Covance, 1:5000), rabbit anti-β-tubulin (Santa Cruz, sc-9104, 1:2000), rabbit anti-pSER (Cell Signaling, #9651, 1:5000), mouse anti-TuJ1 (Covance, 1:5000), rabbit anti-FoxP1 (abcam, ab16645, 1:1000) and anti-pCREB (Cell Signaling, 9198S, 1:1000) antibodies.

Differentiation of ESC lines, RNA extraction and Quantitative RT-PCR

The mouse A172L ESC line (Iacovino et al., 2011; Lee et al., 2012) was maintained in an undifferentiated state on 0.1% gelatin-coated dishes in the ESC growth media that consist of knockout DMEM, 10% FBS, 0.1 mM nonessential amino acids, 2 mM L-glutamine, 0.1 mM β-mercaptoethanol, and recombinant leukemia inhibitory factor (LIF) (1000 units/ml, Chemicon). Cell line was tested periodically for Mycoplasma contamination and has not shown evidence of Mycoplasma. For MN differentiation assays, ESCs were trypsinized and plated in ADFNK media containing advanced DMEM with F-12:neurobasal medium, 200 mM L-glutamine, 1% penicillin/streptomycin, 10% knockout serum replacement, and 0.1% β-mercaptoethanol in suspension as cell aggregates for 2 days. The ESC aggregates (embryoid bodies, EBs) were treated with all-trans RA (1 μM) and Shh agonist, SAG (0.25 μM, Calbiochem) for 2 days. Then, RA and SAG-treated EBs were cultured without or with AKT1/2 inhibitor, iAKT1/2 (10 μM, Sigma Aldrich) in the presence of RA and SAG for another 2 days. At Day6, cells were harvested for RNA extraction, IHC and immunoblotting assays. Total RNAs were extracted using the Trizol (Invitrogen) and reverse-transcribed using the SuperScript III First-Strand Synthesis System (Invitrogen). The levels of mRNAs were determined with quantitative RT-PCR using SYBR-Green kit (Enzynomics) and CFX Connect Real-Time PCR detection system (Biorad). The following primers were used; mouse Arhgap36, 5’-TGG GAT CCA AGA GGA AGA TG, 5’-CAG CCA CAT CAT GGA CAT TC, and Cyclophilin A, 5’-GTC TCC TTC GAG CTG TTT GC, 5’-GAT GCC AGG ACC TGT ATG CT. Data are shown as the mean of duplicate values obtained from representative experiments. Error bars represent standard deviation.

ChIP assays

ChIP was performed in mouse embryonic spinal cords as described previously (Nam and Lee, 2016; Cho et al., 2014; Lee et al., 2013). The spinal cords were microdissected from E12.5 mouse embryos and cells were dissociated and subjected to ChIP assays. Cells were washed with PBS buffer, fixed in 1% formaldehyde for 10 min at room temperature, and quenched by 125 mM glycine. Cells were washed with Buffer I (0.25% Triton X-100, 10 mM EDTA, 0.5 mM EGTA, 10 mM Hepes, pH 6.5) and Buffer II (200 mM NaCl, 1 mM EDTA, 0.5 mM EGTA, 10 mM Hepes, pH 6.5) sequentially. Then, cells were lysed with lysis buffer (0.5% SDS, 5 mM EDTA, 50 mM Tris·HCl, pH 8.0, protease inhibitor mixture) and were subjected to sonication for DNA shearing. Next, cell lysates were diluted 1:10 in ChIP buffer (0.5% Triton X-100, 2 mM EDTA, 100 mM NaCl, 50 mM Tris·HCl, pH 8.0, protease inhibitor mixture) and, for immunoclearing, were incubated with IgG and protein A agarose beads for 1 hr at 4°C. The supernatant was collected after quick spin and incubated with anti- IgG (Santa Cruz), anti-Isl1 (Tsuchida et al., 1994) and anti-Lhx3 (Sharma et al., 1998) antibodies and protein A agarose beads to precipitate the Isl1-Lhx3 complex/chromatin complex for overnight at 4°C. The purified final DNA samples were subjected to quantitative PCR reactions. The primers that were used for ChIP-PCR are; Arhgap36 enhancer, forward, 5’-ACC TTG TAG CAG GAC TGG GGT, reverse, 5’-AGC CAT TAG TGC GAT TGC TCT, and Untr6, forward, 5’-TCA GGC ATG AAC CAC CAT AC, reverse, 5’-AAC ATC CAC ACG TCC AGT GA.

Acknowledgements

We thank Dr. Jae W Lee (OHSU, Portland, USA) for reagents and many helpful suggestions during the course of this work as well as for critically reading the manuscript. This research was supported by grants of the Korea Health Technology R and D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health and Welfare, Republic of Korea (grant number: HI17C0447) to S Lee and NIH/NINDS (R01NS100471) to S-KL.

Funding Statement

The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Contributor Information

Seunghee Lee, Email: leeseung@snu.ac.kr.

Marianne E Bronner, California Institute of Technology, United States.

Francois Guillemot, The Francis Crick Institute, United Kingdom.

Funding Information

This paper was supported by the following grants:

  • Korea Health Industry Development Institute HI17C0447 to Seunghee Lee.

  • National Institute of Neurological Disorders and Stroke R01NS100471 to Soo-Kyung Lee.

Additional information

Competing interests

No competing interests declared.

Author contributions

Data curation, Formal analysis, Validation, Writing—original draft.

Resources, Validation, Methodology.

Data curation, Formal analysis, Writing—review and editing, Generated new images with Shh KD experiment for the revision and counted cell numbers for quantification.

Data curation, Formal analysis, Writing—review and editing, Generated new images for early stage Arhgap36 KO embryos for the revision and counted cell numbers for quantification.

Data curation, Formal analysis, Writing—review and editing, Generated new images of Shh-cKO embryos with BrdU incorporation, Ki67, MN columnar marker stainings for the revision and counted cell numbers for quantification.

Conceptualization, Supervision, Funding acquisition, Writing—review and editing.

Conceptualization, Formal analysis, Supervision, Funding acquisition, Investigation, Writing—original draft, Project administration, Writing—review and editing.

Ethics

Animal experimentation: All mouse works were performed under an approved protocol (SNU-150123-1-2) by the Institutional Animal Care and Use Committee (IACUC) at Seoul National University.

Additional files

Transparent reporting form
DOI: 10.7554/eLife.46683.032

Data availability

All data generated or analysed during this study are included in the manuscript and supporting files.

The following previously published datasets were used:

Lee S, Shen R, Cho HH, Kwon RJ, Seo SY, Lee JW, Lee SK. 2013. STAT3 promotes motor neuron differentiation by collaborating with motor neuron-specific LIM complex. NCBI Gene Expression Omnibus. GSE50993

Mazzoni EO, Mahony S, Closser M, Morrison CA, Nedelec S, Williams DJ, An D, Gifford DK, Wichterle H. 2013. Synergistic binding of transcription factors to cell-specific enhancers programs motor neuron identity. NCBI Gene Expression Omnibus. GSE31456

