Multiple connections link FAK to cell motility and invasion

https://doi.org/10.1016/j.gde.2003.12.002Get rights and content

Abstract

The ability of intracellular signaling networks to orchestrate a complex biological response such as cell motility requires that individual signaling proteins must act as integrators, responding to multiple extracellular inputs and regulating multiple signaling pathway outputs. In this review, we highlight recent findings that place focal adhesion kinase (FAK) in an important receptor-proximal position in the regulation of growth factor and integrin-stimulated cell motility. Emphasis is placed on the molecular mechanisms of FAK activation, connections of FAK to focal contact formation as well as turnover, and the potential that FAK function in promoting cell invasion may be distinct from its role in cell motility.

Introduction

Focal adhesion kinase (FAK) was first identified in 1992 as a highly tyrosine-phosphorylated protein associated with the v-Src oncogene and localized within integrin-enriched focal adhesion contact sites in normal cells (see [1] for review). FAK activity is regulated by integrin-mediated cell adhesion as well as being downstream of growth factor and G-protein-linked receptor activation 2., 3.. In many cells, FAK activation leads to the Src homology 2 (SH2) domain-mediated binding to Src-family protein tyrosine kinases (PTKs) to the motif surrounding the FAK Tyr-397 phosphorylation site. It is this dual FAK–Src complex that promotes the tyrosine phosphorylation of substrates such as paxillin and p130Cas and FAK–Src-associated signaling can lead to the activation of multiple protein kinase cascades 2., 3., 4..

Whereas early studies linked FAK activation to the formation of cell–substratum contact sites, knockout studies showed that FAK-null fibroblasts exhibit an increased numbers of focal contact sites and cell-motility defects [5]. Reconstitution of FAK-null cells, FAK overexpression studies, and the inhibition of FAK activity have all supported a role for FAK–Src signaling in promoting cell migration [2]. In transformed cells and in clinical analyses of human tumors, elevated FAK expression and activity have been correlated with the progression to a highly malignant and metastatic phenotype 6.••, 7., 8.. To this end, the possibility that FAK-mediated signaling may promote increased tumorigenicity has generated much interest to determine the mechanisms of FAK activation.

Section snippets

Multiple mechanisms of FAK activation

Even though the initial cloning of FAK showed that it was activated by integrin clustering, the precise mechanism of FAK activation has eluded investigators for over twelve years. This is likely due to the fact that FAK can be activated by multiple inputs and in different manners. One constant is that cloning efforts have revealed that the FAK central kinase domain is highly conserved from humans to Drosophila and Caenorhabditis elegans. The FAK N-terminal region harbors a FERM (band four point

New roles elucidated for FAK phosphorylation sites

Src SH2-mediated binding to FAK Tyr-397 leads to the generation of an activated Src–FAK signaling complex. Src facilitates maximal FAK activation through phosphorylation at Tyr-576 and Tyr-577 within the FAK kinase domain activation loop and Src can phosphorylate additional sites within the FAK C-terminal domain at Tyr-861 and Tyr-925, the latter serving as a high affinity Grb2 SH2 binding site. Efforts to make constitutively-active FAK mutants by plasma membrane targeting result in elevated

FAK FERM domain interacting proteins

As FAK connects to integrins through various C-terminal domain-mediated interactions, FAK is also functionally linked to various other proteins through N-terminal FERM domain-mediated interactions. Structural analyses and sequence comparisons reveal that FERM domains comprise three lobes containing multiple sites for both lipid and protein binding. Recent studies have begun to identify FAK FERM targets and how these interactions may affect FAK function. For instance, ezrin binding to the FAK

FAK connections to focal contact formation

Many research groups have observed correlations between FAK activation, paxillin tyrosine phosphorylation, and the subsequent formation of actin stress fibers and focal contact sites 1., 3.. These events are often connected to Rho-family GTPase activation that act as switches existing in either an inactive GDP-bound or an active GTP-bound form. Guanine nucleotide exchange factors (GEFs) stimulate the GDP to GTP exchange reaction to facilitate an active RhoGTPase conformation. FAK can promote

FAK-null fibroblasts as a model system to elucidate FAK function

Although FAK is linked to the formation of focal contacts as discussed above, FAK-null fibroblasts exhibit motility defects in part as a result of elevated Rho activity, increased focal contact formation, and the inability to remodel contact sites in response to various motility stimuli 2., 41., 42.•. This cellular response is unique to the loss of FAK as null mutations in other FAK- and focal contact associated proteins such as paxillin [9], p130Cas [43], or Src-family PTKs [44] do not yield a

A mechanism by which FAK promotes focal contact turnover

In addition to functioning as an integrator and amplifier of signaling to ERK2 and JNK, FAK needs to be localized appropriately to focal contact sites to reverse the motility defects of FAK-null cells [45]. This raises the possibility that there may be critical target(s) or substrate(s) of FAK localized within focal contact sites that are not being regulated appropriately in the absence of FAK expression. In addition to the contributions of FAK signaling to JNK with respect to gene-expression

