Stimulation of α1-adrenoceptor or angiotensin type 1 receptor enhances DNA synthesis in human-induced pluripotent stem cells via Gq-coupled receptor-dependent signaling pathways

2013 ◽  
Vol 714 (1-3) ◽  
pp. 202-209 ◽  
Author(s):  
Toshiaki Ishizuka ◽  
Hazuki Goshima ◽  
Ayako Ozawa ◽  
Yasuhiro Watanabe
PLoS ONE ◽  
2012 ◽  
Vol 7 (1) ◽  
pp. e30234 ◽  
Author(s):  
Luca Orlando ◽  
Yolanda Sanchez-Ripoll ◽  
James Foster ◽  
Heather Bone ◽  
Claudia Giachino ◽  
...  

2015 ◽  
Vol 24 (12) ◽  
pp. 2491-2504 ◽  
Author(s):  
Chika Kikuchi ◽  
Martin Bienengraeber ◽  
Scott Canfield ◽  
Andrew Koopmeiner ◽  
Richard Schäfer ◽  
...  

2016 ◽  
Vol 16 (2) ◽  
pp. 233-235 ◽  
Author(s):  
Jean Ann Maguire ◽  
Lin Lu ◽  
Jason A. Mills ◽  
Lisa M. Sullivan ◽  
Alyssa Gagne ◽  
...  

2016 ◽  
Vol 4 (13) ◽  
pp. 2369-2376 ◽  
Author(s):  
Xixi Dong ◽  
Haiyan Li ◽  
Yanling Zhou ◽  
Long Ou ◽  
Junkai Cao ◽  
...  

We report the stimulation of osteogenic differentiation of embryoid body (EB) cells derived from human induced pluripotent stem cells (iPSCs) by akermanite bioceramics.


2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Rui Liu ◽  
Dong Li ◽  
Fangxu Sun ◽  
Antonio Rampoldi ◽  
Joshua T. Maxwell ◽  
...  

Abstract Background Treatment-induced cardiotoxicity is a leading noncancer-related cause of acute and late onset morbidity and mortality in cancer patients on antineoplastic drugs such as melphalan—increasing clinical case reports have documented that it could induce cardiotoxicity including severe arrhythmias and heart failure. As the mechanism by which melphalan impairs cardiac cells remains poorly understood, here, we aimed to use cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) to investigate the cellular and molecular mechanisms of melphalan-induced cardiotoxicity. Methods hiPSC-CMs were generated and treated with clinically relevant doses of melphalan. To characterize melphalan-induced cardiotoxicity, cell viability and apoptosis were quantified at various treatment durations. Ca2+ transient and contractility analyses were used to examine the alterations of hiPSC-CM function. Proteomic analysis, reactive oxygen species detection, and RNA-Sequencing were conducted to investigate underlying mechanisms. Results Melphalan treatment of hiPSC-CMs induced oxidative stress, caused Ca2+ handling defects and dysfunctional contractility, altered global transcriptomic and proteomic profiles, and resulted in apoptosis and cell death. The antioxidant N-acetyl-l-cysteine attenuated these genomic, cellular, and functional alterations. In addition, several other signaling pathways including the p53 and transforming growth factor-β signaling pathways were also implicated in melphalan-induced cardiotoxicity according to the proteomic and transcriptomic analyses. Conclusions Melphalan induces cardiotoxicity through the oxidative stress pathway. This study provides a unique resource of the global transcriptomic and proteomic datasets for melphalan-induced cardiotoxicity and can potentially open up new clinical mechanism-based targets to prevent and treat melphalan-induced cardiotoxicity.


Sign in / Sign up

Export Citation Format

Share Document