References

  1. Arber S. FoxP1: conducting the hox symphony in spinal motor neurons. Nature Neuroscience. 2008;11:1122–1124. doi: 10.1038/nn1008-1122. [DOI] [PubMed] [Google Scholar]
  2. Asaoka Y. Phosphorylation of Gli by cAMP-dependent protein kinase. Vitamins and Hormones. 2012;88:293–307. doi: 10.1016/B978-0-12-394622-5.00013-4. [DOI] [PubMed] [Google Scholar]
  3. Bitgood MJ, McMahon AP. Hedgehog and Bmp genes are coexpressed at many diverse sites of cell-cell interaction in the mouse embryo. Developmental Biology. 1995;172:126–138. doi: 10.1006/dbio.1995.0010. [DOI] [PubMed] [Google Scholar]
  4. Briscoe J, Thérond PP. The mechanisms of hedgehog signalling and its roles in development and disease. Nature Reviews Molecular Cell Biology. 2013;14:416–429. doi: 10.1038/nrm3598. [DOI] [PubMed] [Google Scholar]
  5. Buss RR, Sun W, Oppenheim RW. Adaptive roles of programmed cell death during nervous system development. Annual Review of Neuroscience. 2006;29:1–35. doi: 10.1146/annurev.neuro.29.051605.112800. [DOI] [PubMed] [Google Scholar]
  6. Cho HH, Cargnin F, Kim Y, Lee B, Kwon RJ, Nam H, Shen R, Barnes AP, Lee JW, Lee S, Lee SK. Isl1 directly controls a cholinergic neuronal identity in the developing forebrain and spinal cord by forming cell type-specific complexes. PLOS Genetics. 2014;10:e1004280. doi: 10.1371/journal.pgen.1004280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Dasen JS, Tice BC, Brenner-Morton S, Jessell TM. A hox regulatory network establishes motor neuron pool identity and target-muscle connectivity. Cell. 2005;123:477–491. doi: 10.1016/j.cell.2005.09.009. [DOI] [PubMed] [Google Scholar]
  8. Dasen JS, De Camilli A, Wang B, Tucker PW, Jessell TM. Hox repertoires for motor neuron diversity and connectivity gated by a single accessory factor, FoxP1. Cell. 2008;134:304–316. doi: 10.1016/j.cell.2008.06.019. [DOI] [PubMed] [Google Scholar]
  9. Dasen JS, Jessell TM. Hox networks and the origins of motor neuron diversity. Current Topics in Developmental Biology. 2009;88:169–200. doi: 10.1016/S0070-2153(09)88006-X. [DOI] [PubMed] [Google Scholar]
  10. Dessaud E, Yang LL, Hill K, Cox B, Ulloa F, Ribeiro A, Mynett A, Novitch BG, Briscoe J. Interpretation of the sonic hedgehog morphogen gradient by a temporal adaptation mechanism. Nature. 2007;450:717–720. doi: 10.1038/nature06347. [DOI] [PubMed] [Google Scholar]
  11. Eccles RL, Czajkowski MT, Barth C, Müller PM, McShane E, Grunwald S, Beaudette P, Mecklenburg N, Volkmer R, Zühlke K, Dittmar G, Selbach M, Hammes A, Daumke O, Klussmann E, Urbé S, Rocks O. Bimodal antagonism of PKA signalling by ARHGAP36. Nature Communications. 2016;7:12963. doi: 10.1038/ncomms12963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Grewal SS, Fass DM, Yao H, Ellig CL, Goodman RH, Stork PJ. Calcium and cAMP signals differentially regulate cAMP-responsive element-binding protein function via a Rap1-extracellular signal-regulated kinase pathway. Journal of Biological Chemistry. 2000;275:34433–34441. doi: 10.1074/jbc.M004728200. [DOI] [PubMed] [Google Scholar]
  13. Halacli SO, Dogan AL. FOXP1 regulation via the PI3K/Akt/p70S6K signaling pathway in breast Cancer cells. Oncology Letters. 2015;9:1482–1488. doi: 10.3892/ol.2015.2885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Hanley O, Zewdu R, Cohen LJ, Jung H, Lacombe J, Philippidou P, Lee DH, Selleri L, Dasen JS. Parallel Pbx-Dependent pathways govern the coalescence and fate of motor columns. Neuron. 2016;91:1005–1020. doi: 10.1016/j.neuron.2016.07.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Harfe BD, Scherz PJ, Nissim S, Tian H, McMahon AP, Tabin CJ. Evidence for an expansion-based temporal Shh gradient in specifying vertebrate digit identities. Cell. 2004;118:517–528. doi: 10.1016/j.cell.2004.07.024. [DOI] [PubMed] [Google Scholar]
  16. Harrison KA, Thaler J, Pfaff SL, Gu H, Kehrl JH. Pancreas dorsal lobe agenesis and abnormal islets of Langerhans in Hlxb9-deficient mice. Nature Genetics. 1999;23:71–75. doi: 10.1038/12674. [DOI] [PubMed] [Google Scholar]
  17. Hollyday M. Motoneuron histogenesis and the development of limb innervation. Current Topics in Developmental Biology. 1980a;15 Pt 1:181–215. doi: 10.1016/S0070-2153(08)60120-9. [DOI] [PubMed] [Google Scholar]
  18. Hollyday M. Organization of motor pools in the chick lumbar lateral motor column. The Journal of Comparative Neurology. 1980b;194:143–170. doi: 10.1002/cne.901940108. [DOI] [PubMed] [Google Scholar]
  19. Iacovino M, Bosnakovski D, Fey H, Rux D, Bajwa G, Mahen E, Mitanoska A, Xu Z, Kyba M. Inducible cassette exchange: a rapid and efficient system enabling conditional gene expression in embryonic stem and primary cells. Stem Cells. 2011;29:1580–1588. doi: 10.1002/stem.715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Ingham PW, Nakano Y, Seger C. Mechanisms and functions of hedgehog signalling across the metazoa. Nature Reviews Genetics. 2011;12:393–406. doi: 10.1038/nrg2984. [DOI] [PubMed] [Google Scholar]
  21. Itou J, Kawakami H, Quach T, Osterwalder M, Evans SM, Zeller R, Kawakami Y. Islet1 regulates establishment of the posterior hindlimb field upstream of the Hand2-Shh morphoregulatory gene network in mouse embryos. Development. 2012;139:1620–1629. doi: 10.1242/dev.073056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Iyer GH, Garrod S, Woods VL, Taylor SS. Catalytic independent functions of a protein kinase as revealed by a kinase-dead mutant: study of the Lys72His mutant of cAMP-dependent kinase. Journal of Molecular Biology. 2005;351:1110–1122. doi: 10.1016/j.jmb.2005.06.011. [DOI] [PubMed] [Google Scholar]
  23. Jessell TM. Neuronal specification in the spinal cord: inductive signals and transcriptional codes. Nature Reviews Genetics. 2000;1:20–29. doi: 10.1038/35049541. [DOI] [PubMed] [Google Scholar]
  24. Ji SJ, Zhuang B, Falco C, Schneider A, Schuster-Gossler K, Gossler A, Sockanathan S. Mesodermal and neuronal retinoids regulate the induction and maintenance of limb innervating spinal motor neurons. Developmental Biology. 2006;297:249–261. doi: 10.1016/j.ydbio.2006.05.015. [DOI] [PubMed] [Google Scholar]
  25. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337:816–821. doi: 10.1126/science.1225829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Jung H, Lacombe J, Mazzoni EO, Liem KF, Grinstein J, Mahony S, Mukhopadhyay D, Gifford DK, Young RA, Anderson KV, Wichterle H, Dasen JS. Global control of motor neuron topography mediated by the repressive actions of a single hox gene. Neuron. 2010;67:781–796. doi: 10.1016/j.neuron.2010.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Kanda S, Mochizuki Y, Suematsu T, Miyata Y, Nomata K, Kanetake H. Sonic hedgehog induces capillary morphogenesis by endothelial cells through phosphoinositide 3-kinase. Journal of Biological Chemistry. 2003;278:8244–8249. doi: 10.1074/jbc.M210635200. [DOI] [PubMed] [Google Scholar]
  28. Kotani T. Protein kinase A activity and hedgehog signaling pathway. Vitamins and Hormones. 2012;88:273–291. doi: 10.1016/B978-0-12-394622-5.00012-2. [DOI] [PubMed] [Google Scholar]
  29. Landmesser L. The distribution of motoneurones supplying chick hind limb muscles. The Journal of Physiology. 1978;284:371–389. doi: 10.1113/jphysiol.1978.sp012545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Lee SK, Jurata LW, Funahashi J, Ruiz EC, Pfaff SL. Analysis of embryonic motoneuron gene regulation: derepression of general activators function in concert with enhancer factors. Development. 2004;131:3295–3306. doi: 10.1242/dev.01179. [DOI] [PubMed] [Google Scholar]
  31. Lee S, Lee B, Joshi K, Pfaff SL, Lee JW, Lee SK. A regulatory network to segregate the identity of neuronal subtypes. Developmental Cell. 2008;14:877–889. doi: 10.1016/j.devcel.2008.03.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Lee S, Cuvillier JM, Lee B, Shen R, Lee JW, Lee SK. Fusion protein Isl1-Lhx3 specifies motor neuron fate by inducing motor neuron genes and concomitantly suppressing the interneuron programs. PNAS. 2012;109:3383–3388. doi: 10.1073/pnas.1114515109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Lee S, Shen R, Cho HH, Kwon RJ, Seo SY, Lee JW, Lee SK. STAT3 promotes motor neuron differentiation by collaborating with motor neuron-specific LIM complex. PNAS. 2013;110:11445–11450. doi: 10.1073/pnas.1302676110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Lee SK, Pfaff SL. Transcriptional networks regulating neuronal identity in the developing spinal cord. Nature Neuroscience. 2001;4:1183–1191. doi: 10.1038/nn750. [DOI] [PubMed] [Google Scholar]
  35. Lee SK, Pfaff SL. Synchronization of neurogenesis and motor neuron specification by direct coupling of bHLH and homeodomain transcription factors. Neuron. 2003;38:731–745. doi: 10.1016/S0896-6273(03)00296-4. [DOI] [PubMed] [Google Scholar]
  36. Lewis PM, Dunn MP, McMahon JA, Logan M, Martin JF, St-Jacques B, McMahon AP. Cholesterol modification of sonic hedgehog is required for long-range signaling activity and effective modulation of signaling by Ptc1. Cell. 2001;105:599–612. doi: 10.1016/S0092-8674(01)00369-5. [DOI] [PubMed] [Google Scholar]
  37. Lin AW, Carpenter EM. Hoxa10 and Hoxd10 coordinately regulate lumbar motor neuron patterning. Journal of Neurobiology. 2003;56:328–337. doi: 10.1002/neu.10239. [DOI] [PubMed] [Google Scholar]
  38. Maden M. Retinoids and spinal cord development. Journal of Neurobiology. 2006;66:726–738. doi: 10.1002/neu.20248. [DOI] [PubMed] [Google Scholar]
  39. Mali RS, Peng GH, Zhang X, Dang L, Chen S, Mitton KP. FIZ1 is part of the regulatory protein complex on active photoreceptor-specific gene promoters in vivo. BMC Molecular Biology. 2008;9:87. doi: 10.1186/1471-2199-9-87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Martí E, Bovolenta P. Sonic hedgehog in CNS development: one signal, multiple outputs. Trends in Neurosciences. 2002;25:89–96. doi: 10.1016/S0166-2236(02)02062-3. [DOI] [PubMed] [Google Scholar]
  41. Mazzoni EO, Mahony S, Closser M, Morrison CA, Nedelec S, Williams DJ, An D, Gifford DK, Wichterle H. Synergistic binding of transcription factors to cell-specific enhancers programs motor neuron identity. Nature Neuroscience. 2013;16:1219–1227. doi: 10.1038/nn.3467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Nam H, Lee S. Identification of STAM1 as a novel effector of ventral projection of spinal motor neurons. Development. 2016;143:2334–2343. doi: 10.1242/dev.135848. [DOI] [PubMed] [Google Scholar]
  43. Nichols M, Weih F, Schmid W, DeVack C, Kowenz-Leutz E, Luckow B, Boshart M, Schütz G. Phosphorylation of CREB affects its binding to high and low affinity sites: implications for cAMP induced gene transcription. The EMBO Journal. 1992;11:3337–3346. doi: 10.1002/j.1460-2075.1992.tb05412.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Oppenheim RW. Cell death during development of the nervous system. Annual Review of Neuroscience. 1991;14:453–501. doi: 10.1146/annurev.ne.14.030191.002321. [DOI] [PubMed] [Google Scholar]
  45. Oppenheim RW, Homma S, Marti E, Prevette D, Wang S, Yaginuma H, McMahon AP. Modulation of early but not later stages of programmed cell death in embryonic avian spinal cord by sonic hedgehog. Molecular and Cellular Neuroscience. 1999;13:348–361. doi: 10.1006/mcne.1999.0755. [DOI] [PubMed] [Google Scholar]
  46. Pan Y, Wang C, Wang B. Phosphorylation of Gli2 by protein kinase A is required for Gli2 processing and degradation and the sonic Hedgehog-regulated mouse development. Developmental Biology. 2009;326:177–189. doi: 10.1016/j.ydbio.2008.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Persson M, Stamataki D, te Welscher P, Andersson E, Böse J, Rüther U, Ericson J, Briscoe J. Dorsal-ventral patterning of the spinal cord requires Gli3 transcriptional repressor activity. Genes & Development. 2002;16:2865–2878. doi: 10.1101/gad.243402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Peviani M, Tortarolo M, Battaglia E, Piva R, Bendotti C. Specific induction of Akt3 in spinal cord motor neurons is neuroprotective in a mouse model of familial amyotrophic lateral sclerosis. Molecular Neurobiology. 2014;49:136–148. doi: 10.1007/s12035-013-8507-6. [DOI] [PubMed] [Google Scholar]
  49. Pfaff SL, Mendelsohn M, Stewart CL, Edlund T, Jessell TM. Requirement for LIM homeobox gene Isl1 in motor neuron generation reveals a motor neuron-dependent step in interneuron differentiation. Cell. 1996;84:309–320. doi: 10.1016/S0092-8674(00)80985-X. [DOI] [PubMed] [Google Scholar]
  50. Pfaff SL. Developmental neuroscience: hox and fox. Nature. 2008;455:295–297. doi: 10.1038/455295a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Rack PG, Ni J, Payumo AY, Nguyen V, Crapster JA, Hovestadt V, Kool M, Jones DT, Mich JK, Firestone AJ, Pfister SM, Cho YJ, Chen JK. Arhgap36-dependent activation of Gli transcription factors. PNAS. 2014;111:11061–11066. doi: 10.1073/pnas.1322362111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F. Genome engineering using the CRISPR-Cas9 system. Nature Protocols. 2013;8:2281–2308. doi: 10.1038/nprot.2013.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Riobó NA, Lu K, Ai X, Haines GM, Emerson CP. Phosphoinositide 3-kinase and Akt are essential for sonic hedgehog signaling. PNAS. 2006;103:4505–4510. doi: 10.1073/pnas.0504337103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Rittinger K, Walker PA, Eccleston JF, Nurmahomed K, Owen D, Laue E, Gamblin SJ, Smerdon SJ. Crystal structure of a small G protein in complex with the GTPase-activating protein rhoGAP. Nature. 1997;388:693–697. doi: 10.1038/41805. [DOI] [PubMed] [Google Scholar]
  55. Rousso DL, Gaber ZB, Wellik D, Morrisey EE, Novitch BG. Coordinated actions of the forkhead protein Foxp1 and hox proteins in the columnar organization of spinal motor neurons. Neuron. 2008;59:226–240. doi: 10.1016/j.neuron.2008.06.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Sagner A, Gaber ZB, Delile J, Kong JH, Rousso DL, Pearson CA, Weicksel SE, Melchionda M, Mousavy Gharavy SN, Briscoe J, Novitch BG. Olig2 and hes regulatory dynamics during motor neuron differentiation revealed by single cell transcriptomics. PLOS Biology. 2018;16:e2003127. doi: 10.1371/journal.pbio.2003127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Sharma K, Sheng HZ, Lettieri K, Li H, Karavanov A, Potter S, Westphal H, Pfaff SL. LIM homeodomain factors Lhx3 and Lhx4 assign subtype identities for motor neurons. Cell. 1998;95:817–828. doi: 10.1016/S0092-8674(00)81704-3. [DOI] [PubMed] [Google Scholar]
  58. Shirasaki R, Pfaff SL. Transcriptional codes and the control of neuronal identity. Annual Review of Neuroscience. 2002;25:251–281. doi: 10.1146/annurev.neuro.25.112701.142916. [DOI] [PubMed] [Google Scholar]
  59. Sockanathan S, Perlmann T, Jessell TM. Retinoid receptor signaling in postmitotic motor neurons regulates rostrocaudal positional identity and axonal projection pattern. Neuron. 2003;40:97–111. doi: 10.1016/S0896-6273(03)00532-4. [DOI] [PubMed] [Google Scholar]
  60. Stifani N. Motor neurons and the generation of spinal motor neuron diversity. Frontiers in Cellular Neuroscience. 2014;8:293. doi: 10.3389/fncel.2014.00293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Tanabe Y, William C, Jessell TM. Specification of motor neuron identity by the MNR2 homeodomain protein. Cell. 1998;95:67–80. doi: 10.1016/S0092-8674(00)81783-3. [DOI] [PubMed] [Google Scholar]
  62. Taylor SS, Buechler JA, Yonemoto W. cAMP-dependent protein kinase: framework for a diverse family of regulatory enzymes. Annual Review of Biochemistry. 1990;59:971–1005. doi: 10.1146/annurev.bi.59.070190.004543. [DOI] [PubMed] [Google Scholar]
  63. Thaler J, Harrison K, Sharma K, Lettieri K, Kehrl J, Pfaff SL. Active suppression of interneuron programs within developing motor neurons revealed by analysis of homeodomain factor HB9. Neuron. 1999;23:675–687. doi: 10.1016/S0896-6273(01)80027-1. [DOI] [PubMed] [Google Scholar]
  64. Thaler JP, Lee SK, Jurata LW, Gill GN, Pfaff SL. LIM factor Lhx3 contributes to the specification of motor neuron and interneuron identity through cell-type-specific protein-protein interactions. Cell. 2002;110:237–249. doi: 10.1016/S0092-8674(02)00823-1. [DOI] [PubMed] [Google Scholar]
  65. Thiebes KP, Nam H, Cambronne XA, Shen R, Glasgow SM, Cho HH, Kwon JS, Goodman RH, Lee JW, Lee S, Lee SK. miR-218 is essential to establish motor neuron fate as a downstream effector of Isl1-Lhx3. Nature Communications. 2015;6:7718. doi: 10.1038/ncomms8718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Tosney KW, Landmesser LT. Development of the major pathways for neurite outgrowth in the chick hindlimb. Developmental Biology. 1985a;109:193–214. doi: 10.1016/0012-1606(85)90360-4. [DOI] [PubMed] [Google Scholar]
  67. Tosney KW, Landmesser LT. Growth cone morphology and trajectory in the lumbosacral region of the chick embryo. The Journal of Neuroscience. 1985b;5:2345–2358. doi: 10.1523/JNEUROSCI.05-09-02345.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Tsuchida T, Ensini M, Morton SB, Baldassare M, Edlund T, Jessell TM, Pfaff SL. Topographic organization of embryonic motor neurons defined by expression of LIM homeobox genes. Cell. 1994;79:957–970. doi: 10.1016/0092-8674(94)90027-2. [DOI] [PubMed] [Google Scholar]
  69. Vermot J, Schuhbaur B, Le Mouellic H, McCaffery P, Garnier JM, Hentsch D, Brûlet P, Niederreither K, Chambon P, Dollé P, Le Roux I. Retinaldehyde dehydrogenase 2 and Hoxc8 are required in the murine brachial spinal cord for the specification of Lim1+ motoneurons and the correct distribution of Islet1+ motoneurons. Development. 2005;132:1611–1621. doi: 10.1242/dev.01718. [DOI] [PubMed] [Google Scholar]
  70. Wichterle H, Lieberam I, Porter JA, Jessell TM. Directed differentiation of embryonic stem cells into motor neurons. Cell. 2002;110:385–397. doi: 10.1016/s0092-8674(02)00835-8. [DOI] [PubMed] [Google Scholar]
  71. Wu Y, Wang G, Scott SA, Capecchi MR. Hoxc10 and Hoxd10 regulate mouse columnar, divisional and motor pool identity of lumbar motoneurons. Development. 2008;135:171–182. doi: 10.1242/dev.009225. [DOI] [PubMed] [Google Scholar]
  72. Xue Y, Zhou F, Zhu M, Ahmed K, Chen G, Yao X. For phosphorylation sites prediction. Nucleic Acids Research. 2005;33:W184–W187. doi: 10.1093/nar/gki393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Yamamoto Y, Henderson CE. Patterns of programmed cell death in populations of developing spinal motoneurons in chicken, mouse, and rat. Developmental Biology. 1999;214:60–71. doi: 10.1006/dbio.1999.9413. [DOI] [PubMed] [Google Scholar]
  74. Yang L, Cai CL, Lin L, Qyang Y, Chung C, Monteiro RM, Mummery CL, Fishman GI, Cogen A, Evans S. Isl1Cre reveals a common Bmp pathway in heart and limb development. Development. 2006;133:1575–1585. doi: 10.1242/dev.02322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Yang H, Wang H, Jaenisch R. Generating genetically modified mice using CRISPR/Cas-mediated genome engineering. Nature Protocols. 2014;9:1956–1968. doi: 10.1038/nprot.2014.134. [DOI] [PubMed] [Google Scholar]
  76. Yu F, Sugawara T, Maier CM, Hsieh LB, Chan PH. Akt/Bad signaling and motor neuron survival after spinal cord injury. Neurobiology of Disease. 2005;20:491–499. doi: 10.1016/j.nbd.2005.04.004. [DOI] [PubMed] [Google Scholar]
  77. Zhang B, Zhuang T, Lin Q, Yang B, Xu X, Xin G, Zhu S, Wang G, Yu B, Zhang T, Jiang Q, Zhang C. Patched1–ArhGAP36–PKA–Inversin axis determines the ciliary translocation of Smoothened for Sonic Hedgehog pathway activation. PNAS. 2019;116:874–879. doi: 10.1073/pnas.1804042116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Zhong H, SuYang H, Erdjument-Bromage H, Tempst P, Ghosh S. The transcriptional activity of NF-kappaB is regulated by the IkappaB-associated PKAc subunit through a cyclic AMP-independent mechanism. Cell. 1997;89:413–424. doi: 10.1016/S0092-8674(00)80222-6. [DOI] [PubMed] [Google Scholar]

