Elsevier

Microvascular Research

Volume 83, Issue 1, January 2012, Pages 64-70
Microvascular Research

Focal adhesion kinase regulation of neovascularization

https://doi.org/10.1016/j.mvr.2011.05.002Get rights and content

Abstract

In this review, we discuss the role of focal adhesion kinase (FAK), an intracellular tyrosine kinase, in endothelial cells in relation to neovascularization. Genetic and in vitro studies have identified critical factors, receptor systems, and their intracellular signaling components that regulate the neovasculogenic phenotypes of endothelial cells. Among these factors, FAK appears to regulate several aspects of endothelial cellular behavior, including migration, survival, cytoskeletal organization, as well as cell proliferation. Upon adhesion of endothelial cells to extracellular matrix (ECM) ligands, integrins cluster on the plane of plasma-membrane, while cytoplasmic domains of integrins interact with cytoskeletal proteins and signaling molecules including FAK. However, FAK not only serves as a critical component of integrin signaling, but is also a downstream element of the VEGF/VEGF–receptor and other ligand–receptor systems that regulate neovascularization. A complete understanding of FAK-mediated neovascularization, therefore, should address the molecular and cellular mechanisms that regulate the biology of FAK. Continued research on FAK may, therefore, yield novel therapies to improve treatment modalities for the pathological neovascularization associated with diseases.

Research highlights

► Focal adhesion kinase (FAK) is an intracellular tyrosine kinase that primarily localizes to focal adhesion structures. ► Understanding the biology of FAK in the regulation of angiogenesis is a significant topic. ► FAK appears to regulate multiple aspects of angiogenesis. ► Here we review current literatures as they relate to pathophysiology of angiogenesis.

Introduction

In mammals, the growth of blood vessels occurs mainly by vasculogenesis, angiogenesis, and lymphangiogenesis, together can be termed as neovascularization (Sabin, 1917, Sabine, 1920, Ausprunk and Folkman, 1997, Risau, 1997, Hanahan and Folkman, 1996). Neovascularization is a fundamental biological process that plays a critical role during the development and growth of an organism, and it is required for wound healing and tissue repair in adults (Ausprunk and Folkman, 1997, Hanahan and Folkman, 1996, Hanahan and Weinberg, 2000, Veikkola et al., 2000, Carmeliet, 2003). Angiogenesis is defined as the branching and spreading out of capillaries that are formed from a preformed vessels, primarily through the migration, recruitment, interconnection and lumenization of endothelial cells (Lubarsky and Krasnow, 2003, Kamei et al., 2006, Iruela-Arispe and Davis, 2009). Nevertheless, unwanted angiogenesis has been associated with the expansion of atherosclerotic lesions, diabetic retinopathy, psoriasis, and tumor progression (Wary, 2004, Kalluri et al., 2003, Carmeliet, 2003, Renault and Losordo, 2007, Adams and Alitalo, 2007). On the other hand, ischemic and cardiovascular diseases have been associated with insufficient angiogenesis (Renault and Losordo, 2007, Adams and Alitalo, 2007, Becker and D'Amato, 2007, Fukumura and Jain, 2007, Aird, 2008).

