Family with sequence similarity 5, member C (FAM5C) increases leukocyte adhesion molecules in vascular endothelial cells: implication in vascular inflammation

PLoS One. 2014 Sep 24;9(9):e107236. doi: 10.1371/journal.pone.0107236. eCollection 2014.

Abstract

Identification of the regulators of vascular inflammation is important if we are to understand the molecular mechanisms leading to atherosclerosis and consequent ischemic heart disease, including acute myocardial infarction. Gene polymorphisms in family with sequence similarity 5, member C (FAM5C) are associated with an increased risk of acute myocardial infarction, but little is known about the function of this gene product in blood vessels. Here, we report that the regulation of the expression and function of FAM5C in endothelial cells. We show here that FAM5C is expressed in endothelial cells in vitro and in vivo. Immunofluorescence microscopy showed localization of FAM5C in the Golgi in cultured human endothelial cells. Immunohistochemistry on serial sections of human coronary artery showed that FAM5C-positive endothelium expressed intercellular adhesion molecule-1 (ICAM-1) or vascular cell adhesion molecule-1 (VCAM-1). In cultured human endothelial cells, the overexpression of FAM5C increased the reactive oxygen species (ROS) production, nuclear factor-κB (NF-κB) activity and the expression of ICAM-1, VCAM-1 and E-selectin mRNAs, resulting in enhanced monocyte adhesion. FAM5C was upregulated in response to inflammatory stimuli, such as TNF-α, in an NF-κB- and JNK-dependent manner. Knockdown of FAM5C by small interfering RNA inhibited the increase in the TNF-α-induced production of ROS, NF-κB activity and expression of these leukocyte adhesion molecule mRNAs, resulting in reduced monocyte adhesion. These results suggest that in endothelial cells, when FAM5C is upregulated in response to inflammatory stimuli, it increases the expression of leukocyte adhesion molecules by increasing ROS production and NF-κB activity.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Cell Adhesion / genetics
  • Cells, Cultured
  • DNA-Binding Proteins / genetics
  • DNA-Binding Proteins / metabolism*
  • E-Selectin / genetics
  • E-Selectin / metabolism
  • Endothelial Cells / drug effects
  • Endothelial Cells / metabolism*
  • Gene Expression
  • Golgi Apparatus / metabolism
  • Humans
  • Immunoblotting
  • Immunohistochemistry
  • Inflammation / genetics
  • Inflammation / metabolism
  • Intercellular Adhesion Molecule-1 / genetics
  • Intercellular Adhesion Molecule-1 / metabolism*
  • Male
  • Mice, Inbred C57BL
  • Microscopy, Fluorescence
  • NF-kappa B / metabolism
  • RNA Interference
  • Reactive Oxygen Species / metabolism
  • Reverse Transcriptase Polymerase Chain Reaction
  • Tumor Necrosis Factor-alpha / pharmacology
  • Up-Regulation / drug effects
  • Vascular Cell Adhesion Molecule-1 / genetics
  • Vascular Cell Adhesion Molecule-1 / metabolism*

Substances

  • BRINP3 protein, human
  • DNA-Binding Proteins
  • E-Selectin
  • NF-kappa B
  • Reactive Oxygen Species
  • Tumor Necrosis Factor-alpha
  • Vascular Cell Adhesion Molecule-1
  • Intercellular Adhesion Molecule-1

Grants and funding

This work was supported by a Grant-in-Aid for Scientific Research from the Japan Society for the Promotion of Science (C) (25461128) (https://www.jsps.go.jp/english/e-grants/) (to Y. Rikitake), and grants from the Global Center of Excellence (http://www.jsps.go.jp/english/e-globalcoe/index.html) (to M. Kinugasa, K. Hirata, Y. Rikitake), the Takeda Science Foundation (http://www.takeda-sci.or.jp), the Mochida Memorial Foundation for Medical and Pharmaceutical Research (http://www.mochida.co.jp/zaidan/), and the SENSHIN Medical Research Foundation (http://www.mt-pharma.co.jp/zaidan/) (to Y. Rikitake). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.