Decision letter

Editor: Francois Guillemot1

In the interests of transparency, eLife includes the editorial decision letter and accompanying author responses. A lightly edited version of the letter sent to the authors after peer review is shown, indicating the most substantive concerns; minor comments are not usually included.

[Editors’ note: this article was originally rejected after discussions between the reviewers, but the authors were invited to resubmit after an appeal against the decision.]

Thank you for submitting your article " Critical roles of ARHGAP36 as a signal transduction mediator of Shh pathway in lateral motor columnar specification" for consideration by eLife. Your article has been reviewed by two peer reviewers, one of whom is a member of our Board of Reviewing Editors, and the evaluation has been overseen by Marianne Bronner as the Senior Editor. The reviewers have discussed the reviews with one another and the Reviewing Editor has drafted this decision.

Summary:

This manuscript addresses the role of Sonic hedgehog expressed by postmitotic spinal neurons in chick and mouse embryos. The paper shows that Shh is required for the formation of the lateral motor column at brachial level and that it acts by activating AKT signaling, which stabilises the ARHGAP36 protein, induced in motorneurons by Isl1-Lhx3 complexes. ARHGAP36 in turn inhibits the kinase activity of PKA, which dephosphorylates Gli proteins and favours the formation of activating over repressing forms of Gli proteins.

Comments:

The reviewers found this study novel and interesting. However, reviewer 2 was concerned that electroporation experiments in chick embryos did not rule out an effect of the manipulation on patterning or proliferation of progenitor cells rather than a direct effect on postmitotic motorneurons, because electroporations were performed at a stage when patterning was not fully established and the expression constructs did not contain a motorneuron-specific promoter (reviewer 2, points 7 and 19). Similarly, the reviewer was concerned that Shh deletion in mouse embryos using the Olig2-Cre deleter line did not exclude patterning or proliferation defects due to elimination of Shh in ventral progenitors rather than in postmitotic neurons (reviewer 2 point 11). After consultation between the reviewer, reviewer 1 agrees with these points. As Olig2 is initially expressed in the ventral-most domain of the spinal cord except in the most medial cells (e.g. Dessaud et al., 2007), it cannot be excluded that Shh has been deleted by Olig2-Cre from cells in the early Olig2+ progenitor domain, resulting in the observed phenotype.

Other concerns include the lack of sufficient analysis to exclude patterning, proliferation or survival defects in the manipulated chick and mouse embryos (reviewer point 1; reviewer 2 points 5 and 6), and the insufficient quantification of the data (reviewer 1 point 2; reviewer 2 points 4, 8, 14, 17, 19, 22-25).

