You are on page 1of 13

REVIEWS

FOCAL ADHESION KINASE:


IN COMMAND AND CONTROL
OF CELL MOTILITY
Satyajit K. Mitra, Daniel A. Hanson and David D. Schlaepfer
Abstract | A central question in cell biology is how membrane-spanning receptors transmit
extracellular signals inside cells to modulate cell adhesion and motility. Focal adhesion kinase
(FAK) is a crucial signalling component that is activated by numerous stimuli and functions as a
biosensor or integrator to control cell motility. Through multifaceted and diverse molecular
connections, FAK can influence the cytoskeleton, structures of cell adhesion sites and
membrane protrusions to regulate cell movement.

INTEGRINS Cell migration is a coordinated process that involves highly tyrosine-phosphorylated protein that localized
A large family of heterodimeric rapid changes in the dynamics of actin filaments, to integrin-enriched cell adhesion sites that are known
transmembrane proteins that together with the formation and disassembly of cell as focal contacts (BOX 1). Focal contacts are formed at
function as receptors for cell- adhesion sites1. A complex interplay between the actin ECM–integrin junctions that bring together cytoskele-
adhesion molecules.
cytoskeleton and cell adhesion sites leads to the genera- tal and signalling proteins during the processes of cell
EXTRACELLULAR MATRIX tion of membrane protrusions and traction forces2. adhesion, spreading and migration. Early studies found
(ECM). The complex, multi- External stimuli that control cell migration are trans- that FAK could be activated by either ECM or growth
molecular material that duced into intracellular biochemical signals through the factors, and that tyrosine phosphorylation of FAK was a
surrounds cells. The ECM
interactions of transmembrane INTEGRINS that bind to rapid event that was associated with the formation of
comprises a scaffold on which
tissues are organized, it provides EXTRACELLULAR MATRIX (ECM) proteins, growth factors focal contacts4. Subsequent studies using knockout
cellular microenvironments and that bind to their cognate cell-surface receptors, or mice revealed that null mutation of FAK resulted in
it regulates various cellular mechanical stimuli such as shear stress that promote defective developmental morphogenesis5. As FAK-null
functions. deformation of the actin cytoskeleton. For a cell to fibroblasts show excessive, rather than decreased (as was
process these different environmental motility-pro- initially predicted), formation of focal contacts, FAK
moting stimuli correctly, there must be essential intra- signalling has been associated with the disassembly of
cellular signalling proteins that function as ‘integrators’ integrin-based adhesion sites6. The loss of FAK expres-
— that is, proteins that are stimulated by multiple sion also disrupts microtubule polarization within
extracellular inputs and that function to regulate cells7, and this phenotype, as well as the defect in focal
multiple signalling pathway outputs3. Here, we contact turnover, has been linked to the FAK-mediated
describe the unique molecular connections of focal regulation of RHO-FAMILY GTPases in cells8. Rho-family
adhesion kinase (FAK) that allow this tyrosine kinase GTPases are molecular ‘switches’ within cells, which
The Scripps Research
Institute, Department of to function as an important receptor-proximal regula- control the formation and disassembly of actin
Immunology, IMM21 10550 tor of cell shape, adhesion and motility. cytoskeletal structures (STRESS FIBRES, LAMELLIPODIA and
North Torrey Pines Road, filopodia) and that function to provide the molecular
La Jolla, California 92037, The complexities of FAK framework that supports directed cell motility.
USA.
FAK was independently identified in 1992 by Steve In both normal and transformed cells, FAK signalling
Correspondence to D.D.S.
e-mail: dschlaep@scripps.edu Hanks, Jun-Lin Guan and Michael Schaller as a sub- can promote increased cell motility. The genomic desig-
doi:10.1038/nrm1549 strate of the viral Src oncogene and, in normal cells, as a nation of human FAK is protein-tyrosine kinase-2

56 | JANUARY 2005 | VOLUME 6 www.nature.com/reviews/molcellbio

© 2005 Nature Publishing Group


REVIEWS

many studies have shown that FAK inhibition blocks the


Box 1 | Molecular architecture of focal contacts
response to cell motility cues18, recent and provocative
studies have shown that inhibition of FAK expression or
activity resulted in increased carcinoma cell migration
through the dissolution of N-cadherin-mediated cell–cell
Actin stress fibres
contacts in HeLa CELLS19.
It is possible that this latter observation might be a
cell-type-specific signalling event. However, we specu-
late that the ability of FAK to promote both the matura-

in
tion and turnover of focal contacts is related to its role as

os
My
both a signalling kinase and as an adaptor/scaffold pro-
in

in
os

os
tein, which places FAK in a position to modulate various
My

My
intracellular signalling pathways (FIG. 1). Namely, it is the
association of FAK with both activators and/or
Zyxin
α -A
inhibitors of various small GTPase proteins (Rho, Rac,
c tin α -A Cdc42 and Ras) that enables changes in FAK activity to
in c tin
Vi
nc

in be connected to alterations in the polymerization or sta-


Vi

α -A
ul

nc

α -A
in

ul

c tin bilization of actin and microtubule filaments.


in

in c tin
in Focal contact Additionally, because migrating cells experience changes
proteins
Vi

FAK FAK in forces through integrin contacts that link the ECM
nc
ul

illin P ax with the cytoskeleton, FAK is important in the ‘sensing’


in

Pax in p130 p130 ill


Tali in
Plasma Tl
a Src Cas Cas Src n of mechanical forces that are either generated internally
membrane
or exerted on cells20. FAK activation is therefore involved
in modulating ‘corrective’ cell responses to environmen-
tal stimuli. FAK does this through signal-mediated
effects on actin polymerization, the assembly or disas-
Integrins sembly of focal contacts, and the regulation of protease
activation or secretion16,21,22.
α β β α
The FERM and FAT domains of FAK
Extracellular matrix
FAK is a ubiquitously expressed 125-kDa protein tyro-
sine kinase that is composed of an N-terminal FERM
(protein 4.1, ezrin, radixin and moesin homology)
The extracellular matrix, integrins (α- and β-transmembrane heterodimeric proteins)
domain, a central kinase domain, proline-rich regions
and the cell cytoskeleton interact at sites called focal contacts. Focal contacts are dynamic
groups of structural and regulatory proteins that transduce external signals to the cell and a C-terminal focal-adhesion targeting (FAT)
interior and can also relay intracellular signals to generate an activated integrin state at domain (FIG. 2). The FERM domain of FAK facilitates a
the cell surface113. The integrin-binding proteins paxillin and talin recruit focal adhesion signalling linkage from receptor tyrosine kinases such as
kinase (FAK) and vinculin to focal contacts (see figure). α-Actinin is a cytoskeletal the epidermal growth factor receptor (EGFR) and the
protein that is phosphorylated by FAK, binds to vinculin and crosslinks actomyosin platelet-derived growth factor receptor (PDGFR)23. In
stress fibres and tethers them to focal contacts. Zyxin is an α-actinin- and stress-fibre- analysing cell-motility-promoting signals that are initi-
binding protein that is present in mature contacts. Although the aforementioned proteins ated by G-PROTEIN-COUPLED RECEPTORS (GPCRs), overexpres-
are found in most focal contacts, the membrane-associated protein tyrosine kinase Src sion of the FAK FERM domain blocked FAK activation
and the ADAPTOR PROTEIN p130Cas associate with focal contacts following integrin and resulted in the inhibition of G-protein-stimulated
clustering. Integrin-mediated FAK activation is mediated in part by matrix binding or by cell migration24. It is the FAK FERM domain that can
force-dependent changes in cytoskeletal linkages. Several other proteins such as bind to and promote the integrin- and FAK-mediated
extracellular signal-regulated kinase 2 (ERK2) and calpain are known to be transiently activation of other non-receptor tyrosine kinases such as
present at focal contacts (not shown). The composition of a focal contact is therefore ETK25. Additionally, actin- and membrane-associated
constantly varying depending on external cues and cellular responses. adaptor proteins such as ezrin can bind to the FAK
FERM domain and facilitate increased FAK activation in
an integrin-independent manner26.
(PTK2) and it is located at human chromosome How the FAK FERM domain associates with various
RHO-FAMILY GTPases 8q24term, a locus that is subject to amplification in targets is an active area of research. FAK can become
A subfamily of small (~21 kDa) human cancer cells9. Furthermore, elevated levels of post-translationally modified by the covalent addition
GTP-binding proteins that are
PTK2 mRNA have been found in studies of human car- of a small ubiquitin-related modifier (SUMO) at the
related to Ras and that regulate
the cytoskeleton. The cinoma tumours and in acute lymphoblastic leukaemias, ε-amino position of Lys152 (REF. 27). In most instances,
nucleotide-bound state is as detected by large-scale gene expression profiling10,11. sumoylation is associated with the nuclear import of
regulated by GTPase-activating FAK protein expression is elevated in many highly proteins and, correspondingly, sumoylated FAK was
proteins, which catalyse malignant human cancers12, and studies have shown enriched in the nuclear fraction of cells27. Although
hydrolysis of the bound GTP,
and guanine nucleotide-
that FAK signalling can promote changes in cell shape13,14 blocking nuclear export using leptomycin B promotes
exchange factors, which catalyse and the formation of podosomes or invadopodia15, the nuclear accumulation of FAK, and exogenous
GDP–GTP exchange. which leads to an invasive cell phenotype16,17. Whereas expression of the FAK FERM domain exhibits strong

