SlideShare uma empresa Scribd logo
1 de 14
Baixar para ler offline
Biosci. Rep. (2011) / 31 / 231–244 (Printed in Great Britain) / doi 10.1042/BSR20100094
Signalling to actin: role of C3G, a multitasking
guanine-nucleotide-exchange factor
Vegesna RADHA1
, Aninda MITRA, Kunal DAYMA and Kotagiri SASIKUMAR
Centre for Cellular and Molecular Biology (CSIR), Uppal Road, Hyderabad 500 007, India
'
&
$
%
Synopsis
C3G (Crk SH3-domain-binding guanine-nucleotide-releasing factor) is a ubiquitously expressed member of a class of
molecules called GEFs (guanine-nucleotide-exchange factor) that activate small GTPases and is involved in pathways
triggered by a variety of signals. It is essential for mammalian embryonic development and many cellular functions
in adult tissues. C3G participates in regulating functions that require cytoskeletal remodelling such as adhesion,
migration, maintenance of cell junctions, neurite growth and vesicle traffic. C3G is spatially and temporally regulated
to act on Ras family GTPases Rap1, Rap2, R-Ras, TC21 and Rho family member TC10. Increased C3G protein levels
are associated with differentiation of various cell types, indicating an important role for C3G in cellular differentiation.
In signalling pathways, C3G serves functions dependent on catalytic activity as well as protein interaction and can
therefore integrate signals necessary for the execution of more than one cellular function. This review summarizes
our current knowledge of the biology of C3G with emphasis on its role as a transducer of signals to the actin
cytoskeleton. Deregulated C3G may also contribute to pathogenesis of human disorders and therefore could be a
potential therapeutic target.
Key words: actin cytoskeleton, C3G (Crk SH3-domain-binding guanine-nucleotide-releasing factor), differentiation,
embryonic development, GTPase, signalling
INTRODUCTION
The ability of small GTPases to switch between active GTP and
inactive GDP-bound states enables them to function as hubs in
signalling pathways. A small number of GTPases can respond
to a multitude of signals and also activate multiple downstream
effectors, resulting in diverse and specific responses. GTPases
are activated by GEFs (guanine-nucleotide-exchange factors) and
inhibited by GTPase-activating proteins that accelarate GTP hy-
drolysis [1]. Most upstream signals target GEFs to act on specific
GTPases and therefore GEFs serve to link activated receptors
to downstream signalling cascades and provide signalling spe-
cificity [2]. GEFs are classified on the basis of which family of
GTPases they act on, namely Ras GEFs, Rho GEFs etc. A com-
mon feature of Ras GEFs is the presence of a CDC25 homology
domain that helps in catalysis along with an REM (Ras exchanger
motif). In addition, Rap GEFs have multiple modular domains
that aid in protein and lipid interactions, and in their regulation
[3]. C3G (Crk SH3-domain-binding guanine nucleotide-releasing
factor) was the first Rap GEF identified with a domain showing
............................................................................................................................................................................................................................................................................................................
Abbreviations used: 3D, three-dimensional: AJ, adherence junction; C3G, Crk SH3-domain-binding guanine nucleotide releasing factor; CML, chronic myelogenous leukaemia; DC3G,
Drosophila C3G; EGF, epidermal growth factor; ERK, extracellular-signal-regulated kinase; GEF, guanine-nucleotide-exchange factor; JNK, c-Jun N-terminal kinase; MAPK,
mitogen-activated protein kinase; NB, neuroblastoma; NGF, nerve growth factor; pC3G, Y504 (Tyr504)-phosphorylated C3G; PDGF, platelet-derived growth factor; PV, pervanadate; REM,
Ras exchanger motif; SFK, Src family kinase; siRNA, small interfering RNA; T2D, Type 2 diabetes.
1 To whom correspondence should be addressed (e-mail: vradha@ccmb.res.in)
homology with the yeast CDC25, and was originally isolated as
an interacting partner of CRK (cellular homologue of the v-Crk
oncoprotein) [4,5]. Alternate names of C3G are Rap GEF1, GRF2
and DKFZ p781P1719. C3G has the catalytic domain along with
REM at the extreme C-terminus and lacks modular protein in-
teraction domains found in most other Rap GEFs (Figure 1A).
