Contraindications of VEGF-based therapeutic angiogenesis: Effects on macrophage density and histology of normal and ischemic brains

https://doi.org/10.1016/j.vph.2006.01.008Get rights and content

Abstract

Therapeutic angiogenesis by vascular endothelial growth factor (VEGF) is advocated as a promising treatment strategy for brain ischemic stroke. However, data in the literature demonstrating the benefit of therapeutic angiogenesis are contradictory. In this paper, we describe the effects of non-angiogenic and angiogenic doses of VEGF165 on macrophage density and histology of normal and ischemic brains of adult rats. VEGF165 was administered intra-arterially for 7 days following temporary occlusion of the middle cerebral artery. In contrast to ischemic brains treated with non-angiogenic doses of VEGF165 which showed preserved neuropil and reduced numbers of macrophages, ischemic brains treated by an angiogenic dose showed phagocytized neuropil and high macrophage density. Though neither non-angiogenic nor angiogenic doses caused macrophage infiltration in normal brains, damage of the brain matrix occurred with the angiogenic dose. These results suggest an angiogenic dose of VEGF165 injures the nervous tissue rather than promote recovery. Angiogenesis by VEGF monotherapy for ischemic stroke should be viewed with caution, or avoided. Since our data show intravascular administration of VEGF165 does not cause macrophage inflammation, in contrast to reports in the literature whereby VEGF165 was applied directly to the brain, our findings also indicate the relationships between VEGF, angiogenesis, and macrophage inflammation are governed by the route VEGF is administered to the brain.

Introduction

Vascular endothelial growth factor (VEGF) is neuroprotective and angiogenic (Sun et al., 2003, Carmeliet and Storkebaum, 2002, Ferrara and Gerber, 2001). VEGF also promotes neurogenesis, learning, memory and inhibition of apoptosis (Cao et al., 2004, Greenberg and Jin, 2004, Jin et al., 2002). Consequently, VEGF earned wide attention as a promising pharmacological agent for the treatment of ischemic stroke (Bellomo et al., 2003, Krupinski et al., 2003, Zhang and Chopp, 2002). VEGF could simultaneously protect the injured neurons and augment blood flow to the ischemic tissue. On the other hand, VEGF is notorious for causing increased vascular permeability, inflammation, vasodilation and alterations of systemic hemodynamics (Croll et al., 2004a, Bates and Harper, 2002, Forstreuter et al., 2002). VEGF-induced angiogenic vessels, particularly in the formative stages, are leaky (Bates et al., 2002, Schoch et al., 2002, Dobrogowska et al., 1998). These effects of VEGF can lead to compromise of the blood–brain barrier (BBB) and edema and damage of the nervous tissue (Schoch et al., 2002, Pettersson et al., 2000, van Bruggen et al., 1999). Thus, it is unclear whether neuroprotection and angiogenesis by exogenous VEGF can simultaneously be induced for the treatment of ischemic stroke (Carmeliet and Storkebaum, 2002, Ferrara, 2002, Zhang and Chopp, 2002). Though angiogenesis by VEGF may show therapeutic value for limb and heart ischemia (Losordo et al., 1998, Isner, 1998, Isner et al., 1996), it is possible that angiogenesis by VEGF monotherapy (i.e., VEGF treatment without inclusion of other pharmacologic agents to counteract the side-effects and/or enhance the positive effects of the growth factor) may not be beneficial for the treatment of brain ischemia. Rather, angiogenesis by VEGF monotherapy may pose the risk of greater harm to the stroke patient. We have shown that neuroprotection and angiogenesis by exogenous VEGF165 monotherapy are not necessarily concurrent events (Manoonkitiwongsa et al., 2004). Instead, the relationship between neuroprotection and angiogenesis appears inversely related. Our findings regarding the inverse relationship between neuroprotection and angiogenesis with VEGF monotherapy are supported by later findings of other investigators (Wang et al., 2005, Valable et al., 2005).

