Elsevier

Gene

Volume 614, 30 May 2017, Pages 1-7
Gene

Gene wiki review
PDGFs and their receptors

https://doi.org/10.1016/j.gene.2017.03.003Get rights and content

Highlights

  • There are multiple modes of activating PDGFRs.

  • Once activated, PDGFRs contribute to various pathological conditions.

  • The PDGF/PDGFR family constitutes largely untapped therapeutic targets.

  • The precise role of PDGFR in pathology along with receptor-specific agonists will enable the development of such therapies.

Abstract

The platelet-derived growth factor (PDGF)/PDGFR receptor (PDGFR) family is essential for a vast array of physiological processes such as migration and proliferation of percityes that contribute to the formation and proper function of blood vessels. While ligand-dependent de-repression of the PDGFR's kinase activity is the major mode by which the PDGFR is activated, there are additional mechanisms to activate PDGFRs. Deregulated PDGFR activity contributes to various pathological conditions, and hence the PDGF/PDGFR family members are viable therapeutic targets. An increased appreciation of which PDGFR contributes to pathology, biomarkers that indicate the amplitude and mode of activation, and receptor-specific antagonists are necessary for the development of next-generation therapies that target the PDGF/PDGFR family.

Section snippets

The PDGF family and their relationships

There are four platelet-derived growth factor (PDGF) genes (PDGFA, PDGFB, PDGFC and PDGFD) that reside on chromosomes 7, 22, 4 and 11 in humans, and chromosomes 5, 15, 3 and 9 in mice, respectively. Biologically active (able to activate a PDGF receptor (PDGFR)) PDGF is a dimer of two PDGF protein chains. Extracellular, proteolytic processing is required for activation of some isoforms of PDGF (PDGF-C and PDGF-D), while this step occurs intracellularly for PDGF-A, PDGF-B and PDGF-AB (Fredriksson

PDGF-mediated

The best-known mechanism to activated PDGFRs is the PDGF-mediated (direct) mode of activation (Fig. 1). PDGF assembles two PDGFR molecules and thereby intensely activates the receptor's intrinsic tyrosine kinase activity. The activated PDGFR tyrosine phosphorylates substrates and thereby engages signaling cascades that drive subsequent cellular responses. For instance, both PDGFRα and PDGFRβ autophosphorylate and thereby create a docking sites for SH2 domain-containing proteins such as

PDGF-mediated

As mentioned above, similarities between certain PDGF and PDGFR knock out mice indicate that PDGF-dependent activation of PDGFRs is essential for normal development. Eliminating either PDGFRB, or PDGF-B reduces the number of pericytes and vascular smooth muscle cells, and thereby compromises the integrity and/or functionality of the vasculature in multiple organs, including the brain, heart, kidney, skin and eye (Soriano, 1994, Lindahl et al., 1997, Lindahl et al., 1998, Leveen et al., 1994). A

Therapeutic options

Existing ligand-directed therapies include aptamers and antibodies that selectively recognize a desired PDGF isoform (Lewandowski et al., 2016a, Heldin, 2014, Heldin, 2013). They are typically highly selective, i.e. do not target growth factors outside of the PDGF family, and distinguish amongst the PDGF isoforms. The disadvantage of targeting a specific PDGF isoform is that it may not prevent activation of PDGFRs because multiple isoforms activate a given PDGFR (Table 1). For example, it is

Conclusions

We currently appreciate that the PDGF/PDGFR family makes multiple contributions to physiology in distinct anatomical locations, and that the two PDGFRs have distinct functions. Furthermore, while ligand-driven activation is the major mode of de-repressing the kinase activity of PDGFRs, there are additional mechanisms. Activated PDGFRs trigger signaling events that direct a spectrum of cellular responses that underlie both physiology and pathology. Two types of advances will set the stage for a

Acknowledgements

This review and the corresponding Gene Wiki article are written as part of the Gene Wiki Review series–a series resulting from a collaboration between the journal GENE and the Gene Wiki Initiative. The Gene Wiki Initiative is supported by National Institutes of Health (GM089820). Additional support for Gene Wiki Reviews is provided by Elsevier, the publisher of GENE.

The corresponding Gene Wiki entry for this review can be found here:

https://en.wikipedia.org/wiki/PDGFRA

//en.wikipedia.org/wiki/PDGFRB

References (76)

  • S. Pennock et al.

    A novel strategy to develop therapeutic approaches to prevent proliferative vitreoretinopathy

    Am. J. Pathol.

    (2011)
  • S. Pennock et al.

    Ranibizumab is a potential prophylaxis for proliferative vitreoretinopathy, a nonangiogenic blinding disease

    Am. J. Pathol.

    (2013)
  • S. Pennock et al.

