Home Back

Identification and characterization of intermediate states in mammalian neural crest cell epithelial to mesenchymal transition and delamination

elifesciences.org 2 days ago

eLife assessment

Identification of intermediate stages during mouse cranial NCC EMT and delamination

Mouse cranial NCC undergo EMT and delamination in S or G2/M phase cell cycle

Cell cycle regulation is critical for mouse cranial NCC EMT and delamination

Spatiotemporal localization of intermediate stage NCC in vivo

EMT intermediate stage marker gene Dlc1 regulates NCC delamination

Signal amplification by exchange reaction for multiplexed fluorescent in situ hybridization (SABER-FISH) of epithelial to mesenchymal transition (EMT) intermediate stage markers pinpoints the location of EMT intermediate neural crest cell (NCC) within the dorsal most region of the neural fold. (A) Dotplot showing the expression of selected EMT intermediate NCC markers in early migratory NCC subclusters (resolution 2.0). (B) SABER-FISH staining of premigratory, EMT intermediate stage, and migratory NCC marker genes on the same section. Higher magnification insets of the left side neural fold (box) showing that Wnt1 is expressed in the neuroepithelium and Sox10 is expressed in migratory NCC populating the underlying mesenchyme. Dlc1, Sp5, and Pak3 are expressed in the dorsolateral most region of the neuroepithelium. (C) 2D map showing the number of transcripts per cell, calculated from the SABER-FISH staining. To evaluate the expression of each gene within and across tissues, a polyline kymograph was generated along the track indicated by the arrows at a width of 100 pixels. The polyline kymograph can be seen to the right of each neural fold map it depicts. At the beginning of the track, Wnt1 expression is highest, demarcating the dorsal lateral domain of the neuroepithelium. Toward the middle of the track, at the location of the most dorsolateral region of the neuroepithelium, Wnt1 is expressed along with the intermediate stage markers Dlc1, Sp5, and Pak3. As the track progresses to just outside of the neuroepithelium, Sox10 expression appears and increases as the track continues through the migratory NCC population. Scale bars: embryo sections 100 µm; section insets 50 µm.

EMT is a cellular process that converts epithelial cells to mesenchymal cells. EMT is essential for normal development and is a key driver of disease pathogenesis, particularly cancer metastasis. Although classically considered to be a binary event, studies of EMT in cancer identified multiple intermediate states within the EMT spectrum, a phenomenon termed EMP. However, it remained to be determined whether developmental EMT is also a developmental EMP process. Our goal therefore was to determine whether intermediate stages of NCC development during EMT could be transcriptionally and spatially defined, and then test whether transitional stage-associated genes are functionally required for NCC EMT and delamination.

Through scRNA-seq analysis of mouse cranial tissues at E8.5, which coincided with the onset of NCC EMT and delamination, we identified two populations of NCC, whose gene expression profiles or signatures were representatives of intermediate stages between premigratory and migratory NCC. Furthermore, we determined that the two intermediate populations could be defined by their distinct transcriptional states which were consistent with being in either S phase or G2/M phase of the cell cycle. Although it is possible that the two intermediate populations represent a single population that is temporarily bifurcated due to cell cycle asynchrony, the pseudotime trajectory analysis suggests that these intermediate stage cranial NCC populations can undergo EMT and delaminate in either S phase or G2/M phase, simultaneously, and independently of each other. This is further supported by the observation that S phase cell cycle inhibition failed to completely eliminate NCC delamination since NCC delaminating in G2/M phase were observed. The two intermediate populations later merge into a single or common pool of early migratory NCC suggesting they do not have distinct fates following migration that can be tied to their cell cycle status as the time of delamination. This is not indicative of a common progenitor of both ectomesenchyme and neuro/glial/pigment derivatives, but rather, is consistent with the known plasticity and potency of migrating NCC (Sandell and Trainor, 2006; Trainor and Krumlauf, 2000a; Trainor and Krumlauf, 2000b; Trainor and Krumlauf, 2001).