Connections of FAK to cell invasion extend beyond cell motility

Tumor cell invasion through matrix and tissue barriers requires the combined effects of increased cell motility and regulated proteolytic degradation of the matrix. Though FAK expression is elevated in invasive humans cancers 6.••, 7., 8., assessments of FAK function within tumor cells have often relied upon measurements of cell motility in two dimensions rather than cell invasion in three dimensions [61]. Src-mediated cell-transformation studies revealed that naturally occurring v-Src SH3

Conclusions

FAK has been demonstrated to play important yet differing roles in signaling pathways associated with normal and tumor cell movement. As FAK functions as both a scaffold and as a kinase, the future development of pharmacological inhibitors to FAK may be used to discriminate whether normal cell motility or malignant cell invasion events differentially depend on FAK catalytic activation. The continued characterization of FAK-interacting proteins will provide a better understanding of the

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

  • of special interest

  • ••

    of outstanding interest

Acknowledgements

DDS is supported by grants from the National Cancer Institute (CA75240, CA87038, and CA102310) and SKM is supported by a fellowship (12FT-0122) from the California Tobacco-Related Disease Research Program. This is manuscript 16074-IMM from the Scripps Research Institute.

References (68)

  • X. Ren et al.

    Focal adhesion kinase suppresses Rho activity to promote focal adhesion turnover

    J. Cell Sci.

    (2000)
  • J. Huang et al.

    Differential regulation of cell migration, actin stress fiber organization, and cell transformation by functional domains of Crk-associated substrate

    J. Biol. Chem.

    (2002)
  • C.R. Hauck et al.

    Inhibition of focal adhesion kinase expression or activity disrupts epidermal growth factor-stimulated signaling promoting the migration of invasive human carcinoma cells

    Cancer Res.

    (2001)
  • J.K. Slack-Davis et al.

    PAK1 phosphorylation of MEK1 regulates fibronectin-stimulated MAPK activation

    J. Cell Biol.

    (2003)
  • C. Huang et al.

    JNK phosphorylates paxillin and regulates cell migration

    Nature

    (2003)
  • B.D. Cuevas et al.

    MEKK1 regulates calpain-dependent proteolysis of focal adhesion proteins for rear-end detachment of migrating fibroblasts

    EMBO J.

    (2003)
  • N.O. Carragher et al.

    A Novel role for FAK as a protease-targeting adaptor protein. Regulation by p42 ERK and Src

    Curr. Biol.

    (2003)
  • J.T. Parsons

    Focal adhesion kinase: the first ten years

    J. Cell Sci.

    (2003)
  • S. Abbi et al.

    Focal adhesion kinase: protein interactions and cellular functions

    Histol. Histopathol.

    (2002)
  • S.K. Hanks et al.

    Focal adhesion kinase signaling activities and their implications in the control of cell survival and motility

    Front Biosci.

    (2003)
  • V. Gabarra-Niecko et al.

    FAK regulates biological processes important for the pathogenesis of cancer

    Cancer Metastasis Rev.

    (2003)
  • T.P. Hecker et al.

    Focal adhesion kinase in cancer

    Front Biosci.

    (2003)
  • G.W. McLean et al.

    Focal adhesion kinase as a potential target in oncology

    Expert Opin. Pharmacother.

    (2003)
  • M. Hagel et al.

    The adaptor protein paxillin is essential for normal development in the mouse and is a critical transducer of fibronectin signaling

    Mol. Cell Biol.

    (2002)
  • M. Toutant et al.

    Alternative splicing controls the mechanisms of FAK autophosphorylation

    Mol. Cell Biol.

    (2002)
  • D.J. Sieg et al.

    FAK integrates growth-factor and integrin signals to promote cell migration

    Nat. Cell Biol.

    (2000)
  • P. Poullet et al.

    Ezrin interacts with focal adhesion kinase and induces its activation independently of cell-matrix adhesion

    J. Biol. Chem.

    (2001)
  • Q.G. Medley et al.

    Signaling between focal adhesion kinase and trio

    J. Biol. Chem.

    (2003)
  • S. Abbi et al.

    Regulation of focal adhesion kinase by a novel protein inhibitor FIP200

    Mol. Biol. Cell

    (2002)
  • P. Chiarugi et al.

    Reactive oxygen species as essential mediators of cell adhesion: the oxidative inhibition of a FAK tyrosine phosphatase is required for cell adhesion

    J. Cell Biol.

    (2003)
  • M.A. Schwartz

    Integrin signaling revisited

    Trends Cell Biol.

    (2001)
  • B.Z. Katz et al.

    Targeting membrane-localized focal adhesion kinase to focal adhesions: roles of tyrosine phosphorylation and SRC family kinases

    J. Biol. Chem.

    (2003)
  • I. Hayashi et al.

    The focal adhesion targeting (FAT) region of focal adhesion kinase is a four-helix bundle that binds paxillin

    Nat. Struct. Biol.

    (2002)
  • G. Liu et al.

    Structural insight into the mechanisms of targeting and signaling of focal adhesion kinase

    Mol. Cell Biol.

    (2002)
  • Cited by (0)

    View full text