Addressing these different concerns would require new experiments, which would likely take substantially longer than the two months normally allocated to the revision of eLife manuscripts. We have therefore regretfully decided to reject your manuscript.

Reviewer #1:

This paper reports a novel role of Shh, produced by postmitotic motor neurons, in the specification of the lateral motor column (LMC) in the chick and mouse spinal cord. The authors show that AKT stabilises the Rho GAP protein ARHGAP36 in the LMC region, and that ARHGAP36 in turn inhibits the activity of PKA and thereby enhances the production of activating forms of Gli proteins.

The identification of a role for Shh in motor neuron specification and the elucidation of the downstream pathway involving a novel regulator and multiple signaling molecules is of significant interest. The data supports the conclusions of the authors and the work is of excellent quality and well presented. I have a few requests to improve the data, the analysis and discussion to strengthen the conclusions of the paper.

1) Shh acts as a mitogen in many systems but the authors rule out that it is the case here by labelling MN progenitors with antibodies to Olig2 and Sox2 in Figure 2C. However the Shh LOF phenotype in Figure 2A only affects the generation of a small minority of MNs, which could be due to a proliferation defect affecting only a small fraction of progenitors which may not be picked up by the current analysis. To rule this out, the authors should examine proliferating cells more directly, eg with a short pulse of EdU. This should detect a putative proliferation defect with more sensitivity than examining total progenitor numbers.

2) The ectopic expression of ARHGAP36 in the ventral spinal cord in Figure 5B increases visibly the number of FoxP1+ LMC neurons while Lhx3+ Hb9+ MMCm neurons do not appear affected. However to conclude that ARHGAP36 induces LMC neurons rather than converting MMCm into LMC, requires quantification of the data.

3) The conclusion of the experiment in Figure 6A that AKT stabilises ARHGAP36 protein should be strengthened by measuring ARHGAP36 half-life in HEK293T cells in the presence and absence of AKT, and also by showing that manipulating AKT activity does not affect ARHGAP36 transcription.

4) The regulation of ARHGAP36 expression may need further discussion. Figure 6 shows that AKT controls ARHGAP36 protein stability, but how its transcription is regulated is less clear. Figure 2 S1, shows a reduction of its expression in Shh cKO mice. How does Shh regulates its expression and are there other signals involved? Along the same line, the arrow between Shh and P-AKT in Figure 7D may need more explanation.

Reviewer #2:

The manuscript by Nam and cols, entitled "Critical roles of ARHGAP36 as a signal transduction mediator of Shh pathway in lateral motor columnar specification" proposes a novel role for Shh signalling in spinal cord development.

A putative role for Shh activity as a signal instructing the selection of specific MNs subpopulations is novel and attractive. It is based in the observation of Shh expression in postmitotic MNs, which is not fully novel, but has not been well characterized previously.

Authors also find that the Rho GAP family member Arhgap36 is expressed in postmitotic MNs, and propose that it might act as a Shh effector in MN subpopulations selection. In different cell contexts, Arhgap36 has previously been shown to inhibit PKA activity, hence regulating Shh signalling. However, it would be nice to fully define its in vivo relevance, in the context of selection of specific MNs subpopulations. In summary, the manuscript contains some novel and interesting data, however it lacks definitive demonstration of some of the conclusions raised by the authors.

Hence, I believe that, in the present format, the manuscript has important gaps that need to be fulfilled.

Specific points:

In order to propose a new role for Shh signalling as an instructive signal the selection of specific MNs subpopulations, I believe that authors should improve the characterization of the MN subpopulations expressing Shh.

1) In the chick embryo spinal cord, Shh expression has previously been reported in postmitotic motor neurons of the LMC (at stage HH15), extending later to the MMC (Stage HH35) (Figure 1 in Oppenheim et al., 1999). However, Nam and cols, in this manuscript shows Shh expression restricted to LMC neurons at the brachial level. The selected section showed in Figure 1A shows Shh expression restricted to the mLMC (and excluded from lLMC), as well as excluded from MMC. Could authors clarify these contradictory issues?

2) Panel B of this figure (expression in mouse embryos) do not show this restricted expression to mLMC. Are these specie specific differences? Or developmental stage differences?

3) A detailed temporo-spatial expression of Shh in postmitotic MNs should be provided. These should be easy to clarify combining with the available and well defined markers for each MN column (see for example Adams et al., 2015)

In order to analyse the role of Shh in MN subtype specification; loss-of-function experiments in chick embryo NT were performed by electroporation at HH stage 13 chick embryos (Materials and methods), at which patterning of the NT is not definitely established (see for example Cayuso et al., 2006).

4) Controls showing quantification of Shh- loss are required, particularly since defects appeared to be mild, despite the length of the experiments (96 hours post-electroporation at HH13). For example, Gli-bs-luc experiments to show the efficiency of the Shh-LOF vector.

5) Moreover, the role of Shh signalling in proliferation and survival of neural progenitors are also well stablished, hence controls ruling out these defects are required.

6) Controls showing markers for progenitors should be included in order to rule out patterning defects.

7) I strongly believe that, in order to analyse a specific role for Shh signalling in postmitotic MN subtype generation, experiments should have been done using a MN specific driver (for example the well characterized HB9) to avoid patterning and proliferation defects.

8) Quantitative data indicating the total number of MNs, versus sub-type specific MNs should be provided. It is important to understand whether LMNs have switch their fate, of whether LMNs are not being generated.

9) Quantitative data in Figure 1A panel D shows FoxP1+ cells, are these islet1+? Or islet1-? Same for Hb9, these analyses will discriminate between lLMB vs mLMC. Quantitative data in Figure 1A panel E shows Lhx3+ cells, same for the combination with additional marker to restrict the phenotype to specific MN subtype

10) A schematic drawing of the MN columns, including markers and phenotype would be of great help.

11) The same holds for the analysis of the Shh-Olig2C mutant phenotype. These experiments should have been done using a MN specific driver (for example the well characterized HB9CRE line, available at Jackson) to avoid patterning and proliferation defects.

12) Please indicate the total number of MNs, versus sub-type specific MNs. Please indicate whether FoxP1+ cells are islet1 and Hb9 +/-.

To identify the candidate effector genes, authors searched for target genes of the Isl1-Lhx3 transcriptional complex, well characterized previously by this group. What is the link between this searching strategy and Shh signalling, it is not clear to me, but they identify the Rho GAP family member Arhgap36, which is known to activate Gli transcriptional responses through the inhibition of PKA activities.

13) It would be nice to see, among the many genes regulated by the Isl1-Lhx3 transcriptional complex, whether there might be a cluster of HH-signalling components?

14) Authors identify a HxRE of the Arhgap36 gene, and test the in vivo activity by electroporation experiments in chick embryo NT. Please provide quantitative data indicating number of embryos analysed. Provide quantitative data indicating HB9 ectopic cells induced. Please provide electroporation time course, as well as negative control experiments.

15) I believe that the data provided here demonstrate that the Isl1-Lhx3 complex is sufficient to induce Arhgap36 expression, whether this induction is direct is not fully demonstrated.

Next authors analyse Arhgap36 expression in postmitotic neurons. This is an important issue, since Shh might be acting either in a paracrine manner, or in a cell autonomous manner, to instruct MNs subtype identity.

16) Please provide a better characterization of Arhgap36 in MN subtypes (co-localization with MN subtype markers) and co-localization (or not) with Shh, with cellular resolution.

17) Please provide Arhgap36 expression data in chick embryos MNs. It would be interesting to see the conservation of it expression in MNs.

Next authors test whether the Shh-pathway is activated upon over-expression of Arhgap36. Experiments in chick embryo NT were performed by electroporation of ARHGAP36 at HH stage 13 chick embryos (Materials and methods). These gain-of-function experiments produce NT overgrowth, corresponding to the over-activation of Shh signalling, in addition to ventralization of the entire NT. Quantitative data indicating number of embryos analysed is missing, electroporation time course, as well as quantification of marker-specific ectopic cells should be provided.

18) I believe that these experiments are controls of the capacity of ARHGAP36 in activating Shh-mediated responses in vivo. Since this has been previously documented in other systems, these experiments can go as supplementary information.

19) Authors claim that over-expression of ARHGAP36 in ventral NT, harvested at 4 dpe the number of FoxP1+ LMC neurons increased drastically, suggest that ARHGAP36 is sufficient to direct LMC fate-determination. Again here, I believe that these experiments should have been done using a MN HB9 specific driver to avoid patterning and proliferation defects. With the present experimental design, it is very difficult to assign a direct role for ARHGAP36 in postmitotic MN subpopulations selection, other that the role in MN progenitors

Again in these experiments, quantitative data indicating number of embryos analysed is required, electroporation time course is missing. Quantification of marker-specific ectopic cells should be provided.

Next, authors show that PKA activity is inhibited by ARHGAP36. However, this observation is not novel, since Eccles et al. (2016) showed that ARHGAP36 combines two distinct inhibitory mechanisms to antagonise PKA signalling; it blocks PKA catalytic activity and it targets PKAc for ubiquitin-mediated degradation, resulting in activation of the Shh pathway.

20) I don't see the point in analysing other targets but PKA in the context of MN specification, hence I believe that panels C,D,E from Figure 5 are not relevant in this context.

21) Again, in the context of Shh/ ARHGAP36 signalling in MN subtype selection, I do not see the relevance of the AKT experiments shown if Figure 6. I believe that should be removed.

Moreover, Zhang et al. (2019) find that Patched1 interacts with ArhGAP36 to the centrosome and stabilizes the PKA negative regulator (this paper should be quoted in the manuscript).

Zhang et al., Patched1-ArhGAP36-PKA-Inversin axis determines the ciliary translocation of Smoothened for Sonic Hedgehog pathway activation. Proc Natl Acad Sci U S A. 2019 Jan 15;116(3):874-879

22) Authors show reduced expression of ARHGAP36 in Shh-KO mice (Figure S4), again without any quantitative data.

Finally, authors generate an ARHGAP36 mutant mice. Authors claim that at early developmental stages, there was no defect in MN generation. Quantitative data on this control observation should be provided.

23) Analysis of the phenotype generated by ARHGAP36_CRISPR mutant in MNs at the brachial and the thoracic levels of the developing spinal cord, again require quantitative data indicating that the total number of MNs, have not changed at thoracic levels.

24) Quantitative data indicating that the total number of MMC and the total number of V2 interneurons, have not changed at brachial levels.

25) Quantitative data indicating the total MN numbers versus sub-type specific MNs should be provided. It is important to understand whether, in the absence of ARHGAP36, LMNs have switch their fate, of whether LMNs are not being generated.

eLife. 2019 Jul 15;8:e46683. doi: 10.7554/eLife.46683.039

Author response


[…] Addressing these different concerns would require new experiments, which would likely take substantially longer than the two months normally allocated to the revision of eLife manuscripts. We have therefore regretfully decided to reject your manuscript.