Domains found in Rap GEFs generally are DEP (disheveled-
EGL-10-pleckstrin domain), cNB-L (cyclic nucleotide-binding
domain-like) and PDZ (PSD-95/Dig/ZO-1). These are shown in
Figure 1(B) along with the primary structure of C3G. In humans,
the two primary protein products of about 140 kDa are over 1000
residues in length and have multiple proline-rich sequences in
the central region through which they interact with proteins con-
taining SH3 domains. Crk, Hck, c-Abl and Cas are molecules
known to interact directly with the central domain of C3G [4–8].
Residues in the N-terminus are responsible for interaction with
E-cadherin, indicating that the N-terminal sequences may also
aid in protein interaction [9]. The two isoforms arise due to al-
ternate splicing, and primarily differ in the N-terminus where
three amino acids of isoform a are replaced by 21 amino acids in
isoform b (Figures 2A and 2B).
www.bioscirep.org / Volume 31 (4) / Pages 231–244 231
BioscienceReportswww.bioscirep.org
V. Radha and others
Figure 1 Domain organization of C3G and comparison with other Rap GEFs
(A) Schematic diagram showing the domain organization of C3G protein. The C-terminal catalytic domain of C3G is
homologous with CDC25 and is responsible for target G protein activation. The N-terminal region has a domain that
interacts with E-cadherin. The central protein interaction domain (also known as Crk-binding region, CBR) contains multiple
proline-rich sequences that bind SH3 domains of Crk, Cas, c-Abl and Hck. The non-catalytic sequences negatively regulate
the catalytic activity of C3G. (B) Domain organization of different Rap GEFs. C3G is a unique Rap GEF member that
lacks modular protein interaction domains found in other Rap GEFs. cAMP: cAMP-binding site; EF, EF-hand calcium-binding
domain; Ras GEF or CDC25 homology domain; RA, Ras-association domain; Y504, Tyr504
.
A single-copy gene at chromosomal location 9q34.3 en-
codes C3G [10]. Other proteins encoded from the same region
are c-Abl, nucleoporin 214, laminin γ 3, NET39, protein-O-
mannosyltransferase, uridine-cytidine kinase 1, mediator com-
plex subunit 27 and sarcosine dehydrogenase. The human C3G
gene comprises 24 exons spanning 163 kb (Figure 2A). Tran-
script size of isoform a is 6085 bp and that of isoform b, 6256 bp.
Its homologues have been cloned from several organisms and
show a high degree of conservation in the catalytic domain
(Figure 3). The proline-rich stretches and E-cadherin-binding
domain are conserved among all the vertebrates from which
C3G has been cloned. In invertebrates, some putative proline-
rich SH3-binding stretches could be identified in the primary
sequence, but the E-cadherin-binding domain shows poor con-
servation. C3G may therefore have evolved to perform a broader
range of functions in the vertebrates. A variety of stimuli such
as growth factors, cytokines, integrins, neurotrophins, hormones
and mechanical stress have been shown to engage C3G-mediated
signalling (Table 1).
The GTPases known to be regulated by C3G are Ras
family members Rap1, Rap2, R-Ras, TC-21 and the Rho
family member TC10 leading to the activation of MAPK
(mitogen-activated protein kinase) and other effector pathways
[11–19]. Generally, GEFs do not show promiscuity in targeting
members of the various G-protein subfamilies and act within
their specific family of G-proteins. C3G is an example of
a GEF that targets a Rho family member in addition to the
Ras family GTPases. Over the past 15 years, several studies
have thrown light on the involvement of C3G in multiple
signalling pathways and its role in regulating diverse cellular
functions. Through the activation of ERK (extracellular-signal-
regulated kinase), JNK (c-Jun N-terminal kinase) and Rac
signalling, C3G plays a crucial role in integrin-mediated cell
adhesion and migration and also regulates cell proliferation,
differentiation and apoptosis. These cellular properties are often
associated with, or are a consequence of, cytoskeletal reorganiz-
ation. In the present review, we highlight these properties of C3G
and describe the consequence of its deregulation in cells as well
................................................................... ............................................................ .................................................................. ............................................................. ........................................................ ....................................................