NATURE REVIEWS | MOLECUL AR CELL BIOLOGY VOLUME 6 | JANUARY 2005 | 5 7

© 2005 Nature Publishing Group


REVIEWS

directly to the cytoplasmic tails of integrins33, accu-


Extracellular
matrix Growth mulated evidence supports an indirect association of
α β factor receptors FAK with integrins through binding to integrin-
Integrins associated proteins such as paxillin and talin18. The
FAK FAT domain also binds directly to an activator
of Rho-family GTPases that is known as p190
RhoGEF, and FAK-mediated tyrosine phosphoryla-
tion of p190 RhoGEF might be a direct link to RhoA
activation34.
Assembly
Cadherins FAK activation and phosphorylation
Focal FAK The best-characterized FAK phosphorylation event is
contacts
AUTOPHOSPHORYLATION at Tyr397, which can occur in
Disassembly
?
either cis or trans 35. Phosphorylation of FAK at Tyr397
Rho-family GTPases
creates a motif that is recognized by various SH2-DOMAIN-
containing proteins, such as SRC-FAMILY KINASES (SFKs),
mDia RhoA Rac Cdc42 phospholipase Cγ (PLCγ), suppressor of cytokine sig-
nalling (SOCS), growth-factor-receptor-bound protein-7
(GRB7), the Shc adaptor protein, p120 RasGAP, and
Microtubule Stress Lamellipodia Filopodia
stabilization fibres the p85 subunit of phosphatidylinositol 3-kinase
(PI3K)4,18,31,33 (FIG. 2). It is not known whether these
different signalling proteins differentially bind to
Cell migration Tyr397-phosphorylated FAK in response to particular
cell stimuli or whether simultaneously there are differ-
Figure 1 | Focal adhesion kinase integrates signals to promote cell migration. Focal
ent complexes with a larger pool of activated FAK. In
adhesion kinase (FAK) is activated by growth factors and integrins during migration, and functions as
a receptor-proximal regulator of cell motility. At contacts between cells and the extracellular matrix,
this respect, we favour a sequential association model
FAK functions as an adaptor protein to recruit other focal contact proteins or their regulators, which whereby the binding of cellular Src (hereafter referred to
affects the assembly or disassembly of focal contacts. FAK activity and downstream signalling can as Src) to FAK initiates signalling (discussed below) and
promote changes in actin and microtubule structures, and FAK signalling can affect the formation the association of SOCS with FAK is a terminal event
and disassembly of cell–cell (cadherin-based) contacts. The Rho-family GTPases (RhoA, Rac and that leads to ubiquitin-mediated degradation of FAK36.
Cdc42) direct local actin assembly into stress fibres, lamellipodia and filopodia, respectively. FAK can For integrin-, growth factor- and G-protein-linked
influence the activity of Rho-family GTPases through a direct interaction with, or phosphorylation of,
protein activators or inhibitors of Rho GTPases. RhoA can also influence the stability of microtubules
stimuli that promote cell motility, it is the transient
through its effector Diaphanous (mDia). recruitment of SFKs into a signalling complex with FAK
that is one of the first events associated with FAK activa-
tion18. Proline-rich tyrosine kinase-2 (PYK2) is related
to FAK and shares a similar domain structure (FERM,
nuclear localization28, it is not known whether these kinase, proline-rich and FAT domains) as well as com-
events are dependent on sumoylation. As sumoylated mon phosphorylation sites (BOX 2). The binding of SFKs
FAK showed elevated activity27, and FAK signalling has to PYK2 that is phosphorylated at Tyr402 is also associ-
been linked to enhanced gene transcription29 and cell- ated with PYK2 activation. However, FAK and PYK2
STRESS FIBRES
cycle progression30, it is possible that sumoylation of possess distinct signalling roles in cells, partly owing to
Also termed ‘actin- FAK might facilitate a direct signalling route between differential binding of target proteins to the FERM and
microfilament bundles’, these are focal contacts and the nucleus. FAT domains of FAK and PYK2, respectively.
bundles of parallel filaments that The C-terminal domain of FAK contains two Additionally, PYK2 is preferentially expressed in cells of
contain F-actin and other
proline-rich regions that function as binding sites for the endothelium, central nervous system and
contractile molecules, and often
SRC-HOMOLOGY (SH)3-DOMAIN-containing proteins (FIG. 2). haematopoietic lineages; PYK2 activation is sensitive
stretch between cell attachments
as if under stress. SH3-domain-mediated binding of the adaptor protein to intracellular Ca2+ signals; and PYK2 is only weakly
p130Cas to FAK is important in promoting cell migra- activated in response to the binding of α5β1-integrin
LAMELLIPODIA tion through the coordinated activation of Rac at mem- to fibronectin, whereas FAK is strongly activated37.
Broad, flat protrusions at the
leading edge of a moving cell
brane extensions31,32. The SH3-mediated binding of The difference in α5β1-mediated activation of FAK
that are enriched with a other proteins, such as GRAF (GTPase regulator associ- versus PYK2 is directly related to the FAT-mediated
branched network of actin ated with FAK) and ASAP1 (Arf GTPase-ACTIVATING PROTEIN localization of FAK at focal contacts compared with a
filaments. (GAP) containing SH3, ankyrin repeat and pleckstrin perinuclear distribution of PYK2 in cells38. Although
homology (PH) domains-1), connects FAK to the regu- PYK2 and FAK can bind SFKs and can activate com-
HeLa CELLS
An established tissue-culture lation of cytoskeletal dynamics and focal contact assem- mon signalling pathways, the differential binding
strain of human epidermoid bly. However, the downstream connections of GRAF activities of the FERM and FAT domains might limit
carcinoma cells, containing and ASAP1 remain undefined4. the functional redundancy of these PTKs in cells.
70–80 chromosomes per cell. The C-terminal domain of FAK also encompasses The activity of FAK is dependent on integrin-
These cells were originally
derived from tissue taken from a
the FAT region, which promotes the colocalization of mediated cell adhesion. Models of integrin-mediated
patient named Henrietta Lacks FAK with integrins at focal contacts (BOX 1). Whereas intermolecular FAK activation are based on the fact that
in 1951. it was first hypothesized that FAK might bind FAK mutants can compete for integrin association and

58 | JANUARY 2005 | VOLUME 6 www.nature.com/reviews/molcellbio

© 2005 Nature Publishing Group


REVIEWS

p120 RasGAP, thereby reinforced the role of integrins in the regu-


GRB7, Shc, PLCγ, GRB2, p190 RhoGEF, lation of FAK signalling. Results showing that FAK
p85, Src, SOCS talin, paxillin
Ezrin
catalytic activity can be modulated by either post-
PDGF receptor, translational or mutational changes in activation-loop
ETK, P P P P P residues are consistent with crystal structure analysis of
EGF receptor
Tyr397 Tyr576 Tyr577 Tyr861 Tyr925 the ATP-bound kinase domain of FAK, which shows a
disordered activation-loop conformation41. As the
FAK FERM Kinase domain FAT
crystal structure of the kinase domain of FAK also
Lys152 PRR1 FIP200 PRR2 PRR3 showed the presence of an unusual disulphide bond
SUMO between Cys456 and Cys459 in a regulatory region,
p130Cas, ASAP1, GRAF
conformational changes or protein-binding interac-
Figure 2 | Focal adhesion kinase domain structure and phosphorylation sites. Focal tions might also function to modulate the activation
adhesion kinase (FAK) contains a FERM (protein 4.1, ezrin, radixin and moesin homology) state of FAK.
domain, a kinase domain and a focal adhesion targeting (FAT) domain. The FERM domain This model is supported by findings that cellular
mediates interactions of FAK with the epidermal growth factor (EGF) receptor, platelet-derived
proteins such as FAK-interacting protein of 200 kDa
growth factor (PDGF) receptor, the ETK tyrosine kinase and ezrin, and the FERM domain can be
conjugated to SUMO (small ubiquitin-related modifier) at Lys152. The FAT domain recruits FAK to
(FIP200) bind to the kinase domain of FAK and inhibit
focal contacts by associating with integrin-associated proteins such as talin and paxillin. It also FAK activity42. Additionally, evidence is accumulating
links FAK to the activation of Rho GTPases by binding to guanine nucleotide-exchange factors that intramolecular constraints also have a role in the
(GEFs) such as p190 RhoGEF. FAK contains three proline-rich regions (PRR1–3), which bind Src- regulation of FAK activity. There are alternatively spliced
homology-3 (SH3) domain-containing proteins such as p130Cas, the GTPase regulator isoforms of FAK in which amino-acid additions sur-
associated with FAK (GRAF) and the Arf-GTPase-activating protein ASAP1. FAK is rounding the Tyr397 site promote a change in the
phosphorylated (P) on several tyrosine residues, including Tyr397, 407, 576, 577, 861 and 925.
kinetics of FAK activation (as measured by Tyr397
Tyrosine phosphorylation on Tyr397 creates a Src-homology-2 (SH2) binding site for Src,
phospholipase Cγ (PLCγ), suppressor of cytokine signalling (SOCS), growth-factor-receptor- phosphorylation) from a primarily trans-intermolecular
bound protein 7 (GRB7), the Shc adaptor protein, p120 RasGAP and the p85 subunit of reaction to a cis-intramolecular reaction35. Although
phosphatidylinositol 3-kinase (PI3K). Phosphorylation of Tyr576 and Tyr577 within the kinase alternative splicing of FAK does not alter the FERM
domain is required for maximal FAK catalytic activity, whereas the binding of FAK-family domain residues of FAK, truncation or removal of the
interacting protein of 200 kDa (FIP200) to the kinase region inhibits FAK catalytic activity. FAK FAK FERM domain does result in enhanced FAK cat-
phosphorylation at Tyr925 creates a binding site for GRB2.
alytic activity35. As binding of proteins such as ezrin or
the GUANINE NUCLEOTIDE-EXCHANGE FACTOR (GEF) TRIO to
the FAK FERM domain result in enhanced FAK activ-
can inhibit endogenous FAK activity4 and that kinase- ity26,43, and as the FAK FERM domain can bind in trans
inactive FAK can become transphosphorylated on to the FAK catalytic domain, resulting in the inhibition
Tyr397 in cells23. FAK Tyr397 phosphorylation pro- of FAK activity44, it is possible that binding interactions
motes Src binding, which leads to the conformational or conformational changes in the FAK FERM domain
activation of Src and results in a dual-activated FAK–Src might function to release cis-inhibitory constraints on
signalling complex18. Within this FAK–Src complex, Src FAK catalytic activation.
ADAPTOR PROTEINS
Proteins that augment cellular phosphorylates FAK at Tyr861, and this is associated The activity of FAK can also be modulated posi-
responses by recruiting other with an increase in SH3-domain-mediated binding of tively45 or negatively 46 by the action of protein-tyrosine
proteins to a complex. They p130Cas to the FAK C-terminal proline-rich regions39. phosphatases (PTPs). Studies using PTPα-deficient
usually contain several Activated Src also phosphorylates FAK at Tyr925, which fibroblasts showed that this phosphatase was required
protein–protein interaction
domains.
creates an SH2-binding site for the GRB2 adaptor pro- for maximal stimulation of Src catalytic activity by
tein. GRB2 binding to FAK is one of several connections β1-integrins, and that PTPα functioned as an upstream
G-PROTEIN-COUPLED that lead to the activation of Ras and the extracellular regulator of FAK Tyr397 phosphorylation45. This result
RECEPTOR signal-regulated kinase-2 (ERK2)/mitogen-activated is consistent with the potential intermolecular activa-
A seven-helix membrane-
protein kinase (MAPK) cascade18. ERK2 phosphoryla- tion of FAK by Src. As Src can also become activated
spanning cell-surface receptor
that signals through tion and the subsequent activation of myosin light chain through direct interaction with the cytoplasmic
heterotrimeric GTP-binding and kinase can modulate focal contact dynamics in motile domains of β-integrins47, these types of result reinforce
GTP-hydrolysing G-proteins to cells3, as well as generate both proliferative and survival the fact that Tyr397 phosphorylation of FAK might not
stimulate or inhibit the activity signals inside cells31. always reflect FAK catalytic activity that is mediated by
of a downstream enzyme.
autophosphorylation.
SRC-HOMOLOGY (SH)3-DOMAIN Regulation of FAK catalytic activity
A protein sequence of 50 amino Src-mediated transphosphorylation of FAK within the p130Cas and paxillin as targets of FAK
acids that recognizes and binds kinase domain ACTIVATION LOOP at Tyr576 and Tyr577 In addition to promoting maximal FAK activation, the
sequences that are rich in proline.
promotes maximal FAK catalytic activation31. Mutation recruitment of Src into a FAK–Src signalling complex
GTPase-ACTIVATING PROTEIN
of FAK within this loop produces FAK mutants with functions to facilitate the phosphorylation of various
(GAP). A protein that stimulates either enhanced or refractory activities. One such FAK-associated proteins, as many FAK targets are also
the intrinsic ability of a GTPase mutant, ‘superFAK’, contains a Lys to Glu substitution independent binding partners and phosphorylation tar-
to hydrolyse GTP to GDP. at residues 578 and 581, and results in a FAK protein gets of Src. Two of the best-characterized target proteins
Therefore, GAPs negatively
regulate GTPases by converting
with adhesion-independent activity40. However, the of FAK–Src-mediated phosphorylation are p130Cas and
them from active (GTP-bound) phosphorylation of downstream targets in superFAK- paxillin31,32,48. SH3-mediated binding of p130Cas to FAK
to inactive (GDP-bound). expressing cells remained adhesion dependent, which is linked to enhanced tyrosine phosphorylation of

