Review
Platelet-derived growth factors and their receptors: Structural and functional perspectives

https://doi.org/10.1016/j.bbapap.2012.10.015Get rights and content

Abstract

The four types of platelet-derived growth factors (PDGFs) and the two types of PDGF receptors (PDGFRs, which belong to class III receptor tyrosine kinases) have important functions in the development of connective tissue cells. Recent structural studies have revealed novel mechanisms of PDGFs in propeptide loading and receptor recognition/activation. The detailed structural understanding of PDGF–PDGFR signaling has provided a template that can aid therapeutic intervention to counteract the aberrant signaling of this normally silent pathway, especially in proliferative diseases such as cancer. This review summarizes the advances in the PDGF system with a focus on relating the structural and functional understandings, and discusses the basic aspects of PDGFs and PDGFRs, the mechanisms of activation, and the insights into the therapeutic antagonism of PDGFRs. This article is part of a Special Issue entitled: Emerging recognition and activation mechanisms of receptor tyrosine kinases.

Highlights

► Recent advances on PDGFs and their receptors PDGFRs. ► Dissection of PDGF structural/functional understandings. ► Mechanistic insights into PDGF:propeptide recognition, receptor recognition and activation. ► Perspectives on therapeutic modulation of PDGFR signaling.

Introduction

Platelet-derived growth factors (PDGFs) are a family of four cystine-knot-type growth factors (PDGF-A, -B, -C and -D) which control the growth of connective tissue cells such as fibroblasts and smooth muscle cells [1], [2]. By acting on these mesenchymal cells, PDGFs critically regulate embryonic development, especially the formation of vessels and organs (reviewed in [3]). There are two types of receptors for PDGFs, PDGFRα and PDGFRβ, which belong to the class III receptor tyrosine kinases (RTKs), and have different expression patterns and physiological roles. PDGFRα signaling controls gastrulation and the development of several organs such as lung, intestine, skin, testis, kidney, bones, and neuroprotective tissues. PDGFRβ signaling is better recognized as an essential regulator of early hematopoiesis and blood vessel formation [3]. While PDGF–PDGFR signaling plays important roles during developmental stages, the expression of both PDGFs and PDGFRs is tightly controlled in adulthood. Enhanced PDGF–PDGFR signaling, except when happening briefly during wound repair, is generally considered abnormal, and is an important feature in a number of diseases involving proliferation, including many types of cancers, inflammation, pulmonary fibrosis and restenosis, and notably atherosclerosis [4].

PDGF signaling through PDGFRs utilizes the general strategy for RTKs, which involves ligand-induced receptor dimerization, and the subsequent receptor conformational changes that are coupled to the activation of intracellular tyrosine kinase domain (reviewed in [5]). The activation of PDGFR signaling pathways is built on structural platforms of both the ligands and the receptors, which are predicted to be conserved family-wise based on the sequence similarities between different PDGF and PDGFR subtypes. However, for decades, except for a crystal structure of the PDGF-B ligand [6], detailed mechanistic and structural understanding of PDGFR recognition and activation had been hampered until the recent elucidation of the PDGF-B:PDGFRβ complex structure. Inhibiting PDGF–PDGFR signaling, especially by selectively blocking the extracellular assembly through antibodies, ligand decoy or receptor decoys, is actively pursued in anti-cancer drug development, as these ligands and receptors serve in multiple aspects of tumor progression, such as mediating tumor growth in an autocrine fashion, recruiting fibroblast-rich tumor stroma, and regulating tumor vasculature [4]. The therapeutic efforts targeting PDGF/PDGFR are well compatible with strategies that can be derived from structural templates of both ligands and receptors. This review will summarize some current structural and functional understandings of the four PDGF ligands and the two PDGFRs.

Section snippets

The four types of PDGFs

PDGF(s) were discovered in 1970s as a platelet-dependent serum factor that stimulates the proliferation of fibroblasts, arterial smooth muscle cells, and glial cells [1], [2], [7]. There are four types of PDGF polypeptide chains as encoded by four genes. Of these four genes, PDGF-B was first characterized by amino acid sequencing to reveal its surprisingly close homology to the simian sarcoma virus oncogene v-sis [8], [9]. The cDNA of PDGF-A was subsequently cloned and its chromosomal

The propeptides of PDGFs and their association with the growth factor domains

As mentioned above, all PDGFs have pro-sequences, but PDGF-A and PDGF-B's pro-sequences contain only ~ 60 amino acids, whereas PDGF-C and PDGF-D have over 200 amino acids preceding the cysteine-knot growth factor domains. Of the long pro-sequences of PDGF-C and PDGF-D, the N-terminal part is a CUB domain which spans ~ 110 residues. A three-dimension structures of the PDGF-C/-D CUB domain is not yet available, but by analogy to known CUB domain structures [32], they should fold in a β-sandwich of

