Elsevier

Peptides

Volume 28, Issue 2, February 2007, Pages 405-412
Peptides

Review
Neuropeptide Y (NPY) in neuroblastoma: Effect on growth and vascularization

https://doi.org/10.1016/j.peptides.2006.08.038Get rights and content

Abstract

Neuroblastomas are pediatric tumors of sympathetic origin, expressing neuronal markers, such as NPY and its receptors. Due to this, neuroblastomas are often associated with elevated plasma levels of NPY, which correlates with poor clinical outcome of the disease. This clinical data corroborates the recent discovery of growth-promoting actions of NPY in neuroblastomas. The peptide has been shown to stimulate proliferation of neuroblastoma cells in an autocrine manner and induce tumor vascularization. Since both processes are mediated by the same Y2 and Y5 receptors, targeting this pathway may be a potential bidirectional therapy for these children's tumors.

Introduction

Neuroblastoma cells are commonly used in neuropeptide Y (NPY) research to study the regulation of peptide expression, its receptor signaling and trafficking [37], [60]. The best known neuroblastoma cell lines include SK-N-BE, SHSY5Y, SMS-KAN, SK-N-AS. In contrast, the SK-N-MC cell line, which has been initially described as a neuroblastoma and widely used to characterize NPY-Y1 receptors, in fact belongs to the Ewing's sarcoma family of tumors [7], [8], [38], [59], [64]. Due to this, SK-N-MC cells have a different biology and, consequently, a different NPY receptor pattern than neuroblastoma cells.

The purpose of this review is to show that neuroblastoma cells are not just models to study NPY and its receptors. They are derived from real patients, who often died despite intense treatment, and represent a real disease, which lacks an adequate treatment. Moreover, the review summarizes the evidence that NPY is not only a marker of neuronal differentiation in neuroblastomas, but in fact the NPY system serves as an important autocrine pathway, which sustains tumor growth and, consequently, may be a potential target for new therapies for these pediatric tumors.

Section snippets

Biology of neuroblastoma

Neuroblastoma, together with rhabdosarcoma and Ewing's sarcoma, belongs to a family of aggressive, small blue round cell tumors of childhood. Neuroblastoma is the most common malignant disease of infancy and is responsible for approximately 15% of all childhood cancer deaths [5], [10], [40]. It is one of the most diverse and enigmatic diseases among all known malignancies. The low grade neuroblastoma (stages 1 and 2) is a localized disease, has a good prognosis and does not require intensive

NPY in neuroblastoma

As tumors derived from sympathetic precursors, neuroblastomas synthesize and release various neuronal proteins, such as NPY, which is considered a marker of their neuronal differentiation. Due to this, neuroblastoma is often associated with elevated plasma levels of the peptide, which normalize after treatment [8], [26], [50], [51]. However, the first attempts to use NPY as a general diagnostic marker for neuroblastoma failed due to the high variability of the peptide concentrations. Further

NPY as a growth factor for neuroblastoma cells

The fact that neuroblastoma cells express both NPY and its receptors suggests that the peptide can act here as an autocrine growth factor. Indeed, exogenous NPY stimulates the proliferation of SK-N-BE(2) neuroblastoma cells, which is associated with transient activation of p44/42 MAPK (Fig. 2B) [26]. This effect is mimicked by both Y2 and Y5 agonists and blocked by Y2 and Y5 antagonists, suggesting that both receptors are involved in this process. Interestingly, the NPY receptor antagonists not

Role of NPY in tumor vascularization

Another aspect of NPYs role in neuroblastomas is associated with its angiogenic effect. The angiogenic properties of NPY have been established using variety of models. In vitro, the peptide stimulates endothelial cell proliferation and migration [68]. In vivo, it is involved in the revascularization of ischemic tissues, wound healing and retinopathy [20], [22], [32], [35], [66]. The angiogenic activities of the peptide are severely impaired in Y2−/− mice, indicating that Y2 is the main receptor

NPY in the neuroblastoma microenvironment

Taken together, studies on in vitro and in vivo models indicate that NPY may be an important growth factor for neuroblastoma, which stimulates their growth by two distinct inter-dependent mechanisms: an autocrine effect on neuroblastoma cell proliferation and paracrine angiogenic effect (Fig. 3) [26]. However, to understand the role of NPY in neuroblastoma, it is essential to study its function in the context of the entire tumor microenvironment and establish interactions of the NPY system with

NPY and its receptors as potential targets in neuroblastoma therapy

As described above, NPY is not just a marker of neuronal differentiation in neuroblastoma, but also a stimulator of their growth [26]. The fact that both NPY-induced neuroblastoma cell proliferation and angiogenesis seem to be Y2/Y5 receptor mediated (Fig. 3) suggests that blocking this pathway can be a bi-directional therapy for neuroblastoma, targeting both the growth of tumor cells and the formation of new blood vessels. Neuroblastomas are very well vascularized, angiogenesis-dependent