The purpose of this report is to evaluate the tissues of our earlier work (Manoonkitiwongsa et al., 2004) to characterize changes in the histology of the brain matrix and density of macrophages of normal and ischemic brains exposed to non-angiogenic and angiogenic doses of VEGF165. This information is vital if angiogenesis by VEGF is to be applied for the treatment of ischemic stroke. The histopathologic endpoints are indispensable for elucidating the therapeutic and adverse effects of VEGF on the nervous tissue. With respect to macrophage inflammation, it is currently believed that administration of exogenous VEGF165 to the brain causes migration of macrophages (Croll et al., 2004b, Proescholdt et al., 1999). However, in these reports, VEGF165 was applied directly to the brain through invasive techniques. Thus, it is unclear whether VEGF administered intravascularly would produce similar relationships between exogenous VEGF, angiogenesis and macrophage inflammation. In clinical practice, presumably, VEGF would be administered intravascularly rather than directly to the brain through invasive approaches such as cortical penetration, intraventricular or topical. The presence of the blood–brain barrier (BBB), which VEGF would encounter when administered intravascularly, could change the manners in which exogenous VEGF influences the migration of macrophages. The route through which VEGF is administered to the brain can produce different outcomes (Kaya et al., 2005). For our study, we targeted the macrophages as markers of inflammation because they are the characteristic inflammatory cells of ischemic brain (Croll et al., 2004b, Tanaka et al., 2003, Persson et al., 1989). It is known that VEGF attracts macrophages and that macrophages secrete VEGF and express VEGF receptors (Malaguarnera et al., 2004, Sawano et al., 2001, Moore et al., 2001, Heil et al., 2000, Waltenberger et al., 2000).

Section snippets

Animals, occlusion of middle cerebral artery, and VEGF doses

The animal experiments were approved by the Animal Research Committee of the Department of Veterans Affairs Medical Center, San Diego, CA, and conducted according to the National Research Council's Guide for the Care and Use of Laboratory Animals. Adult male Sprague–Dawley rats, weighing between 250–320 g, were subjected to middle cerebral artery occlusion (MCAO) for 4 h using a modification of the Longa et al. (1989) procedure for MCAO. After MCAO, the occluding suture was removed to allow

Macrophage density data

The MD means ± SD of all the 10 groups are shown in Table 1. No macrophages were present in Groups 1–5, and 8. Though Group 9 showed a few macrophages, the number was not statistically different from Groups 1–5, and 8. In contrast, the MD of Groups 6, 7 and 10 were significantly increased compared to Groups 1–5, 8 and 9 (p < 0.001). There were no differences between Groups 6, 7 and 10.

Histology data of normal brains (Groups 1–5)

Fig. 2A–D show the histology of untreated normal brain (2A), normal brain treated by the low (non-angiogenic) dose

VEGF-based angiogenic monotherapy for brain ischemic stroke

It is currently proposed that therapeutic angiogenesis by VEGF is a promising treatment method for ischemic brain (Chen et al., 2005, Rosell-Novel et al., 2004, Wang et al., 2004, Greenberg, 1998). VEGF could augment blood flow to the ischemic brain and simultaneously preserve the injured neurons. In essence, VEGF could promote neuroprotection through indirect (functional angiogenesis) and direct neuroprotection mechanisms. However, the findings of our study suggest that safe and effective

Acknowledgements

Funding for this project was provided by RO1-NS043300, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA, and the Veterans Affairs Medical Research Department, San Diego, CA, USA. We thank Dr. Douglas B. McCreery, Director of the Neural Engineering Program, Huntington Medical Research Institutes (HMRI), Pasadena, CA, USA, for his valuable constructive criticisms of the paper. The review of the manuscript by Dr. Albert S. Lossinsky,

References (61)

  • J.M. Isner et al.

    Clinical evidence of angiogenesis after arterial gene transfer of ph VEGF165 in patient with ischaemic limb

    Lancet

    (1996)
  • S. Kanda et al.

    Fibroblast growth factor-2-mediated capillary morphogenesis of endothelial cells requires signals via Flt-1/vascular endothelial growth factor receptor-1: possible involvement of c-Akt

    J. Biol. Chem.

    (2004)
  • A. Pettersson et al.

    Heterogeneity of the angiogenic response induced in different normal adult tissues by vascular permeability factor/vascular endothelial growth factor

    Lab. Invest.

    (2000)
  • A. Sawano et al.

    Flt-1, vascular endothelial growth factor receptor 1, is a novel cell surface marker for the lineage of monocyte-macrophages in humans

    Blood

    (2001)
  • R. Tanaka et al.

    Migration of enhanced green fluorescent protein expressing bone marrow-derived microglia/macrophage into the mouse brain following permanent focal ischemia

    Neurosci.

    (2003)
  • M.J. Tolentino et al.

    Intravitreous injections of vascular endothelial growth factor produce retinal ischemia and microangiopathy in an adult primate

    Ophthalmology

    (1996)
  • D. Wu

    Neuroprotection in experimental stroke with targeted neurotrophins

    NeuroRx

    (2005)
  • Z. Zhang et al.