Two types of PDGFRs

The two receptors for PDGFs, PDGFRα and PDGFRβ, belong to the class III receptor tyrosine kinases (RTKs), a clan of five members including PDGFRα and PDGFRβ, KIT, FMS and FLT3 [5]. Like all RTKs, the PDGFR family of receptors have a modular architecture that utilize the extracellular domain to recognize ligands, a single transmembrane helix to pass structural/informational input from outside the cell, and an effector tyrosine kinase domain that respond to the extracellular signals, and

PDGF:PDGFR recognition

The PDGF dimer, as mentioned above, has a flat shape, with all β-strands forming a super-sheet, leaving the inter-strand loops at the ends of these strands. These loops are not only used for propeptide binding, but also for receptor binding. The binding of receptor to PDGFs is sterically incompatible with the simultaneous binding of propeptides to PDGFs. When the PDGF-A/propeptide complex and the PDGF-B/PDGFRβ complex are superimposed with the backbones of the growth factor domains overlaid, it

Recognition specificity between PDGFs and PDGFRs

PDGF-A and PDGF-B are often expressed in the same types of cells (reviewed in [64]) and the assembly of PDGF-AB heterodimers has been observed [65], [66], [67]. A study even suggested that the PDGF-AB heterodimer is preferentially formed over the homodimers [68]. There have been, however, no evidence that PDGF-C and PDGF-D form heterodimers. Therefore, considering all homodimers and heterodimers, five types of PDGF dimer proteins currently exist (Fig. 6). Given that PDGFRs are dimerized by the

Activation of PDGFRs

The activation of PDGFRs is not entirely understood mechanistically, but it likely requires receptor conformational changes in multiple steps. Initially, the bivalent nature of PDGF ligands brings two receptor protomers into proximity with one another. Importantly, activation further requires contacts between the receptor membrane-proximal regions [71], so that the intracellular kinase domains can be brought together for transphosphorylation. Although the molecular details of the interactions

Perspectives in PDGFR targeting

PDGF–PDGFR signaling plays essential roles in development, but once adulthood is reached, its function is usually detrimental rather than constructive in human physiology. An exception is in tissue repair and wound healing. PDGF-BB and PDGFRβ appear to be involved in the formational of new vessels and collagen production [77]. For this reason recombinant human PDGF-B has been used in clinical studies. However, the general need is to block PDGF–PDGFR signaling to stop or reverse proliferative

Conclusions

In the last a few years we have gained significant insight into the recognition and activation mechanisms of PDGFRs, a class of import receptor tyrosine kinase that is involved not only in physiological functions such as organogenesis and vessel formation, but also in several widespread diseases such as cancer and atherosclerosis. The significant roles of PDGFRs in cancer and vascular diseases underline the significant efforts in both academia and industry in search for specific, high-affinity,

Acknowledgements

The authors wish to thank Heli Liu and Ann Shim for comments and help in the preparation of some figures. This work in the authors’ laboratory is supported by the NIH grant 5R01GM098259.

References (84)

  • S. Yuzawa et al.

    Structural basis for activation of the receptor tyrosine kinase KIT by stem cell factor

    Cell

    (2007)
  • K. Verstraete et al.

    Structural insights into the extracellular assembly of the hematopoietic Flt3 signaling complex

    Blood

    (2011)
  • X. Ma et al.

    Structural basis for the dual recognition of helical cytokines IL-34 and CSF-1 by CSF-1R

    Structure

    (2012)
  • H. Liu et al.

    The mechanism of shared but distinct CSF-1R signaling by the non-homologous cytokines IL-34 and CSF-1

    Biochim. Biophys. Acta

    (2012)
  • J. Elegheert et al.

    Extracellular complexes of the hematopoietic human and mouse CSF-1 receptor are driven by common assembly principles

    Structure

    (2011)
  • J. Oates et al.

    Strong oligomerization behavior of PDGFbeta receptor transmembrane domain and its regulation by the juxtamembrane regions

    Biochim. Biophys. Acta

    (2010)
  • M. Walter et al.

    The 2.7 A crystal structure of the autoinhibited human c-Fms kinase domain

    J. Mol. Biol.

    (2007)
  • J. Griffith et al.

    The structural basis for autoinhibition of FLT3 by the juxtamembrane domain

    Mol. Cell

    (2004)
  • C.M. Rohde et al.

