Elsevier

Experimental Cell Research

Volume 312, Issue 5, 10 March 2006, Pages 584-593
Experimental Cell Research

Review Article
Neuropilins in neoplasms: Expression, regulation, and function

https://doi.org/10.1016/j.yexcr.2005.11.024Get rights and content

Abstract

Neuropilins (NRP) are membranous receptors capable of binding two disparate ligands, class 3 semaphorins (SEMA) and vascular endothelial growth factors (VEGF), and regulating two diverse systems, neuronal guidance and angiogenesis. The neuropilin genes, NRP1 and NRP2, share similar protein structure, but differ in their expression patterns, regulation, and ligand-binding specificities. NRPs vary in their expression patterns; for example, endothelial cells express both NRP1 and NRP2, lymphatic endothelial cells predominantly express NRP2, and epidermal cells predominantly express NRP1. NRP expression can be differentially regulated by transcription factors, e.g. prox-1 induces NRP2 while suppressing NRP1, or by growth factors, e.g. epidermal growth factor (EGF) induces NRP1 but not NRP2. Nearly all tumor cells express NRP1, NRP2, or both. Carcinomas express NRP1, whereas neuronal tumors and melanomas predominantly express NRP2. SEMAs play a role in neoplasms as angiogenesis inhibitors. For example, SEMA3F, which binds specifically to NRP2, inhibits tumor angiogenesis and metastasis. Metastatic tumor cells lose SEMA3F expression during progression. Therefore, SEMA3F may have therapeutic potential. This article focuses on the role of NRPs and SEMAs in tumor progression and angiogenesis.

Introduction

Neuropilin (NRP) is a 130- to 140-kDa transmembrane glycoprotein. Fujisawa and colleges first identified NRP in 1987 as the antigen to a monoclonal antibody named A5 that specifically bound to the superficial neuropile in the Xenopus laevis optic tectum [1]. Subsequently, NRP was found to be a receptor for the class 3 semaphorins (SEMA3), a family of chemorepulsive guidance molecules capable of collapsing axonal growth cones and repelling axons of ganglia. NRP2 was discovered in 1997 [2], [3]. The two neuropilin genes, NRP1 and NRP2, share 45% homology and map to different chromosomes [4]. In humans, NRP1 is located on chromosome 10, and NRP2 is located on chromosome 2 [4]. Both genes are composed of 17 exons, but NRP2 is expressed as several alternatively spliced transcripts [4]. Both NRPs have similar overall structure but differ significantly in their expression patterns, regulation, and ligand binding specificities. The extracellular regions of NRPs are composed of separate subdomains: the a1a2 domain is involved in SEMA3-binding, the b1b2 domain is involved in both SEMA3- and vascular endothelial growth factor (VEGF)-binding (discussed in more detail below), and the c domain is involved in dimerization [1], [5]. The cytoplasmic domain of either NRP is relatively short, consisting of only 40 amino acids, and contains no known signaling motif. NRP1 and NRP2 sequences end in the same three C-terminal amino acids, SEA. This region has been shown to bind to GIPC (GAIP interacting protein at the C terminus) [6]. GIPC, which is also called NIP (neuropilin interacting protein), is a 40 kDa protein containing a PDZ domain that interacts with RGS-GAIP, a GTPase-activating protein (GAP) for Gαi subunits. The signaling function of GIPC in response to NRP ligands has not been elucidated [6]. In addition to the transmembrane forms, neuropilins also exist as soluble isoforms containing only the extracellular ligand binding a1a2 and b1b2 subdomains but lacking the c, transmembrane, and cytoplasmic domains [4], [7]. Premature truncation within introns results in these smaller isoforms that contain C-terminal intron-derived amino acids. To date, four soluble neuropilin-1 (sNRP1) species and one sNRP2 species have been reported [8], [9]. These sNRPs can function as competitive antagonists for NRP ligands, for example, VEGF [5], [7].