NATURE REVIEWS | MOLECUL AR CELL BIOLOGY VOLUME 6 | JANUARY 2005 | 5 9

© 2005 Nature Publishing Group


REVIEWS

of FAK with β1-integrin and the localization of FAK to


Box 2 | The FAK-related kinase PYK2
focal contacts38. Paxillin binding is mediated by two
Src GRB2 leucine-rich peptide regions in paxillin that are known
↑ ↑
Tyr397 Tyr576 Tyr577 Tyr861 Tyr925 as LD MOTIFS, which interface with hydrophobic surface
grooves on the FAT domain54,55. Interestingly, the SH2
FAK FERM Kinase domain FAT
binding site for GRB2 at FAK Tyr925 partially overlaps
PRR1 PRR2 PRR3 with one of the two paxillin LD-motif binding sites in
Sequence the FAT domain54, and localization studies of phospho-
similarity <10% ~40% ~60% ~40%
rylated FAK have shown that Tyr925-phosphorylated
FAK might be selectively excluded from focal contact
PYK2 FERM Kinase domain FAT sites56. Overexpression of a Tyr925Phe mutant of FAK
PRR1 Tyr402 Tyr579 Tyr580 PRR2 PRR3 Tyr881 resulted in strong focal contact distribution56, and in
↓ ↓ activated Src-expressing cells, Tyr925Phe FAK blocks
Src GRB2 the turnover of focal contacts (V. Brunton, personal
Proline-rich tyrosine kinase-2 (PYK2) shares a similar domain arrangement with focal communication; see note added in proof). As NMR
adhesion kinase (FAK) (see figure), with 60% sequence identity in the central kinase analyses have shown that the FAT domain can
domain, conservation of proline-rich regions (PRRs), and identical positions of four undergo conformational rearrangements that might
tyrosine phosphorylation sites. PYK2 tyrosines 402, 579, 580 and 881 correspond to FAK selectively promote either Tyr925 phosphorylation
tyrosines 397, 576, 577 and 925, respectively. Phosphorylation of PYK2 Tyr402 and and/or paxillin binding57, it is possible that Src-medi-
Tyr881 create Src-homology-2 (SH2) binding sites for Src and growth-factor-receptor- ated phosphorylation of FAK on Tyr925, and subse-
bound-2 (GRB2), respectively. PYK2 contains a C-terminal focal adhesion targeting quent GRB2 binding, could displace paxillin, promote
(FAT) domain that binds to paxillin53. However, PYK2 shows perinuclear distribution the dissociation of FAK from focal contacts, and sub-
and is not strongly localized to focal contacts in many cells37. The substitution of the FAK sequently lead to focal contact turnover through
C-terminal domain to PYK2 facilitated the colocalization of this PYK2–FAK chimaera to undefined mechanisms (FIG. 3).
β1-integrin-containing focal contacts38, which indicates that there are biologically relevant Ser910 within the FAT domain is phosphorylated
binding differences between FAK and PYK2. For instance, the FAK C-terminal domain during mitosis58 and after growth factor stimulation of
uniquely binds the integrin-associated protein talin114, and PYK2 — but not FAK — cells. Ser910 is phosphorylated by ERK2 and this is also
binds the actin-associated protein gelsolin115. Although PYK2 can be activated by associated with reduced paxillin binding to FAK59. So,
integrins, this is dependent on integrin-mediated activation of Src-family kinases116,117. Src-mediated phosphorylation of Tyr925 on FAK and
The 40% sequence similarity between the N-terminal FERM (protein 4.1, ezrin, radixin GRB2 binding leading to ERK2 activation, coupled
and moesin homology) domains of PYK2 and FAK also accounts for differential association
with the feedback of ERK2-mediated Ser910 phospho-
with target proteins118.What remains unknown is why PYK2 activity is highly dependent
rylation, could potentiate the release of FAK from focal
on intracellular Ca2+ levels and how PYK2 associates with members of the Janus kinase
contacts. Alternatively, FAK–Src-mediated phosphory-
family37,119 — properties that are not shared by FAK.As PYK2 regulates several signalling
lation of paxillin at Tyr118 promotes the binding of
events that are crucial for macrophage120 and monocyte morphology121, and the Pyk2-null
phenotype results in a MARGINAL ZONE B-cell developmental defect122, there is probably a ERK2 to paxillin60, and ERK2-mediated phosphoryla-
unique role for PYK2 in mediating haematopoietic cell responses to chemokine stimuli. tion of paxillin can facilitate FAK binding to paxillin
and can enhance FAK activation60,61. Therefore, we
speculate that there might be a regulatory cycle in
which FAK–Src activation and signalling to ERK2 can
p130Cas at multiple sites, which promotes SH2-medi- function first to promote FAK release from existing
ated binding of the Crk adaptor protein to p130Cas. focal contacts and then, through ERK2-mediated phos-
Signalling downstream of p130Cas results in increased phorylation of paxillin, to promote FAK re-binding
activity of Rac, enhanced MEMBRANE RUFFLING or lamel- and activation at new or different focal contacts in a
lipodia formation, and the promotion of cell motility or migrating cell (FIG. 3).
invasion17,49,50 (FIG. 3). Paxillin is phosphorylated by
AUTOPHOSPHORYLATION
The transfer of a phosphate FAK–Src on Tyr31 and Tyr118, and this can also pro- Regulation of focal contact dynamics
group by a protein kinase either mote SH2-mediated binding of Crk to paxillin48,51. Lessons from FAK–/– and SHP2 –/– cells. In analyses mon-
to a residue in the same kinase Overexpressing paxillin that is mutated at these phos- itoring the formation of focal contacts, FAK was found
molecule (cis) or to a residue in a phorylation sites inhibits the turnover of focal contacts6 to be one of the first signalling proteins to be recruited to
different kinase molecule but of
the same type (trans).
and cell motility52, which therefore supports the pres- these sites62. Although FAK recruitment to focal con-
ence of multiple routes for FAK–Src-mediated sig- tacts is associated with increased FAK tyrosine phos-
SH2 DOMAIN nalling in modulating the dynamics of cell adhesion phorylation63, focal contacts readily form in FAK-null
A protein motif that recognizes sites. (FAK–/–) fibroblasts, which indicates that FAK activity is
and binds tyrosine-
not essential for the process of focal-adhesion forma-
phosphorylated sequences, and
thereby has a key role in relaying Regulated targeting of FAK to focal contacts tion5. However, focal contacts in FAK–/– cells form pri-
cascades of signal transduction. It is the C-terminal FAT domain of FAK that facilitates marily around the cell periphery, enmeshed in a cortical
the linkage to integrins and focal contacts. The FAT actin ring, and do not undergo a normal maturation
SRC-FAMILY KINASES domain adopts a four-helix bundle structure that con- cycle64. In normal fibroblasts, peripheral immature focal
Kinases that belong to the Src
family of tyrosine kinases, the
tains binding sites for integrin-associated proteins such contacts become connected to longitudinal stress fibres
largest of the non-receptor- as paxillin53. Point mutations in the FAT domain of FAK in cells and undergo actin contractility-mediated matu-
tyrosine-kinase families. that disrupt paxillin binding also prevent the association ration during cell polarization63. FAK re-expression in

60 | JANUARY 2005 | VOLUME 6 www.nature.com/reviews/molcellbio

© 2005 Nature Publishing Group


REVIEWS

phosphatase SHP2 (SH2-domain-containing protein


α β
Migration Integrins
tyrosine phosphatase 2). SHP2–/– cells have increased
Membrane
FAK activity, but they also show an accumulation of
Rac immature focal contacts and similar refractory migra-
tion defects to FAK–/– cells67. Hyperactive FAK in SHP2–/–
Src
Crk cells results in an increase in the levels of Tyr12-phos-
Focal contacts phorylated α-actinin, which thereby reduces the
SH2 p130Cas P
crosslinking of stress fibres and prevents the maturation
P P P SH3
of focal contacts68. In SHP2–/– cells, there is a high level of
Tyr397 Tyr576 Tyr577 P-X-X-P Tyr925
Paxillin focal contact turnover68, whereas in FAK–/– cells, focal
FAK FERM Kinase domain FAT ERK2 contact turnover and maturation are inhibited8. So, both
P Tyr118 SHP2–/– and FAK–/– cells show an accumulation of
P immature focal contacts, but through different mecha-
nisms. These studies support the importance of FAK
Release expression and the precise temporal regulation of
ERK2
FAK activity as important factors that control the
dynamics of focal contacts.
P GRB2
Tyr925 Paxillin–/–, p130Cas–/– and Src –/– cells. In addition to
FAK FERM Kinase domain FAT FAK–/– and SHP2–/– cells, fibroblasts that contain null
mutations for various other focal-contact-associated
Ser910
proteins also show altered dynamics of focal contact
P
maturation, cell spreading defects and refractory cell
motility responses (TABLE 1). Time-lapse analyses showed
Figure 3 | Focal adhesion kinase (FAK)–Src signals that regulate cell motility and focal
contact localization. Integrin clustering promotes FAK autophosphorylation (P) at Tyr397, which
that the incorporation of labelled paxillin into focal con-
creates a binding site for the Src-homology (SH)2 domain of Src. Src-mediated phosphorylation tacts of FAK–/–, paxillin–/–, p130Cas–/– or SYF–/– (Src–/–,
of FAK at Tyr576 and Tyr577 promotes maximal FAK catalytic activity. Active FAK–Src facilitates Yes–/– and Fyn–/–) cells did not significantly differ com-
SH3-mediated binding of p130Cas to FAK and its subsequent phosphorylation. Crk binding to pared to normal fibroblasts6. However, the rate of focal
phosphorylated p130Cas facilitates Rac activation, lamellipodia formation and cell migration. contact disassembly was much slower in these null cells.
Paxillin binding to the FAK focal adhesion targeting (FAT) domain is important for FAK focal Analyses of FAK–/– cells showed that disassembly was
contact localization. Src-mediated phosphorylation of FAK at Tyr925 creates an SH2 binding site
for the growth-factor-receptor-bound protein 2 (GRB2) adaptor protein, which leads to the
dependent on FAK Tyr397 phosphorylation; SYF–/– cells
activation of Ras and the extracellular signal-regulated kinase-2 (ERK2) cascade. The GRB2 and showed that Src kinase activity was required; and studies
paxillin binding sites within the FAT domain overlap and Tyr925-phosphorylated FAK might be with paxillin–/– cells indicated that the integrity of the
selectively released from focal contacts. ERK2 activation promotes FAK phosphorylation at Tyr31 and Tyr118 paxillin phosphorylation sites were
Ser910, which is also associated with decreased paxillin binding to FAK. Within focal contacts, needed to promote focal contact turnover6. As the
FAK–Src-mediated phosphorylation of paxillin at Tyr118 promotes ERK2 binding. ERK2-mediated expression of constitutively active Src in FAK–/– cells can
phosphorylation of paxillin can facilitate FAK binding to paxillin and enhances FAK activation. So,
promote focal contact turnover and increased cell
there might be a cycle whereby Src- and ERK2-mediated phosphorylation of FAK promotes its
release from focal contacts and ERK2-mediated phosphorylation of paxillin promotes the motility17,69, these combined analyses support the con-
association of unphosphorylated FAK with paxillin at new or growing focal contact sites. clusion that, in normal cells, integrin- and FAK-medi-
ated control of Src activity is a key event that promotes
focal contact dynamics.