One of the main concerns of reviewers was if Shh deletion in mouse embryos using the Olig2-Cre deleter line could exclude patterning or proliferation defects due to the elimination of Shh in ventral progenitors rather than in postmitotic neurons. The reviewer pointed out that "As Olig2 is initially expressed in the ventral-most domain of the spinal cord except in the most medial cells [1], it cannot be excluded that Shh has been deleted by Olig2-Cre from cells in the early Olig2+ progenitor domain, resulting in the observed phenotype."

We understand why the reviewer expressed the concern as Shh secreted from the floor plate is critical for the patterning of the ventral neural tube, thus affecting the subsequent motor neuron differentiation. However, the data from my lab and other groups indicate that the expression of Shh in the floor plate is not compromised in our Shh-cKO;Olig2-Cre mouse model, excluding the possibility that the observed phenotype is caused by Shh deletion in the floor plate.

First, it is important to note that Shh is specifically expressed in the floor plate, but not in neurons, at the early stage of spinal cord development when the ventral progenitors undergo Shh-dependent patterning. Our in situ hybridization data (Figure 1A,B) are consistent with the previous reports. Further supporting the floor plate-specific expression pattern of Shh, Shh-Cre activity is also found exclusively in the floor plate [2] (see Figure 1N', N" of Varadarajan et al., 2017 Neuron [3]).

Second, Olig2-Cre is not active in the floor plate. For instance, Varadarajan et al. 2017 Neuron [3] depicted Olig2-Cre active cells and their progeny by crossing Olig2-Cre deleter with Rosa26-gfp reporter mice, and the data show that GFP is not expressed in the floor plate (see Figure 2FF of Varadarajan et al., 2017 Neuron [3]). We have also crossed Olig2-Cre with the reporter allele detecting Cre-mediated recombination and found that Olig2-Cre is not active in the floor plate.

As the reviewer described, Figure 1H of Dessaud et al., 2007 [1] clearly shows that Olig2 is not expressed in the MOST medial cells, which are the floor plate. The ventral most domain of the spinal cord in which Olig2 is initially expressed in this paper [1] is the p3 domain, which gives rise to V3 interneurons. Importantly, V3 interneurons do not express Shh.

Taken together, when the patterning of the ventral neural progenitors occurs, Olig2-Cre is not active in the floor plate cells, which are the only cell types that express Shh at this stage. Thus, the deletion of the Shh gene by Olig2-Cre should affect neither the expression of Shh in the neural tube nor Shh-dependent patterning of the ventral progenitor domains, including pMN domain.

Finally, the phenotype of Shh-cKO;Olig2-Cre mice further supports the notion that the observed phenotype is not caused by Shh deletion in the floor plate. Shh in the floor plate is required for the patterning of the ventral spinal cord, the production of pMN cells, and the generation of motor neurons and ventral interneurons [4]. Thus, if Shh in the floor plate is absent or reduced in Shh-cKO;Olig2-Cre embryos, it would lead to a complete loss or a drastic reduction of Olig2+ pMN progenitors, all types of motor neurons, and V2 interneurons. However, we found no significant difference in the number of Olig2+ pMN progenitors in the analyzed Shh-cKO;Olig2-Cre and control embryos (Figure 3B,C), indicating that Shh level secreted from the floor plate was adequate to drive the pMN patterning and production. In the revised manuscript, we included the quantification data of the number of Olig2+ cells in Shh-cKO;Olig2-Cre and control embryos (Figure 3B,C). We also found that only FoxP1+ LMC motor neurons, but neither Lhx3+Hb9+ MMC, Isl1+Hb9+ HMC motor neurons nor Lhx3+Hb9- V2 interneurons, was reduced in Shh-cKO;Olig2-Cre mice (Figure 3A,C). These data further support the notion that Shh secreted from the floor plate in Shh-cKO;Olig2-Cre mice was adequate to drive the patterning of the ventral spinal cord and the production of pMN cells and that the reduced LMC motor neuron is not a secondary outcome of the defects in neural progenitors.

We hope these address the reviewer’s concern that the observed phenotype can be caused by the deletion of Shh in Olig2+ progenitors.

Next, the reviewer #2 stressed to use a motor neuron-specific driver, such as Hb9-Cre line. We have already purchased Hb9-Cre for this purpose, but Hb9-Cre turned out to be problematic for our experiments, because Hb9 is expressed (therefore Hb9-Cre is active) in the notochord, which secretes Shh required for the neural tube development [5]. As Hb9-Cre deletes Shh in the notochord and eliminates Shh required for the ventral neural tube patterning, Hb9-Cre cannot be used for our experiment. To circumvent this issue, we also used Isl1-Cre, whose Cre expression occurs as motor neurons emerge from the progenitors. We generated Shh-cKO with Isl1-Cre and found that Shh-cKO;Isl1-Cre mice have severe deficits in the limb development as Isl1-Cre inactivates Shh in the developing limb [2, 6, 7]. The deficits of limb development in Shh-cKO;Isl1-Cre compounded our analyses of LMC development, given the observed defects of LMC motor neurons can be caused secondarily by the severe developmental defects of the limbs. In summary, Hb9-Cre is inadequate for deleting Shh only in motor neurons, and Olig2-Cre was the best line that we could use to inactive Shh in motor neurons. Finally, our data indicate that the observed phenotypes in Shh-cKO;Olig2-Cre mice are caused by inactivation of Shh in motor neurons, not by the removal of Shh in the floor plate which does not occur in Shh-cKO;Olig2-Cre mice, as described above.

Other concerns include the lack of sufficient analysis to exclude patterning, proliferation or survival defects in the manipulated chick and mouse embryos (reviewer 1 point 1; reviewer 2 points 5 and 6), and the insufficient quantification of the data (reviewer 1 point 2; reviewer 2 points 4, 8, 14, 17, 19, 22-25). We addressed these concerns as explained below. In particular, as for the concern regarding the chick electroporation of Shh knockdown construct, the electroporation efficiency of the floor plate cells is generally very low. In our electroporation scheme, all electroporated cells are labeled by GFP. To better address reviewer's concern, we analyzed the chick embryos that do not express GFP in the floor plate (therefore no inactivation of Shh), which was confirmed by in situ hybridization analyses of Shh expression. We also confirmed that the patterning and cell proliferation are normal in chick embryos analyzed (Figure 2A,C). These rigorous analyses exclude the possibility that any observed phenotypes are caused by the effect of Shh deletion in the progenitors.

With the successful completion of all the requested revisions, we hope that you and the reviewers find this manuscript exciting and convincing. In particular, we hope to emphasize that this story presents at least two highly innovative findings; a novel function for Shh in postmitotic neurons and its critical downstream effector ARHGAP36 in directing the development of the specific type of MNs (LMC-MNs) in the developing spinal cord. Our results identify a new regulatory axis consisting of AKT-ARHGAP36-PKA, which plays crucial roles in Shh signal transduction for LMC specification.

Reviewer #1:

This paper reports a novel role of Shh, produced by postmitotic motor neurons, in the specification of the lateral motor column (LMC) in the chick and mouse spinal cord. The authors show that AKT stabilises the Rho GAP protein ARHGAP36 in the LMC region, and that ARHGAP36 in turn inhibits the activity of PKA and thereby enhances the production of activating forms of Gli proteins.

The identification of a role for Shh in motor neuron specification and the elucidation of the downstream pathway involving a novel regulator and multiple signaling molecules is of significant interest. The data supports the conclusions of the authors and the work is of excellent quality and well presented. I have a few requests to improve the data, the analysis and discussion to strengthen the conclusions of the paper.

Thank you for acknowledging that our findings provide new insights into the role of Shh signaling in motor neuron fate specification through a novel downstream pathway involving multiple signaling molecules. We would also like to thank the reviewer for many insightful suggestions. We are pleased that we were able to address all issues that the reviewer #1 raised, as described below each of the reviewer's point.

1) Shh acts as a mitogen in many systems but the authors rule out that it is the case here by labelling MN progenitors with antibodies to Olig2 and Sox2 in Figure 2C. However the Shh LOF phenotype in Figure 2A only affects the generation of a small minority of MNs, which could be due to a proliferation defect affecting only a small fraction of progenitors which may not be picked up by the current analysis. To rule this out, the authors should examine proliferating cells more directly, eg with a short pulse of EdU. This should detect a putative proliferation defect with more sensitivity than examining total progenitor numbers.

Following the reviewer’s suggestion, we performed a short pulse of BrdU labeling and Ki67 staining. Our analyses revealed that Shh-cKO mice exhibited no significant change in the proliferation of progenitors as monitored by the four independent markers, such as Olig2, Ki67, Sox2 and BrdU (Figure 3B,C and Figure 3—figure supplement 1B).

2) The ectopic expression of ARHGAP36 in the ventral spinal cord in Figure 5B increases visibly the number of FoxP1+ LMC neurons while Lhx3+ Hb9+ MMCm neurons do not appear affected. However to conclude that ARHGAP36 induces LMC neurons rather than converting MMCm into LMC, requires quantification of the data.

We included the quantification data for FoxP1+ LMC and MMC (Lhx3+Hb9+) neurons (Figure 6B).

3) The conclusion of the experiment in Figure 6A that AKT stabilises ARHGAP36 protein should be strengthened by measuring ARHGAP36 half-life in HEK293T cells in the presence and absence of AKT, and also by showing that manipulating AKT activity does not affect ARHGAP36 transcription.

We included the data showing the half-life of ARHGAP36 protein was prolonged in the presence of AKT when treated with cycloheximide that blocks the protein translation (Figure 7—figure supplement 1).

We also confirmed that AKT inhibitor reduced the protein level of ARHGAP36 by western blotting (Figure 7B, Figure 7—figure supplement 2C) but not the transcription of Arhgap36 by RT-PCR (Figure 7—figure supplement 2D) in mouse embryonic stem cells (mESCs).

4) The regulation of ARHGAP36 expression may need further discussion. Figure 6 shows that AKT controls ARHGAP36 protein stability, but how its transcription is regulated is less clear. Figure 2—figure supplement 1, shows a reduction of its expression in Shh cKO mice. How does Shh regulates its expression and are there other signals involved? Along the same line, the arrow between Shh and P-AKT in Figure 7D may need more explanation.

In Figure 7, to test the effect of AKT activity in the regulation of ARHGAP36 protein stability, we ectopically expressed ARHGAP36 together with different forms of AKT by transfecting HEK293T cells with plasmids that encode ARHGAP36 and AKT genes. As ARHGAP36 is not expressed endogenously in HEK293T cells, in these cells, ARHGAP36 proteins are expressed only from the ARHGAP36-encoding plasmid, in which CMV promoter drives the transcription of ARHGAP36. These experiments show that active AKT increases ARHGAP36 protein levels, suggesting that AKT increases ARHGAP36 proteins likely by stabilizing ARHGAP36 protein, rather than activating the ARHGAP36 promoter transcriptionally, in this experimental setting.

