Role of A2B adenosine receptors in regulation of paracrine functions of stem cell antigen 1-positive cardiac stromal cells

J Pharmacol Exp Ther. 2012 Jun;341(3):764-74. doi: 10.1124/jpet.111.190835. Epub 2012 Mar 19.

Abstract

The existence of multipotent cardiac stromal cells expressing stem cell antigen (Sca)-1 has been reported, and their proangiogenic properties have been demonstrated in myocardial infarction models. In this study, we tested the hypothesis that stimulation of adenosine receptors on cardiac Sca-1(+) cells up-regulates their secretion of proangiogenic factors. We found that Sca-1 is expressed in subsets of mouse cardiac stromal CD31(-) and endothelial CD31(+) cells. The population of Sca-1(+)CD31(+) endothelial cells was significantly reduced, whereas the population of Sca-1(+)CD31(-) stromal cells was increased 1 week after myocardial infarction, indicating their relative functional importance in this pathophysiological process. An increase in adenosine levels in adenosine deaminase-deficient mice in vivo significantly augmented vascular endothelial growth factor (VEGF) production in cardiac Sca-1(+)CD31(-) stromal cells but not in Sca-1(+)CD31(+) endothelial cells. We found that mouse cardiac Sca-1(+)CD31(-) stromal cells predominantly express mRNA encoding A(2B) adenosine receptors. Stimulation of adenosine receptors significantly increased interleukin (IL)-6, CXCL1 (a mouse ortholog of human IL-8), and VEGF release from these cells. Using conditionally immortalized Sca-1(+)CD31(-) stromal cells obtained from wild-type and A(2B) receptor knockout mouse hearts, we demonstrated that A(2B) receptors are essential for adenosine-dependent up-regulation of their paracrine functions. We found that the human heart also harbors a population of stromal cells similar to the mouse cardiac Sca-1(+)CD31(-) stromal cells that increase release of IL-6, IL-8, and VEGF in response to A(2B) receptor stimulation. Thus, our study identified A(2B) adenosine receptors on cardiac stromal cells as potential targets for up-regulation of proangiogenic factors in the ischemic heart.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adenosine / metabolism
  • Adenosine Deaminase / deficiency
  • Adenosine Deaminase / metabolism
  • Agammaglobulinemia / metabolism
  • Animals
  • Antigens, Ly / metabolism*
  • Cells, Cultured
  • Chemokine CXCL1 / metabolism
  • Female
  • Interleukin-6 / metabolism
  • Male
  • Membrane Proteins / metabolism*
  • Mesenchymal Stem Cells / metabolism*
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Mice, Transgenic
  • Myocardial Infarction / metabolism
  • Myocardium / cytology
  • Paracrine Communication / physiology*
  • Platelet Endothelial Cell Adhesion Molecule-1 / metabolism
  • RNA, Messenger / metabolism
  • Receptor, Adenosine A2B / physiology*
  • Severe Combined Immunodeficiency / metabolism
  • Stem Cells / metabolism
  • Up-Regulation
  • Vascular Endothelial Growth Factor A / metabolism

Substances

  • Antigens, Ly
  • Chemokine CXCL1
  • Cxcl1 protein, mouse
  • Interleukin-6
  • Ly6a protein, mouse
  • Membrane Proteins
  • Platelet Endothelial Cell Adhesion Molecule-1
  • RNA, Messenger
  • Receptor, Adenosine A2B
  • Vascular Endothelial Growth Factor A
  • vascular endothelial growth factor A, mouse
  • Adenosine Deaminase
  • Adenosine

Supplementary concepts

  • Severe combined immunodeficiency due to adenosine deaminase deficiency