Vascular endothelial growth factor of the lung: friend or foe

https://doi.org/10.1016/j.coph.2008.03.003Get rights and content

The discovery of vascular endothelial growth factor (VEGF) changed the field of angiogenesis. We have learned that VEGF has broader actions than merely a driver of tumor angiogenesis, particularly that VEGF controlled several fundamental functions and properties of endothelial cells and nonendothelial cells. The lung is one of the main organs where VEGF controls several crucial physiological functions. These actions rely on tightly regulated temporal and concentration gradients of VEGF and VEGF receptor expression in the lung. Excessive or diminished VEGF have been linked to abnormal lung phenotypes and, in humans, linked to several diseases. The beneficial and detrimental actions of VEGF underscore that therapeutic targeting of VEGF in disease has to carefully consider the lung biology of VEGF.

Introduction

VEGF-A, also known as vascular permeability factor (VPF) [1••], plays a fundamental role in physiological and pathophysiological forms of angiogenesis and regulation of endothelial cell differentiation (Figure 1). In contrast to fibroblast growth factor, which requires cell damage or basement membrane proteolysis for its release and binding to multiple cell targets, vascular endothelial growth factor (VEGF) is actively secreted and has high specificity for endothelial cells. The lung contains the highest level of transcripts [2] amongst a wide range of organs that express VEGF. VEGF is necessary for the formation of vascular beds of several organs during embryo development, as demonstrated by the lethality of VEGF knockout mice and abnormal vasculogenesis of the heart and large vessels with the loss of only a single copy of the VEGF gene [3••]. VEGF contributes to endothelial cell nitric oxide (NO) production in coronary arteries and cultured umbilical vein endothelial cells [4]. The increase in endothelial cell NO synthase activity relies on the activation of Src and MAP kinase [5] and the PI3K/Akt pathway [6]. VEGF has an anti-inflammatory action, decreasing leukocyte adhesion in an NO-dependent manner [7].

VEGF prevents death of endothelial cells, both in vitro and in animal models of oxygen-mediated retinopathy [8]. VEGF-dependent survival of endothelial cells relies on the activation of PI3 kinase, Akt, and Src [5]. The discovery of the parent VEGF-A molecule led to the subsequent identification of the subforms B–E. In this review, we will focus on VEGF-A (designated hereafter as VEGF).

VEGF binds to two tyrosine-kinase receptors present on endothelial cells, Flt or VEGF receptor 1 (VEGFR-1) and KDR or VEGFR-2. Dominant negative mutant forms of VEGFR-2 can abrogate VEGF-induced signal transduction in vitro and reduce blood vessel proliferation in vivo models of brain tumors [9]. VEGFR-1 binds VEGF with approximately 10-fold higher affinity than KDR [10], undergoes receptor autophosphorylation, and stimulates Ca++ influx. VEGF binding to VEGFR-2 results in cell ruffling, mitosis, chemotaxis, and actin rearrangement [11]. VEGFR-2 undergoes autophosphorylation more efficiently than receptor 1 upon ligand binding. Inhibition of VEGFR-2 blocks proliferation of cultured umbilical vein endothelial cells, in vivo angiogenesis, and vascular permeability [12]. VEGFR-2 is also stimulated in an autocrine manner by endothelial VEGF, which was recently found to be essential for endothelial survival [13]. VEGFR-1 plays a role in the organization of development of embryonic blood vessels [14] and in enhanced monocyte adhesion to endothelial cells [15]. There is the potential for crossinteraction of both VEGFRs as they are approximately 70% homologous and possibly heterodimerize in vivo. VEGFR-1 might act as a silent receptor for VEGF since it has a poor kinase activity. However, its downstream cell signaling remains poorly delineated [16]. It may also serve as a decoy for VEGF [17], as documented by the excess numbers of endothelial cells in amniotic membrane vessels of embryonic bodies lacking VEGFR-1 in the presence of intact VEGFR-2 [18]. On the contrary, VEGFR-1 enhances VEGF-induced VEGFR-2 signaling during abnormal angiogenesis, because it prevents endothelial cell apoptosis. As there are no conditional knockouts of VEGFR-1 and VEGFR-2, loss-of-function experiments have relied on neutralizing antibodies (such as DC101 against VEGFR-2 and MF1 against VEGFR-1), soluble chimeric molecules with the ligand-binding domain of VEGFR-1 (VEGF traps), or chemical inhibition with small-molecule inhibitors such as SU5416, which prevents VEGF-induced phosphorylation of VEGFR-2 [12] and, subsequently shown, to also block VEGFR-1 [19] (Figure 1).