To further test whether endogenous ARHGAP36 transcription is upregulated by AKT in differentiating motor neurons, we used mESC system where ARHGAP36 is induced under motor neuron differentiation condition. In this condition, AKT inhibitor did not affect the mRNA level of ARHGAP36 (see the answer above and Figure 7—figure supplement 2D), suggesting that AKT is not involved in the transcriptional activation of the ARHGAP36 gene.

Then, how does Shh increase ARHGAP36 levels? It is noteworthy that Shh is capable of inducing the activation of phosphoinositide 3-kinase (PI3-kinase)-dependent AKT phosphorylation in cell lines (LIGHT cells and HUBEC cells) [8, 9]. We proposed that Shh expressed in the motor neurons triggers AKT activation, which in turn stabilizes the protein level of ARHGAP36 in motor neurons. Our model predicts that the ARHGAP36 protein level would be decreased in Shh-cKO mice.

Reviewer #2:

The manuscript by Nam and cols, entitled "Critical roles of ARHGAP36 as a signal transduction mediator of Shh pathway in lateral motor columnar specification" proposes a novel role for Shh signalling in spinal cord development.

A putative role for Shh activity as a signal instructing the selection of specific MNs subpopulations is novel and attractive. It is based in the observation of Shh expression in postmitotic MNs, which is not fully novel, but has not been well characterized previously.

Authors also find that the Rho GAP family member Arhgap36 is expressed in postmitotic MNs, and propose that it might act as a Shh effector in MN subpopulations selection. In different cell contexts, Arhgap36 has previously been shown to inhibit PKA activity, hence regulating Shh signalling. However, it would be nice to fully define its in vivo relevance, in the context of selection of specific MNs subpopulations. In summary, the manuscript contains some novel and interesting data, however it lacks definitive demonstration of some of the conclusions raised by the authors.

Hence, I believe that, in the present format, the manuscript has important gaps that need to be fulfilled.

We appreciate the acknowledgment that our findings provide a novel role for Shh signaling and its effector ARHGAP36 in generation of specific MNs subpopulation, and we would like to thank the reviewer for many excellent suggestions. As described below, now we provided all quantification, control, and other data, which the reviewer asked to make the manuscript more complete.

Specific points:

In order to propose a new role for Shh signalling as an instructive signal the selection of specific MNs subpopulations, I believe that authors should improve the characterization of the MN subpopulations expressing Shh.

1) In the chick embryo spinal cord, Shh expression has previously been reported in postmitotic motor neurons of the LMC (at stage HH15), extending later to the MMC (Stage HH35) (Figure 1 in Oppenheim et al., 1999). However, Nam and cols, in this manuscript shows Shh expression restricted to LMC neurons at the brachial level. The selected section showed in Figure 1A shows Shh expression restricted to the mLMC (and excluded from lLMC), as well as excluded from MMC. Could authors clarify these contradictory issues?

As the reviewer suggested, we re-examined the expression of Shh more thoroughly with well-defined markers for motor columns in chick embryos (stage HH29) (Figure 1B). Our analyses show that Shh is expressed in LMCm (Isl1+/FoxP1+) but not LMCl (Isl1-/FoxP1+) at brachial level, and interestingly, it is expressed in LMCl (Isl1-/FoxP1+) at lumbar level. Shh expression is clearly excluded from MMC (Lhx3+ MNs) at all axial levels, as demonstrated by converging analyses of Lhx3 and Shh (Figure 1B). Overall, our Shh expression analysis results are highly similar to the data in the referenced paper (Oppenheim et al., 1999) [10], despite the minor differences likely due to detection sensitivity variations of Shh antisense probes.

Oppenheim et al. showed nice expression of Shh in chick embryos at different developmental stages. At stage HH15, Shh is expressed in notochord and floor plate but not in postmitotic motor neurons of the LMC (Figure 1A in Oppenheim et al. [10]), which is similar to what we have observed. There is a low level detection of Shh in the ventral horn at stage HH23 (Figure 1C in Oppenheim et al. [10]), which may result from higher level sensitivity of Shh antisense probe. Oppenheim et al. also described that at stage HH35, Shh is expressed in two distinct motoneuron (MN) populations (medial and lateral) and the referee 2 may be referring this medial MNs as MMC. However, this Shh-expressing medial MN population is likely to be LMCm, judging by the cell body location. It is hard to conclude the identity of motor columns without co-expression analyses with motor column markers. Thus, we performed these needed co-expression analyses in Figure 1B.

2) Panel B of this figure (expression in mouse embryos) do not show this restricted expression to mLMC. Are these specie specific differences? Or developmental stage differences?

The expression level of Shh in mouse motor neurons is much lower than that in chick motor neurons. As we examined the comparable developmental stages in mouse and chick embryos, we do not think that the difference in expression levels reflects developmental stage differences. We have observed that overall in situ hybridization analyses tend to work more effectively in chick embryos than in mouse embryos not only for Shh but also for many other genes. This sensitivity differences in in situ hybridization outcomes may reflect species differences, but we are yet to understand what factors exactly contribute to this observation. Nonetheless, our analyses in both mouse and chick embryos demonstrate the expression of Shh in postmitotic motor neurons.

3) A detailed temporo-spatial expression of Shh in postmitotic MNs should be provided. These should be easy to clarify combining with the available and well defined markers for each MN column (see for example Adams et al., 2015)

Following the reviewer’s advice, we re-examined the expression of Shh more thoroughly with well-defined markers for motor columns in chick embryos (stage HH29) (Figure 1B). Our analyses show that Shh is expressed in LMCm (Isl1+/FoxP1+) but not LMCl (Isl1-/FoxP1+) at brachial level, and interestingly, it is expressed in LMCl (Isl1-/FoxP1+) at lumbar level. Shh expression is clearly excluded from MMC (Lhx3+ MNs) at all axial levels, as demonstrated by converging analyses of Lhx3 and Shh (Figure 1B).

In order to analyse the role of Shh in MN subtype specification; loss-of-function experiments in chick embryo NT were performed by electroporation at HH stage 13 chick embryos (Materials and methods), at which patterning of the NT is not definitely established (see for example Cayuso et al., 2006).

4) Controls showing quantification of Shh- loss are required, particularly since defects appeared to be mild, despite the length of the experiments (96 hours post-electroporation at HH13). For example, Gli-bs-luc experiments to show the efficiency of the Shh-LOF vector.

In the revised manuscript, we quantified the intensity of Shh ISH signal and confirmed the significant reduction of Shh expression in motor neurons (Figure 2C). Of note, in our electroporation condition, the electroporation efficiency of the floor plate cells is generally very low, likely due to the depth and location of the electrodes. Thus, we expected that Shh knockdown would be mostly effective in motor neurons but not in the floor plate, allowing us to focus on analyzing the effect of Shh-LOF on motor columnar specification without significant changes in progenitor proliferation or early patterning of the neural tube. Our analyses including careful quantification indeed revealed that there’s no significant change in proliferation (BrdU+ cells) or ventral neural patterning (Olig2+, Nkx2.2+ cells in Figure 2A,C). We also found a significant reduction of LMCl motor neurons (Figure 2B,C).

5) Moreover, the role of Shh signalling in proliferation and survival of neural progenitors are also well stablished, hence controls ruling out these defects are required.

In the revised manuscript, we provided the data of BrdU incorporation for proliferation and cleaved caspase 3 staining for apoptotic cells and these results show no significant change in proliferation and survival of neural progenitors (Figure 2A,C).

6) Controls showing markers for progenitors should be included in order to rule out patterning defects.

Now we provided the data that ventral progenitor cells are established properly by staining with Olig2 and Nkx2.2, markers of p2 and p3 domain, respectively, whose expression is dependent on Shh levels in the floor plate (Figure 2A,C).

7) I strongly believe that, in order to analyse a specific role for Shh signalling in postmitotic MN subtype generation, experiments should have been done using a MN specific driver (for example the well characterized HB9) to avoid patterning and proliferation defects.

We have already attempted to use Hb9-Cre for this purpose, but Hb9-Cre turned out to be problematic for our experiments, because Hb9 is expressed (therefore Hb9-Cre is active) in the notochord, which secretes Shh required for the neural tube development [5]. As Hb9-Cre deletes Shh in the notochord and eliminates Shh required for the ventral neural tube patterning, deleting Shh with Hb9-Cre is expected to cause severe patterning and proliferation defects in the neural tube. Thus, Hb9-Cre cannot be used for our experiment. To circumvent this issue, we also used Isl1-Cre, whose Cre expression occurs as motor neurons emerge from the progenitors. We generated Shh-cKO with Isl1-Cre and found that Shh-cKO;Isl1-Cre mice have severe deficits in the limb development as Isl1-Cre inactivates Shh in the developing limb [2, 6, 7]. The deficits of limb development in Shh-cKO;Isl1-Cre compounded our analyses of LMC development, given the observed defects of LMC motor neurons can be caused secondarily by the severe developmental defects of the limbs. In summary, Hb9-Cre is inadequate for deleting Shh only in motor neurons, and Olig2-Cre was the best line that we could use to inactive Shh in motor neurons.

To complement Shh-cKO mouse analysis, we also analyzed chick embryos in which Shh is knocked down in motor neurons, but not in the floor plate (Figure 2A). In our chick electroporation experiments, the electroporation efficiency of the floor plate cells is generally very low and all electroporated cells are labeled by GFP. We analyzed the chick embryos that do not express GFP in the floor plate (therefore no inactivation of Shh), which was further confirmed by in situ hybridization analyses of Shh expression. Our data show that the cell proliferation is normal in chick embryos analyzed. We also examined ventral progenitor markers, such as Nkx2.2 and Olig2, whose expression is dependent on Shh levels in the floor plate to further probe the presence of Shh in the floor plate. These rigorous analyses exclude the possibility that any observed phenotypes are caused by the effect of Shh deletion in the progenitors.

8) Quantitative data indicating the total number of MNs, versus sub-type specific MNs should be provided. It is important to understand whether LMNs have switch their fate, of whether LMNs are not being generated.

Now we provided the quantification data for total number of MNs and sub-type specific MNs in Figure 2C. Knockdown of Shh resulted in reduction of LMCl motor neurons without affecting other motor column subtypes and consequently, the total number of MNs was reduced compared to the uninjected side (Figure 2C). This result indicates that LMC motor neurons are not properly generated or maintained rather than switching to other MN subtypes in the absence of Shh.

9) Quantitative data in Figure 1A panel D shows FoxP1+ cells, are these islet1+? Or islet1-? Same for Hb9, these analyses will discriminate between lLMB vs mLMC.