FAK–/– cells promotes the reorganization of the ‘imma- FAK–Src and proteolysis. In addition to signalling events
ture’ focal contacts, which allows for their connection to that are associated with the phosphorylation of α-actinin,
MARGINAL ZONE
actin stress fibres, therefore mediating cell contractility p130Cas or paxillin, FAK–Src signalling can affect focal
A region in the spleen in which
white blood cell precursors such and cell polarization64. contact dynamics through the regulation of both
as B-cells, granulocytes, Mechanistically, these alterations in focal contacts extracellular and intracellular proteolytic events.
macrophages and plasma-cells and actin structures involve the regulation of the activity Inhibition of FAK activity in human carcinoma cells or
reside or transit through during of α-actinin, a protein that promotes actin crosslinking Src-transformed cells, or stable FAK re-expression in
primary or secondary immune
responses.
and that has an important role in maintaining the link- FAK–/– cells, can alter the expression and activation of
age between focal contacts and stress fibres1,65 (FIG. 4). MATRIX METALLOPROTEINASES (MMPs)
16,17,22
. The influence
ACTIVATION LOOP FAK phosphorylates α-actinin at Tyr12, which results in of FAK on MMP regulation is associated with sig-
A conserved structural motif in reduced α-actinin binding to actin66. α-Actinin is not nalling from Ras to ERK2 and from Rac to Jun N-ter-
kinase domains, which needs to
phosphorylated in FAK–/– cells, so this FAK signalling minal kinase (JNK). Activation of MMPs at the LEADING
be phosphorylated for full
activation of the kinase. linkage to α-actinin might underlie some of the matu- EDGE of migrating cells functions to promote matrix
ration defects, as well as turnover dynamics, of focal proteolysis, which leads to the extracellular release of
GUANINE NUCLEOTIDE- contacts in FAK–/– cells66. It is possible that the lack of integrin–matrix contacts and thereby facilitates focal
EXCHANGE FACTOR α-actinin phosphorylation might be associated with the contact turnover70.
A protein that facilitates the
exchange of GDP for GTP in the
presence of focal contacts enmeshed in a cortical actin The intracellular linkage of focal contacts to the
nucleotide-binding pocket of a ring at the FAK–/– cell periphery. This hypothesis is fur- actin cytoskeleton is also regulated by calpain-mediated
GTP-binding protein. ther supported by studies of cells that lack the tyrosine proteolysis71. Calpain can cleave constituents of focal

NATURE REVIEWS | MOLECUL AR CELL BIOLOGY VOLUME 6 | JANUARY 2005 | 6 1

© 2005 Nature Publishing Group


REVIEWS

Actin stress fibres

Actin stress fibres

Myosin Myosin

Assembly

Myosin Myosin
MLCK ↑
ARP2/3 Reduced
α-actinin
Increased MLC crosslinking
α-actinin phosphatase
crosslinking in
ctin
α-A
in ROCK
Cdc42
P ctin
α-A Tyr12
Tyr256 P
in Tyr256
ctin Rho GRAF Rho
Cdc42 N-WASP α-A p190 p190
RhoGEF N-WASP RhoGEF
FAK FAK
P
Tyr256

N-WASP Focal contact Focal contact


Nucleus

Figure 4 | Focal adhesion kinase promotes cytoskeletal fluidity. Stress fibres and cortical actin are continuously
destabilized/stabilized by focal adhesion kinase (FAK)-regulated processes. Normally, the actin cytoskeleton exists in a semi-solid
state, owing to a high degree of α-actinin-mediated crosslinking of stress fibres, which are tethered and exert tension at focal
contacts (left panel). Conversion to a more soluble state (right panel) is promoted by FAK phosphorylation (P) on Tyr12 of α-actinin,
which results in reduced crosslinking and the release of actin stress fibres from focal contacts. Cytoskeletal fluidity is also regulated
by the effects of FAK on Rho-family GTPases and on the neuronal Wiskott–Aldrich syndrome protein (N-WASP). FAK
phosphorylates Cdc42-activated N-WASP at Tyr256, thereby retaining phosphorylated N-WASP in the cytoplasm where it can
affect ARP2/3-mediated actin polymerization. Through associations with Rho GTPase-activating proteins (GAPs) and Rho guanine
nucleotide-exchange factors (GEFs), FAK can regulate actomyosin stress fibre polymerization. Reduced tension can be attributed in
part to increased RhoGAP activity of GTPase regulator associated with FAK (GRAF). Conversely, FAK can promote cytoskeletal
tension through phosphorylation and activation of p190 RhoGEF. Subsequent Rho activation indirectly regulates myosin light chain
(MLC) phosphorylation through Rho-associated kinase (ROCK) phosphorylation of MLC phosphatase, which leads to increased
MLC kinase (MLCK) activity through the downregulation of MLC phosphatase activity.
MEMBRANE RUFFLE
A process that is formed by the
movement of lamellipodia that
are in the dynamic process of
folding back onto the cell body contacts, such as talin and FAK, and calpain-4–/– cells FAK effects on GTPases and actin
from which they previously have an increased number of peripheral focal contacts72. The activity of Ras and the Rho-family GTPases Rho,
extended. Calpain is not appropriately activated in FAK–/– cells21, Rac and Cdc42 is positively regulated by GEFs and neg-
and this defect might be due in part to ERK2 activa- atively regulated by GAPs. As mentioned above, a num-
LD MOTIF
A short sequence found within tion being required for calpain function73. Notably, ber of studies have shown that FAK–Src-mediated
proteins that has the consensus FAK re-expression in FAK–/– cells promotes the forma- phosphorylation events can lead to the activation of
sequence LDXLLXXL and tion of a complex between calpain, ERK2 and activated Ras–ERK2 and Rac–JNK signalling cascades to pro-
functions as a protein-binding Src21. Restoration of calpain activity in FAK–/– cells mote increased cell migration and invasion18. In a
interface.
requires specific FAK phosphorylation events: a form recently discovered mechanism, FAK overexpression
MATRIX METALLOPROTEINASES of FAK that is mutated at several phosphorylation sites facilitated the SH2-mediated binding and sequestering
Proteolytic enzymes that can form a complex with calpain and ERK2, but it of p120 RasGAP, which diminished the association of
degrade the extracellular matrix does not restore full calpain activity in contrast to cells p120 RasGAP with active Ras76 and thereby led to Ras
and have important roles in
in which wild-type FAK is re-expressed74. So, FAK sig- activation.
tissue remodelling and tumour
metastasis. nalling is connected to the increased turnover of focal In FAK –/– cells, the intrinsic GTPase activity of
contacts through calpain activation. As calpain is a RhoA is elevated8, and pharmacological inhibitors of
LEADING EDGE Ca2+-dependent protease, and FAK activation is associ- Rho-associated kinase (ROCK) — a substrate of Rho
The thin margin of a ated with local Ca2+-flux-induced disassembly of focal — partially reverse the polarization defects of FAK–/–
lamellipodium that spans the
area of the cell from the plasma
contacts75, the FAK–calpain linkage might be selec- cells77. Integrin signalling can suppress RhoA activity
membrane to a depth of about tively activated at either cell protrusions or tail retrac- by tyrosine phosphorylation of p190 RhoGAP
1 µm into the lamellipodium. tion sites in motile cells. (which increases its GAP activity)78. Likewise, stable

62 | JANUARY 2005 | VOLUME 6 www.nature.com/reviews/molcellbio

© 2005 Nature Publishing Group


REVIEWS

Table 1 | Phenotypes associated with null mutations in focal contact proteins


Cell Embryonic Focal contact Focal contact Integrin-stimulated FAK tyrosine Reference
phenotypes (lethality) day formation turnover migration phosphorylation
FAK–/– (p53–/–) 8.5 Increased immature Inhibited Inhibited NA 5
SYF–/– 9.5 No change Inhibited Reduced pTyr397 reduced 123
p130Cas–/– 11.5–12.5 No change Inhibited Reduced No difference 124
Paxillin–/– 9.5 Increased size Decreased Inhibited pTyr397 reduced 125
Vinculin–/– 10.0 Decreased size ND Stimulated Increased activity 126
–/–
PTPα None Delayed ND Reduced pTyr397 reduced 45
SHP2–/– 8.5–10.5 Increased immature Elevated Inhibited Increased activity 67,68
in suspension
Calpain-4–/– 10.0 Larger Reduced Decreased No change 72
This table summarizes the phenotypes of fibroblasts that are derived from mice that are null for various proteins associated with focal contacts. FAK and paxillin are involved in
the formation of focal contacts, whereas FAK, Src-family kinases (Src, Yes and Fyn; SYF), p130Cas and calpain are also involved in focal contact turnover. Except for vinculin-
null cells, the lack of any of the above proteins results in impaired integrin-stimulated cell migration. FAK, focal adhesion kinase; NA, not applicable; ND, not determined;
PTPα, protein tyrosine phosphatase-α; pTyr, phosphotyrosine; SHP2, Src-homology (SH)2-containing phosphotyrosine phosphatase-2.