    A juxtamembrane tyrosine in the colony stimulating factor-1 receptor regulates ligand-induced Src association, receptor kinase function, and down-regulation

    J. Biol. Chem.

    (2004)
  • C.D. Mol et al.

    Structure of a c-kit product complex reveals the basis for kinase transactivation

    J. Biol. Chem.

    (2003)
  • A. Kazlauskas et al.

    Autophosphorylation of the PDGF receptor in the kinase insert region regulates interactions with cell proteins

    Cell

    (1989)
  • J. Lennartsson et al.

    Alix facilitates the interaction between c-Cbl and platelet-derived growth factor beta-receptor and thereby modulates receptor down-regulation

    J. Biol. Chem.

    (2006)
  • T. Davis-Smyth et al.

    Mapping the charged residues in the second immunoglobulin-like domain of the vascular endothelial growth factor/placenta growth factor receptor Flt-1 required for binding and structural stability

    J. Biol. Chem.

    (1998)
  • G. Fuh et al.

    Requirements for binding and signaling of the kinase domain receptor for vascular endothelial growth factor

    J. Biol. Chem.

    (1998)
  • C. Wiesmann et al.

    Crystal structure at 1.7 A resolution of VEGF in complex with domain 2 of the Flt-1 receptor

    Cell

    (1997)
  • A. Hammacher et al.

    A major part of platelet-derived growth factor purified from human platelets is a heterodimer of one A and one B chain

    J. Biol. Chem.

    (1988)
  • R.A. Klinghoffer et al.

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

    Dev. Cell

    (2002)
  • M. Tallquist et al.

    PDGF signaling in cells and mice

    Cytokine Growth Factor Rev.

    (2004)
  • T. Erpel et al.

    Src family protein tyrosine kinases and cellular signal transduction pathways

    Curr. Opin. Cell Biol.

    (1995)
  • A. Levitzki

    PDGF receptor kinase inhibitors for the treatment of PDGF driven diseases

    Cytokine Growth Factor Rev.

    (2004)
  • R. Ross et al.

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

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

    (1974)
  • J. Andrae et al.

    Role of platelet-derived growth factors in physiology and medicine

    Genes Dev.

    (2008)
  • C. Oefner et al.

    Crystal structure of human platelet-derived growth factor BB

    EMBO J.

    (1992)
  • M.D. Waterfield et al.

    Platelet-derived growth factor is structurally related to the putative transforming protein p28sis of simian sarcoma virus

    Nature

    (1983)
  • R.F. Doolittle et al.

    Simian sarcoma virus onc gene, v-sis, is derived from the gene (or genes) encoding a platelet-derived growth factor

    Science

    (1983)
  • C. Betsholtz et al.

    cDNA sequence and chromosomal localization of human platelet-derived growth factor A-chain and its expression in tumour cell lines

    Nature

    (1986)
  • X. Li et al.

    PDGF-C is a new protease-activated ligand for the PDGF alpha-receptor

    Nat. Cell Biol.

    (2000)
  • E. Bergsten et al.

    PDGF-D is a specific, protease-activated ligand for the PDGF beta-receptor

    Nat. Cell Biol.

    (2001)
  • W.J. LaRochelle et al.

    PDGF-D, a new protease-activated growth factor

    Nat. Cell Biol.

    (2001)
  • A.H. Shim et al.

    Structures of a platelet-derived growth factor/propeptide complex and a platelet-derived growth factor/receptor complex

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

    (2010)
  • F. Rorsman et al.

    Structural characterization of the human platelet-derived growth factor A-chain cDNA and gene: alternative exon usage predicts two different precursor proteins

    Mol. Cell. Biol.

    (1988)
  • A. Abramsson et al.

    Defective N-sulfation of heparan sulfate proteoglycans limits PDGF-BB binding and pericyte recruitment in vascular development

    Genes Dev.

    (2007)
  • Cited by (169)

    • Estrogen receptor β affects hypoxia response in colorectal cancer cells

      2024, Biochimica et Biophysica Acta - Molecular Basis of Disease
    • Receptor tyrosine kinases: an overview

      2023, Receptor Tyrosine Kinases in Neurodegenerative and Psychiatric Disorders
    • Receptor tyrosine kinases (RTKs): from biology to pathophysiology

      2023, Receptor Tyrosine Kinases in Neurodegenerative and Psychiatric Disorders
    View all citing articles on Scopus

    This article is part of a Special Issue entitled: Emerging recognition and activation mechanisms of receptor tyrosine kinases.

    View full text