SCF-KIT signaling induces endothelin-3 synthesis and secretion: Thereby activates and regulates endothelin-B-receptor for generating temporally- and spatially-precise nitric oxide to modulate SCF- and or KIT-expressing cell functions

PLoS One. 2017 Sep 7;12(9):e0184154. doi: 10.1371/journal.pone.0184154. eCollection 2017.

Abstract

We demonstrate that SCF-KIT signaling induces synthesis and secretion of endothelin-3 (ET3) in human umbilical vein endothelial cells and melanoma cells in vitro, gastrointestinal stromal tumors, human sun-exposed skin, and myenteric plexus of human colon post-fasting in vivo. This is the first report of a physiological mechanism of ET3 induction. Integrating our finding with supporting data from literature leads us to discover a previously unreported pathway of nitric oxide (NO) generation derived from physiological endothelial NO synthase (eNOS) or neuronal NOS (nNOS) activation (referred to as the KIT-ET3-NO pathway). It involves: (1) SCF-expressing cells communicate with neighboring KIT-expressing cells directly or indirectly (cleaved soluble SCF). (2) SCF-KIT signaling induces timely local ET3 synthesis and secretion. (3) ET3 binds to ETBR on both sides of intercellular space. (4) ET3-binding-initiated-ETBR activation increases cytosolic Ca2+, activates cell-specific eNOS or nNOS. (5) Temporally- and spatially-precise NO generation. NO diffuses into neighboring cells, thus acts in both SCF- and KIT-expressing cells. (6) NO modulates diverse cell-specific functions by NO/cGMP pathway, controlling transcriptional factors, or other mechanisms. We demonstrate the critical physiological role of the KIT-ET3-NO pathway in fulfilling high demand (exceeding basal level) of endothelium-dependent NO generation for coping with atherosclerosis, pregnancy, and aging. The KIT-ET3-NO pathway most likely also play critical roles in other cell functions that involve dual requirement of SCF-KIT signaling and NO. New strategies (e.g. enhancing the KIT-ET3-NO pathway) to harness the benefit of endogenous eNOS and nNOS activation and precise NO generation for correcting pathophysiology and restoring functions warrant investigation.

MeSH terms

  • Atherosclerosis / pathology
  • Cell Line, Tumor
  • Endothelin-3 / metabolism*
  • Endothelium, Vascular / metabolism
  • Enzyme-Linked Immunosorbent Assay
  • Gastrointestinal Motility
  • Gastrointestinal Stromal Tumors / metabolism
  • Gastrointestinal Stromal Tumors / pathology
  • Gastrointestinal Stromal Tumors / physiopathology
  • Homeostasis
  • Human Umbilical Vein Endothelial Cells / metabolism
  • Humans
  • Immunohistochemistry
  • Melanoma / pathology
  • Myenteric Plexus / metabolism
  • Neoplasm Invasiveness
  • Nitric Oxide / metabolism*
  • Nitric Oxide Synthase Type I / metabolism
  • Nitric Oxide Synthase Type III / metabolism
  • Oligonucleotide Array Sequence Analysis
  • Proto-Oncogene Proteins c-kit / metabolism*
  • Receptor, Endothelin B / metabolism*
  • Signal Transduction
  • Skin / metabolism
  • Stem Cell Factor / metabolism*
  • Sunlight
  • Time Factors
  • Up-Regulation / genetics
  • Vasodilation

Substances

  • Endothelin-3
  • Receptor, Endothelin B
  • Stem Cell Factor
  • Nitric Oxide
  • Nitric Oxide Synthase Type I
  • Nitric Oxide Synthase Type III
  • Proto-Oncogene Proteins c-kit

Grants and funding

The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. The commercial affiliations provided support in the form of salaries for authors [JS, SRT, AJ, MAV], but did not have any additional role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.