232 C The Authors Journal compilation C 2011 Biochemical Society
C3G signals to actin remodelling
Figure 2 Isoforms of C3G
(A) Schematic diagram showing the genome organization of C3G gene. The human C3G gene comprises 24 exons, spanning
163 kb on chromosome 9q34.3. Human C3G has two predominant isoforms, a and b, which arise due to alternate splicing
and differ in their N-termini. Isoform a has 6085 bp of transcript length, whereas isoform b has 6256 bp of transcript
length. (B) Characterized mammalian isoforms of C3G. The two isoforms of C3G protein differ such that 3 aa (amino acids)
of Isoform a are replaced by 21 aa in isoform b. A truncated isoform is expressed in CML cells (K562), p87C3G which
arises from a 4.5 kb transcript. An alternate isoform in rat, which is expressed only in testis and brain, has a 51 aa (153
bp) insertion, just after the proline-rich domain. In mouse an additional isoform is found which has a deletion of 38 aa at
the N-terminal.
as during embryonic development. The examples of defective
signalling due to C3G leading to pathological states are also
presented.
Isoforms and expression
Although C3G is ubiquitously expressed, some tissue-specific
differences in expression levels have been seen. C3G transcripts
are subject to alternate splicing and variant isoforms have been
cloned from different species (Figure 2B). Rat tissues have shown
the presence of a major ubiquitously expressed 7 kb transcript
and a 4 kb transcript in some tissues [20]. An isoform contain-
ing a 153 bp insert after the fifth proline-rich region has also
been cloned from rat testis [20]. This isoform is predominantly
expressed in the testis and to some extent in the brain unlike
all other tissues which show predominant expression of only the
isoform without this insert. Specific functions served by these
isoforms have not been studied yet.
In human tissues, too, C3G shows ubiquitous expression, but
levels of a 7.5-kb transcript were high in adult skeletal muscle
and placenta, fetal heart and brain and low in the liver [4]. A short
87-kDa isoform encoded by a 4.4-kb transcript is expressed in
myeloid leukaemic cells, and lacks N-terminal 305 amino acids
of the full-length C3G [21]. This lacks the first two polyproline
regions and interacts with Bcr-Abl through the third proline-rich
sequence. The p87 isoform showed differences in expression
levels depending on disease remission during treatment, suggest-
ing a role for C3G in CML (chronic myelogenous leukaemia)
...................................................................... .......................................................... .................................................................... ........................................................... ........................................................... ................................................
www.bioscirep.org / Volume 31 (4) / Pages 231–244 233
V. Radha and others
Figure 3 C3G protein from various species
A comparative analysis of C3G protein from various species is shown with human (H. sapiens) C3G. Human C3G (isoform
b) shows E-cadherin-binding domain (red), proline-rich regions (light blue), REM (dark blue) and Ras GEF domain (black).
Amino acid residues belonging to each domain are mentioned and the position of amino acid residue at the start of
each proline-rich region is also mentioned. The longest isoform of C3G from each species is depicted along with all the
domains and the regions showing significant homology with human C3G with percentage identity. Corresponding regions
are indicated (in red). Analysis reveals the presence of putative proline-rich SH3 binding sequences in C3G protein from
various species. The N-terminal region in C3G protein from rat (R. norvegicus), mouse (M. musculus), Xenopus (X. laevis)
and zebrafish (D. rerio) showing high identity with E-cadherin-binding domain of human C3G is also depicted.
pathogenesis. In mouse tissues, two transcripts with and without
a 114 bp insertion in the N-terminal were expressed in most tis-
sues. C3G expression was high in brain, heart, liver and muscle
and low in adipose tissue, kidney and spleen [22].
Regulation
Currently, very little information is available on regulation of
C3G expression. Difference in relative expression of the two
mouse isoforms was seen during adipocyte differentiation [22].
................................................................... ............................................................ .................................................................. ............................................................. ........................................................ ....................................................
234 C The Authors Journal compilation C 2011 Biochemical Society
C3G signals to actin remodelling
Table 1 Stimuli that engage C3G
JAK, Janus kinase; STAT, signal transducer and activator of transcription; TIMP2 tissue inhibitor of metallo-
proteinases 2.
