FGF binding protein 3 is required for spinal cord motor neuron development and regeneration in zebrafish

2021 ◽  
pp. 113944
Author(s):  
Guangmin Xu ◽  
Zigang Huang ◽  
Jiajing Sheng ◽  
Xiang Gao ◽  
Wang Xin ◽  
...  
2020 ◽  
Vol 13 ◽  
Author(s):  
Jie Gong ◽  
Songqun Hu ◽  
Zigang Huang ◽  
Yuebo Hu ◽  
Xiaoning Wang ◽  
...  

2016 ◽  
Vol 122 (3) ◽  
pp. 730-737 ◽  
Author(s):  
Esperanza Recio-Pinto ◽  
Jose V. Montoya-Gacharna ◽  
Fang Xu ◽  
Thomas J. J. Blanck

2020 ◽  
Author(s):  
Adèle Salin-Cantegrel ◽  
Rola Dali ◽  
Jae Woong Wang ◽  
Marielle Beaulieu ◽  
Mira Deshmukh ◽  
...  

ABSTRACTSpinal cord motor neuron diversity and the ensuing variety of motor circuits allow for the processing of elaborate muscular behaviours such as body posture and breathing. Little is known, however, about the molecular mechanisms behind the specification of axial and hypaxial motor neurons controlling postural and respiratory functions respectively. Here we show that the Groucho/TLE (TLE) transcriptional corepressor is a multi-step regulator of axial and hypaxial motor neuron diversification in the developing spinal cord. TLE first promotes axial motor neuron specification at the expense of hypaxial identity by cooperating with non-canonical WNT5A signalling within the motor neuron progenitor domain. TLE further acts during post-mitotic motor neuron diversification to promote axial motor neuron topology and axonal connectivity whilst suppressing hypaxial traits. These findings provide evidence for essential and sequential roles of TLE in the spatial and temporal coordination of events regulating the development of motor neurons influencing posture and controlling respiration.HIGHLIGHTSGroucho/TLE mediates non-canonical WNT signalling in developing motor neuronsNon canonical WNT:TLE pathway regulates thoracic motor neuron diversificationTLE promotes axial while inhibiting hypaxial motor neuron developmentTLE influences developing motor neuron topology and muscle innervationIN BRIEFSalin-Cantegrel et al use in ovo engineered approaches to show that a non-canonical WNT:TLE pathway coordinates temporally and spatially separated elements of motor neuron diversification, repressing hypaxial motor neuron development to promote the axial fate.GRAPHICAL ABSTRACTTLE contribution to the development of thoracic somatic motor columnsProgenitor cells in the ventral pMN domain are exposed to higher concentrations of non-canonical WNTs and express more TLE. Cooperation of non-canonical WNTs and TLE renders ventral pMN progenitors refractory to a respiratory MN fate, thereby contributing to the separation of MMC and RMC MN lineages. Differentiating MNs that maintain high TLE expression also maintain LHX3 expression, adopt axial motor neuron topology and connect to axial muscles. TLE activity in differentiating MMC MNs prevents the acquisition of respiratory MN topology and innervation traits.


2019 ◽  
Vol 7 (23) ◽  
Author(s):  
Siraj Patwa ◽  
Curtis A. Benson ◽  
Lauren Dyer ◽  
Kai‐Lan Olson ◽  
Lakshmi Bangalore ◽  
...  

Neuroreport ◽  
1991 ◽  
Vol 2 (9) ◽  
pp. 505-508 ◽  
Author(s):  
Edgar F. Salazar-Grueso ◽  
Sandra Kim ◽  
Howard Kim

2020 ◽  
Author(s):  
Vincent Mouilleau ◽  
Célia Vaslin ◽  
Simona Gribaudo ◽  
Rémi Robert ◽  
Nour Nicolas ◽  
...  

SUMMARYRostro-caudal patterning of vertebrates depends on the temporally progressive activation of HOX genes within axial stem cells that fuel axial embryo elongation. Whether HOX genes sequential activation, the “HOX clock”, is paced by intrinsic chromatin-based timing mechanisms or by temporal changes in extrinsic cues remains unclear. Here, we studied HOX clock pacing in human pluripotent stem cells differentiating into spinal cord motor neuron subtypes which are progenies of axial progenitors. We show that the progressive activation of caudal HOX genes in axial progenitors is controlled by a dynamic increase in FGF signaling. Blocking FGF pathway stalled induction of HOX genes, while precocious increase in FGF alone, or with GDF11 ligand, accelerated the HOX clock. Cells differentiated under accelerated HOX induction generated appropriate posterior motor neuron subtypes found along the human embryonic spinal cord. The HOX clock is thus dynamically paced by exposure parameters to secreted cues. Its manipulation by extrinsic factors alleviates temporal requirements to provide unprecedented synchronized access to human cells of multiple, defined, rostro-caudal identities for basic and translational applications.


Sign in / Sign up

Export Citation Format

Share Document