Structural basis for fibroblast growth factor receptor activation

https://doi.org/10.1016/j.cytogfr.2005.01.008Get rights and content

Abstract

FGF signaling plays a ubiquitous role in human biology as a regulator of embryonic development, homeostasis and regenerative processes. In addition, aberrant FGF signaling leads to diverse human pathologies including skeletal, olfactory, and metabolic disorders as well as cancer. FGFs execute their pleiotropic biological actions by binding, dimerizing and activating cell surface FGF receptors (FGFRs). Proper regulation of FGF–FGFR binding specificity is essential for the regulation of FGF signaling and is achieved through primary sequence variations among the 18 FGFs and seven FGFRs. The severity of human skeletal syndromes arising from mutations that violate FGF–FGFR specificity is a testament to the importance of maintaining precision in FGF–FGFR specificity. The discovery that heparin/heparan sulfate (HS) proteoglycans are required for FGF signaling led to numerous models for FGFR dimerization and heralded one of the most controversial issues in FGF signaling. Recent crystallographic analyses have led to two fundamentally different models for FGFR dimerization. These models differ in both the stoichiometry and minimal length of heparin required for dimerization, the quaternary arrangement of FGF, FGFR and heparin in the dimer, and in the mechanism of 1:1 FGF–FGFR recognition and specificity. In this review, we provide an overview of recent structural and biochemical studies used to differentiate between the two crystallographic models. Interestingly, the structural and biophysical analyses of naturally occurring pathogenic FGFR mutations have provided the most compelling and unbiased evidences for the correct mechanisms for FGF–FGFR dimerization and binding specificity. The structural analyses of different FGF–FGFR complexes have also shed light on the intricate mechanisms determining FGF–FGFR binding specificity and promiscuity and also provide a plausible explanation for the molecular basis of a large number craniosynostosis mutations.

Section snippets

Identification and characterization of FGF ligands

The inception of the fibroblast growth factor (FGF) signaling field can be traced to year the 1939, when bovine brain extracts were shown to promote proliferation of fibroblast cell lines in vitro [1], [2]. Biochemical characterization of this mitogenic activity (termed FGF) from bovine pituitary and brain tissue [3], [4], showed that FGF activity was due to a ∼15 kDa molecule, which was termed basic FGF (bFGF) reflecting on its high isoelectric point [5], [6]. A second molecule with FGF

Models for FGFR dimerization

Ligand-induced receptor dimerization is a prerequisite for RTK activation. Receptor dimerization brings the cytoplasmic domains of the receptors in close vicinity of each other providing the opportunity for receptor trans autophosphorylation, subsequent tyrosine kinase activation and initiation of downstream signaling pathways [93]. Using a dominant negative approach initially applied to PDGFR and EGFR [94], [95], it was shown that FGFR dimerization is required for signal transduction [96], [97]

Analysis of pathogenic gain-of-function FGFR mutations provides insight into the mode of FGF–FGFR binding and dimerization

Gain-of-function missense mutations in FGFR1–3 are responsible for craniosynostosis [55], [137], [138], [139] and chondrodysplasia syndromes [56]. Craniosynostosis, the premature fusion of one or more cranial sutures, affects approximately 1 in 2500 individuals and is the clinical finding of over 100 distinct syndromes, including Apert syndrome (AS), Crouzon syndrome (CS), Muenke syndrome (MS), and Pfeiffer syndrome (PS). Chondrodysplasia syndromes, including achondroplasia and thanatophoric

Mutational studies confirm the physiological relevance of secondary ligand–receptor interface observed only in the symmetric “two-end” model

As described in Sections 2.3.1 Crystal structure of a 2:2 FGF2–FGFR1c symmetric dimer (PDB ID: 1CVS) reveals a heparin binding canyon; first crystallographic insight into the mechanism of FGFR dimerization, 2.3.2 Crystal structure of FGF2–FGFR1c–heparin ternary complex (PDB ID 1FQ9) reveals a symmetric “two-end” model for FGFR dimerization, dimerization in the symmetric “two-end” model is also promoted by secondary FGF–FGFR contacts. The secondary FGF–FGFR contact site in the symmetric

A canyon dimer also forms in the FGF1–FGFR2c–heparin crystal

In lieu of the incorrect conformation of linker proline in the asymmetric model, we analyzed the crystal packing in 1E0O to understand what other features of the asymmetric model may have been affected by the incorrect cis configuration of the invariant linker proline. Importantly, Blundell and coworkers also crystallized their FGF1–FGFR2c–heparin complex (PDB ID: 1E0O) under a high sulfate ion condition (1 M lithium sulfate), which we know facilitates canyon dimer formation. Indeed, examination

What is the stoichiometry of heparin in the FGF–FGFR dimerization?