Stimulus Molecules involved in the pathway Reference(s)
Integrin binding VLA-4/VLA-5/R-Ras [33,78,98]
T-cell receptor c-Cbl/CrkL [99]
B-cell receptor c-Cbl/CrkL [100]
Insulin Cbl/Crk/C3G/TC10 [17,36,47]
EGF EGF/Crk/C3G/Rap1/B-Raf [47,48]
NGF FRS2/Crk/C3G /Rap1/B-Raf [14,48]
Interferon γ c-Cbl/CrkL [101]
Erythropoetin, interleukin-3 and interleukin-5 CrkL/STAT5 [102,103]
Hepatocyte growth factor Gab1-CrkL [34]
Growth hormone JAK2 and c-Src [18]
Reelin stimulation via Dab1/CrkL [104]
Mechanical force CrkII/Cas [60]
Nectin c-Src/Crk [66]
Cadherins c-Src/Vav2/Crk [105]
Bombesin Crk/CrkL [106]
TIMP2 Crk [81]
Figure 4 Differentiated human monocytes express higher levels
of C3G protein
Two human monocytic cell lines U937 and HL-60 were induced to dif-
ferentiate to a macrophage lineage by treating with 10 ng of PMA for
48 h or 1 % DMSO for 24 and 48 h respectively. Whole-cell lysates
were prepared along with UT (untreated) cells and subjected to Western
blotting by using indicated antibodies. Hck was used as a marker of
differentiation and Cdk2 as a protein loading control.
C3G protein levels also increase on differentiation of NB (neur-
oblastoma) cells [23]. Similarly, enhancement in C3G protein
was seen on differentiation of human monocytic cells to a mac-
rophage lineage (Figure 4). A several-fold increase in C3G gene
expression was observed on keratinocyte growth factor treatment
of human airway epithelia, indicating that C3G expression may
be regulated transcriptionally [24]. No information is available
on the promoter of C3G. In silico analysis carried out by us
using web-based software, Promo 3.0 and BKL TRANSFAC,
has shown binding sites for multiple transcription factors in the
upstream regulatory region, but they require experimental valida-
tion. Decreased C3G expression was found in cervical squamous
cell carcinomas due to hypermethylation of upstream regulat-
ory sequences [25]. There appears to be an inter-relationship in
the expression of some GEFs. Knocking down of DOCK-180,
a GEF for Rac resulted in an increase in C3G levels leading to
changes in many cellular properties such as reduced proliferation
and attenuated migration in ovarian carcinoma cells [26].
Most of the Rap GEFs are multidomain proteins and their
activation is regulated by protein–lipid interaction, binding of
second messengers, post-translational modification and subcel-
lular localization. C3G activation has been shown to be regulated
by tyrosine phosphorylation at Y504 and membrane targeting, en-
abled through its interaction with the adaptor protein Crk [27]. c-
Src, Hck, Fyn and c-Abl are kinases known to phosphorylate C3G
at Y504 [7,28–30]. The sequence surrounding Y504 of human
C3G is not totally conserved in rat and mouse, indicating species-
specific differences in C3G regulation. In addition to Y504, C3G
is phosphorylated on other tyrosine residues, but their contri-
bution to C3G regulation has not been studied [30]. The SH2
domain in Crk enables translocation of the Crk–C3G complex
to tyrosine-phosphorylated molecules [such as receptor tyrosine
kinases, p130Cas, Cbl, ARMS (ankyrin repeat-rich membrane
spanning), IRS-1 (insulin receptor substrate-1) and paxillin] in
response to extracellular stimuli [31–34]. Complex formation
between Crk and C3G is influenced by Crk phosphorylation
and the tyrosine phosphatase PTP1B regulates this modification
[6,35–37].
C3G regulation to activate specific GTPases may be complex.
C3G shows constitutive membrane binding upon v-Crk trans-
formation [38]. C3G expression enhances JNK activation and
transformation in v-Crk NIH 3T3 cells. In this case, localization
to the plasma membrane was not sufficient for JNK activation.
The catalytic domain was required but was independent of Rap1
indicating that, under these conditions, C3G targeted other GT-
Pases. Constitutive association of C3G with Crk has been de-
scribed. This interaction seems to vary in an adhesion-dependent
...................................................................... .......................................................... .................................................................... ........................................................... ........................................................... ................................................
www.bioscirep.org / Volume 31 (4) / Pages 231–244 235
V. Radha and others
Figure 5 C3G localizes to filopodia tips
Left panel: HeLa cells transfected with C3G expression vector were
stained for C3G and F-actin. C3G expressing cells show stable filopodial
extensions with C3G localized to their tips. Right panel: HeLa cells
treated with 50 mM PV, for 20 min show filopodia extensions. Staining
for p-C3G and F-actin showed pC3G localized predominantly at the Golgi
and filopodia tips. Images were captured using a confocal microscope.
