Abstract

After its publication in 1999 as a DNA-binding and SMAD-binding transcription factor (TF) that co-determines cell fate in amphibian embryos, ZEB2 was from 2003 studied by embryologists mainly by documenting the consequences of conditional, cell-type specific Zeb2 knockout (cKO) in mice. In between, it was further identified as causal gene causing Mowat-Wilson Syndrome (MOWS) and novel regulator of epithelial–mesenchymal transition (EMT). ZEB2’s functions and action mechanisms in mouse embryos were first addressed in its main sites of expression, with focus on those that helped to explain neurodevelopmental and neural crest defects seen in MOWS patients. By doing so, ZEB2 was identified in the forebrain as the first TF that determined timing of neuro-/gliogenesis, and thereby also the extent of different layers of the cortex, in a cell non-autonomous fashion, i.e., by its cell-intrinsic control within neurons of neuron-to-progenitor paracrine signaling. Transcriptomics-based phenotyping of Zeb2 mutant mouse cells have identified large sets of intact-ZEB2 dependent genes, and the cKO approaches also moved to post-natal brain development and diverse other systems in adult mice, including hematopoiesis and various cell types of the immune system. These new studies start to highlight the important adult roles of ZEB2 in cell–cell communication, including after challenge, e.g., in the infarcted heart and fibrotic liver. Such studies may further evolve towards those documenting the roles of ZEB2 in cell-based repair of injured tissue and organs, downstream of actions of diverse growth factors, which recapitulate developmental signaling principles in the injured sites. Evident questions are about ZEB2’s direct target genes, its various partners, and ZEB2 as a candidate modifier gene, e.g., in other (neuro)developmental disorders, but also the accurate transcriptional and epigenetic regulation of its mRNA expression sites and levels. Other questions start to address ZEB2’s function as a niche-controlling regulatory TF of also other cell types, in part by its modulation of growth factor responses (e.g., TGFβ/BMP, Wnt, Notch). Furthermore, growing numbers of mapped missense as well as protein non-coding mutations in MOWS patients are becoming available and inspire the design of new animal model and pluripotent stem cell-based systems. This review attempts to summarize in detail, albeit without discussing ZEB2’s role in cancer, hematopoiesis, and its emerging roles in the immune system, how intense ZEB2 research has arrived at this exciting intersection.

Highlights

  • ZEB2, originally named SIP1, was discovered in a yeast 2-hybrid screening together with other candidate SMAD-interacting proteins (SIPs) from mid-gestation mouse embryos, using the transcription activation (i.e., MH2) domain of BMP-SMAD1 as bait [1]

  • Al least half of the Mowat-Wilson Syndrome (MOWS) patients suffer from Hirschsprung disease (HSCR), which manifests by gastrointestinal defects as megacolon and constipation and/or obstruction of the bowel; about half of the patients develop seizures and epilepsy [12,22,23,24] (Figure 1B)

  • Because we focus here on Zeb2 functional analysis of central nervous system (CNS)/PNS development in the mouse and include adult neurogenesis, and because of the connection to certain aspects of MOWS, we recall here EGFP-based confirmation of ZEB2 presence in pyramidal neurons of the hippocampal dentate gyrus, and in neurons in the brain cortex, where in layer 5 Zeb2-driven EGFP was complementary to CTIP2+ neurons, possibly in corticospinal neurons

Read more

Summary

Discovery of ZEB2

ZEB2, originally named SIP1 (and ZFHX1B), was discovered in a yeast 2-hybrid screening together with other candidate SMAD-interacting proteins (SIPs) from mid-gestation mouse embryos, using the transcription activation (i.e., MH2) domain of BMP-SMAD1 as bait [1]. The homozygous Zeb2-cKO mouse models provided explanations for multiple defective cellular and developmental aspects of MOWS in humans This was the case for CNS deficiencies, such as brain neuro/gliogenesis and guided migration of forebrain interneurons [47,64], as well as for eye lens malformation and for the resulting imbalance between cell types in the early and late retina [77,79,86], neurocristopathies resulting from aberrant craniofacial and ENS, as well as trunk NCCs [8,59,87], and reduced pain sensitivity [73,74]. For an expected growing number of mutations in mild cases of MOWS that do not affect ZEB2 protein, but do affect the timing and level of ZEB2 transcription, cellular rather than animal models may be developed first

ZEB2 Gene and Protein Organization
ZEB2 Regulation
MOWS and Neurodevelopmental Relevant Zeb2 Expression Domains in Mouse Embryos
Zeb2-cKO in the Neural Crest Cell Lineage
Neurocristopathies Reminiscent of MOWS
Dorsal Root Ganglia and Pain Sensing
The ENS and HSCR
Zeb2-cKO in the Developing Forebrain
ZEB2 in the Formation and Output of the Adult Neurogenic V-SVZ Compartment
Emerging Cellular Models for Studying ZEB2
Findings
General Conclusion and Future Perspectives
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call