    Is neutralizing vitreal growth factors a viable strategy to prevent proliferative vitreoretinopathy?

    Prog. Retin. Eye Res.

    (2014)
  • S. Pennock et al.

    Vascular endothelial growth factor acts primarily via platelet-derived growth factor receptor alpha to promote proliferative vitreoretinopathy

    Am. J. Pathol.

    (2014)
  • L.M. Petti et al.

    A single amino acid substitution converts a transmembrane protein activator of the platelet-derived growth factor beta receptor into an inhibitor

    J. Biol. Chem.

    (2013)
  • K.G. Sharp et al.

    A re-assessment of treatment with a tyrosine kinase inhibitor (imatinib) on tissue sparing and functional recovery after spinal cord injury

    Exp. Neurol.

    (2014)
  • S. Svegliati et al.

    Stimulatory autoantibodies to PDGF receptor in patients with extensive chronic graft-versus-host disease

    Blood

    (2007)
  • M. Tallquist et al.

    PDGF signaling in cells and mice

    Cytokine Growth Factor Rev.

    (2004)
  • X. Wang et al.

    Targeting the PDGF-B/PDGFR-beta Interface with destruxin A5 to selectively block PDGF-BB/PDGFR-betabeta signaling and attenuate liver fibrosis

    EBioMedicine

    (2016)
  • D. Windisch et al.

    Hydrophobic mismatch drives the interaction of E5 with the transmembrane segment of PDGF receptor

    Biophys. J.

    (2015)
  • M.B. Abrams et al.

    Imatinib enhances functional outcome after spinal cord injury

    PLoS One

    (2012)
  • M.V. Adzemovic et al.

    Imatinib ameliorates neuroinflammation in a rat model of multiple sclerosis by enhancing blood-brain barrier integrity and by modulating the peripheral immune response

    PLoS One

    (2013)
  • J. Andrae et al.

    Role of platelet-derived growth factors in physiology and medicine

    Genes Dev.

    (2008)
  • H.N. Antoniades et al.

    Purification of human platelet-derived growth factor

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

    (1979)
  • A. Armulik et al.

    Pericytes regulate the blood-brain barrier

    Nature

    (2010)
  • E. Balada et al.

    Anti-PDGFR-alpha antibodies measured by non-bioactivity assays are not specific for systemic sclerosis

    Ann. Rheum. Dis.

    (2008)
  • S.G. Ball et al.

    Vascular endothelial growth factor can signal through platelet-derived growth factor receptors

    J. Cell Biol.

    (2007)
  • S. Banerjee et al.

    Multiplex bead analysis of vitreous humor of patients with vitreoretinal disorders

    Invest. Ophthalmol. Vis. Sci.

    (2007)
  • S.S. Baroni et al.

    Stimulatory autoantibodies to the PDGF receptor in systemic sclerosis

    N. Engl. J. Med.

    (2006)
  • M. Citirik et al.

    Vitreous vascular endothelial growth factor concentrations in proliferative diabetic retinopathy versus proliferative vitreoretinopathy

    Ophthalmic Res.

    (2012)
  • J.F. Classen et al.

    Lack of evidence of stimulatory autoantibodies to platelet-derived growth factor receptor in patients with systemic sclerosis

    Arthritis Rheum.

    (2009)
  • S.C. Dieudonne et al.

    Balance of vascular endothelial growth factor and pigment epithelial growth factor prior to development of proliferative vitreoretinopathy

    Ophthalmic Res.

    (2007)
  • H. Ding et al.

    A specific requirement for PDGF-C in palate formation and PDGFR-alpha signaling

    Nat. Genet.

    (2004)
  • A.P. Edwards et al.

    Compensatory mutants of the bovine papillomavirus E5 protein and the platelet-derived growth factor beta receptor reveal a complex direct transmembrane interaction

    J. Virol.

    (2013)
  • J.A. Engelman et al.

    The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism

    Nat. Rev. Genet.

    (2006)
  • T.P. Fleming et al.

    Amplification and/or overexpression of platelet-derived growth factor receptors and epidermal growth factor receptor in human glial tumors

    Cancer Res.

    (1992)
  • D.J. Goldstein et al.

    The bovine papillomavirus type 1 E5 transforming protein specifically binds and activates the beta-type receptor for the platelet-derived growth factor but not other related tyrosine kinase-containing receptors to induce cellular transformation

    J. Virol.

    (1994)
  • T.R. Golub et al.

    Fusion of PDGF receptor b to a novel ets-like gene, tel, in chronic myelomoncytic leukemia with t(5;12) chromosomal translocation

    Cell

    (1994)
  • Cited by (139)

    View all citing articles on Scopus
    View full text