FAK re-expression in FAK–/– cells decreased RhoA lipid-raft marker, GANGLIOSIDE GM1, to the leading edge
activity 8 and enhanced p190 RhoGAP tyrosine phos- of motile cells. It is hypothesized that the lipid environ-
phorylation17. In other cell types, FAK activation and ment at the leading edge preferentially localizes micro-
tyrosine phosphorylation are associated with RhoA tubule capping or bridging proteins, which stabilize the
activation and the formation of stress fibres18. This con- association of microtubules with cortical receptors82.
nection could be mediated by FAK binding to, and This regulation of a distinct membrane lipid environ-
phosphorylating, p190 RhoGEF34. In neuronal develop- ment by FAK or integrin signalling83 also functions to
ment, FAK signalling through p190 RhoGEF controls promote Rac signalling by maintaining a suitable lipid
axonal branching and synapse formation79. Although environment that facilitates the interaction of Rac and
FAK-mediated activation of p190 RhoGEF is a direct effectors such as p21-activated kinase (PAK)84.
route to RhoA activation, the formation of distinct sig- Interestingly, FAK-stimulated phosphorylation of
nalling complexes will probably influence whether FAK Ser298 in MAPK/ERK kinase-1 (MEK1, also known as
activation leads to increased or decreased RhoA activity MAPK kinase-1) by PAK is a secondary route that leads
in cells (FIG. 4). to ERK2/MAPK activation85.
In addition to affecting the activity of Ras, Rac and In neuronal cells, a fraction of FAK colocalizes with
Rho, FAK can influence the function of Cdc42 through a distinct microtubule structure that arises from
binding and phosphorylation of the Cdc42 effector microtubule-organizing centres and that extends
Wiskott–Aldrich syndrome protein N-WASP (neuronal around the nucleus in a branched fork-like form
WASP)80. N-WASP, which, in contrast to its name, is termed a microtubule fork86. Microtubule forks are
ubiquitously expressed, regulates the actin cytoskeleton believed to promote nuclear re-positioning in the
through activation of the ARP2/3 COMPLEX3. Interestingly, direction of cell movement. Cyclin-dependent kinase-5
FAK only associates with Cdc42-activated N-WASP, and (CDK5) phosphorylates FAK at Ser732 in post-mitotic
does not itself activate N-WASP. Although FAK neurons, and antibodies that recognize Ser732-phospho-
phosphorylation of N-WASP at Tyr256 does not rylated FAK specifically stain microtubule fork structures
ARP2/3 COMPLEX affect N-WASP activity towards ARP2/3, it does seem near the nucleus86. Neurons that are devoid of CDK5 or
A complex that consists of two
important for maintaining a cytoplasmic distribution of that express a FAK mutant in which Ser732 cannot be
actin-related proteins ARP2 and
ARP3, along with five smaller N-WASP and for promoting cell motility80. As Cdc42 phosphorylated show a malformed microtubule fork,
proteins. When activated, the regulates actin dynamics in cellular projections, the impaired nuclear movement and altered neuronal devel-
ARP2/3 complex binds to the interaction of FAK with Cdc42-activated N-WASP opment positioning in vivo 86. Whereas the molecular
side of an existing actin filament might couple actin polymerization with membrane mechanisms that link FAK Ser732 phosphorylation to the
and nucleates the assembly of a
new actin filament. The resulting
protrusion during cell motility (FIG. 4). localization and organization of microtubule fork struc-
branch structure is Y-shaped. tures remain to be defined, this observation is the first of
FAK and microtubules its kind and supports studies that link FAK phosphoryla-
GANGLIOSIDE Integrating factors coordinate the regulation of micro- tion to enhanced neuronal cell migration87.
An anionic glycosphingolipid
tubule structures and the actin cytoskeleton during cell
that carries, in addition to other
sugar residues, one or more sialic motility. Microtubules are important in the establishment FAK and membrane composition
acid residues. and maintenance of cell polarity, and the Rho effector The polarization of migrating cells requires membrane
Diaphanous (mDia) functions to stabilize microtubules modification as well as changes in the underlying
LIPID RAFTS at the leading edge of migrating cells81. Integrin-mediated cytoskeleton. In addition to the role of FAK in the
Lateral aggregates of cholesterol
and sphingomyelin that are
activation of FAK is required for microtubule stabiliza- translocation of LIPID RAFT components7, FAK–Src sig-
thought to occur in the plasma tion by the Rho–mDia signalling pathway7 (FIG. 5). This is nalling is involved in the modification of phosphatidyli-
membrane. partly the result of the FAK-regulated localization of a nositol lipids, and differentially phosphorylated lipid

NATURE REVIEWS | MOLECUL AR CELL BIOLOGY VOLUME 6 | JANUARY 2005 | 6 3

© 2005 Nature Publishing Group


REVIEWS

phosphorylated by a FAK–Src complex, which facilitates


increased PIPKIγ activity (and, therefore, increased pro-
duction of PtdIns(4,5)P2) and increased PIPKIγ associa-
tion with talin91. In this manner, FAK signalling is con-
nected to the formation of focal contacts and the spatial
PtdIns Formation of α regulation of PtdIns(4,5)P2 generation. However, the
membrane Integrins β integrin and PIPKIγ binding sites within the talin
proximal contacts
PtdIns(4)P FERM domain overlap, which implies that PIPKIγ
Talin Vinculin binding might displace talin from integrin tails92. To this
PIPKIγ
end, FAK-enhanced Src-mediated phosphorylation of
PtdIns(4,5)P2 FAK
Paxillin PIPKIγ on Tyr644 creates a high affinity binding site for
PI3K p85 the talin FERM domain, which displaces β-integrin
PtdIns(3,4,5)P3 binding from talin FERM93. So, although FAK–Src
New Membrane activity could promote the production of PtdIns(4,5)P2
membranes component mDia GM1 and the formation of focal contacts by enhancing the
activity of PIPKIγ, subsequent phosphorylation of
Phospholipid
PIPKIγ by activated Src might break the talin–integrin
Rac

modification by Stable
phosphorylation Lipid lamellipodia linkage and promote the turnover of focal contacts.
rafts
Rac

FAK and intercellular contacts


Increased Another biological context in which FAK signalling
Microtubule
membrane has been associated with the formation or turnover of
fluidity
stabilization contacts is cadherin-based cell–cell junctions.
+
Cadherins are transmembrane proteins that mediate
+
Ca2+-dependent homophilic protein–protein attach-
+
ments between cells and that are also linked to the
actin cytoskeleton through interaction with α- or β-
catenins94. Downregulation of E-cadherin-based
ADHERENS JUNCTIONS is a hallmark of malignant and

Figure 5 | Focal adhesion kinase influences phospholipid and microtubule structures. invasive carcinomas, and the activity of the FAK–Src
The phospholipid kinases that have a role in the modification of phosphatidylinositol (PtdIns) complex promotes the disruption of colon carcinoma
cooperate with focal adhesion kinase (FAK) at several levels. Type I PtdIns phosphate kinase-γ cell homotypic adhesions95. Importantly, expression of
(PIPKIγ) associates with FAK and talin, and promotes the conversion of PtdIns-4-phosphate a FAK protein that is mutated at five tyrosine phospho-
(PtdIns(4)P) to PtdIns-4,5-bisphosphate (PtdIns(4,5)P2). PIPKIγ is phosphorylated by FAK, which
rylation sites (Tyr407, 576, 577, 861 and 925) blocked
leads to increased PIPKIγ activity and increased generation of PtdIns(4,5)P2. The binding of
PtdIns(4,5)P2 to talin and vinculin is associated with the formation of focal contacts. PtdIns(4,5)P2 the Src-mediated disruption of colon carcinoma E-cad-
can be converted to PtdIns-3,4,5-trisphosphate (PtdIns(3,4,5)P3) by PtdIns 3-kinase (PI3K). The herin-based contacts, thereby implying that phosphory-
regulatory p85 subunit of PI3K binds to FAK at Tyr397, which leads to PI3K activation by FAK112. lation-dependent signalling through FAK was
Directional motility requires the generation of phospholipid components such as PtdIns(4,5)P2 and required96. In an opposite manner, overexpression of a
PtdIns(3,4,5)P3. Integrin and FAK signalling also promote the translocation of specific kinase-defective mutant of FAK blocked the accumula-
components of lipid rafts to membranes. The stabilization of lipid rafts through integrin signalling
tion of peripheral E-cadherin in endothelial cells that
facilitates the coupling of Rac to target proteins. FAK-mediated translocation of the lipid
ganglioside GM1 to the membrane, which is mediated through the activation of the Rho GTPase
were subjected to a hyperosmolar challenge (a stimu-
effector Diaphanous (mDia), regulates microtubule polarity at the leading edge of motile cells. lus that promotes increased E-cadherin-based TIGHT-
97
Microtubule polarization and Rac activation contribute to the formation of membrane ruffles and JUNCTION barrier formation) . These results imply that
stable lamellipodia. FAK signalling has a role in both the formation and
turnover of E-cadherin-based contacts.
As opposed to E-cadherin function, N-cadherin
expression in carcinoma cells is generally associated
intermediates function as binding sites for signalling with a scattered morphology and a migratory or inva-
proteins that are involved in the formation of focal con- sive phenotype. Antisense and DOMINANT-NEGATIVE inhibi-
tacts (FIG. 5). Phosphatidylinositol-4,5-bisphosphate tion of FAK showed that FAK expression and activity
ADHERENS JUNCTION (PtdIns(4,5)P2) binds to and controls the assembly of were needed for the formation of N-cadherin-based
A cell–cell adhesion complex proteins such as α-actinin, vinculin and talin into focal cell–cell contacts in HeLa cells19. However, in contradic-
that contains classical cadherins contacts2. As the binding of the talin FERM domain to tion, the above study also found that cells with less FAK
and catenins that are attached to
β-integrin cytoplasmic tails is enhanced by expression and reduced N-cadherin-mediated cell–cell
cytoplasmic actin filaments.
PtdIns(4,5)P2 (REF. 88), and the talin rod domain binds contacts exhibited ‘increased’ motility when plated as
TIGHT JUNCTION vinculin and actin89, a link between integrins, focal individual cells on a collagen matrix. Whereas much
A circumferential ring at the contact formation and the actin cytoskeleton is estab- remains to be determined about the molecular role of
apex of epithelial cells that seals lished. The type I phosphatidylinositol phosphate FAK in either the dissolution or formation of cadherin-
adjacent cells to one another.
Tight junctions regulate solute
kinase-γ (PIPKIγ) is an enzyme that makes based contacts, it is intriguing that the findings so far are
and ion flux between adjacent PtdIns(4,5)P2 and it is targeted to focal contacts by an somewhat similar to the bi-functional role of FAK in
epithelial cells. association with the talin FERM domain90. PIPKIγ is focal contact dynamics.