Quantitative data in Figure 1A panel E shows Lhx3+ cells, same for the combination with additional marker to restrict the phenotype to specific MN subtype

Now we provided the quantification data for each motor column such as LMCl (Hb9+/FoxP1+ or Isl1-/FoxP1+), LMCm (Hb9-/FoxP1+ or Isl1+/FoxP1+), HMC (Hb9+/Isl1+) and MMC (Lhx3+/Hb9+) in Figure 2C.

10) A schematic drawing of the MN columns, including markers and phenotype would be of great help.

We included the schematic drawing of the MN columns with markers and phenotype in Figure 8D.

11) The same holds for the analysis of the Shh-Olig2C mutant phenotype. These experiments should have been done using a MN specific driver (for example the well characterized HB9CRE line, available at Jackson) to avoid patterning and proliferation defects.

We chose Olig2-Cre driver for our conditional knockout experiments because the previous work has shown that Olig2-Cre does not delete the gene in the floor plate cells [1, 11]. Shh in the floor plate is required for the patterning of the ventral spinal cord, the production of pMN cells, and the generation of motor neurons and ventral interneurons [4]. Thus, if Shh in the floor plate is absent or reduced in Shh-cKO;Olig2-Cre embryos, we expect that it would lead to a complete loss or a drastic reduction of Olig2+ pMN progenitors, all types of motor neurons, and V2 interneurons. However, there was no significant difference in the number of Olig2+ pMN progenitors in the analyzed Shh-cKO;Olig2-Cre and control embryos, suggesting that Shh level secreted from the floor plate was adequate to drive the pMN patterning and production. We included the quantification data of the number of Olig2+ cells in Shh-cKO;Olig2-Cre and control embryos (Figure 3C). We also found that only FoxP1+ LMC motor neurons including both LMCm and LMCl, but neither MMC (Lhx3+Hb9+) motor neurons nor HMC (Isl1+Hb9+) motor neurons, was reduced in Shh-cKO;Olig2-Cre mice (Figure 3C), further indicating that Shh secreted from the floor plate was adequate to drive the patterning of the ventral spinal cord and the production of pMN cells.

Also please see our response to the question 7, regarding why Hb9-Cre can’t be used for Shh-cKO analyses.

12) Please indicate the total number of MNs, versus sub-type specific MNs. Please indicate whether FoxP1+ cells are islet1 and Hb9 +/-.

We included the quantification data for each motor column and total number of MNs in Figure 3C. The number of FoxP1+ LMC motor neurons including both LMCm and LMCl was reduced, but neither MMC (Lhx3+Hb9+) motor neurons nor Isl1+Hb9+ HMC motor neurons, was changed in Shh-cKO;Olig2-Cre mice (Figure 3C), which results in reduction of total number of MNs in Shh-cKO compared to control littermates.

To identify the candidate effector genes, authors searched for target genes of the Isl1-Lhx3 transcriptional complex, well characterized previously by this group. What is the link between this searching strategy and Shh signalling, it is not clear to me, but they identify the Rho GAP family member Arhgap36, which is known to activate Gli transcriptional responses through the inhibition of PKA activities.

While we were analyzing the function of Shh in the columnar specification, ARHGAP36 was reported to activate Gli transcriptional responses through the inhibition of PKA activities. We have examined the expression pattern of putative target genes of the Isl1-Lhx3 complex in the developing spinal cord and found ARHGAP36 as one of the potential MN genes as its expression is very high and specific in MNs.

13) It would be nice to see, among the many genes regulated by the Isl1-Lhx3 transcriptional complex, whether there might be a cluster of HH-signalling components?

We do not see a cluster of HH-signaling components regulated by the Isl1-Lhx3 complex other than ARHGAP36.

14) Authors identify a HxRE of the Arhgap36 gene, and test the in vivo activity by electroporation experiments in chick embryo NT. Please provide quantitative data indicating number of embryos analysed. Provide quantitative data indicating HB9 ectopic cells induced. Please provide electroporation time course, as well as negative control experiments.

We provided quantitative data for the number of embryos injected, electroporation time course in the figure legend. It is very well established that electroporation of Isl1+Lhx3 induces ectopic Hb9+ cells in the dorsal spinal cord [12-14], and we routinely do this experiment in the lab. From this set of experiment, we would like to see the induction of GFP reporter driven by Arghap36-enhancer activated by Isl1+Lhx3 in the dorsal spinal cord. Indeed, we could detect very nice GFP expression as well as ectopic Hb9+ cells in the dorsal spinal cord by Isl1+Lhx3 expression. We included the counting data of the ectopic Hb9+ cells in the dorsal spinal cord (Figure 4F) and negative control experiment with TATA-GFP vector construct that has no HxRE and was not activated by Isl1+Lhx3 (Figure 4E).

15) I believe that the data provided here demonstrate that the Isl1-Lhx3 complex is sufficient to induce Arhgap36 expression, whether this induction is direct is not fully demonstrated.

ChIP assay using mouse spinal cord extracts showed that Isl1 and Lhx3 are strongly recruited to the enhancer region of Arhgap36 but not to the negative control region of Untr6 (Figure 4C), indicating that the induction of Arhgap36 by Isl1-Lhx3 complex is direct indeed.

Next authors analyse Arhgap36 expression in postmitotic neurons. This is an important issue, since Shh might be acting either in a paracrine manner, or in a cell autonomous manner, to instruct MNs subtype identity.

16) Please provide a better characterization of Arhgap36 in MN subtypes (co-localization with MN subtype markers) and co-localization (or not) with Shh, with cellular resolution.

We now provided the detailed expression data of Arhgap36 combining with defined markers for each MN column such as Arhgap36/Isl1/FoxP1, Arhgap36/Isl1/Hb9, and Arhgap36/Lhx3/Hb9 in mouse embryos (Figure 5C). Arhgap36 is most highly expressed in LMCl (Isl1-/FoxP1+) region, some in MMC-rhomboideus (Hb9+/Lhx3low) and a very little in the most medial part of MMC but not in LMCm (Isl1+/FoxP1+) at cervical level. At thoracic level, Arhgap36 is also expressed in PGC (FoxP1+/Isl1+) and HMC (Isl1+/Hb9+) MNs, although the expression level is relatively lower. At lumbar level, Arhgap36 is relatively highly enriched in LMCl (Isl1-/FoxP1+) and show very low level in the most medial part of MMC. To examine the co-localization of Arhgap36 with Shh, we performed in situ hybridization of Shh and IHC of Arhgap36 in mouse E12.5 spinal cord at cervical level. Shh is co-localized with Arhgap36 mostly in LMCl region (Figure 5D).

In summary, mouse Shh is mainly expressed in LMCl of the mouse spinal cord that is co-localized with Arhgap36, while chick Shh is expressed in LMCm at cervical level and LMCl at lumbar level of chick spinal cord where chick Arhgap36 is broadly expressed within the spinal cord. Although there is a discrepancy in the expression pattern of Shh in mouse and chick spinal cord, Arhgap36 seems to be coexpressed with Shh in both species and the regulatory axis of Shh-ARHGAP36 can still function in both mouse and chick spinal cord.

17) Please provide Arhgap36 expression data in chick embryos MNs. It would be interesting to see the conservation of it expression in MNs.

In situ hybridization of chick Arhgap36 showed rather broad but specific and high expression within the spinal cord compared to other tissues in the chick embryo (Figure 7—figure supplement 3).

Next authors test whether the Shh-pathway is activated upon over-expression of Arhgap36. Experiments in chick embryo NT were performed by electroporation of ARHGAP36 at HH stage 13 chick embryos (Materials and methods). These gain-of-function experiments produce NT overgrowth, corresponding to the over-activation of Shh signalling, in addition to ventralization of the entire NT. Quantitative data indicating number of embryos analysed is missing, electroporation time course, as well as quantification of marker-specific ectopic cells should be provided.

We provided quantitative data for the number of embryos injected, electroporation time course in the figure legend and included the quantification data for FoxP1+ LMC and MMC (Lhx3+/Hb9+) neurons (Figure 6B).

18) I believe that these experiments are controls of the capacity of ARHGAP36 in activating Shh-mediated responses in vivo. Since this has been previously documented in other systems, these experiments can go as supplementary information.

We moved these data to supplementary information (Figure 6—figure supplement 1).

19) Authors claim that over-expression of ARHGAP36 in ventral NT, harvested at 4 dpe the number of FoxP1+ LMC neurons increased drastically, suggest that ARHGAP36 is sufficient to direct LMC fate-determination. Again here, I believe that these experiments should have been done using a MN HB9 specific driver to avoid patterning and proliferation defects. With the present experimental design, it is very difficult to assign a direct role for ARHGAP36 in postmitotic MN subpopulations selection, other that the role in MN progenitors

We used Gal4/UAS system to drive the motor neuron specific expression of Arhgap36. We generated Hb9-Gal4 construct where Gal4 DNA binding transactivation domain is expressed driven by motor neuron specific Hb9 promoter and UAS-Arhgap36 construct where Arhgap36 is expressed through the binding of Gal4 to the UAS response element. Using this system, we could direct the expression of Arhgap36 into postmitotic MNs and this experiment also resulted in the specific increase of FoxP1+ LMC neurons but had no effect on MMC (Lhx3+/Hb9+) neurons (Figure 6).

Again in these experiments, quantitative data indicating number of embryos analysed is required, electroporation time course is missing. Quantification of marker-specific ectopic cells should be provided.

We now provided the quantitative data of number of embryos in the figure legend and quantification data for marker+ cell numbers in Figure 6.

Next, authors show that PKA activity is inhibited by ARHGAP36. However, this observation is not novel, since Eccles et al. (2016) showed that ARHGAP36 combines two distinct inhibitory mechanisms to antagonise PKA signalling; it blocks PKA catalytic activity and it targets PKAc for ubiquitin-mediated degradation, resulting in activation of the Shh pathway.

20) I don't see the point in analysing other targets but PKA in the context of MN specification, hence I believe that panels C,D,E from Figure 5 are not relevant in this context.

We moved these data to supplementary figures (Figure 6—figure supplement 1).

21) Again, in the context of Shh/ ARHGAP36 signalling in MN subtype selection, I do not see the relevance of the AKT experiments shown if Figure 6. I believe that should be removed.

Moreover, Zhang et al. (2019) find that Patched1 interacts with ArhGAP36 to the centrosome and stabilizes the PKA negative regulator (this paper should be quoted in the manuscript).

Zhang et al., Patched1-ArhGAP36-PKA-Inversin axis determines the ciliary translocation of Smoothened for Sonic Hedgehog pathway activation. Proc Natl Acad Sci U S A. 2019 Jan 15;116(3):874-879

We cited this paper in the introduction. As Shh pathway plays critical roles in embryonic development, tissue homeostasis and tumorigenesis, it is important to define the regulatory network modulating multiple components involved in the pathway. We found that AKT stabilizes the Arhgpa36 protein level and further potentiates the Shh activity in MN subtype generation, which is quite novel and this regulatory axis can be broadly implicated in various Shh-dependent conditions.

