Systemic influences contribute to prolonged microvascular rarefaction after brain irradiation: a role for endothelial progenitor cells

Am J Physiol Heart Circ Physiol. 2014 Sep 15;307(6):H858-68. doi: 10.1152/ajpheart.00308.2014. Epub 2014 Jul 18.

Abstract

Whole brain radiation therapy (WBRT) induces profound cerebral microvascular rarefaction throughout the hippocampus. Despite the vascular loss and localized cerebral hypoxia, angiogenesis fails to occur, which subsequently induces long-term deficits in learning and memory. The mechanisms underlying the absence of vessel recovery after WBRT are unknown. We tested the hypotheses that vascular recovery fails to occur under control conditions as a result of loss of angiogenic drive in the circulation, chronic tissue inflammation, and/or impaired endothelial cell production/recruitment. We also tested whether systemic hypoxia, which is known to promote vascular recovery, reverses these chronic changes in inflammation and endothelial cell production/recruitment. Ten-week-old C57BL/6 mice were subjected to a clinical series of fractionated WBRT: 4.5-Gy fractions 2 times/wk for 4 wk. Plasma from radiated mice increased in vitro endothelial cell proliferation and adhesion compared with plasma from control mice, indicating that WBRT did not suppress the proangiogenic drive. Analysis of cytokine levels within the hippocampus revealed that IL-10 and IL-12(p40) were significantly increased 1 mo after WBRT; however, systemic hypoxia did not reduce these inflammatory markers. Enumeration of endothelial progenitor cells (EPCs) in the bone marrow and circulation indicated that WBRT reduced EPC production, which was restored with systemic hypoxia. Furthermore, using a bone marrow transplantation model, we determined that bone marrow-derived endothelial-like cells home to the hippocampus after systemic hypoxia. Thus, the loss of production and homing of EPCs have an important role in the prolonged vascular rarefaction after WBRT.

Keywords: angiogenesis; cytokines; endothelial progenitor cells; hypoxia; radiation therapy.

Publication types

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

MeSH terms

  • Animals
  • Bone Marrow Transplantation
  • Brain Injuries / etiology*
  • Brain Injuries / metabolism
  • Brain Injuries / pathology
  • Brain Injuries / physiopathology
  • Cell Adhesion / radiation effects
  • Cell Proliferation / radiation effects
  • Cells, Cultured
  • Disease Models, Animal
  • Dose Fractionation, Radiation
  • Endothelial Cells / pathology
  • Endothelial Cells / radiation effects*
  • Endothelial Cells / transplantation
  • Green Fluorescent Proteins / genetics
  • Green Fluorescent Proteins / metabolism
  • Hippocampus / blood supply*
  • Hippocampus / pathology
  • Hippocampus / radiation effects*
  • Hypoxia / metabolism
  • Hypoxia / pathology
  • Hypoxia / physiopathology
  • Inflammation Mediators / metabolism
  • Interleukin-10 / metabolism
  • Interleukin-12 Subunit p40 / metabolism
  • Male
  • Mice
  • Mice, Inbred C57BL
  • Mice, Transgenic
  • Microvessels / pathology
  • Microvessels / physiopathology
  • Microvessels / radiation effects*
  • Neovascularization, Physiologic / radiation effects*
  • Radiation Injuries / etiology*
  • Radiation Injuries / metabolism
  • Radiation Injuries / pathology
  • Radiation Injuries / physiopathology
  • Stem Cell Niche
  • Stem Cells / drug effects*
  • Stem Cells / pathology
  • Time Factors
  • Whole-Body Irradiation*

Substances

  • IL10 protein, mouse
  • Inflammation Mediators
  • Interleukin-12 Subunit p40
  • Interleukin-10
  • Green Fluorescent Proteins