manner and in response to other stimuli [36,39]. The ability of
molecules such as Cbl to alter CrkL–C3G interaction affects
C3G activation [40]. Cbl-b plays a negative role since Cbl− / −
T-cells show better interaction and higher Rap1 activation. In
response to insulin receptor signalling in skeletal muscle cells,
translocation of C3G to lipid rafts regulates its activation, and
disruption of flotillin-based membrane domains prevents C3G
activation [41]. In neutrophils, the bacterial chemoattractant pro-
tein fMLP (fMet-Leu-Phe) causes membrane targeting of C3G
dependent on function of the cytoskeletal regulator protein VASP
(vasodilator-stimulated phosphoprotein) [42]. Expression of pro-
teins like Bcr-Abl reduces the interaction of C3G and CrkL and
inhibits tyrosine phosphorylation of C3G upon cell spreading
and attachment of NIH 3T3 cells [43]. Bcr-Abl has been found
in a complex containing C3G dependent on CrkL [44].
C3G is also subject to autoregulation. It is known that C3G
enzyme activity is regulated negatively by its non-catalytic se-
quence since deletion of non-catalytic residues results in con-
stitutive catalytic activity [27]. The activation of C3G in the cells
may also be regulated through targeting to specific intracellu-
lar domains [45,46]. All studies so far have shown that C3G
localizes to the cytoplasmic compartment. In epithelial cells,
overexpressed C3G induces filopodia and localizes to filopodia
tips (Figure 5). PV (pervanadate)-induced filopodia show pC3G
(Y504-phosphorylated C3G) localized to their tips indicating a
role for C3G in filopodia functions (Figure 5). C3G, upon being
phosphorylated by SFKs (Src family kinases) or c-Abl, has been
shown to localize to the subcortical actin cytoskeleton, Golgi and
retracting lamellipodia of cells undergoing apoptosis [23,28,30].
Multimolecular complex formation involving C3G in response
to stimuli is a major means of activating C3G. Several proteins
that are capable of interacting with C3G directly or indirectly
have been identified and their involvement in pathways leading
to specific functions are shown in Figure 6. Components of mul-
timolecular complexes containing C3G also vary depending on
the stimulus [39,47]. Stimulation of PC12 cells by EGF (epi-
dermal growth factor), results in the formation of a short-lived
complex containing Crk, C3G, Rap1 and B-Raf. NGF (nerve
growth factor) stimulation causes formation of a stable complex
containing FRS2 (fibroblast growth factor receptor substrate 2),
Crk, C3G, Rap1 and B-Raf leading to prolonged MAPK activa-
tion [48]. In response to cell adhesion, Cas association with C3G
brings it into proximity of Src and focal adhesion kinase at focal
adhesions leading to the activation of JNK by integrins in fibro-
blasts [49]. In response to the activation of Fcγ R1 of myeloid
cells, complex formation is seen with the cytoskeletal protein
Hef-1, Crk, Cbl and C3G [50]. In Ba/F3 haematopoietic cells,
CrkL was found in a complex with C3G, Sos (Son of seven-
less) and c-Abl, but upon Bcr-Abl expression this complex is
disrupted [51]. In NIH 3T3 cells, PDGF (platelet-derived growth
factor) induces formation of complexes containing Necl-5, Integ-
rin α1βIII, PDGF-R (PDGF receptor), Rap1, Crk, C3G and Ral
GDS that enable cell movement [52].
FUNCTIONS
Role in embryonic development
The in vivo function of mammalian C3G has been studied by de-
veloping mice lacking C3G expression (knockout) or having very
low expression from a hypomorphic allele. C3G− / −
homozyg-
ous mice died before embryonic day 7.5, suggesting a significant
role for C3G during mammalian development [53]. The lethality
was rescued by expression of the human C3G transgene. Em-
bryonic fibroblasts from C3G knockout mouse embryos showed
impaired cell adhesion, delayed cell spreading and accelerated
cell migration. These effects were suppressed by expression
of active Rap1, Rap2 or R-Ras. This suggested the requirement of
C3G-dependent activation of GTPase targets for adhesion and
spreading of embryonic fibroblasts and for early embryogenesis
[53]. The fact that other Rap GEFs do not compensate for em-
bryonic lethality indicated that spatial and temporal functions
of C3G other than Rap1 activation may be required during em-
bryonic development.