NRPs are receptors for two different ligand families, the SEMA family of axonal guidance regulators [3] and the VEGF family of angiogenesis factors [4]. The semaphorins are divided into seven classes consisting of transmembrane proteins, glycosylphosphatidylinosotol (GIP)-linked proteins, as well as soluble proteins (class 3). Semaphorin 3A (SEMA3A, human protein; Sema3A, murine or chick protein; originally called collapsin-1) [10] was the first NRP1 ligand identified [3], [11]. The class 3 semaphorin family is comprised of 6 secreted proteins, SEMA3A, B, C, D, E, and F. Although the ligand binding domains of NRP1 and NRP2 are similar, there is a degree of specificity in SEMA/NRP binding and activity. SEMA3A binds NRP1; SEMA3F binds NRP2; whereas SEMA3B binds both NRP1 and NRP2 [12]. Sensory neurons such as dorsal root ganglia (DRG) express NRP1 and bind SEMA3A, resulting in functional collapse of axonal growth cones [1]. SEMA3F binds NRP2 and repels sympathetic neurons such as superior cervical ganglia (SCG) [13], [25]. The “sema”-domain of Sema3A binds to the a1a2 domain of NRP1, and the carboxy-terminal C-domain of Sema3A binds the b1b2 domain of NRP1 [14]. The Sema3A protein dimerizes via a cysteine at residue 723, between the Ig domain and basic tail. Only dimerized Sema3A protein can functionally collapse axonal growth cones [15]. The sema-domain alone is inactive since it cannot dimerize. In the neuronal guidance pathway, NRPs act as co-receptors with plexins, transmembrane receptors that transduce the semaphorin signal [16]. Recently, Plexin-A4 has been found to signal via NRP1, while Plexin-A3 signals via NRP2 [17].

In 1998, NRP1 was identified as a functional receptor for VEGF165 [18]. Exon 7 of VEGF165 binds to the b1b2 domain of NRP1. VEGF121 has no exon 7-domain and therefore cannot bind NRP1 [18], [19]. Both NRP1 and NRP2 bind VEGF165, but differ in their binding affinities to other family members. For example, NRP1 binds VEGF-B, VEGF-E, and placenta growth factor-2 (PlGF2, a splice isoform of PlGF), whereas NRP2 binds VEGF145 and VEGF-C [4]. In the angiogenesis pathway, NRP1 functions as a co-receptor with VEGFR-2 that increases the binding of VEGF165 to VEGFR-2 and that enhancesVEGF165-mediated chemotaxis of EC [18], [20]. NRP1 also regulates VEGF-mediated permeability in cultured EC [21]. Transgenic mice overexpressing sNRP1 in the skin have decreased vascular permeability [22]. VEGF and SEMA3A have overlapping binding domains on NRP1 and thus are competitive inhibitors of one another [4]. The fact that these two disparate ligand families can bind to the same receptor and yet mediate two different processes, neuronal guidance and angiogenesis, suggests common molecular mechanisms in these processes. These results have sparked new areas of investigation and raise the question of whether other neuronal guidance mediators may affect angiogenesis [23], [24].

Section snippets

Neuropilin expression patterns

Although NRPs initially received their name from their neuronal localization, it is now known that NRPs are expressed in adult human organs [18] by a variety of tissue types [7]. NRP1 and NRP2 have specific and overlapping expression patterns in several tissues. For example, both sensory and sympathetic neurons express NRP1, making them susceptible to the repulsive effects of SEMA3A, whereas NRP2 is found only on sympathetic, but not on sensory neurons [25]. As a consequence, only sympathetic

Concluding remarks

In summary, semaphorins and neuropilins play a dual role as mediators of both neuronal guidance and angiogenesis. The regulation of NRP1 and NRP2 is complex and independently mediated. NRP1 and NRP2 are expressed in different tissues in a largely non-overlapping pattern. Carcinomas typically overexpress NRP1, whereas neuronal tumors largely express NRP2. NRPs can promote angiogenesis through their VEGF ligands or inhibit angiogenesis through their SEMA ligands. The NRP expression pattern in

Acknowledgments

This work was supported by NIH grants CA37392 and CA45548 (M. Klagsbrun); the SPORE in Skin Cancer Developmental Project Award (NIH, Department of Dermatology, Brigham and Women's Hospital, Boston, MA) (D. R. Bielenberg) and Grants-in-Aid for Scientific Research from the Japanese Ministry of Education, Culture, Sports, Science, and Technology (S. Takashima). We thank Dr. Akio Shimizu for PCR primers and Dr. Jay Harper for the murine ISH probe. We thank Dr. Isaiah Fidler for providing cell

References (83)

  • M. Klagsbrun et al.

    A role for axon guidance receptors and ligands in blood vessel development and tumor angiogenesis

    Cytokine Growth Factor Rev.

    (2005)
  • A. Eichmann et al.

    Guidance of vascular and neural network formation

    Curr. Opin. Neurobiol.

    (2005)
  • H. Chen et al.

    Semaphorin-neuropilin interactions underlying sympathetic axon responses to class III semaphorins

    Neuron

    (1998)
  • Y. Herzog et al.

    Differential expression of neuropilin-1 and neuropilin-2 in arteries and veins

    Mech. Dev.

    (2001)
  • S. Kwei et al.

    Early adaptive responses of the vascular wall during venous arterialization in mice

    Am. J. Pathol.