    Vascular endothelial growth factor and angiopoietins in focal cerebral ischemia

    Trends Cardiovasc. Med.

    (2002)
  • A. Al-Khaldi et al.

    Postnatal bone marrow stromal cells elicit a potent VEGF-dependent neoangiogenic response in vivo

    Gene Ther.

    (2003)
  • J.A. Alvarez et al.

    Localization of basic fibroblast growth factor and vascular endothelial growth factor in human glial neoplasms

    Mod. Path.

    (1992)
  • D.O. Bates et al.

    Regulation of microvascular permeability by vascular endothelial growth factors

    J. Anat.

    (2002)
  • J.B. Bederson et al.

    Rat middle cerebral artery occlusion: evaluation of the model and development of a neurologic examination

    Stroke

    (1986)
  • X.W. Bian et al.

    Correlation of bFGF, FGFR-1 and VEGF expression with vascularity and malignancy of human astrocytomas

    Anal. Quant. Cytol. Histol.

    (2000)
  • J. Bogousslavsky et al.

    Fiblast (trafermin) in acute stroke: results of the European–Australian phase II/III safety and efficacy trial

    Cerebrovasc. Dis.

    (2002)
  • L. Cao et al.

    VEGF links hippocampal activity with neurogenesis, learning and memory

    Nat. Genet.

    (2004)
  • J. Chen et al.

    Atorvastatin induction of VEGF and BDNF promotes brain plasticity after stroke in mice

    J. Cereb. Blood Flow Metab.

    (2005)
  • Z.Z. Chong et al.

    Angiogenesis and plasticity: role of erythropoietin in vascular systems

    J. Hematother. Stem Cell Res.

    (2002)
  • S.D. Croll et al.

    Vascular endothelial growth factor (VEGF) in seizures: a double-edged sword

    Adv. Exp. Med. Biol.

    (2004)
  • G. D'Angelo et al.

    cAMP-dependent protein kinase inhibits the mitogenic action of vascular endothelial growth factor and fibroblast growth factor in capillary endothelial cells by blocking Raf activation

    J. Cell. Biochem.

    (1997)
  • D.H. Dobrogowska et al.

    Increased blood–brain barrier permeability and endothelial abnormalities induced by vascular endothelial growth factor

    J. Neurocytol.

    (1998)
  • Cited by (42)

    • Electrical preconditioning of stem cells with a conductive polymer scaffold enhances stroke recovery

      2017, Biomaterials
      Citation Excerpt :

      Further studies of the observed vasculature changes could evaluate integrity of the vessels or determine if the increased vasculature has additional benefits for the behavioral improvement of the animals. Additional effects of VEGF-A after stroke would also be interesting to explore in future studies, including its role in the immune response and blood brain barrier regulation [37,38]. With our in vitro system, we can seed the hNPCs onto the conductive scaffold, stimulate the cells and then implant the conductive scaffold into the post-stroke environment.

    • Background on Imaging Structural Imaging

      2015, Image-Guided Neurosurgery
    • Advanced imaging of adult brain tumors with MRI and PET

      2010, Blue Books of Neurology
      Citation Excerpt :

      Intermediate grade gliomas may produce varying degrees of upregulation of vascular growth factor/receptor signaling—including VEGF, PDGF, EGFR and IL-8 among others—and expression of AQP4 and other aquaporins that disrupt endothelial tight junctions. These factors act directly on existing brain capillaries to increase permeability of the blood-brain barrier (BBB), resulting in varying degrees of edema; they may produce mild contrast enhancement.92 On the other hand, high density, morphologically abnormal, tortuous, high-density neocapillaries are seen nearly exclusively in HGG and often result in markedly elevated local cerebral capillary blood volume (CBV).

    • Enhanced angiogenesis with dl-3n-butylphthalide treatment after focal cerebral ischemia in RHRSP

      2009, Brain Research
      Citation Excerpt :

      Its level is elevated during hypoxia, resulting in the transcriptional stimulation of various factors that can accelerate angiogenesis and neuroprotection (Freeman and Barone, 2005; Mu et al., 2003). Vascular endothelial growth factor (VEGF), known as a key factor favoring angiogenesis, is one of the targets of HIF-1α (Li et al., 2007a; Manoonkitiwongsa et al., 2006). Basic fibroblast growth factor (bFGF) is also an important neurotrophic factor that supports angiogenesis (Guo et al., 2006; Issa et al., 2005), whose induction during hypoxia is also HIF-1α dependent (Calvani et al., 2006).

    View all citing articles on Scopus
    View full text