64 | JANUARY 2005 | VOLUME 6 www.nature.com/reviews/molcellbio

© 2005 Nature Publishing Group


REVIEWS

FAK as a biosensor 2D focal contacts105, functional experiments with


Early studies with integrins found that they ‘sensed’ human lung fibroblasts106, breast carcinoma cells107 or
environmental cues and functioned to control anchorage- normal CYTOTROPHOBLASTS108 have supported the impor-
dependent cell proliferation and survival98. Integrins are tance of FAK Tyr397 phosphorylation in promoting cell
also intimately involved in the conversion of physical survival, cell proliferation and cell invasion, respectively,
signals, such as contractile forces or external mechanical in 3D cell culture. As increased FAK phosphorylation of
perturbations, into chemical signalling events, and FAK Tyr397 is associated with the elevated rigidity of a 3D
activation is an important component of this collagen matrix107, it is likely that factors such as integrin
‘mechanosensing’ by cells20. Early observations showed engagement and cell tension are needed to promote
that FAK could be activated by TANGENTIAL FLUID SHEAR FAK activity in 3D. Integrin signalling is also important
STRESS of endothelial cells and that this was associated in the formation of tumour cell invadopodia — 3D cell
with the formation of a FAK–Src signalling complex, extensions that are enriched in MMPs and that promote
FAK Tyr925 phosphorylation, and the downstream acti- tumour cell invasion through matrix barriers109.
vation of ERK2 and JNK18. In a process known as FAK–Src signalling has an important role in promoting
mechanotaxis, endothelial cells that encounter shear invadopodia formation110 and lung carcinoma cell inva-
stress will initiate focal contact remodelling and cell sion22, in part through the induction of Rac activity17,49.
migration in the direction of the flow. Under these con- FAK–Src signalling leads to the elevated expression and
ditions, phosphorylation of FAK is enhanced at the secretion of MMPs and is associated with a metastatic
leading edge of motile cells99. tumour cell phenotype16. As FAK expression is corre-
Another means of ‘mechanoperception’ is the ability lated with increased tumour cell malignancy12, the
of cells to sense the ‘rigidity’ of the surrounding ECM, as importance of FAK catalytic activity in promoting cell
cells will preferentially migrate towards areas of higher motility and invasion17,64 make it an attractive target for
substrate rigidity in a process known as durotaxis. potential therapeutic intervention.
Studies have shown that this response requires FAK
expression and that FAK–/– cells are insensitive to Conclusions and perspectives
changes in substrate flexibility100. Interestingly, whereas The number of different signalling connections that
expression of a Tyr397Phe mutant of FAK does not res- have been characterized for FAK has expanded greatly
cue the overall migration speed or directional motility over the past 5 years. In this review, we have high-
persistence defects of FAK–/– cells23,64,101, cells expressing lighted the diversity of FAK-signalling inputs and out-
this mutant exhibited similar durotaxis responses to puts and the molecular mechanisms that connect FAK
wild-type cells100. Although it remains to be determined to both the assembly and disassembly of focal contacts.
whether intrinsic FAK catalytic activity or the role of It is in this unique signalling position that FAK can exert
FAK as a scaffolding protein at focal contacts is the control over cytoskeletal or cell adhesion dynamics and,
determining factor for the durotaxis response, this therefore, cell motility.
observation remains one of the few examples in which Continued efforts will be needed to understand the
Tyr397 FAK phosphorylation was not required for a sig- regulatory factors that influence whether FAK–Src sig-
nalling response. nalling is coupled to the assembly or disassembly of cell
adhesion sites in 2D and 3D, and how FAK targeting to
Moving in three dimensions different cellular locations influences these processes. As
As adherent cells in culture readily make focal contacts, such, it is likely that the many interactions and phospho-
these points of cell adhesion undergo a maturation rylation events that are associated with FAK are tran-
DOMINANT NEGATIVE process to form fibrillar adhesions and they will form sient events and possibly occur in a defined sequence as
A defective protein that retains three-dimensional (3D) adhesions when cells are placed adhesions transit from assembly to disassembly during
interaction capabilities and so in a 3D environment102. FAK–/– fibroblasts fail to prop- cell migration. Additionally, as structural studies have
competes with normal proteins,
erly remodel focal contacts into fibrillar adhesions103 provided important information about the function of
thereby impairing protein
function. and FAK–/– endothelial cells do not form tubule struc- the FAT domain of FAK, similar studies are needed to
tures when grown in a 3D matrix environment13. understand the role of the FAK FERM domain. The
TANGENTIAL FLUID SHEAR Interfering with FAK activity prevents tubule forma- finding that exogenous overexpression of the FAK
STRESS tion in human brain microvascular endothelial cells14, FERM domain can inhibit cell motility24 might be asso-
A planar force exerted by the
friction of a flowing substance
and gain-of-function experiments with FAK–/– fibrob- ciated with FERM-mediated inhibition of FAK catalytic
— for example, forces lasts show that the formation of fibrillar adhesions activity44, so it is also possible that the FAK FERM
experienced by endothelial cells requires phosphorylation of Tyr397, FAK catalytic domain functions to target FAK to distinct cellular sites
as blood flows through activity and FAK scaffolding functions103. Similar to the that are involved in growth factor signalling or GPCR
capillaries.
maturation process of fibrillar and 3D adhesions, FAK- signalling events23,24. To this end, sumoylation can
CYTOTROPHOBLAST
dependent matrix organization is also observed during occur within the FAK FERM domain, which promotes
The inner trophoblastic layer of dorsal forebrain development in mice, as glial cells that FAK activation27, a fraction of FAK can translocate to
cells that give rise to the lack FAK exhibit basement-membrane formation the nucleus28,111, and FAK signalling can influence the
syncytiotrophoblast facing the defects104. expression of transcription factors30. How — if at all —
maternal circulation and
constitute a layer through which
Although immunostaining of cells that were grown these events are related to the ability of FAK to promote
all substances must pass from in 3D did not reveal enhanced Tyr397 phosphorylation cell polarization and motility is the subject of active
the mother to the fetus. of FAK compared with the levels found in cells forming investigation.

NATURE REVIEWS | MOLECUL AR CELL BIOLOGY VOLUME 6 | JANUARY 2005 | 6 5

© 2005 Nature Publishing Group


REVIEWS

It is also notable that the FERM domain of FAK can probably yield valuable insights into the role of FAK as
localize to cell–cell junctions111, whereas the FAT an essential scaffolding protein or as an active contribu-
domain is associated with focal contacts in epithelial tor to the formation of a FAK–Src signalling complex.
cells. It is possible that the FAK-mediated regulation of
cadherin-based cell contacts might be distinguished Note added in proof
through the differential FERM- or FAT-mediated target- V. Brunton’s personal communication has now been
ing of FAK to distinct intracellular sites. As many of the accepted for publication: Brunton, V. G. et al.
phenotypes that are associated with FAK have been elu- Identification of Src-specific phosphorylation site on
cidated using overexpression studies, the development FAK: dissection of the role of Src SH2 and catalytic
of pharmacological inhibitors to FAK and analyses of functions and their consequences for tumour cell
catalytically-defective FAK mutants in FAK–/– cells will behaviour. Cancer Res. (in the press).

1. Brakebusch, C. & Fassler, R. The integrin–actin 18. Schlaepfer, D. D., Mitra, S. K. & Ilic, D. Control of motile Together with reference 79, shows that FAK can
connection, an eternal love affair. EMBO J. 22, and invasive cell phenotypes by focal adhesion kinase. directly activate Rho through binding and
2324–2333 (2003). Biochim. Biophys. Acta 1692, 77–102 (2004). phosphorylation of a GEF, and that this activation
2. DeMali, K. A., Wennerberg, K. & Burridge, K. Integrin Provides a solid review on the role of FAK during regulates axonal branching.
signaling to the actin cytoskeleton. Curr. Opin. Cell Biol. embryonic development. 35. Toutant, M. et al. Alternative splicing controls the
15, 572–582 (2003). 19. Yano, H. et al. Roles played by a subset of integrin mechanisms of FAK autophosphorylation. Mol. Cell. Biol.
3. Ridley, A. J. et al. Cell migration: integrating signals from signaling molecules in cadherin-based cell–cell adhesion. 22, 7731–7743 (2002).
front to back. Science 302, 1704–1709 (2003). J. Cell Biol. 166, 283–295 (2004). 36. Liu, E., Cote, J. F. & Vuori, K. Negative regulation of FAK
4. Parsons, J. T. Focal adhesion kinase: the first ten years. 20. Katsumi, A., Orr, A. W., Tzima, E. & Schwartz, M. A. signaling by SOCS proteins. EMBO J. 22, 5036–5046
J. Cell Sci. 116, 1409–1416 (2003). Integrins in mechanotransduction. J. Biol. Chem. 279, (2003).
Provides a good overview of the early studies on 12001–12004 (2004). This paper established a link between FAK
FAK. 21. Carragher, N. O., Westhoff, M. A., Fincham, V. J., Schaller, activation, phosphorylation of Tyr397 and
5. Ilic, D. et al. Reduced cell motility and enhanced focal M. D. & Frame, M. C. A novel role for FAK as a protease- subsequent degradation of FAK.
adhesion contact formation in cells from FAK-deficient targeting adaptor protein. Regulation by p42 ERK and 37. Avraham, H., Park, S. Y., Schinkmann, K. & Avraham, S.
mice. Nature 377, 539–544 (1995). Src. Curr. Biol. 13, 1442–1450 (2003). RAFTK/Pyk2-mediated cellular signalling. Cell. Signal 12,
Shows that null mutation of FAK results in defects in 22. Hauck, C. R. et al. Inhibition of focal adhesion kinase 123–133 (2000).
embryonic morphogenesis, and that FAK-null cells expression or activity disrupts epidermal growth factor- 38. Klingbeil, C. K. et al. Targeting Pyk2 to β1-integrin-
show enhanced focal-contact formation and cell stimulated signaling promoting the migration of invasive containing focal contacts rescues fibronectin-stimulated
motility defects in culture. human carcinoma cells. Cancer Res. 61, 7079–7090 signaling and haptotactic motility defects of focal
6. Webb, D. J. et al. FAK–Src signalling through paxillin, ERK (2001). adhesion kinase-null cells. J. Cell Biol. 152, 97–110 (2001).
and MLCK regulates adhesion disassembly. Nature Cell 23. Sieg, D. J. et al. FAK integrates growth-factor and integrin 39. Lim, Y. et al. Phosphorylation of focal adhesion kinase at
Biol. 6, 154–161 (2004). signals to promote cell migration. Nature Cell Biol. 2, tyrosine 861 is crucial for Ras transformation of
7. Palazzo, A. F., Eng, C. H., Schlaepfer, D. D., Marcantonio, 249–256 (2000). fibroblasts. J. Biol. Chem. 279, 29060–29065 (2004).
E. E. & Gundersen, G. G. Localized stabilization of 24. Streblow, D. N. et al. Human cytomegalovirus chemokine 40. Gabarra-Niecko, V., Keely, P. J. & Schaller, M. D.
microtubules by integrin- and FAK-facilitated Rho receptor US28-induced smooth muscle cell migration is Characterization of an activated mutant of focal adhesion
signaling. Science 303, 836–839 (2004). mediated by focal adhesion kinase and Src. J. Biol. kinase: ‘SuperFAK’. Biochem. J. 365, 591–603 (2002).
Provides evidence that FAK promotes cell Chem. 278, 50456–50465 (2003). 41. Nowakowski, J. et al. Structures of the cancer-related
polarization through the stabilization of Together with reference 23, this paper shows that Aurora-A, FAK, and EphA2 protein kinases from
microtubules at leading edges of motile cells. the FAK FERM domain has important roles in nanovolume crystallography. Structure (Camb.) 10,
8. Ren, X. et al. Focal adhesion kinase suppresses Rho promoting growth-factor-stimulated and G-protein- 1659–1667 (2002).
activity to promote focal adhesion turnover. J. Cell Sci. stimulated cell motility. 42. Abbi, S. et al. Regulation of focal adhesion kinase by a
113, 3673–3678 (2000). 25. Chen, R. et al. Regulation of the PH-domain-containing novel protein inhibitor FIP200. Mol. Biol. Cell 13,
9. Agochiya, M. et al. Increased dosage and amplification of tyrosine kinase Etk by focal adhesion kinase through 3178–3191 (2002).
the focal adhesion kinase gene in human cancer cells. the FERM domain. Nature Cell Biol. 3, 439–444 (2001). 43. Medley, Q. G. et al. Signaling between focal adhesion
Oncogene 18, 5646–5653 (1999). 26. Poullet, P. et al. Ezrin interacts with focal adhesion kinase and Trio. J. Biol. Chem. 278, 13265–13270
10. Bhattacharjee, A. et al. Classification of human lung kinase and induces its activation independently of (2003).
carcinomas by mRNA expression profiling reveals distinct cell–matrix adhesion. J. Biol. Chem. 276, 37686–37691 44. Cooper, L. A., Shen, T. L. & Guan, J. L. Regulation of focal
adenocarcinoma subclasses. Proc. Natl Acad. Sci. USA (2001). adhesion kinase by its amino-terminal domain through an
98, 13790–13795 (2001). 27. Kadare, G. et al. PIAS1-mediated sumoylation of focal autoinhibitory interaction. Mol. Cell. Biol. 23, 8030–8041
11. Yeoh, E. J. et al. Classification, subtype discovery, and adhesion kinase activates its autophosphorylation. J. Biol. (2003).
prediction of outcome in pediatric acute lymphoblastic Chem. 278, 47434–47440 (2003). 45. Zeng, L. et al. PTPα regulates integrin-stimulated FAK
leukemia by gene expression profiling. Cancer Cell 1, Shows that sumoylation of FAK within the FERM autophosphorylation and cytoskeletal rearrangement in
133–143 (2002). domain is associated with catalytic activation and cell spreading and migration. J. Cell Biol. 160, 137–146
12. Cance, W. G. et al. Immunohistochemical analyses of preferential nuclear localization. (2003).
focal adhesion kinase expression in benign and malignant 28. Jones, G. & Stewart, G. Nuclear import of N-terminal FAK 46. Chiarugi, P. et al. Reactive oxygen species as essential
human breast and colon tissues: correlation with by activation of the FcεRI receptor in RBL-2H3 cells. mediators of cell adhesion: the oxidative inhibition of a
preinvasive and invasive phenotypes. Clin. Cancer Res. 6, Biochem. Biophys. Res. Comm. 314, 39–45 (2004). FAK tyrosine phosphatase is required for cell adhesion.
2417–2423 (2000). 29. McKean, D. M. et al. FAK induces expression of Prx1 to J. Cell Biol. 161, 933–944 (2003).
13. Ilic, D. et al. Focal adhesion kinase is required for blood promote tenascin-C-dependent fibroblast migration. 47. Arias-Salgado, E. G. et al. Src kinase activation by direct
vessel morphogenesis. Circ. Res. 92, 300–307 (2003). J. Cell Biol. 161, 393–402 (2003). interaction with the integrin β cytoplasmic domain.
14. Haskell, H. et al. Focal adhesion kinase is expressed in 30. Zhao, J. et al. Identification of transcription factor KLF8 as Proc. Natl Acad. Sci. USA 100, 13298–13302 (2003).
the angiogenic blood vessels of malignant astrocytic a downstream target of focal adhesion kinase in its Shows that selected β-integrin subunits can bind
tumors in vivo and promotes capillary tube formation of regulation of cyclin D1 and cell cycle progression. and activate Src in the absence of a contribution
brain microvascular endothelial cells. Clin. Cancer Res. 9, Mol. Cell 11, 1503–1515 (2003). from FAK.
2157–2165 (2003). 31. Hanks, S. K., Ryzhova, L., Shin, N. Y. & Brabek, J. Focal 48. Turner, C. E. Paxillin and focal adhesion signalling. Nature
15. Hauck, C. R., Hsia, D. A., Ilic, D. & Schlaepfer, D. D. v-Src adhesion kinase signaling activities and their implications Cell Biol. 2, 231–236 (2000).
SH3-enhanced interaction with focal adhesion kinase at in the control of cell survival and motility. Front. Biosci. 8, 49. Cho, S. Y. & Klemke, R. L. Purification of pseudopodia
β1 integrin-containing invadopodia promotes cell invasion. 982–996 (2003). from polarized cells reveals redistribution and activation of
J. Biol. Chem. 277, 12487–12490 (2002). 32. Chodniewicz, D. & Klemke, R. L. Regulation of integrin- Rac through assembly of a CAS/Crk scaffold. J. Cell Biol.
16. Hauck, C. R., Hsia, D. A., Puente, X. S., Cheresh, D. A. & mediated cellular responses through assembly of a 156, 725–736 (2002).
Schlaepfer, D. D. FRNK blocks v-Src-stimulated invasion CAS/Crk scaffold. Biochim. Biophys. Acta. 1692, 63–76 50. Brabek, J. et al. CAS promotes invasiveness of
and experimental metastases without effects on cell (2004). Src-transformed cells. Oncogene 23, 7406–7415
motility or growth. EMBO J. 21, 6289–6302 (2002). 33. Schaller, M. D. Biochemical signals and biological (2004).
17. Hsia, D. A. et al. Differential regulation of cell motility and responses elicited by the focal adhesion kinase. Biochim. 51. Schaller, M. D. Paxillin: a focal adhesion-associated
invasion by FAK. J. Cell Biol. 160, 753–767 (2003). Biophys. Acta. 1540, 1–21 (2001). adaptor protein. Oncogene 20, 6459–6472 (2001).
This reference, together with reference 69, shows 34. Zhai, J. et al. Direct interaction of focal adhesion kinase 52. Subauste, M. C. et al. Vinculin modulation of paxillin–FAK
that constitutively active Src can bypass the need with p190RhoGEF. J. Biol. Chem. 278, 24865–24873 interactions regulates ERK to control survival and motility.
for FAK in promoting the turnover of focal contacts. (2003). J. Cell Biol. 165, 371–381 (2004).