    (2004)
  • H. Fujita et al.

    Expressions of neuropilin-1, neuropilin-2 and semaphorin 3A mRNA in the rat brain after middle cerebral artery occlusion

    Brain Res.

    (2001)
  • E. Giraudo et al.

    Tumor necrosis factor-alpha regulates expression of vascular endothelial growth factor receptor-2 and of its co-receptor neuropilin-1 in human vascular endothelial cells

    J. Biol. Chem.

    (1998)
  • A.A. Parikh et al.

    Neuropilin-1 in human colon cancer: expression, regulation, and role in induction of angiogenesis

    Am. J. Pathol.

    (2004)
  • J.M. Coulson

    Transcriptional regulation: cancer, neurons and the REST

    Curr. Biol.

    (2005)
  • D. Watanabe et al.

    Transcription factor Ets-1 mediates ischemia- and vascular endothelial growth factor-dependent retinal neovascularization

    Am. J. Pathol.

    (2004)
  • S.C. Heffelfinger et al.

    Inhibition of VEGFR2 prevents DMBA-induced mammary tumor formation

    Lab. Invest.

    (2004)
  • D. Bates et al.

    Neurovascular congruence results from a shared patterning mechanism that utilizes Semaphorin3A and Neuropilin-1

    Dev. Biol.

    (2003)
  • S. Kusy et al.

    Selective suppression of in vivo tumorigenicity by semaphorin SEMA3F in lung cancer cells

    Neoplasia

    (2005)
  • E. Brambilla et al.

    Semaphorin SEMA3F localization in malignant human lung and cell lines: a suggested role in cell adhesion and cell migration

    Am. J. Pathol.

    (2000)
  • P. Nasarre et al.

    Semaphorin SEMA3F and VEGF have opposing effects on cell attachment and spreading

    Neoplasia

    (2003)
  • Y.K. Hong et al.

    Prox1, master regulator of the lymphatic vasculature phenotype

    Cell Tissue Res.

    (2003)
  • H. Fujisawa

    From the discovery of neuropilin to the determination of its adhesion sites

    Adv. Exp. Med. Biol.

    (2002)
  • M. Klagsbrun et al.

    The role of neuropilin in vascular and tumor biology

    Adv. Exp. Med. Biol.

    (2002)
  • H. Cai et al.

    Cloning and characterization of neuropilin-1-interacting protein: a PSD-95/Dlg/ZO-1 domain-containing protein that interacts with the cytoplasmic domain of neuropilin-1

    J. Neurosci.

    (1999)
  • M.L. Gagnon et al.

    Identification of a natural soluble neuropilin-1 that binds vascular endothelial growth factor: in vivo expression and antitumor activity

    Proc. Natl. Acad. Sci. U. S. A.

    (2000)
  • Unified nomenclature for the semaphorins/collapsins

    Cell

    (1999)
  • A. Bagri et al.

    Neuropilins as Semaphorin receptors: in vivo functions in neuronal cell migration and axon guidance

    Adv. Exp. Med. Biol.

    (2002)
  • R.H. Adams et al.

    The chemorepulsive activity of secreted semaphorins is regulated by furin-dependent proteolytic processing

    EMBO J.

    (1997)
  • A.W. Puschel

    The function of neuropilin/plexin complexes

    Adv. Exp. Med. Biol.

    (2002)
  • S. Soker et al.

    Inhibition of vascular endothelial growth factor (VEGF)-induced endothelial cell proliferation by a peptide corresponding to the exon 7-encoded domain of VEGF165

    J. Biol. Chem.

    (1997)
  • S. Soker et al.

    VEGF165 mediates formation of complexes containing VEGFR-2 and neuropilin-1 that enhance VEGF165-receptor binding

    J. Cell. Biochem.

    (2002)
  • P.M. Becker et al.

    Neuropilin-1 regulates vascular endothelial growth factor-mediated endothelial permeability

    Circ. Res.

    (2005)
  • R. Mamluk et al.

    Soluble neuropilin targeted to the skin inhibits vascular permeability

    Angiogenesis

    (2005)
  • T. Kawasaki et al.

    A requirement for neuropilin-1 in embryonic vessel formation

    Development

    (1999)
  • L. Yuan et al.

    Abnormal lymphatic vessel development in neuropilin 2 mutant mice

    Development

    (2002)
  • F. le Noble et al.

    Flow regulates arterial–venous differentiation in the chick embryo yolk sac

    Development

    (2004)
  • Cited by (0)

    Please see the link to Supplemental Materials on the electronic version of this paper for a complete description of materials and methods.

    View full text