The symmetric “two-end” model and asymmetric model differ with respect to both the stoichiometry and the minimal oligosaccharide length required to promote FGF–FGFR dimerization. In an effort to differentiate between these two models, Rosenberg and coworkers have recently studied the stoichiometry of the dimeric FGF–FGFR–HS complex using native gel analysis and concluded that the stoichiometry of the dimeric FGF1–FGFR1c–HS complex is 2:2:2 [155], as observed in the symmetric “two-end” model. In

A role for D1 and the D1–D2 linker region in receptor autoinhibition

A large body of structural and biochemical data show that D2, D3 and the interconnecting linker harbor all of the determinants for ligand binding and specificity. In addition, the recent crystal structure of the D1–D3 portion of FGFR3c in complex with FGF1, shows that D1 and the D1–D2 linker are completely disordered [131], providing solid structural evidence that these regions are dispensable for ligand binding. In contrast, the role of D1 and the D1–D2 linker, which contains a stretch of

The symmetric “two-end” model provides a molecular basis for HS specificity

The composition of HS in the periplasm has been shown to undergo major dynamic changes during development and recent data suggest that these spatial and temporal changes impact FGF signaling. Emerging data show that distinct HS motifs are required to promote binding and activation of different FGF–FGFR pairs. Interestingly, these data reveal that the HS requirements of different FGF–FGFR complexes do not simply reflect the sum of HS requirements of individual FGF and FGFR components, rather,

Epilogue

Receptor dimerization is a universal mechanism in RTK activation. The structural basis for receptor dimerization has been elucidated for several RTK subfamilies including EGFR, VEGF, TRK. These studies reveal that the mode of receptor dimerization varies greatly among RTK subfamilies and is likely tailored to meet their individual biological functions. The mechanism by which FGFs transduce their signal across cell membranes has been intensely studied over the past 30 years. Recent X-ray

Acknowledgements

We thank Jinghong Ma for help in figure preparation. This work was funded by NIH grant DE13686 to M.M.

References (171)

  • H. Kirikoshi et al.

    Molecular cloning and characterization of human FGF-20 on chromosome 8p21.3–p22

    Biochem Biophys Res Commun

    (2000)
  • Y. Nakatake et al.

    Identification of a novel fibroblast growth factor, FGF-22, preferentially expressed in the inner root sheath of the hair follicle

    Biochim Biophys Acta

    (2001)
  • T. Yamashita et al.

    Identification of a novel fibroblast growth factor, FGF-23, preferentially expressed in the ventrolateral thalamic nucleus of the brain

    Biochem Biophys Res Commun

    (2000)
  • J. Gemel et al.

    Structure and sequence of human FGF8

    Genomics

    (1996)
  • J. Schoorlemmer et al.

    Fibroblast growth factor homologous factors and the islet brain-2 scaffold protein regulate activation of a stress-activated protein kinase

    J Biol Chem

    (2002)
  • S.K. Olsen et al.

    Fibroblast growth factor (FGF) homologous factors share structural but not functional homology with FGFs

    J Biol Chem

    (2003)
  • R.L. Glaser et al.

    The paternal-age effect in Apert syndrome is due, in part, to the increased frequency of mutations in sperm

    Am J Hum Genet

    (2003)
  • J.S. Colvin et al.

    Male-to-female sex reversal in mice lacking fibroblast growth factor 9

    Cell

    (2001)
  • C. Yu et al.

    Elevated cholesterol metabolism and bile acid synthesis in mice lacking membrane tyrosine kinase receptor FGFR4

    J Biol Chem

    (2000)
  • T. Yamashita et al.

    Fibroblast growth factor (FGF)-23 inhibits renal phosphate reabsorption by activation of the mitogen-activated protein kinase pathway

    J Biol Chem

    (2002)
  • M.J. Econs

    New insights into the pathogenesis of inherited phosphate wasting disorders

    Bone

    (1999)
  • M.V. Cronauer et al.

    Fibroblast growth factors and their receptors in urological cancers: basic research and clinical implications

    Eur Urol

    (2003)
  • J.S. Huang et al.

    Bovine brain-derived growth factor. Purification and characterization of its interaction with responsive cells

    J Biol Chem

    (1986)
  • S.R. Coughlin et al.