66 | JANUARY 2005 | VOLUME 6 www.nature.com/reviews/molcellbio

© 2005 Nature Publishing Group


REVIEWS

53. Hayashi, I., Vuori, K. & Liddington, R. C. The focal 72. Dourdin, N. et al. Reduced cell migration and disruption of colon cancer and transformed rodent cells. Cancer Res.
adhesion targeting (FAT) region of focal adhesion kinase is the actin cytoskeleton in calpain-deficient embryonic 62, 2669–2674 (2002).
a four-helix bundle that binds paxillin. Nature Struct. Biol. fibroblasts. J. Biol. Chem. 276, 48382–48388 (2001). 96. Avizienyte, E. et al. Src-induced de-regulation of
9, 101–106 (2002). 73. Cuevas, B. D. et al. MEKK1 regulates calpain-dependent E-cadherin in colon cancer cells requires integrin
54. Liu, G., Guibao, C. D. & Zheng, J. Structural insight into proteolysis of focal adhesion proteins for rear-end signalling. Nature Cell Biol. 4, 632–638 (2002).
the mechanisms of targeting and signaling of focal detachment of migrating fibroblasts. EMBO J. 22, 97. Quadri, S. K., Bhattacharjee, M., Parthasarathi, K.,
adhesion kinase. Mol. Cell. Biol. 22, 2751–2760 (2002). 3346–3355 (2003). Tanita, T. & Bhattacharya, J. Endothelial barrier
55. Gao, G. et al. NMR solution structure of the focal 74. Westhoff, M. A., Serrels, B., Fincham, V. J., Frame, M. C. strengthening by activation of focal adhesion kinase.
adhesion targeting domain of focal adhesion kinase in & Carragher, N. O. Src-mediated phosphorylation of focal J. Biol. Chem. 278, 13342–13349 (2003).
complex with a paxillin LD peptide: evidence for a two-site adhesion kinase couples actin and adhesion dynamics to 98. Miranti, C. K. & Brugge, J. S. Sensing the environment:
binding model. J. Biol. Chem. 279, 8441–8451 (2004). survival signaling. Mol. Cell. Biol. 24, 8113–8133 (2004). a historical perspective on integrin signal transduction.
56. Katz, B. Z. et al. Targeting membrane-localized focal 75. Giannone, G. et al. Calcium rises locally trigger focal Nature Cell Biol. 4, E83–E90 (2002).
adhesion kinase to focal adhesions: roles of tyrosine adhesion disassembly and enhance residency of focal 99. Li, S. et al. The role of the dynamics of focal adhesion
phosphorylation and SRC family kinases. J. Biol. Chem. adhesion kinase at focal adhesions. J. Biol. Chem. 279, kinase in the mechanotaxis of endothelial cells. Proc. Natl
278, 29115–29120 (2003). 28715–28723 (2004). Acad. Sci. USA 99, 3546–3551 (2002).
57. Prutzman, K. C. et al. The focal adhesion targeting 76. Hecker, T. P., Ding, Q., Rege, T. A., Hanks, S. K. & 100. Wang, H. B., Dembo, M., Hanks, S. K. & Wang, Y. Focal
domain of focal adhesion kinase contains a hinge region Gladson, C. L. Overexpression of FAK promotes Ras adhesion kinase is involved in mechanosensing during
that modulates tyrosine 926 phosphorylation. Structure activity through the formation of a FAK/p120RasGAP fibroblast migration. Proc. Natl Acad. Sci. USA 98,
(Camb.) 12, 881–891 (2004). complex in malignant astrocytoma cells. Oncogene 23, 11295–11300 (2001).
References 53, 54, 55 and 57 provide structural 3962–3971 (2004). Shows that FAK functions as an important
analyses of the FAK FAT domain and the paxillin LD 77. Chen, B. H., Tzen, J. T., Bresnick, A. R. & Chen, H. C. environmental biosensor in promoting directional
peptide binding, and show that Tyr925 Roles of Rho-associated kinase and myosin light chain motility signals in response to changes in substrate
phosphorylation might require conformational kinase in morphological and migratory defects of focal flexibility.
alterations in the FAT domain. adhesion kinase-null cells. J. Biol. Chem. 277, 101. Owen, J. D., Ruest, P. J., Fry, D. W. & Hanks, S. K.
58. Ma, A., Richardson, A., Schaefer, E. M. & Parsons, J. T. 33857–33863 (2002). Induced focal adhesion kinase (FAK) expression in
Serine phosphorylation of focal adhesion kinase in 78. Arthur, W. T., Petch, L. A. & Burridge, K. Integrin FAK-null cells enhances cell spreading and migration
interphase and mitosis: a possible role in modulating engagement suppresses RhoA activity via a c-Src- requiring both auto- and activation loop phosphorylation
binding to p130Cas. Mol. Biol. Cell 12, 1–12 (2001). dependent mechanism. Curr. Biol. 10, 719–722 (2000). sites and inhibits adhesion-dependent tyrosine
59. Hunger-Glaser, I., Fan, R. S., Perez-Salazar, E. & 79. Rico, B. et al. Control of axonal branching and synapse phosphorylation of Pyk2. Mol. Cell. Biol. 19, 4806–4818
Rozengurt, E. PDGF and FGF induce focal adhesion formation by focal adhesion kinase. Nature Neurosci. 7, (1999).
kinase (FAK) phosphorylation at Ser-910: dissociation 1059–1069 (2004). 102. Cukierman, E., Pankov, R. & Yamada, K. M. Cell
from Tyr-397 phosphorylation and requirement for ERK 80. Wu, X., Suetsugu, S., Cooper, L. A., Takenawa, T. & interactions with three-dimensional matrices. Curr. Opin.
activation. J. Cell Physiol. 200, 213–222 (2004). Guan, J. L. Focal adhesion kinase regulation of N-WASP Cell Biol. 14, 633–639 (2002).
60. Liu, Z. X., Yu, C. F., Nickel, C., Thomas, S. & Cantley, L. G. subcellular localization and function. J. Biol. Chem. 279, 103. Ilic, D. et al. FAK promotes organization of fibronectin
Hepatocyte growth factor induces ERK-dependent 9565–9576 (2004). matrix and fibrillar adhesions. J. Cell Sci. 117, 177–187
paxillin phosphorylation and regulates paxillin–focal 81. Palazzo, A. F., Cook, T. A., Alberts, A. S. & Gundersen, G. G. (2004).
adhesion kinase association. J. Biol. Chem. 277, mDia mediates Rho-regulated formation and orientation 104. Beggs, H. E. et al. FAK deficiency in cells contributing to
10452–10458 (2002). of stable microtubules. Nature Cell Biol. 3, 723–729 the basal lamina results in cortical abnormalities
61. Ishibe, S., Joly, D., Zhu, X. & Cantley, L. G. (2001). resembling congenital muscular dystrophies. Neuron 40,
Phosphorylation-dependent paxillin–ERK association 82. Gundersen, G. G., Gomes, E. R. & Wen, Y. Cortical 501–514 (2003).
mediates hepatocyte growth factor-stimulated epithelial control of microtubule stability and polarization. Curr. References 103 and 104 show that FAK has crucial
morphogenesis. Mol. Cell 12, 1275–1285 (2003). Opin. Cell Biol. 16, 106–112 (2004). roles in promoting 3D-matrix assembly and/or
62. Kirchner, J., Kam, Z., Tzur, G., Bershadsky, A. D. & 83. del Pozo, M. A. et al. Integrins regulate Rac targeting by remodelling during development and in cell culture
Geiger, B. Live-cell monitoring of tyrosine phosphorylation internalization of membrane domains. Science 303, model systems.
in focal adhesions following microtubule disruption. 839–842 (2004). 105. Cukierman, E., Pankov, R., Stevens, D. R. & Yamada, K. M.
J. Cell Sci. 116, 975–986 (2003). 84. del Pozo, M. A., Price, L. S., Alderson, N. B., Ren, X. D. & Taking cell–matrix adhesions to the third dimension.
63. Zaidel-Bar, R., Ballestrem, C., Kam, Z. & Geiger, B. Early Schwartz, M. A. Adhesion to the extracellular matrix Science 294, 1708–1712 (2001).
molecular events in the assembly of matrix adhesions at regulates the coupling of the small GTPase Rac to its 106. Xia, H., Nho, R. S., Kahm, J., Kleidon, J. & Henke, C. A.
the leading edge of migrating cells. J. Cell Sci. 116, effector PAK. EMBO J. 19, 2008–2014 (2000). Focal adhesion kinase is upstream of
4605–4613 (2003). 85. Slack-Davis, J. K. et al. PAK1 phosphorylation of MEK1 phosphatidylinositol 3-kinase/Akt in regulating
64. Sieg, D. J., Hauck, C. R. & Schlaepfer, D. D. Required role regulates fibronectin-stimulated MAPK activation. J. Cell fibroblast survival in response to contraction of
of focal adhesion kinase (FAK) for integrin-stimulated cell Biol. 162, 281–291 (2003). type I collagen matrices via a β1 integrin viability
migration. J. Cell Sci. 112, 2677–2691 (1999). 86. Xie, Z. et al. Serine 732 phosphorylation of FAK by Cdk5 signaling pathway. J. Biol. Chem. 279, 33024–33034
65. Rajfur, Z., Roy, P., Otey, C., Romer, L. & Jacobson, K. is important for microtubule organization, nuclear (2004).
Dissecting the link between stress fibres and focal movement, and neuronal migration. Cell 114, 469–482 107. Wozniak, M. A., Desai, R., Solski, P. A., Der, C. J. & Keely,
