scholarly journals ZFP217 regulates adipogenesis by controlling mitotic clonal expansion in a METTL3-m6A dependent manner

RNA Biology ◽  
2019 ◽  
Vol 16 (12) ◽  
pp. 1785-1793 ◽  
Author(s):  
Qing Liu ◽  
Yuanling Zhao ◽  
Ruifan Wu ◽  
Qin Jiang ◽  
Min Cai ◽  
...  
Endocrinology ◽  
2007 ◽  
Vol 148 (4) ◽  
pp. 1598-1604 ◽  
Author(s):  
Erin R. Cernkovich ◽  
Jianbei Deng ◽  
Kunjie Hua ◽  
Joyce B. Harp

Mitotic clonal expansion is believed to be necessary for 3T3-L1 adipocyte formation. Signal transducer and activator of transcription 3 (STAT3), a mitogenic signaling protein, is activated through tyrosine phosphorylation during the proliferative phases of adipogenesis. We hypothesize that this signaling protein plays a key role in mitotic clonal expansion and differentiation. Here we determined that the adipocyte differentiation cocktail containing isobutylmethylxanthine, dexamethasone, and insulin (MDI) induced STAT3 tyrosine phosphorylation indirectly through the synthesis of an autocrine/paracrine factor. We further determined that the factor has heparin binding properties and identified the factor as midkine, a pleiotrophic growth factor previously associated with neuronal development and oncogenesis. Recombinant midkine induced STAT3 tyrosine phosphorylation in a time- and dose-dependent manner and stimulated the proliferation of postconfluent 3T3-L1 cells. Midkine neutralizing antibodies inhibited differentiation-induced STAT3 tyrosine phosphorylation as well as adipogenesis. These results show that MDI-induced synthesis and release of midkine explains the delayed activation of STAT3 during adipogenesis and that the midkine-STAT3 signaling pathway plays a necessary role in mitotic clonal expansion and differentiation.


2012 ◽  
Vol 190 (1) ◽  
pp. 174-183 ◽  
Author(s):  
Sonia Feau ◽  
Stephen P. Schoenberger ◽  
Amnon Altman ◽  
Stéphane Bécart

BMB Reports ◽  
2021 ◽  
Vol 54 (7) ◽  
pp. 374-379
Author(s):  
Seulgi Go ◽  
Jihyun Park ◽  
Safikur Rahman ◽  
Juno Jin ◽  
Inho Choi & Jihoe Kim

2019 ◽  
Vol 10 (1) ◽  
Author(s):  
Zhenzhen Pan ◽  
Zixin Zhou ◽  
Huiying Zhang ◽  
Hui Zhao ◽  
Peixuan Song ◽  
...  

Abstract Background White adipose tissue includes subcutaneous and visceral adipose tissue (SAT and VAT) with different metabolic features. SAT protects from metabolic disorders, while VAT promotes them. The proliferative and adipogenic potentials of adipose-derived stem cells (ADSCs) are critical for maintaining adipose tissue homeostasis through driving adipocyte hyperplasia and inhibiting pathological hypertrophy. However, it remains to be elucidated the critical molecules that regulate different potentials of subcutaneous and visceral ADSCs (S-ADSCs, V-ADSCs) and mediate distinct metabolic properties of SAT and VAT. CD90 is a glycosylphosphatidylinositol-anchored protein on various cells, which is also expressed on ADSCs. However, its expression patterns and differential regulation on S-ADSCs and V-ADSCs remain unclear. Methods S-ADSCs and V-ADSCs were detected for CD90 expression. Proliferation, colony formation, cell cycle, mitotic clonal expansion, and adipogenic differentiation were assayed in S-ADSCs, V-ADSCs, or CD90-silenced S-ADSCs. Glucose tolerance test and adipocyte hypertrophy were examined in mice after silencing of CD90 in SAT. CD90 expression and its association with CyclinD1 and Leptin were analyzed in adipose tissue from mice and humans. Regulation of AKT by CD90 was detected using a co-transfection system. Results Compared with V-ADSCs, S-ADSCs expressed high level of CD90 and showed increases in proliferation, mitotic clonal expansion, and adipogenic differentiation, together with AKT activation and G1-S phase transition. CD90 silencing inhibited AKT activation and S phase entry, thereby curbing proliferation and mitotic clonal expansion of S-ADSCs. In vivo CD90 silencing in SAT inhibited S-ADSC proliferation, which caused adipocyte hypertrophy and glucose intolerance in mice. Furthermore, CD90 was highly expressed in SAT rather than in VAT in human and mouse, which had positive correlation with CyclinD1 but negative correlation with Leptin. CD90 promoted AKT activation through recruiting its pleckstrin homology domain to plasma membrane. Conclusions CD90 is differentially expressed on S-ADSCs and V-ADSCs, and plays critical roles in ADSC proliferation, mitotic clonal expansion, and hemostasis of adipose tissue and metabolism. These findings identify CD90 as a crucial modulator of S-ADSCs and V-ADSCs to mediate distinct metabolic features of SAT and VAT, thus proposing CD90 as a valuable biomarker or target for evaluating ADSC potentials, monitoring or treating obesity-associated metabolic disorders.


2009 ◽  
Vol 284 (16) ◽  
pp. 10601-10609 ◽  
Author(s):  
Haemi Lee ◽  
Yoo Jeong Lee ◽  
Hyeonjin Choi ◽  
Eun Hee Ko ◽  
Jae-woo Kim

Sign in / Sign up

Export Citation Format

Share Document