Your browser doesn't support javascript.
loading
Reticulon 3 regulates sphingosine-1-phosphate synthesis in endothelial cells to control blood pressure.
Jin, Jie-Yuan; Chang, Si-Hua; Chen, Ya-Qin; Liu, Meng-Wei; Dong, Yi; Liu, Ji-Shi; Wang, Qin; Huang, Hao; Fan, Liang-Liang; Xiang, Rong.
Afiliación
  • Jin JY; Department of Orthopaedics Microsurgery & Reconstruction Research Center Xiangya Hospital Central South University Changsha China.
  • Chang SH; Department of Cell Biology, School of Life Sciences Central South University Changsha China.
  • Chen YQ; National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha China.
  • Liu MW; Department of Cell Biology, School of Life Sciences Central South University Changsha China.
  • Dong Y; Department of Cardiology The Second Xiangya Hospital of Central South University Changsha China.
  • Liu JS; Department of Cell Biology, School of Life Sciences Central South University Changsha China.
  • Wang Q; College of Basic Medical Xinjiang Medical University Urumqi China.
  • Huang H; Department of Cell Biology, School of Life Sciences Central South University Changsha China.
  • Fan LL; Department of Nephrology The Third Xiangya Hospital of Central South University Changsha China.
  • Xiang R; Department of Cell Biology, School of Life Sciences Central South University Changsha China.
MedComm (2020) ; 5(2): e480, 2024 Feb.
Article en En | MEDLINE | ID: mdl-38352050
ABSTRACT
The discovery of the endothelium as a major regulator of vascular tone triggered intense research among basic and clinical investigators to unravel the physiologic and pathophysiologic significance of this phenomenon. Sphingosine-l-phosphate (S1P), derived from the vascular endothelium, is a significant regulator of blood pressure. However, the mechanisms underlying the regulation of S1P biosynthetic pathways in arteries remain to be further clarified. Here, we reported that Reticulon 3 (RTN3) regulated endothelial sphingolipid biosynthesis and blood pressure. We employed public datasets, patients, and mouse models to explore the pathophysiological roles of RTN3 in blood pressure control. The underlying mechanisms were studied in human umbilical vein endothelial cells (HUVECs). We reported that increased RTN3 was found in patients and that RTN3-null mice presented hypotension. In HUVECs, RTN3 can regulate migration and tube formation via the S1P signaling pathway. Mechanistically, RTN3 can interact with CERS2 to promote the selective autophagy of CERS2 and further influence S1P signals to control blood pressure. We also identified an RTN3 variant (c.116C>T, p.T39M) in a family with hypertension. Our data provided the first evidence of the association between RTN3 level changes and blood pressure anomalies and preliminarily elucidated the importance of RTN3 in S1P metabolism and blood pressure regulation.
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Idioma: En Revista: MedComm (2020) Año: 2024 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Idioma: En Revista: MedComm (2020) Año: 2024 Tipo del documento: Article