Section snippets

Role as a crucial lung endothelial cell morphogenetic and maintenance factor

Lung morphogenesis requires the continuous physical and molecular interaction between the mesenchymal stroma and epithelial elements [20]. During airway growth, the lung progressively acquires a rich blood supply through the growth of endothelial cells and vascular cells in the pulmonary mesenchyme. This growth is paralleled by the expression of VEGF and its receptors [21], which play central morphogenetic functions throughout fetal lung maturation. Lung VEGF is synthesized by alveolar

Role in acute lung injury (ALI)

ALI consists of an acute clinical syndrome caused by alveolar leakage of plasma proteins, alveolar epithelial cell necrosis, scattered infiltration by neutrophils, and characteristic hyaline membranes, which impair oxygen diffusion, leading to hypoxemia. This syndrome can be caused by a variety of fulminant events, including sepsis, extensive trauma, oxygen or drug toxicity, viral infections, blood transfusions, and pancreatitis, among others. Its idiopathic form is known as acute interstitial

Conclusion

The lung's requirement on VEGF is more complex than that documented with most organs. For the past 13 years, we have learned that VEGF is both a friend and a foe. Too much or too little of VEGF is catastrophic to the lung  an observation that is shared with manipulations of other highly important molecules. However, the final verdict for VEGF still awaits clear therapeutic interventions, which will fulfill the central elements of Koch's postulate on the role of VEGF in lung diseases. At the

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

Grant support: this work has been supported by the CMREF Center for Tissue Processing Center of the Pulmonary Hypertension Breakthrough Initiative, P150 HL 084946-01 (Pathology Core and Project 5), and R01HL 66554 to RMT.

References (59)

  • B.N. Feltis et al.

    Effects of inhaled fluticasone on angiogenesis and vascular endothelial growth factor in asthma

    Thorax

    (2007)
  • R.M. Tuder et al.

    Expression of angiogenesis-related molecules in plexiform lesions in severe pulmonary hypertension: evidence for a process of disordered angiogenesis

    J Pathol

    (2001)
  • J.L. Wright et al.

    Pulmonary hypertension in chronic obstructive pulmonary disease: current theories of pathogenesis and their implications for treatment

    Thorax

    (2005)
  • D.R. Senger et al.

    Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid

    Science

    (1983)
  • W.T. Monacci et al.

    Expression of vascular permeability factor/vascular endothelial growth factor in normal rat tissues

    Am J Physiol

    (1993)
  • P. Carmeliet et al.

    Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele

    Nature

    (1996)
  • Y. Tsurumi et al.

    Reciprocal relation between VEGF and NO in the regulation of endothelial integrity

    Nat Med

    (1997)
  • H. He et al.

    Vascular endothelial growth factor signals endothelial cell production of nitric oxide and prostacyclin through flk-1/KDR activation of c-Src

    J Biol Chem

    (1999)
  • Y. Chen et al.

    Mechanisms of activation of eNOS by 20-HETE and VEGF in bovine pulmonary artery endothelial cells

    Am J Physiol Lung Cell Mol Physiol

    (2006)
  • T. Alon et al.

    Vascular endothelial growth factor acts as a survival factor for newly formed retinal vessels and has implications for retinopathy of prematurity

    Nat Med

    (1995)
  • B. Millauer et al.

    Glioblastoma growth inhibited in vivo by a dominant-negative Flk-1 mutant

    Nature

    (1994)
  • N. Vaisman et al.

    Characterization of the receptors for vascular endothelial growth factor

    J Biol Chem

    (1990)
  • T.P. Quinn et al.

    Fetal liver kinase 1 is a receptor for vascular endothelial growth factor and is selectively expressed in vascular endothelium

    Proc Natl Acad Sci U S A

    (1993)
  • S. Lee et al.

    Autocrine VEGF signaling is required for vascular homeostasis

    Cell

    (2007)
  • F. Shalaby et al.

    Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice

    Nature

    (1995)
  • J. Waltenberger et al.

    Different signal transduction properties of KDR and Flt1, two receptors for vascular endothelial growth factor

    J Biol Chem

    (1994)
  • J.M. Wood et al.

    PTK787/ZK 222584, a novel and potent inhibitor of vascular endothelial growth factor receptor tyrosine kinases, impairs vascular endothelial growth factor-induced responses and tumor growth after oral administration

    Cancer Res

    (2000)
  • M.J. Acarregui et al.

    Vascular endothelial growth factor gene expression in human fetal lung in vivo

    Am J Respir Cell Mol Biol

    (1999)
  • M. Mura et al.

    Vascular endothelial growth factor and related molecules in acute lung injury

    J Appl Physiol

    (2004)
  • Cited by (64)

    • Exhaled nitric oxide and vascular endothelial growth factor as predictors of cold symptoms after stress

      2018, Biological Psychology
      Citation Excerpt :

      However, it should be noted in this context that prior exposure has not been shown to be a critical protective factor in experimental rhinovirus infections (Cohen et al., 1991; Pedersen et al., 2010). VEGF has been shown to have multiple roles in health and disease and both beneficial and detrimental effects have been outlined (Tuder & Yun, 2008) depending on its concentrations in various tissues. With relevance to this study, VEGF also orchestrates the migration of immune cells to mucosa and thereby strengthens the immune defense (Puxeddu, Ribatti, Crivellato, & Levi-Schaffer, 2005; Zhang et al., 2010) and, on the other hand, increases membrane permeability, thus facilitating the penetration and translocation of pathogens (Söderholm et al., 2002).

    • Angiogenesis and pulmonary fibrosis

      2023, Zhonghua Jiehe he Huxi Zazhi / Chinese Journal of Tuberculosis and Respiratory Diseases
    View all citing articles on Scopus
    View full text