adhesions by CALI with EGFP fusion proteins. Nature Cell (2003). P. J. ROCK-generated contractility regulates breast
Biol. 4, 286–293 (2002). 87. Ivankovic-Dikic, I., Gronroos, E., Blaukat, A., Barth, B.-U. epithelial cell differentiation in response to the physical
66. Izaguirre, G. et al. The cytoskeletal/non-muscle isoform of & Dikic, I. Pyk2 and FAK regulate neurite outgrowth properties of a three-dimensional collagen matrix.
α-actinin is phosphorylated on its actin-binding domain by induced by growth factors and integrins. Nature Cell Biol. J. Cell Biol. 163, 583–595 (2003).
the focal adhesion kinase. J. Biol. Chem. 276, 2, 574–581 (2000). 108. Ilic, D. et al. Plasma membrane-associated
28676–28685 (2001). 88. Calderwood, D. A. Integrin activation. J. Cell Sci. 117, pY397FAK is a marker of cytotrophoblast invasion
67. Yu, D. H., Qu, C. K., Henegariu, O., Lu, X. & Feng, G. S. 657–666 (2004). in vivo and in vitro. Am. J. Pathol. 159, 93–108
Protein-tyrosine phosphatase Shp-2 regulates cell 89. Papagrigoriou, E. et al. Activation of a vinculin-binding site (2001).
spreading, migration, and focal adhesion. J. Biol. Chem. in the talin rod involves rearrangement of a five-helix 109. Bowden, E. T., Coopman, P. J. & Mueller, S. C.
273, 21125–21131 (1998). bundle. EMBO J. 23, 2942–2951 (2004). Invadopodia: unique methods for measurement of
68. Von Wichert, G., Haimovich, B., Feng, G. S. & 90. Di Paolo, G. et al. Recruitment and regulation of extracellular matrix degradation in vitro. Methods Cell Biol.
Sheetz, M. P. Force-dependent integrin–cytoskeleton phosphatidylinositol phosphate kinase type 1γ by the 63, 613–627 (2001).
linkage formation requires downregulation of focal FERM domain of talin. Nature 420, 85–89 (2002). 110. Hauck, C. R., Hunter, T. & Schlaepfer, D. D. The v-Src
complex dynamics by Shp2. EMBO J. 22, 5023–5035 91. Ling, K., Doughman, R. L., Firestone, A. J., Bunce, M. W. SH3 domain facilitates a cell adhesion-independent
(2003). & Anderson, R. A. Type Iγ phosphatidylinositol phosphate association with focal adhesion kinase. J. Biol. Chem.
Together with reference 67, this study shows that kinase targets and regulates focal adhesions. Nature 420, 276, 17653–17662 (2001).
null mutation of SHP2 results in FAK 89–93 (2002). 111. Stewart, A., Ham, C. & Zachary, I. The focal adhesion
hyperactivation, elevated α-actinin phosphorylation, 92. Barsukov, I. L. et al. Phosphatidylinositol phosphate kinase amino-terminal domain localises to nuclei and
and the failure to promote the maturation of kinase type 1γ and β1-integrin cytoplasmic domain bind to intercellular junctions in HEK 293 and MDCK cells
integrin–cytoskeletal linkages. the same region in the talin FERM domain. J. Biol. Chem. independently of tyrosine 397 and the carboxy-terminal
69. Moissoglu, K. & Gelman, I. H. v-Src rescues actin-based 278, 31202–31209 (2003). domain. Biochem. Biophys. Res. Comm. 299, 62–73
cytoskeletal architecture and cell motility and induces 93. Ling, K. et al. Tyrosine phosphorylation of type Iγ (2002).
enhanced anchorage independence during oncogenic phosphatidylinositol phosphate kinase by Src regulates an 112. Chen, H. C., Appeddu, P. A., Isoda, H. & Guan, J. L.
transformation of focal adhesion kinase-null fibroblasts. integrin–talin switch. J. Cell Biol. 163, 1339–1349 (2003). Phosphorylation of tyrosine 397 in focal adhesion
J. Biol. Chem. 278, 47946–47959 (2003). Together with references 91 and 92, this reference kinase is required for binding phosphatidylinositol 3-
70. Visse, R. & Nagase, H. Matrix metalloproteinases and shows that FAK–Src phosphorylation events kinase. J. Biol. Chem. 271, 26329–26334
tissue inhibitors of metalloproteinases: structure, function to control the composition of membrane (1996).
function, and biochemistry. Circ. Res. 92, 827–839 lipids and the dynamics of focal contacts. 113. Calderwood, D. A. & Ginsberg, M. H. Talin forges the links
(2003). 94. Wheelock, M. J. & Johnson, K. R. Cadherins as between integrins and actin. Nature Cell Biol. 5, 694–697
71. Bhatt, A., Kaverina, I., Otey, C. & Huttenlocher, A. modulators of cellular phenotype. Ann. Rev. Cell Dev. Biol. (2003).
Regulation of focal complex composition and 19, 207–235 (2003). 114. Zheng, C. et al. Differential regulation of Pyk2 and focal
disassembly by the calcium-dependent protease calpain. 95. Irby, R. B. & Yeatman, T. J. Increased Src activity disrupts adhesion kinase (FAK). J. Biol. Chem. 273, 2384–2389
J. Cell Sci. 115, 3415–3425 (2002). cadherin/catenin-mediated homotypic adhesion in human (1998).

NATURE REVIEWS | MOLECUL AR CELL BIOLOGY VOLUME 6 | JANUARY 2005 | 6 7

© 2005 Nature Publishing Group


REVIEWS

115. Wang, Q. et al. Regulation of the formation of Shows that null mutation of the FAK-related Acknowledgements
osteoclastic actin rings by proline-rich tyrosine kinase 2 kinase PYK2 results in integrin and chemokine- S. Mitra is supported by a fellowship from the California
interacting with gelsolin. J. Cell Biol. 160, 565–575 stimulated motility defects of macrophages Tobacco-Related Disease Research Program and D. Schlaepfer
(2003). that are not functionally compensated by FAK is supported by grants from the National Cancer Institute. This is
116. Sieg, D. J. et al. Pyk2 and Src-family protein-tyrosine expression. manuscript 16827-IMM from The Scripps Research Institute.
kinases compensate for the loss of FAK in fibronectin- 121. Watson, J. M. et al. Inhibition of the calcium-dependent
stimulated signaling events but Pyk2 does not fully tyrosine kinase (CADTK) blocks monocyte spreading and Competing interests statement
function to enhance FAK– cell migration. EMBO J. 17, motility. J. Biol. Chem. 276, 3536–3542 (2001). The authors declare no competing financial interests.
5933–5947 (1998). 122. Guinamard, R., Okigaki, M., Schlessinger, J. & Ravetch, J. V.
117. Lakkakorpi, P. T., Bett, A. J., Lipfert, L., Rodan, G. A. & Absence of marginal zone B cells in Pyk2-deficient mice
Duong, L. T. Pyk2 autophosphorylation, but not kinase defines their role in the humoral response. Nature Online links
activity, is necessary for adhesion-induced association Immunol. 1, 31–36 (2000).
with c-Src, osteoclast spreading, and bone resorption. 123. Klinghoffer, R. A., Sachsenmaier, C., Cooper, J. A. & DATABASES
J. Biol. Chem. 278, 11502–11512 (2003). Soriano, P. Src family kinases are required for integrin but The following terms in this article are linked online to:
118. Lev, S. et al. Identification of a novel family of targets not PDGFR signal transduction. EMBO J. 18, 2459–2471 Entrez Gene:
of Pyk2 related to Drosophila retinal degeneration B (1999). http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=gene
(rdgB) protein. Mol. Cell. Biol. 19, 2278–2288 124. Honda, H. et al. Cardiovascular anomaly, impaired actin PTK2
(1999). bundling and resistance to Src- induced transformation in Swiss-Prot: http://www.expasy.ch
119. Benbernou, N., Muegge, K. & Durum, S. K. Interleukin mice lacking p130Cas. Nature Genet. 19, 361–365 (1998). calpain | CDK5 | Diaphanous | E-cadherin | ezrin | FAK | FIP200 |
(IL)-7 induces rapid activation of Pyk2, which is bound to 125. Hagel, M. et al. The adaptor protein paxillin is essential for Fyn | N-cadherin | N-WASP | p130Cas | paxillin | PTPα | RhoA |
Janus kinase 1 and IL-7Rα. J. Biol. Chem. 275, normal development in the mouse and is a critical SHP2 | Src | talin| TRIO | Yes
7060–7065 (2000). transducer of fibronectin signaling. Mol. Cell. Biol. 22,
120. Okigaki, M. et al. Pyk2 regulates multiple signaling 901–915 (2002). FURTHER INFORMATION
events crucial for macrophage morphology and 126. Xu, W., Baribault, H. & Adamson, E. D. Vinculin knockout The Schlaepfer laboratory:
migration. Proc. Natl Acad. Sci. USA 100, 10740–10745 results in heart and brain defects during embryonic http://www.scripps.edu/imm/schlaepfer/index1.htm
(2003). development. Development 125, 327–337 (1998). Access to this interactive links box is free online.

68 | JANUARY 2005 | VOLUME 6 www.nature.com/reviews/molcellbio

© 2005 Nature Publishing Group

You might also like