The vascular tree develops early in embryogenesis to nourish and provide oxygen to the developing organism (Risau, 1997, Watson and Cross, 2005, Robb and Elefanty, 1998, Hickey and Simon, 2006). In the mouse at embryonic day 7.5 (E7.5), the extraembryonic mesodermal cells of the yolk sac aggregate into clusters, representing the initial stage of blood island formation and hemoglobin accumulation (Sabine, 1920, Robb and Elefanty, 1998, Hickey and Simon, 2006, Palis et al., 1995, Gerber et al., 2002, Rossant and Hirashima, 2003, Coultas et al., 2005, Hasegawa et al., 2007). Soon after this stage, blood islands differentiate into an external layer of endothelial cells and an inner core of blood cells (Sabine, 1920, Risau, 1997). Simultaneously, embryonic cells of the proximal lateral mesoderm assemble into pre-endocardial tubes that connect anteriorly to generate cardiac endocardium around the anterior intestinal portal (Palis et al., 1995, Gerber et al., 2002, Rossant and Hirashima, 2003, Coultas et al., 2005). Posteriorly, angioblasts organize into paired dorsal aortas, which later assemble in the midline to form a single tube. Correspondingly, mesodermal cells in the allantois generate cords that produce the umbilical blood vessels (Gerber et al., 2002, Rossant and Hirashima, 2003, Coultas et al., 2005, Hasegawa et al., 2007). The allantois develops rapidly inside the exocoelomic cavity and fuses with the chorionic extraembryonic mesoderm (Palis et al., 1995, Gerber et al., 2002, Rossant and Hirashima, 2003, Coultas et al., 2005, Hasegawa et al., 2007). This fusion results in a signal that initiates the differentiation of the chorionic vasculature, which indirectly connects to the maternal vascular system of the placenta. These early events are described as “vasculogenesis”, which signifies the de novo formation of blood vessels from angiogenic precursor cells. In the mouse, vasculogenesis is usually complete by E7.5 days. Later in embryogenesis, the vascular tree grows by the sprouting, cell division, migration and assembly of endothelial cells derived from pre-existing vessels through a process termed angiogenesis. At the cellular level, Brachyury acts as a switch to induce formation of the mesodermal lineage. Mesodermally derived mesenchymal stem cells give rise to hemangioblasts (Fig. 1). Hemangioblasts are bipotential stem cells, which are characterized as CD133+/CD34+ and fetal liver kinase-1 (Flk1+) cells that can give rise to hematopoietic cells (HSCs) and angioblasts. The vascular tree grows by sprouting endothelial cells derived from angioblasts. Concurrent with VEGFR activation, cell–cell and cell–matrix interactions provide positional information related to when endothelial cells will elongate, interconnect, migrate, divide, and assemble into tube-like structures (Risau, 1997, Rossant and Hirashima, 2003, Coultas et al., 2005, Hasegawa et al., 2007). More recently, a subset of VE-cadherin+ and Runx-1+ endothelial cells, derived from dorsal aorta have been termed as “hemogenic endothelial cells”. Hemogenic endothelial cells originating from dorsal aorta between E8.5 and E11.5 days can give rise to hematopoietic cells that directly home into the liver (Chen et al., 2009, Swiers et al., 2010). Nevertheless, productive neovascularization requires not only endothelial cells, but also hematopoietic cells and the recruitment of pericytes and smooth muscle cells.

In vitro transgenic and knockout studies have provided evidence that vascular endothelial growth factor (VEGF) and the VEGF receptor-2 system (also known as fetal liver kinase (Flk-1) and kinase domain receptor (KDR)) play fundamental roles in the formation of blood vessels (Shima et al., 1996, Shalaby et al., 1997, Park et al., 2004). Flk-1 is one of earliest markers of endothelial cells and is first detected during mouse development in the extra-embryonic mesoderm of the yolk sac, where the blood islands form shortly after gastrulation, around day 7 postcoitum (Shima et al., 1996, Shalaby et al., 1997, Park et al., 2004, Lugus et al., 2009, Hellström et al., 2007). In the extraembryonic mesoderm, flk-1 appears shortly after gastrulation, followed sequentially by tie-2 and tie-1 over the next 24 h. In addition, flk-1 is initially observed in the extraembryonic mesoderm prior to its expression in the embryonic mesoderm (Shima et al., 1996, Shalaby et al., 1997, Park et al., 2004). Activation of the VEGFR-2/Flk1 receptor promotes endothelial elongation, motility and proliferation. Specifically, the activation of endothelial cells by VEGF induces expression of Delta like ligand-4 (Dll4) by one of the endothelial cells, which is referred to as the “tip cell”. Tip cells display a highly motile phenotype. Cell surface associated Dll4 expressed by the tip cell then ligate Notch receptor on the adjacent endothelial cell to induce intracellular signaling to activate the Hes and Hey transcriptional target genes (Siekmann and Lawson, 2007, Hofmann and Iruela-Arispe, 2007, Nakatsu et al., 2003, Benedito et al., 2009, Kume, 2009, Jakobsson et al., 2010). Activation of Hes and Hey promotes cell proliferation and these subsets of proliferating cells are termed as the “stalk cells” (Siekmann and Lawson, 2007, Hofmann and Iruela-Arispe, 2007, Nakatsu et al., 2003, Benedito et al., 2009, Kume, 2009, Jakobsson et al., 2010). Proliferative cells at some point receive a cell cycle “arrest” signal which induces morphogenic differentiation, lumenization and tube formation (Hofmann and Iruela-Arispe, 2007, Nakatsu et al., 2003, Benedito et al., 2009, Kume, 2009, Iruela-Arispe and Davis, 2009, Jakobsson et al., 2010).