These results correlate with observations that cell cycle status is also a critical factor regulating NCC delamination in avian and zebrafish embryos. For example, BrdU incorporation, which demarcates proliferating cells in S phase of the cell cycle, was previously used to evaluate the cell cycle status of emigrating trunk NCC, dorsal midline neuroepithelial cells, and surrounding cells at the segmental plate, epithelial somite and dissociating somite axial levels in chicken embryos (Burstyn-Cohen and Kalcheim, 2002). Most emigrating trunk NCC (about 80%) at the epithelial somite and dissociating somite axial levels were in S phase of the cell cycle, while less than 50% of dorsal neuroepithelial cells were BrdU+. Similarly, slice culture of the trunk of chicken embryos also revealed that most premigratory NCC contained basally positioned nuclei indicative of S phase. Interestingly, however, a small proportion of premigratory NCC presented as round mitotic cells, whose daughter cells later became migratory (Ahlstrom and Erickson, 2009). Furthermore, in vivo time-lapse imaging of chicken embryos revealed that half of the delaminating trunk NCC that were tracked displayed signs of cell division (McKinney et al., 2013). However, in most cases, only one progeny of a mitotic premigratory NCC was observed to exit the neural tube and become a migratory NCC. Interestingly, time-lapse imaging of zebrafish embryos also revealed active cell division in dorsal neuroepithelial cells prior to NCC delamination and EMT. The daughter cells of those divisions then translocate into the basal side of the neuroepithelium, where EMT subsequently occurs (Berndt et al., 2008). Whether delaminating NCC undergo proliferation and cell division in zebrafish embryos remains to be investigated, however, active cell division in the dorsal neuroepithelium is a shared feature of NCC delamination in avian and aquatic species. Even though our findings primarily illustrate the association of distinct cell cycle phases with intermediate stage NCC during EMT and delamination, this work has emphasized the importance of further examining the cell division and cell cycle activities of delaminating NCC in mouse embryos as critical contributors to normal development and the pathogenesis of neurocristopathies.

In contrast to delaminating trunk NCC, only around 30% of delaminating cranial NCC in chicken embryos were found to be in S phase (Théveneau et al., 2007) illustrating considerable differences between cranial and trunk NCC and in the correlation between cell cycle phase and delamination. Blocking G1/S transition in the trunk of chicken embryos via in ovo electroporation, or via small molecule inhibitors in explanted neural primordia, prevents the onset of NCC delamination (Burstyn-Cohen and Kalcheim, 2002). Furthermore, BMP and Wnt canonical signaling regulates the G1/S transition and promotes trunk NCC delamination (Burstyn-Cohen et al., 2004). Although it remains to be determined which signaling pathways regulate cell cycle phase progression in the intermediate or transitional populations of cranial NCC in mouse embryos, our results illustrate an evolutionarily conserved mechanistic role for cell cycle progression in NCC delamination in vertebrate embryos.

The intermediate populations of cranial NCC in mouse embryos exhibited transcriptional profiles that were characterized by the downregulation of tight junction and polarity genes. This is consistent with the breakdown of intercellular tight junctions and degradation of apicobasal polarity, which are hallmarks of EMT (Zhao and Trainor, 2023). Further interrogation of genes that were differentially expressed in the intermediate NCC populations revealed Dlc1, Sp5, and Pak3 based on their relatively high expression levels as potentially specific markers, and regulators of intermediate NCC populations. Through SABER-FISH staining of Dlc1, Sp5, and Pak3 in combination with Wnt1 as a marker of premigratory NCC, and Sox10 as a marker of migratory NCC, we spatially resolved the location of intermediate NCC to the most dorsolateral domain of the cranial neural plate in E8.5 mouse embryos. We then prioritized Dlc1 for functional analyses because it is expressed at high levels in all EMT intermediate stage NCC, but not in premigratory NCC. Lentiviral shRNA knockdown of Dlc1 in cultured mouse embryos resulted in a significant reduction in the number of migratory NCC, which may account for the craniofacial and cardiac malformation phenotypes observed in Dlc1 null mutant mice (Sabbir et al., 2010). Gross morphological defects in craniofacial tissues are apparent in E10.5 Dlc1 null mouse embryos. More specifically, Dlc1 null embryos exhibit hyperplastic frontonasal prominences and pharyngeal arches, a neural tube closure defect and underdeveloped atrial and ventricular walls of the heart. Although phenotypes at earlier developmental stages and during NCC development were not examined in these mutants, Dlc1 is primarily expressed in intermediate stage NCC, and plays an important role in NCC EMT, delamination, and the subsequent development of NCC-derived tissues. Dlc1 is a Rho GTPase-activating protein (GAP) that regulates the activity of Rho family GTPases Rho and Cdc42 (Kim et al., 2008). RhoGTPases are known to regulate cell morphology and motility through modulating the activity of the actin cytoskeleton. More specifically, Rho has been shown to facilitate the formation of stress fibers, while Cdc42 is involved in filopodium formation (Kim et al., 2008). RhoGTPases also regulate the organization of tight junctions, which breakdown during EMT (Popoff and Geny, 2009; Terry et al., 2010). Therefore, the effect of Dlc1 knockdown on cranial NCC EMT and delamination may be mediated via disrupted RhoGTPase activity and subsequent downstream cellular changes.