Future directions

Although the study summarized in this review provided a significant insight into understanding the role of NPY in neuroblastoma, several important questions remain unanswered. For example, is NPY involved in neuroblastoma metastases? In advanced neuroblastomas, elevated NPY plasma levels have been associated with poor outcome of the disease, which correlates with metastases and high tumor vascularization [14], [15], [17], [19], [21], [43]. Moreover, the concentration of NPY is significantly

Summary

In summary, NPY is a growth factor for neuroblastomas, stimulating their growth via a direct effect on tumor cell proliferation and indirectly, via its angiogenic activity. However, further studies are required to fully elucidate the functions of the peptide in a neuroblastoma microenvironment and, based on this, design potential treatment strategies for these children's tumors.

Acknowledgements

The author would like to thank Lydia Kuo for critical reading of the manuscript. This work was supported by the grant from Children's Cancer Foundation (Baltimore, MD) and American Cancer Society Institutional Research Grant IRG-97-152-13 to Joanna Kitlinska.

References (69)

  • C.M. McConville et al.

    Neuroblastoma—a developmental perspective

    Cancer Lett

    (2003)
  • C.A. Minth-Worby

    Transcriptional regulation of the human neuropeptide Y gene by nerve growth factor

    J Biol Chem

    (1994)
  • A. Nakagawara

    Trk receptor tyrosine kinases: a bridge between cancer and neural development

    Cancer Lett

    (2001)
  • A. Nakagawara et al.

    Role of neurotrophins and their receptors in human neuroblastomas: a primary culture study

    Eur J Cancer

    (1997)
  • D. Ribatti et al.

    Antiangiogenic strategies in neuroblastoma

    Cancer Treat Rev

    (2005)
  • A. Schramm et al.

    Biological effects of TrkA and TrkB receptor signaling in neuroblastoma

    Cancer Lett

    (2005)
  • M. Schwab

    MYCN in neuronal tumours

    Cancer Lett

    (2004)
  • S. Sheriff et al.

    NPY upregulates genes containing cyclic AMP response element in human neuroblastoma cell lines bearing Y1 and Y2 receptors: involvement of CREB

    Regul Pept

    (1998)
  • S. Shusterman et al.

    Prospects for therapeutic inhibition of neuroblastoma angiogenesis

    Cancer Lett

    (2005)
  • B. Srinivasan et al.

    Microglia-derived pronerve growth factor promotes photoreceptor cell death via p75 neurotrophin receptor

    J Biol Chem

    (2004)
  • A. Tacconelli et al.

    TrkA alternative splicing: a regulated tumor-promoting switch in human neuroblastoma

    Cancer Cell

    (2004)
  • J. Whang-Peng et al.

    Cytogenetic characterization of selected small round cell tumors of childhood

    Cancer Genet Cytogenet

    (1986)
  • Z. Zukowska-Grojec et al.

    Mechanisms of vascular growth-promoting effects of neuropeptide Y: role of its inducible receptors

    Regul Pept

    (1998)
  • Z. Zukowska-Grojec et al.

    Mitogenic effect of neuropeptide Y in rat vascular smooth muscle cells

    Peptides

    (1993)
  • C. Beltinger et al.

    Murine models for experimental therapy of pediatric solid tumors with poor prognosis

    Int J Cancer

    (2001)
  • G. Bergers et al.

    Combining antiangiogenic agents with metronomic chemotherapy enhances efficacy against late-stage pancreatic islet carcinomas in mice

    Cold Spring Harb Symp Quant Biol

    (2002)
  • J.L. Biedler et al.

    Morphology and growth, tumorigenicity, and cytogenetics of human neuroblastoma cells in continuous culture

    Cancer Res

    (1973)
  • J.L. Biedler et al.

    Multiple neurotransmitter synthesis by human neuroblastoma cell lines and clones

    Cancer Res

    (1978)
  • P. Bjellerup et al.

    Limited neuropeptide Y precursor processing in unfavourable metastatic neuroblastoma tumours

    Br J Cancer

    (2000)
  • G.M. Brodeur

    Neuroblastoma: biological insights into a clinical enigma

    Nat Rev Cancer

    (2003)
  • G.M. Brodeur et al.

    Expression of TrkA, TrkB and TrkC in human neuroblastomas

    J Neurooncol

    (1997)
  • T. Browder et al.

    Antiangiogenic scheduling of chemotherapy improves efficacy against experimental drug-resistant cancer

    Cancer Res

    (2000)
  • J. Carnahan et al.

    Regulation of neuropeptide expression in the brain by neurotrophins, Potential role in vivo

    Mol Neurobiol

    (1995)
  • P.S. Cohen et al.

    Neuropeptide Y expression in the developing adrenal gland and in childhood neuroblastoma tumors

    Cancer Res

    (1990)
  • Cited by (0)

    View full text