All of these processes may involve FAK signaling, either directly or indirectly, in the context of key molecules and pathways controlling cell migration, VEGF production, cell proliferation and the formation of new vessels. Below, we review and discuss the structure, expression, and FAK regulation of vasculogenesis and angiogenesis in developmental settings and under pathological conditions.

Section snippets

Regulation of FAK expression

It became clear that upon transformation of fibroblast cells with v-Src, an intracellular tyrosine kinase, v-Src phosphorylated several intracellular proteins in these cells. In the early 1990s, several laboratories generated monoclonal antibodies using proteins derived from v-Src-transformed cells. By screening a chicken embryo cDNA expression library with such an antibody raised by J.T. Parsons' group, an intracellular tyrosine kinase protein, designated p125, was cloned (Schaller et al., 1992

FAK signaling in cell migration and angiogenesis

Structurally, FAK contains a N-terminal FERM domain, a central tyrosine kinase domain, proline-rich (PR) regions, and a focal adhesion-targeting (FAT) domain at the C-terminus (Fig. 2B). Schaller et al. (1994) provided evidence on how FAK mediates integrin signaling. There are two major tyrosine phosphorylation sites, Y397 and Y925, that are central to FAK functioning (Schaller et al., 1993, Schaller et al., 1994) and serve to recruit key signaling and adaptor molecules. For example, Y397

Role of FAK in endothelial cell proliferation and differentiation

Endothelial cell proliferation is one of the defining hallmarks of angiogenesis. However, mature endothelial cells have also been known to exhibit limited turnover, i.e., these cells have a finite proliferative capacity. Although primary endothelial cells proliferate relatively well in vitro, the proliferation of endothelial cells in vivo is difficult to detect. In particular, the detection of endothelial cell proliferation in developing embryos is technically challenging. In contrast, it is

FAK in tumor angiogenesis

Tumor angiogenesis usually indicates increased tumor growth and metastatic phenotypes. Tumor angiogenesis requires both cell adhesion events and VEGF action. The observations that FAK promotes cell migration and proliferation suggested that FAK could be necessary for tumor angiogenesis. Although the overexpression of FAK in tumor cells has been documented in the literature, constitutive overexpression as a result of gene amplification or somatic mutation of FAK has not been observed or linked

Conclusions

Our discussion indicates that FAK is a multifunctional protein in that it regulates non-genomic responses, such as cell migration, cytoskeletal organization, and cell spreading, and genomic responses, including the cell cycle and proliferation, via different mechanisms. The role of FAK in the regulation of cell migration has been studied in vitro and in vivo. Because of the embryonic lethality associated with Fak-knockout mice, the role of FAK in vasculogenesis and angiogenesis is still not

Sources of funding

The authors were supported by the National Institutes of Health (R01HL079356; HL079356-03S1), American Heart Association (GRNT4520014) grants and the University of Illinois at Chicago (UIC) Center for Clinical and Translational Science (CCTS) Award Number UL1RR029879 from the National Center for Research Resources to K.K.W. E.E.K. was supported by T32GM070388 and T32HL072742 NIH training grants.

Competing interest statement

The authors declare no competing financial interests.

Acknowledgments

We would like to apologize for not being able to accommodate many references of our colleagues who have contributed to the understanding of FAK function.

References (73)

  • J.J. Lugus

    Both primitive and definitive blood cells are derived from Flk-1+ mesoderm

    Blood

    (2009)
  • Y. Maru

    The tubulogenic activity associated with an activated form of Flt-1 kinase is dependent on focal adhesion kinase

    Biochim. Biophys. Acta

    (2001)
  • M.N. Nakatsu

    Angiogenic sprouting and capillary lumen formation modeled by human umbilical vein endothelial cells (HUVEC) in fibrin gels: the role of fibroblasts and Angiopoietin-1

    Microvasc. Res.

    (2003)
  • J. Palis

    Initiation of hematopoiesis and vasculogenesis in murine yolk sac explants

    Blood

    (1995)
  • L.K. Phng

    Nrarp coordinates endothelial Notch and Wnt signaling to control vessel density in angiogenesis

    Dev. Cell

    (2009)
  • M.A. Renault et al.

    Therapeutic myocardial angiogenesis

    Microvasc. Res.

    (2007)
  • J. Rossant et al.

    Vascular development and patterning: making the right choices

    Curr. Opin. Genet. Dev.

