1. Academic Validation
  2. Notch1 Inhibits Rosiglitazone-Induced Adipogenic Differentiation in Primary Thymic Stromal Cells

Notch1 Inhibits Rosiglitazone-Induced Adipogenic Differentiation in Primary Thymic Stromal Cells

  • Front Pharmacol. 2018 Nov 12;9:1284. doi: 10.3389/fphar.2018.01284.
Yajun Wang 1 Jianxin Tan 2 Hongmei Du 3 Xue Liu 1 Siliang Wang 3 Simeng Wu 4 Zhe Yuan 4 Xike Zhu 1
Affiliations

Affiliations

  • 1 Research Center, Shengjing Hospital of China Medical University, Shenyang, China.
  • 2 State Key Laboratory of Reproductive Medicine, Department of Prenatal Diagnosis, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.
  • 3 Department of Medical Oncology, Shengjing Hospital of China Medical University, Shenyang, China.
  • 4 Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang, China.
Abstract

Adipocyte deposition is believed to be a primary characteristic of age-related thymic involution. Herein, we cultured primary thymic stromal cells (TSCs), used rosiglitazone, a potent Peroxisome Proliferator-activated Receptor γ (PPARγ) agonist, to induce adipogenic differentiation, and investigated the differentially expressed genes during adipogenic differentiation by using RNA-sequencing analysis. Furthermore, the effects of Notch1 on rosiglitazone-induced adipogenic differentiation of TSCs as well as the underlying mechanisms were also investigated. As a result, we identified a total of 1737 differentially expressed genes, among which 965 genes were up-regulated and 772 genes were down-regulated in rosiglitazone-treated cells compared with control cells. Gene ontology (GO) enrichment analysis showed that the GO terms were enriched in metabolic process, intracellular, and protein binding. Kyoto encyclopedia of genes and genomes (KEGG) analysis showed that a number of pathways, including ubiquitin mediated proteolysis, PPAR signaling pathway, and mammalian target of rapamycin (mTOR) signaling pathway were predominantly over-represented. Meanwhile, overexpression of Notch1 suppressed and inhibition of Notch1 promoted rosiglitazone-induced adipogenic differentiation in TSCs, and the pro-adipogenic effects of the Notch Inhibitor DAPT were associated with the activation of Autophagy. Taken together, our results suggest that Notch1 is a key regulator in thymic adipogenesis and may serve as a potential target to hinder thymic adiposity in age-related thymic involution.

Keywords

Notch1; RNA-seq; autophagy; thymic adipogenesis; thymic stromal cells.

Figures
Products