RegenerativeMedicine.net

Spatiotemporal structure of cell fate decisions in murine neural crest

Authors: Ruslan Soldatov, Marketa Kaucka, Maria Eleni Kastriti, Julian Petersen, Tatiana Chontorotzea, Lukas Englmaier, Natalia Akkuratova, Yunshi Yang, Martin Häring, Viacheslav Dyachuk, Christoph Bock, Matthias Farlik, Michael L. Piacentino, Franck Boismoreau, Markus M. Hilscher, Chika Yokota, Xiaoyan Qian, Mats Nilsson, Marianne E. Bronner, Laura Croci, Wen-Yu Hsiao, Jean-Francois Brunet, Gian Giacomo Consalez, Patrik Ernfors, Kaj Fried, Peter V. Kharchenko, Igor Adameyko

Summary:

Introduction: Multipotent progenitors must choose among multiple downstream fates. In developing embryos, progenitor cells exhibit transcriptional or epigenetic heterogeneity that is related to early biases in cell fate choices, and can be externally induced or stochastic in nature. Molecular assessment of the transient states assumed by cells during these developmental progressions has the potential to illuminate how such fate-specific biases emerge and unfold to ensure fate commitment. With this aim, we examine multipotent neural crest cells—transient embryonic progenitors unique to vertebrates that build the head, teeth, neuroendocrine tissue, and autonomic and sensory nervous systems. Cranial neural crest preferentially gives rise to a multitude of mesenchymal types of facial cartilage and bones, in addition to neuronal, glial, and pigment cell–type progeny. By contrast, trunk neural crest does not form bone or cartilage derivatives in vivo. The logic and molecular mechanisms that allow neural crest to resolve multiple potential cell fates at each axial level remain poorly understood.

Rationale: Here we used single-cell and spatial transcriptomics with statistical analysis of branching trajectories to investigate lineage relationships in mouse neural crest. Combined with lineage tracing and functional perturbations, we addressed spatiotemporal dynamics associated with early cell fate decisions in mouse trunk and cranial neural crest cells with different fate potential.

Results: We find that up to early migration, neural crest cells progress through a sequence of common transcriptional states, followed by fate bifurcations during migration that can be formalized as a series of sequential binary decisions. The first decision separates sensory neuro-glial fate from all other fates, whereas the second decision occurs between autonomic and mesenchymal lineages and reveals a bipotent Phox2b+/Prrx1+ subpopulation. Decision points uncover distinct roles of neural crest regulators: Neurog2 is involved in early repression of melanocytes and activation of sensory fate at later steps. Each decision consists of initial coactivation, gradual biasing, and commitment phases. Early genes of competing cell fate programs coactivate in the same cells, starting from premigratory stage. As cells approach cell fate bifurcation points, increased synchronization of fate-specific programs and repulsion of competing fate programs lead to gradual appearance of cell fate bias, which becomes pronounced upon neural crest migration. Cell fate commitment culminates with activation of mutually exclusive, fate-specific gene expression programs. Early transcriptional patterns reveal that fate biasing of neural crest is already detectable when neural crest cells delaminate from the neural tube. In particular, the neuronal bias of trunk and mesenchymal bias of cranial neural crest emerge during delamination, indicating that this might be the time when the mesenchymal potential, distinct between cranial and trunk neural crest, is installed. In support to this hypothesis, we find that sustained overexpression of a single gene, Twist1, normally activated upon delamination only in the cranial compartment, is sufficient to reverse the trunk crest developmental program to a mesenchymal route.

Conclusion: Our analysis resolved a branching transcriptional trajectory of the differentiating neural crest, illustrating transcriptional implementation of major cell fate decisions and pinpointing the key differences defining cranial versus trunk neural crest potential. Our results show that neural crest cells differentiate through a series of stereotypical lineage-restriction events that involve coexpression and competition of genes driving alternative fate programs.

Source: Science, 2019; 364 (6444): eaas9536