    (2003)
  • M.D. Schaller

    Biochemical signals and biological responses elicited by the focal adhesion kinase

    BBA

    (2001)
  • F. Shalaby

    A requirement for Flk1 in primitive and definitive hematopoiesis and vasculogenesis

    Cell

    (1997)
  • L.M. Shaw

    Activation of phosphoinositide 3-OH kinase by the alpha6beta4 integrin promotes carcinoma invasion

    Cell

    (1997)
  • D.T. Shima

    The mouse gene for vascular endothelial growth factor. Genomic structure, definition of the transcriptional unit, and characterization of transcriptional and post-transcriptional regulatory sequences

    J. Biol. Chem.

    (1996)
  • K.K. Wary

    The adaptor protein Shc couples a class of integrins to the control of cell cycle progression

    Cell

    (1996)
  • K.K. Wary

    A requirement for caveolin-1 and associated kinase Fyn in integrin signaling and anchorage-dependent cell growth

    Cell

    (1998)
  • R.H. Adams et al.

    Molecular regulation of angiogenesis and lymphangiogenesis

    Nat. Rev. Mol. Cell Biol.

    (2007)
  • W.C. Aird

    Endothelium in health and disease

    Pharmacol. Rep.

    (2008)
  • D.H. Ausprunk et al.

    Migration and proliferation of endothelial cells in preformed and newly formed blood vessels during tumor angiogenesis

    Microvasc. Res.

    (1997)
  • R. Braren

    Endothelial FAK is essential for vascular network stability, cell survival, and lamellipodial formation

    J. Cell Biol.

    (2006)
  • P. Carmeliet

    Angiogenesis in health and disease

    Nat. Med.

    (2003)
  • L.A. Cary et al.

    Focal adhesion kinase in integrin-mediated signaling

    Front. Biosci.

    (1999)
  • M.J. Chen

    Runx1 is required for the endothelial to haematopoietic cell transition but not thereafter

    Nature

    (2009)
  • L. Coultas

    Endothelial cells and VEGF in vascular development

    Nature

    (2005)
  • J.M. Enciso

    Endothelial cell migration during murine yolk sac vascular remodeling occurs by means of a Rac1 and FAK activation pathway in vivo

    Dev. Dyn.

    (2010)
  • H.P. Gerber

    VEGF regulates haematopoietic stem cell survival by an internal autocrine loop mechanism

    Nature

    (2002)
  • T. Hasegawa

    The embryonic human choriocapillaris develops by hemo-vasculogenesis

    Dev. Dyn.

    (2007)
  • H. Haskell

    Focal adhesion kinase is expressed in the angiogenic blood vessels of malignant astrocytic tumors in vivo and promotes capillary tube formation of brain microvascular endothelial cells

    Clin. Cancer Res.

    (2003)
  • M. Hellström

    Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis

    Nature

    (2007)
  • Cited by (23)

    • Curcumolide, a unique sesquiterpenoid from Curcuma wenyujin displays anti-angiogenic activity and attenuates ischemia-induced retinal neovascularization

      2019, Phytomedicine
      Citation Excerpt :

      VEGF stimulation leads to receptor dimerization and autophosphorylation (Abhinand et al., 2016). These events subsequently initiate intracellular signal cascades that activate various signal molecules, including Src family kinase(Werdich and Penn, 2006), focal adhesion kinase (FAK) (Wary et al., 2012), extracellular signal-regulated kinases (ERKs) (Kanda et al., 2012), phosphoinositide 3- kinase (PI3K), protein kinase B (PKB/AKT), mammalian target of rapamycin (mTOR), and ribosomal protein S6 kinase (p70S6K) (Sasore et al., 2014). These, among other signal molecules, are involved in the regulation of cell proliferation, migration, and survival, as well as angiogenesis and microvascular permeability as in PDR (Hu et al., 2017).

    • Spinal Cord Injury and Regeneration: A Critical Evaluation of Current and Future Therapeutic Strategies

      2014, Pathobiology of Human Disease: A Dynamic Encyclopedia of Disease Mechanisms
    • Paxillin regulates pulmonary arterial smooth muscle cell function in pulmonary hypertension

      2012, American Journal of Pathology
      Citation Excerpt :

      Hypoxia-induced binding of HIF-1α to the HRE in the paxillin promoter was verified by chromatin immunoprecipitation (ChIP) (Figure 7H, line 3). Cell adhesion, migration, and proliferation are important hallmarks of vascular remodeling, which underlies the development of PH.4 A growing body of evidence supports the involvement of cytoskeletal proteins in these processes.32–34 However, the contribution of cytoskeletal proteins to the pathogenesis of PH is poorly understood.

    View all citing articles on Scopus
    View full text