[go: up one dir, main page]

Evaluation of VEGF-mediated signaling in primary human cells reveals a paracrine action for VEGF in osteoblast-mediated crosstalk to endothelial cells

J Cell Physiol. 2008 Feb;214(2):537-44. doi: 10.1002/jcp.21234.

Abstract

Communication between endothelial and bone cells is crucial for controlling vascular supply during bone growth, remodeling, and repair but the molecular mechanisms coordinating this intercellular crosstalk remain ill-defined. We have used primary human and rat long bone-derived osteoblast-like cells (HOB and LOB) and human umbilical vein endothelial cells (HUVEC) to interrogate the potential autocrine/paracrine role of vascular endothelial cell growth factor (VEGF) in osteoblast:endothelial cell (OB:EC) communication and examined whether prostaglandins (PG), known modulators of both OB and EC behavior, modify VEGF production. We found that the stable metabolite of PGI2, 6-keto-PGF(1alpha) and PGE2, induced a concentration-dependent increase in VEGF release by HOBs but not ECs. In ECs, VEGF promoted early ERK1/2 activation, late cyclooxygenase-2 (COX-2) protein induction, and release of 6-keto-PGF1alpha. In marked contrast, no significant modulation of these events was observed in HOBs exposed to VEGF, but LOBs clearly exhibited COX-dependent prostanoid release (10-fold less than EC) following VEGF treatment. A low level of osteoblast-like cell responsiveness to exogenous VEGF was supported by VEGFR2/Flk-1 immunolabelling and by blockade of VEGF-mediated prostanoid generation by a VEGFR tyrosine kinase inhibitor (TKI). HOB alkaline phosphatase (ALP) activity was increased following long-term non-contact co-culture with ECs and exposure of ECs to VEGF in this system further increased OB-like cell differentiation and markedly enhanced prostanoid release. Our studies confirm a paracrine EC-mediated effect of VEGF on OB-like cell behavior and are the first supporting a model in which prostanoids may facilitate this unidirectional VEGF-driven OB:EC communication. These findings may offer novel regimes for modulating pathological bone remodeling anomalies through the control of the closely coupled vascular supply.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Acromion / cytology
  • Acromion / surgery
  • Alkaline Phosphatase / analysis
  • Animals
  • Cell Differentiation
  • Cells, Cultured
  • Coculture Techniques
  • Collagen Type I / analysis
  • Culture Media / chemistry
  • Culture Media / pharmacology
  • Dinoprostone / analysis
  • Dinoprostone / metabolism
  • Endothelial Cells / cytology
  • Endothelial Cells / metabolism*
  • Endothelium, Vascular / cytology
  • Epidermal Growth Factor / metabolism
  • Epoprostenol / analysis
  • Epoprostenol / metabolism
  • Female
  • Femur / cytology
  • Fluorescein-5-isothiocyanate
  • Fluorescent Antibody Technique, Direct
  • Fluorescent Dyes
  • Humans
  • Interleukin-1alpha
  • Organ Culture Techniques
  • Osteoblasts / cytology
  • Osteoblasts / drug effects
  • Osteoblasts / metabolism*
  • Osteocalcin / analysis
  • Paracrine Communication*
  • Propidium
  • RNA, Messenger / metabolism
  • Rats
  • Rats, Sprague-Dawley
  • Signal Transduction*
  • Time Factors
  • Umbilical Veins / cytology
  • Vascular Endothelial Growth Factor A / analysis
  • Vascular Endothelial Growth Factor A / genetics
  • Vascular Endothelial Growth Factor A / metabolism*

Substances

  • Collagen Type I
  • Culture Media
  • Fluorescent Dyes
  • Interleukin-1alpha
  • RNA, Messenger
  • VEGFA protein, human
  • Vascular Endothelial Growth Factor A
  • vascular endothelial growth factor A, rat
  • Osteocalcin
  • Propidium
  • Epidermal Growth Factor
  • Epoprostenol
  • Alkaline Phosphatase
  • Fluorescein-5-isothiocyanate
  • Dinoprostone