학술논문

RETRACTED ARTICLE: Indoleamine 2,3-Dioxygenase 1 (IDO1) Promotes Cardiac Hypertrophy via a PI3K-AKT-mTOR-Dependent Mechanism
Article
Document Type
Academic Journal
Source
Cardiovascular Toxicology. August 2021, Vol. 21 Issue 8, p655, 14 p.
Subject
Physiological aspects
Angiotensin II -- Physiological aspects
Heart hypertrophy -- Physiological aspects
Myosin -- Physiological aspects
Heart -- Physiological aspects
Natriuretic peptides -- Physiological aspects
Protein synthesis -- Physiological aspects
Protein biosynthesis -- Physiological aspects
Heart enlargement -- Physiological aspects
Language
English
ISSN
1530-7905
Abstract
Author(s): Yang Liu [sup.1], Shuang Li [sup.1], Zhanqun Gao [sup.1], Shuangjia Li [sup.1], Qingyun Tan [sup.2], Yanmei Li [sup.1], Dongwei Wang [sup.2], Qingdong Wang [sup.2] Author Affiliations: (1) grid.452866.b, Emergency [...]
Indoleamine 2,3-dioxygenase 1 (IDO1) is an enzyme for tryptophan metabolism, involved in immune cell differentiation/maturation and cancer biology. IDO1 is also expressed in cardiomyocytes, but its roles in the cardiovascular system are not fully understood. Here, we reported the functions of IDO1 during cardiac hypertrophy. Quantitative real-time PCR and Western blot experiments demonstrated the upregulation of IDO1 mRNA and protein levels in human and hypertrophic mouse hearts, as well as in angiotensin II (Ang II)-induced hypertrophic rat cardiomyocytes. IDO1 activity and metabolite product kynurenine were upregulated in rodent hypertrophic hearts and cardiomyocytes. Inhibition of IDO1 activity with PF-06840003 reduced Ang II-induced cardiac hypertrophy and rescued cardiac function in mice. siRNA-mediated knockdown of Ido1 repressed Ang II-induced growth in cardiomyocyte size and overexpression of hypertrophy-associated genes atrial natriuretic peptide (Anp or Nppa), brain natriuretic peptide (Bnp or Nppb), [beta]-myosin heavy chain ([beta]-Mhc or Myh7). By contrast, adenovirus-mediated rat Ido1 overexpression in cardiomyocytes promoted hypertrophic growth induced by Ang II. Mechanism analysis showed that IDO1 overexpression was associated with PI3K-AKT-mTOR signaling to activate the ribosomal protein S6 kinase 1 (S6K1), which promoted protein synthesis in Ang II-induced hypertrophy of rat cardiomyocytes. Finally, we provided evidence that inhibition of PI3K with pictilisib, AKT with perifosine, or mTOR with rapamycin, blocked the effects of IDO1 on protein synthesis and cardiomyocyte hypertrophy in Ang II-treated cells. Collectively, our findings identify that IDO1 promotes cardiomyocyte hypertrophy partially via PI3K-AKT-mTOR-S6K1 signaling.