scholarly journals Induction of salivary gland-like cells from epithelial tissues transdifferentiated from mouse embryonic fibroblasts

Author(s):  
Ryogo Katada ◽  
Junichi Tanaka ◽  
Koki Takamatsu ◽  
Kenji Hata ◽  
Rika Yasuhara ◽  
...  
PLoS ONE ◽  
2013 ◽  
Vol 8 (12) ◽  
pp. e84062 ◽  
Author(s):  
Yu-Cheng Tu ◽  
Duen-Yi Huang ◽  
Shine-Gwo Shiah ◽  
Jang-Shiun Wang ◽  
Wan-Wan Lin

Cells ◽  
2021 ◽  
Vol 10 (6) ◽  
pp. 1534
Author(s):  
Krystyna Żyżyńska-Galeńska ◽  
Jolanta Karasiewicz ◽  
Agnieszka Bernat

We would like to address the issues raised by Pierre Savatier in “Introduction of Mouse Embryonic Fibroblasts into Early Embryos Causes Reprogramming and (Con)Fusion” [...]


2008 ◽  
Vol 134 (4) ◽  
pp. A-86
Author(s):  
Engda G. Hagos ◽  
Amr Ghaleb ◽  
W Brian Dalton ◽  
Jonathan P. Katz ◽  
Klaus H. Kaestner ◽  
...  

Cells ◽  
2021 ◽  
Vol 10 (4) ◽  
pp. 772
Author(s):  
Pierre Savatier

The reprogramming of somatic cell nuclei to achieve pluripotency is one of the most important biological discoveries of the last few decades [...]


Cells ◽  
2021 ◽  
Vol 10 (4) ◽  
pp. 929
Author(s):  
Aleksandra Hać ◽  
Karolina Pierzynowska ◽  
Anna Herman-Antosiewicz

Autophagy is a specific macromolecule and organelle degradation process. The target macromolecule or organelle is first enclosed in an autophagosome, and then delivered along acetylated microtubules to the lysosome. Autophagy is triggered by stress and largely contributes to cell survival. We have previously shown that S6K1 kinase is essential for autophagic flux under stress conditions. Here, we aimed to elucidate the underlying mechanism of S6K1 involvement in autophagy. We stimulated autophagy in S6K1/2 double-knockout mouse embryonic fibroblasts by exposing them to different stress conditions. Transient gene overexpression or silencing, immunoblotting, immunofluorescence, flow cytometry, and ratiometric fluorescence analyses revealed that the perturbation of autophagic flux in S6K1-deficient cells did not stem from impaired lysosomal function. Instead, the absence of S6K1 abolished stress-induced tubulin acetylation and disrupted the acetylated microtubule network, in turn impairing the autophagosome-lysosome fusion. S6K1 overexpression restored tubulin acetylation and autophagic flux in stressed S6K1/2-deficient cells. Similar effect of S6K1 status was observed in prostate cancer cells. Furthermore, overexpression of an acetylation-mimicking, but not acetylation-resistant, tubulin variant effectively restored autophagic flux in stressed S6K1/2-deficient cells. Collectively, S6K1 controls tubulin acetylation, hence contributing to the autophagic flux induced by different stress conditions and in different cells.


Sign in / Sign up

Export Citation Format

Share Document