22) Authors show reduced expression of ARHGAP36 in Shh-KO mice (Figure S4), again without any quantitative data.

We provided the quantitative data (Figure 3—figure supplement 1).

Finally, authors generate an ARHGAP36 mutant mice. Authors claim that at early developmental stages, there was no defect in MN generation. Quantitative data on this control observation should be provided.

We included the data showing that there’s no defect in proliferation (BrdU), ventral neural patterning (Nkx2.2, Olig2) and overall MN generation (Hb9) at E11.5 and provided quantification data in Figure 8—figure supplement 1.

23) Analysis of the phenotype generated by ARHGAP36_CRISPR mutant in MNs at the brachial and the thoracic levels of the developing spinal cord, again require quantitative data indicating that the total number of MNs, have not changed at thoracic levels.

We included the quantification data of HMC (Hb9+/Isl1+), MMC (Lhx3+/Hb9+), PGC (nNOS+) and total number of MNs at thoracic levels (Figure 8C).

24) Quantitative data indicating that the total number of MMC and the total number of V2 interneurons, have not changed at brachial levels.

We included the quantification data of MMC (Lhx3+/Hb9+) and V2-INs (Chx10+/Lhx3+) at brachial levels (Figure 8C).

25) Quantitative data indicating the total MN numbers versus sub-type specific MNs should be provided. It is important to understand whether, in the absence of ARHGAP36, LMNs have switch their fate, of whether LMNs are not being generated.

We included the quantification data of LMCm (Isl1+/FoxP1+), LMCl (Hb9+/FoxP1+, Lhx1+/FoxP1+), HMC (Hb9+/Isl1+), MMC (Lhx3+/Hb9+) at the brachial and thoracic levels (Figure 8C). In Arhgap36 KO mice, FoxP1+ LMC including LMCm and LMCl neurons reduced, while there is no compensatory increase in non-LMC type motor neurons that we examined, and then as a result this leads to the reduction of total MNs (Figure 8C). The increase of active Caspase3+ cells in the motor neuron area of Arhgap36 KO mice (Figure 8—figure supplement 1) suggests that prospective LMC neurons undergo apoptotic cell death in the absence of Arhgap36. It remains unclear, however, if cell death is a direct outcome of loss of Arhgap36 in promoting cell survival or an indirect outcome of the failure of acquiring or maintaining the correct LMC identity. Importantly, previous studies show that postmitotic motor neuron fate specification and their survival are closely linked and it is difficult to distinguish the phenotypes between the fate specification and survival of postmitotic motor neurons. After cell cycle exit, the 'generic' motor neurons diversify into distinct motor neuron subtypes, including LMC. Retinoic acid (RA) signaling plays a role in the specification and maintenance of forelimb LMC identity [15-21]. The loss of RA from motor neurons leads to a reduced number of LMC neurons without a significant change in the number of MMC neurons [21], similar to our Arhgap36 KO embryos. The ectopic activation of RA signaling in thoracic motor neurons impairs non-LMC type motor neuron differentiation and ultimately triggers apoptotic cell death [19]. One potential mechanism that links the fate specification and cell survival is that motor neurons which did not acquire the correct cell identity fail to receive target-derived neurotrophic signals.

References:

1) Dessaud, E., et al., Interpretation of the sonic hedgehog morphogen gradient by a temporal adaptation mechanism. Nature, 2007. 450(7170): p. 717-20.

2) Harfe, B.D., et al., Evidence for an expansion-based temporal Shh gradient in specifying vertebrate digit identities. Cell, 2004. 118(4): p. 517-28.

3) Varadarajan, S.G., et al., Netrin1 Produced by Neural Progenitors, Not Floor Plate Cells, Is Required for Axon Guidance in the Spinal Cord. Neuron, 2017. 94(4): p. 790-799.e3.

4) Jessell, T.M., Neuronal specification in the spinal cord: inductive signals and transcriptional codes. Nat Rev Genet, 2000. 1(1): p. 20-9.

5) Harrison, K.A., et al., Pancreas dorsal lobe agenesis and abnormal islets of Langerhans in Hlxb9-deficient mice. Nat Genet, 1999. 23(1): p. 71-5.

6) Yang, L., et al., Isl1Cre reveals a common Bmp pathway in heart and limb development. Development, 2006. 133(8): p. 1575-85.

7) Itou, J., et al., Islet1 regulates establishment of the posterior hindlimb field upstream of the Hand2-Shh morphoregulatory gene network in mouse embryos. Development, 2012. 139(9): p. 1620-9.

8) Kanda, S., et al., Sonic hedgehog induces capillary morphogenesis by endothelial cells through phosphoinositide 3-kinase. J Biol Chem, 2003. 278(10): p. 8244-9.

9) Riobo, N.A., et al., Phosphoinositide 3-kinase and Akt are essential for Sonic Hedgehog signaling. Proc Natl Acad Sci U S A, 2006. 103(12): p. 4505-10.

10) Oppenheim, R.W., et al., Modulation of early but not later stages of programmed cell death in embryonic avian spinal cord by sonic hedgehog. Mol Cell Neurosci, 1999. 13(5): p. 348-61.

11) Sagner, A., et al., Olig2 and Hes regulatory dynamics during motor neuron differentiation revealed by single cell transcriptomics. PLoS Biol, 2018. 16(2): p. e2003127.

12) Thaler, J.P., et al., LIM factor Lhx3 contributes to the specification of motor neuron and interneuron identity through cell-type-specific protein-protein interactions. Cell, 2002. 110(2): p. 237-49.

13) Lee, S., et al., STAT3 promotes motor neuron differentiation by collaborating with motor neuron-specific LIM complex. Proc Natl Acad Sci U S A, 2013. 110(28): p. 11445-50.

14) Cho, H.H., et al., Isl1 directly controls a cholinergic neuronal identity in the developing forebrain and spinal cord by forming cell type-specific complexes. PLoS Genet, 2014. 10(4): p. e1004280.

15) Sockanathan, S. and T.M. Jessell, Motor neuron-derived retinoid signaling specifies the subtype identity of spinal motor neurons. Cell, 1998. 94(4): p. 503-14.

16) Solomin, L., et al., Retinoid-X receptor signalling in the developing spinal cord. Nature, 1998. 395(6700): p. 398-402.

17) Diez del Corral, R., et al., Opposing FGF and retinoid pathways control ventral neural pattern, neuronal differentiation, and segmentation during body axis extension. Neuron, 2003. 40(1): p. 65-79.

18) Novitch, B.G., et al., A requirement for retinoic acid-mediated transcriptional activation in ventral neural patterning and motor neuron specification. Neuron, 2003. 40(1): p. 81-95.

19) Sockanathan, S., T. Perlmann, and T.M. Jessell, Retinoid receptor signaling in postmitotic motor neurons regulates rostrocaudal positional identity and axonal projection pattern. Neuron, 2003. 40(1): p. 97-111.

20) Vermot, J., et al., Retinaldehyde dehydrogenase 2 and Hoxc8 are required in the murine brachial spinal cord for the specification of Lim1+ motoneurons and the correct distribution of Islet1+ motoneurons. Development, 2005. 132(7): p. 1611-21.

21) Ji, S.J., et al., Mesodermal and neuronal retinoids regulate the induction and maintenance of limb innervating spinal motor neurons. Dev Biol, 2006. 297(1): p. 249-61.

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Data Citations

    1. Lee S, Shen R, Cho HH, Kwon RJ, Seo SY, Lee JW, Lee SK. 2013. STAT3 promotes motor neuron differentiation by collaborating with motor neuron-specific LIM complex. NCBI Gene Expression Omnibus. GSE50993 [DOI] [PMC free article] [PubMed]
    2. Mazzoni EO, Mahony S, Closser M, Morrison CA, Nedelec S, Williams DJ, An D, Gifford DK, Wichterle H. 2013. Synergistic binding of transcription factors to cell-specific enhancers programs motor neuron identity. NCBI Gene Expression Omnibus. GSE31456 [DOI] [PMC free article] [PubMed]

    Supplementary Materials

    Figure 2—source data 1. Source data for Figure 2C.
    DOI: 10.7554/eLife.46683.004
    Figure 3—source data 1. Source data for Figure 3C.
    DOI: 10.7554/eLife.46683.008
    Figure 3—figure supplement 1—source data 1. Source data for Figure 3—figure supplement 1.
    DOI: 10.7554/eLife.46683.007
    Figure 4—source data 1. Source data for Figure 4C.
    DOI: 10.7554/eLife.46683.010
    Figure 4—source data 2. Source data for Figure 4D.
    DOI: 10.7554/eLife.46683.011
    Figure 4—source data 3. Source data for Figure 4F.
    DOI: 10.7554/eLife.46683.012
    Figure 5—source data 1. Source data for Figure 5A.
    DOI: 10.7554/eLife.46683.014
    Figure 6—source data 1. Source data for Figure 6B.
    DOI: 10.7554/eLife.46683.019
    Figure 6—figure supplement 1—source data 1. Source data for Figure 6—figure supplement 1D.
    DOI: 10.7554/eLife.46683.017
    Figure 7—source data 1. Source data for Figure 7H and 7I.
    DOI: 10.7554/eLife.46683.025
    Figure 7—figure supplement 1—source data 1. Source data for Figure 7—figure supplement 1.
    DOI: 10.7554/eLife.46683.022
    Figure 8—source data 1. Source data for Figure 8C.
    DOI: 10.7554/eLife.46683.031
    Figure 8—figure supplement 1—source data 1. Source data for Figure 8—figure supplement 1C.
    DOI: 10.7554/eLife.46683.028
    Figure 8—figure supplement 2—source data 1. Source data for Figure 8—figure supplement 2C.
    DOI: 10.7554/eLife.46683.030
    Transparent reporting form
    DOI: 10.7554/eLife.46683.032

    Data Availability Statement

    All data generated or analysed during this study are included in the manuscript and supporting files.

    The following previously published datasets were used:

    Lee S, Shen R, Cho HH, Kwon RJ, Seo SY, Lee JW, Lee SK. 2013. STAT3 promotes motor neuron differentiation by collaborating with motor neuron-specific LIM complex. NCBI Gene Expression Omnibus. GSE50993

    Mazzoni EO, Mahony S, Closser M, Morrison CA, Nedelec S, Williams DJ, An D, Gifford DK, Wichterle H. 2013. Synergistic binding of transcription factors to cell-specific enhancers programs motor neuron identity. NCBI Gene Expression Omnibus. GSE31456


    Articles from eLife are provided here courtesy of eLife Sciences Publications, Ltd

    RESOURCES