A similar regulatory role for Dlc1 and other GAP family members has also been observed during NCC EMT and delamination in chicken and zebrafish embryos. In chicken embryos, Dlc1 overexpression results in ectopic trunk NCC delamination, including apically into the neural tube lumen, due to a disruption in apical-basal polarity of dorsal neuroepithelial cells (Liu et al., 2017). Furthermore, NCC overexpressing Dlc1 exhibit a loss of directionality during migration. Conversely, Dlc1 inhibition and depletion results in less NCC emigration and thus fewer migratory NCC. In addition, the downregulation of Dlc1 in migrating NCC restricts their motility. Thus, Dlc1 regulates trunk NCC delamination and migration in chicken embryos. Interestingly, in zebrafish embryos, another GAP family member Arhgap has been shown to modulate NCC EMT and delamination via the localization of Rho activation to designated subcellular compartments in concert with promoting localized actomyosin contraction to trigger directional cell motility (Clay and Halloran, 2013). More specifically, the knockdown of Arhgap in NCC results in Rho activation, which in turn inhibits NCC EMT and delamination. These data suggest that the role of Dlc1 during NCC delamination might be evolutionarily conserved in vertebrate embryos. However, whether Dlc1 regulates mouse cranial NCC delamination through localized activation of Rho remains to be determined. Nevertheless, consistent with similarities in EMT, delamination, and cell migration between NCC and cancer cells, Dlc1 may also play an important role in promoting cell migration during cancer progression. High levels of DLC1 expression are detected in most melanoma tissues, and functional studies have revealed that DLC1 is both necessary and sufficient for melanoma growth and metastasis (Yang et al., 2020b).

Our study focused on the identification and characterization of NCC EMT intermediate states and essential regulatory factors during cranial NCC delamination and EMT. Nevertheless, whether trunk NCC delamination and EMT also involve intermediate stages and are regulated by similar genetic/cellular elements remains to be explored. Differences between cranial and trunk NCC delamination have been previously identified. For example, in mouse embryos, cranial NCC delaminate from the dorsolateral neuroepithelium or neural plate border prior to neural plate closure. Trunk NCC, on the other hand, delaminate from the dorsal neural tube after neural plate closure. In the cranial region, premigratory NCC delaminate in bulk as a collective stream, to give rise to the cranial ganglia and craniofacial bones and cartilages. In the trunk region, premigratory NCC delaminate progressively one after another in a chain migration fashion to form the sympathetic ganglia, the dorsal root ganglia, glial cells, and melanocytes. Additionally, key signaling pathways and regulatory networks governing cranial and trunk NCC in various species have also been shown to differ in these distinct axial populations of NCC (Theveneau and Mayor, 2012). Due to these differences between cranial and trunk NCC, the molecular and cellular mechanisms discovered during cranial NCC delamination and EMT do not necessarily represent the signaling scheme in trunk NCC. To enhance our knowledge of the gene regulatory network governing trunk NCC delamination/EMT and to decipher whether EMT intermediate states also exist in the trunk region of mouse embryos, scRNA-seq must be performed using trunk tissues during trunk NCC delamination. Since trunk NCC delamination/EMT occurs progressively from anterior to posterior as the embryos grow, it is very important to select correct axial domains of the trunk to match the appropriate or required developmental stage.

In conclusion, through scRNA-sequencing we have transcriptionally identified two distinct intermediate stages of NCC during EMT and delamination based primarily on cell cycle status. Delamination in S phase or G2/M phase seems to occur simultaneously but also independently, resulting in a single or common pool of early migratory NCC. Further interrogation of our transcriptomic dataset revealed Dlc1 to be a key molecular marker of intermediate stage NCC, and their location in situ in the dorsolateral neural plate, which we spatially resolved in E8.5 mouse embryos. Lastly, we tested and functionally validated that Dlc1 plays an important role in NCC delamination in mouse embryos. Taken together, our identification and characterization of intermediate stage cranial NCC during their delamination are consistent with NCC EMT being a developmental EMP event. Similar to intermediate EMT states in cancer metastasis, NCC downregulate certain epithelial cell features but maintain co-expression of epithelial cell markers and mesenchymal cell markers during EMT and delamination. Additionally, intermediate stage NCC are localized at the dorsolateral edge of the neural plate border, which is reminiscent of the localization of intermediate EMT states at the leading edge of invasion in several types of primary tumors. However, unlike certain intermediate EMT cells present in the lymph nodes or circulating tumor cells, mouse EMT intermediate NCC represent a transient state and eventually form migratory NCC with mesenchymal character. This suggests that EMP may be a more common developmental phenomenon. Our transcriptional data and signatures of intermediate stage NCC during EMT and delamination can serve as a useful resource for the community. This also now sets the stage for uncovering the gene regulatory networks that govern intermediate stage NCC development and function, and for exploring whether EMP is a feature of other developmental and pathological EMT events such as in gastrulation, wound healing, and fibrosis.

People are also reading