    Acidic and basic fibroblast growth factors stimulate tyrosine kinase activity in vivo

    J Biol Chem

    (1988)
  • R.A. Horlick et al.

    Cloning, expression and tissue distribution of the gene encoding rat fibroblast growth factor receptor subtype 4

    Gene

    (1992)
  • D.M. Ornitz et al.

    Receptor specificity of the fibroblast growth factor family

    J Biol Chem

    (1996)
  • A. Baird et al.

    Fibroblast growth factors are present in the extracellular matrix produced by endothelial cells in vitro: implications for a role of heparinase-like enzymes in the neovascular response

    Biochem Biophys Res Commun

    (1987)
  • J. Schlessinger et al.

    Regulation of growth factor activation by proteoglycans: what is the role of the low affinity receptors?

    Cell

    (1995)
  • A. Yayon et al.

    Cell surface, heparin-like molecules are required for binding of basic fibroblast growth factor to its high affinity receptor

    Cell

    (1991)
  • E. Amaya et al.

    Expression of a dominant negative mutant of the FGF receptor disrupts mesoderm formation in Xenopus embryos

    Cell

    (1991)
  • H. Ueno et al.

    A truncated form of fibroblast growth factor receptor 1 inhibits signal transduction by multiple types of fibroblast growth factor receptor

    J Biol Chem

    (1992)
  • T. Spivak-Kroizman et al.

    Heparin-induced oligomerization of FGF molecules is responsible for FGF receptor dimerization, activation, and cell proliferation

    Cell

    (1994)
  • O.A. Trowell et al.

    J Exp Biol

    (1939)
  • R.S. Hoffman

    Growth

    (1940)
  • H.A. Armelin

    Pituitary extracts and steroid hormones in the control of 3T3 cell growth

    Proc Natl Acad Sci USA

    (1973)
  • D. Gospodarowicz et al.

    Purification of a growth factor for ovarian cells from bovine pituitary glands

    Proc Natl Acad Sci USA

    (1974)
  • T. Maciag et al.

    An endothelial cell growth factor from bovine hypothalamus: identification and partial characterization

    Proc Natl Acad Sci USA

    (1979)
  • S.C. Thornton et al.

    Human endothelial cells: use of heparin in cloning and long-term serial cultivation

    Science

    (1983)
  • Y. Shing et al.

    Heparin affinity: purification of a tumor-derived capillary endothelial cell growth factor

    Science

    (1984)
  • T. Maciag et al.

    Heparin binds endothelial cell growth factor, the principal endothelial cell mitogen in bovine brain

    Science

    (1984)
  • T.A. Libermann et al.

    An angiogenic growth factor is expressed in human glioma cells

    Embo J

    (1987)
  • S.K. Lemmon et al.

    Bovine fibroblast growth factor: comparison of brain and pituitary preparations

    J Cell Biol

    (1982)
  • W.H. Burgess et al.

    Structural evidence that endothelial cell growth factor beta is the precursor of both endothelial cell growth factor alpha and acidic fibroblast growth factor

    Proc Natl Acad Sci USA

    (1986)
  • W.H. Burgess et al.

    The heparin-binding (fibroblast) growth factor family of proteins

    Annu Rev Biochem

    (1989)
  • H. Sakamoto et al.

    Transforming gene from human stomach cancers and a noncancerous portion of stomach mucosa

    Proc Natl Acad Sci USA

    (1986)
  • P. Delli Bovi et al.

    Isolation of a rearranged human transforming gene following transfection of Kaposi sarcoma DNA

    Proc Natl Acad Sci USA

    (1987)
  • X. Zhan et al.

    The human FGF-5 oncogene encodes a novel protein related to fibroblast growth factors

    Mol Cell Biol

    (1988)
  • I. Marics et al.

    Characterization of the HST-related FGF.6 gene, a new member of the fibroblast growth factor gene family

    Oncogene

    (1989)
  • J.S. Rubin et al.

    Purification and characterization of a newly identified growth factor specific for epithelial cells

    Proc Natl Acad Sci USA

    (1989)
  • A. Tanaka et al.

    Cloning and characterization of an androgen-induced growth factor essential for the androgen-dependent growth of mouse mammary carcinoma cells

    Proc Natl Acad Sci USA

    (1992)
  • Cited by (582)

    • FGF12: biology and function

      2023, Differentiation
    • Receptor tyrosine kinases (RTKs): from biology to pathophysiology

      2023, Receptor Tyrosine Kinases in Neurodegenerative and Psychiatric Disorders
    • Regulation of receptor tyrosine kinase gene expression

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