Cancer Letters

Cancer Letters

Volume 232, Issue 2, 8 February 2006, Pages 139-147
Cancer Letters

Mini review
Platelet-derived growth factor (PDGF) and glial tumorigenesis

https://doi.org/10.1016/j.canlet.2005.02.002Get rights and content

Abstract

Platelet-derived growth factor (PDGF) has long been implicated in cancer and is known to be involved in many biological processes. In Central Nervous System (CNS) neoplasms, particularly gliomas, PDGF is often over-expressed. However, what role PDGF plays in tumor progression remains to be fully described. A wide range of work from in vitro studies to mouse models have implicated the PDGF pathway in various processes including proliferation, cellular migration, development, and angiogenesis. Being a secreted factor, PDGF not only has autocrine effects on producing cells but also has potential for paracrine effects on other tumor cells and the tumor microenvironment. The development of small molecules that inhibit the PDGF receptor and various subsequent signaling components promises to introduce new approaches to the treatment of gliomas.

Introduction

Platelet-derived growth factor (PDGF) was first identified as the serum component responsible for the proliferation of arterial smooth muscle cells [1]. It has now been shown to be expressed in a wide range of tissues and be involved in a variety of functions [2]. The link between PDGF and tumor formation was demonstrated early on when the transforming gene in the Simian Sarcoma Virus, sis, was found to encode PDGFB [3]. Expression of PDGF and its receptor was subsequently found in multiple tumor types including central nervous system (CNS) tumors, sarcomas, germ cell tumors, and gastrointestinal system carcinomas. In particular, the PDGF pathway is well established to be involved in gliomas, tumors of the glial support cells (astrocytes and oligodendrocytes) in the CNS. The role that PDGF plays in glial tumorigenesis can be understood from its signal transduction functions, regulation of normal development, expression in tumors, and effects in mouse modeling.

Section snippets

PDGF ligands and receptors

The PDGF family consists of four ligands, PDGFA-D and two receptors, PDGFRα and PDGFRβ. PDGFs are disulfide-linked covalent dimers that are secreted and bind PDGFRs. All PDGF ligands (A–D) form homodimers, but PDGFA and PDGFB can also form a heterodimer (AB). PDGFA and B are secreted as active ligands, while C and D ligands require cleavage of N-terminal CUB domains to become active. The in vivo proteases responsible for activating C and D ligands have not been definitively identified; however,

PDGF in development

The pattern of PDGF and PDGFR expression in the CNS exhibits important modes of regulation. PDGFA is found expressed in neurons starting on E15 and continues in adult stages while PDGFB is also expressed in neurons but in a more limited manner [20], [21]. There is also evidence that the PDGFB message may be regulated by splicing as two different splice forms, differing in their 5′ untranslated regions (UTRs), are identifiable in developing rat brain [22]. The PDGFA message is also known to have

PDGF in glial tumors

Glial tumors of the CNS include astrocytomas, oligodendrogliomas, and mixed glial tumors, e.g. oligoastrocytomas [31], [32], [33]. Astrocytomas, the most common primary adult CNS tumor—making up to 60% of cases, are composed of tumor cells with features characteristic of astrocytes, supportive cells in the brain that carry out multiple functions. These tumors are categorized into different grades from I to IV based on different histological criteria determined through the World Health

PDGF in mouse models of gliomagenesis

The role of PDGF in tumorigenesis has been validated in many mouse models. Two such models for gliomas have used viral delivery of the PDGF gene into the brains of newborn mice. Delivery of PDGFB by Moloney murine leukemia virus (MMLV) was sufficient to induce malignant tumors [47]. In situ analysis demonstrated co-expression of PDGF and PDGFRα, and thus, capacity for autocrine signaling. In newborn mice, overexpression of PDGFB targeted to neural progenitors induces the formation of

Conclusion

The PDGF signaling pathway is able to perform multiple functions in glial tumorigenesis. Its overexpression in glial tumor samples clearly indicates a tendency for this pathway to be activated in tumor progression. Signaling molecules that could be activated in response to PDGF signals have also been shown to contribute to glial tumor formation. However, the causal link between activation of the PDGF pathway and subsequent signaling components in vivo remains to be explored. Which of these

Acknowledgements

Work in the laboratory is supported by the Serrossi Foundation, Kirby Foundation, NIH grant UO1CA894314-1, and NIH grant RO1 CA099489 to ECH. AHS was supported by NIH MSTP grant GM07739.

References (57)

  • R.A. Klinghoffer et al.

    An allelic series at the PDGFalphaR locus indicates unequal contributions of distinct signaling pathways during development

    Dev. Cell

    (2002)
  • P. Guo et al.

    Platelet-derived growth factor-B enhances glioma angiogenesis by stimulating vascular endothelial growth factor expression in tumor endothelia and by promoting pericyte recruitment

    Am. J. Pathol.

    (2003)
  • K. Pietras et al.

    PDGF receptors as cancer drug targets

    Cancer Cell

    (2003)
  • R. Ross et al.

    A platelet-dependent serum factor that stimulates the proliferation of arterial smooth muscle cells in vitro

    Proc. Natl. Acad. Sci. USA

    (1974)
  • C.H. Heldin et al.

    Mechanism of action and in vivo role of platelet-derived growth factor

    Physiol. Rev.

    (1999)
  • R. Dalla-Favera et al.

    A human onc gene homologous to the transforming gene (v-sis) of simian sarcoma virus

    Nature

    (1981)
  • L. Fredriksson et al.

    Tissue plasminogen activator is a potent activator of PDGF-CC

    Eur. Mol. Biol. Org. J.

    (2004)
  • P. Lindblom et al.

    Endothelial PDGF-B retention is required for proper investment of pericytes in the microvessel wall

    Genes Dev.

    (2003)
  • H. Ding et al.

    Astrocyte-specific expression of activated p21-ras results in malignant astrocytoma formation in a transgenic mouse model of human gliomas

    Cancer Res.

    (2001)
  • E.C. Holland et al.

    Combined activation of Ras and Akt in neural progenitors induces glioblastoma formation in mice

    Nat. Genet.

    (2000)
  • R.M. Wolf et al.

    Tyrosine phosphorylation of p190 RhoGAP by Fyn regulates oligodendrocyte differentiation

    J. Neurobiol.

    (2001)
  • S. Khoshyomn et al.

    Inhibition of phospholipase C-gamma1 activation blocks glioma cell motility and invasion of fetal rat brain aggregates

    Neurosurgery

    (1999)
  • A.B. da Rocha et al.

    Targeting protein kinase C: new therapeutic opportunities against high-grade malignant gliomas?

    Oncologist

    (2002)
  • M.L. Vignais et al.

    Platelet-derived growth factor induces phosphorylation of multiple JAK family kinases and STAT proteins

    Mol. Cell Biol.

    (1996)
  • A. Bonni et al.

    Regulation of gliogenesis in the central nervous system by the JAK-STAT signaling pathway

    Science

    (1997)
  • M. Sasahara et al.

    Normal developing rat brain expresses a platelet-derived growth factor B chain (c-sis) mRNA truncated at the 5′ end

    Oncogene

    (1998)
  • F. Rorsman et al.

    Platelet-derived growth factor (PDGF) A-chain mRNA heterogeneity generated by the use of alternative promoters and alternative polyadenylation sites

    Growth Factors

    (1992)
  • P. Horvath et al.

    Multiple elements in the 5′ untranslated region down-regulate c-sis messenger RNA translation

    Cell Growth Differ.

    (1995)
  • Cited by (0)

    View full text