scholarly journals Regulatory roles of ganglioside GQ1b in neuronal cell differentiation of mouse embryonic stem cells

BMB Reports ◽  
2011 ◽  
Vol 44 (12) ◽  
pp. 799-804 ◽  
Author(s):  
Dong-Hoon Kwak ◽  
Jung-Woo Jin ◽  
Jae-Sung Ryu ◽  
Kinram Ko ◽  
So-Dam Lee ◽  
...  
2012 ◽  
Vol 227 (11) ◽  
pp. 3593-3602 ◽  
Author(s):  
Daniel Vittet ◽  
Galina Merdzhanova ◽  
Marie-Hélène Prandini ◽  
Jean-Jacques Feige ◽  
Sabine Bailly

2019 ◽  
Vol 112 (3) ◽  
pp. e101-e102
Author(s):  
Aysha Trout ◽  
Philip Xie ◽  
Alessandra Parrella ◽  
Zev Rosenwaks ◽  
Gianpiero D. Palermo

2021 ◽  
Vol 2021 ◽  
pp. 1-12
Author(s):  
Juan Luo ◽  
Hailin Zou ◽  
Liang Deng ◽  
Xiang Sun ◽  
Ping Yuan ◽  
...  

The RNA-binding protein Lin28 regulates neurogliogenesis in mammals, independently of the let-7 microRNA. However, the detailed regulatory mechanism remains obscured. Here, we established Lin28a or Lin28b overexpression mouse embryonic stem cells (ESCs) and found that these cells expressed similar levels of the core pluripotent factors, such as Oct4 and Sox2, and increased Yap1 but decreased lineage-specific markers compared to the control ESCs. Further differentiation of these ESCs to neuronal and glial lineage cells revealed that Lin28a/b overexpression did not affect the expression of neuronal marker βIII-tubulin, but dramatically inhibited the glial lineage markers, such as Gfap and Mbp. Interestingly, overexpression of Yap1 in mouse ESCs phenocopied Lin28a/b overexpression ESCs by showing defect in glial cell differentiation. Inhibition of Yap1/Tead-mediated transcription with verteporfin partially rescued the differentiation defect of Lin28a/b overexpression ESCs. Mechanistically, we demonstrated that Lin28 can directly bind to Yap1 mRNA, and the induction of Yap1 by Lin28a in mESCs is independent of Let7. Taken together, our results unravel a novel Lin28-Yap1 regulatory axis during mESC to glial lineage cell differentiation, which may shed light on glial cell generation in vitro.


2010 ◽  
Vol 30 (2) ◽  
pp. 243-243
Author(s):  
Ahmed M. Osman ◽  
Sjors H.W. Schulpen ◽  
Dorien A.M. van Dartel ◽  
Edwin Zwart ◽  
Marco Blokland ◽  
...  

2020 ◽  
Vol 3 (5) ◽  
pp. e201900535 ◽  
Author(s):  
Amador Gallardo ◽  
Aldara Molina ◽  
Helena G Asenjo ◽  
Jordi Martorell-Marugán ◽  
Rosa Montes ◽  
...  

Mammals optimize their physiology to the light–dark cycle by synchronization of the master circadian clock in the brain with peripheral clocks in the rest of the tissues of the body. Circadian oscillations rely on a negative feedback loop exerted by the molecular clock that is composed by transcriptional activators Bmal1 and Clock, and their negative regulators Period and Cryptochrome. Components of the molecular clock are expressed during early development, but onset of robust circadian oscillations is only detected later during embryogenesis. Here, we have used naïve pluripotent mouse embryonic stem cells (mESCs) to study the role of Bmal1 during early development. We found that, compared to wild-type cells, Bmal1−/− mESCs express higher levels of Nanog protein and altered expression of pluripotency-associated signalling pathways. Importantly, Bmal1−/− mESCs display deficient multi-lineage cell differentiation capacity during the formation of teratomas and gastrula-like organoids. Overall, we reveal that Bmal1 regulates pluripotent cell differentiation and propose that the molecular clock is an hitherto unrecognized regulator of mammalian development.


Sign in / Sign up

Export Citation Format

Share Document