To help study the role of C3G in other tissues and at later devel-
opmental stages, a mouse strain carrying a hypomorphic C3G al-
lele, C3Ggt
, was developed. Lysates of primary embryonic fibro-
blasts from C3Ggt/gt
mice showed less than 5% protein seen in
cells from wild-type animals, but they survived up to embryonic
day 14.5 [54]. C3Ggt/gt
mutant embryos die due to a blood ves-
sel maturation defect caused by inappropriate development of
vascular supporting cells. C3G-deficient fibroblasts responded to
PDGF-BB abnormally, exhibited cell adhesion defects and lacked
paxillin and integrin-β1-positive cell adhesions. This study elu-
cidated the requirement of C3G for vascular myogenesis, cell
adhesion and response to PDGF, necessary for vascular myogen-
esis [54].
C3Ggt/gt
mice also showed over proliferation of the cortical
neuroepithelium [55]. Neuroepithelial cells from these animals
failed to exit the cell cycle in vivo. C3G mutant neural pre-
cursor cells failed to activate Rap1, exhibited Akt/PKB activation,
................................................................... ............................................................ .................................................................. ............................................................. ........................................................ ....................................................
236 C The Authors Journal compilation C 2011 Biochemical Society
C3G signals to actin remodelling
Figure 6 Interacting partners of C3G and their involvement in pathways leading to specific functions
These members interact with the proline-rich Crk-binding region of C3G through their SH3 domain, except for some
members such as E-cadherin. A direct interaction has been characterized only in case of some members like Crk, Cas,
Hck and Abl. IL3, interleukin 3.
Gsk3β inhibition and β-catenin accumulation, when exposed to
growth factors, in vitro. These findings indicated that the size of
the cortical neural precursor population is controlled by C3G-
mediated inhibition of the Ras signalling pathway [55]. Mutant
embryos also exhibited a cortical neuron migration defect leading
to a failure of preplate splitting into marginal zone and subplate
and a failure to form a cortical plate. The basement membrane
was disrupted and radial glial processes were disorganized indic-
ating the requirement of C3G in neuronal migration and radial
glial attachment during cerebral cortex development [56].
A role for C3G in the development of invertebrates is also
known. During Drosophila eye and wing development, overex-
pression of membrane targeted full-length C3G phenotypically
mimics activation of the Ras-MAPK pathway, suggesting that
DC3G (Drosophila C3G) is involved in MAPK activation in vivo
[57]. The effects of C3G overactivity can be suppressed by re-
ducing the gene dose of components of the Ras-MAPK pathway
and of Rap1. DC3G is likely to stimulate both Ras1 and Rap1 dir-
ectly, which in turn leads to a convergent activation of the MAPK
pathway [57]. Deletion of C3G caused semi-lethality [58]. It is
an accessory component of the Drosophila musculature, essen-
tial for the proper localization of integrins at muscle–muscle and
muscle–epidermis attachment sites and important for maintain-
ing muscle integrity during larval stages.
Cellular functions
Various cellular functions regulated by C3G are mediated either
through changes in gene expression or through signalling to actin
cytoskeletal reorganization. Expression of constitutively active
C3G, or knocking down endogenous C3G have been used to
understand these functions. Changes in gene expression have
been seen under conditions of C3G overexpression as well as
repression [25].
Actin remodelling
Initial evidence that C3G is involved in signalling pathways lead-
ing to actin rearrangement came from studies which showed that
C3G expression resulted in filopodia formation in epithelial cell
lines dependent on an intact actin cytoskeleton [8]. C3G was also
required for c-Abl-induced filopodia formation. It was shown that
C3G could signal to actin by engaging N-Wasp, but independent
of Cdc42, a Rho family GTPase whose activation has generally
been associated with filopodia formation. C3G expressing cells
showed loss of stress fibres suggesting that C3G can alter actin
dynamics in these cells. In response to PV treatment, which is
known to cause filopodia formation [59], pC3G localized to the
subcortical actin cytoskeleton and to the tips of filopodia (Fig-
ure 5). The unique morphology of neuronal cells is achieved and
...................................................................... .......................................................... .................................................................... ........................................................... ........................................................... ................................................
www.bioscirep.org / Volume 31 (4) / Pages 231–244 237
V. Radha and others
maintained through extensive changes in microfilaments and mi-
crotubules. Neurite extension is also dependent on filopodia at
the growth cone. C3G expression in human NB cells resulted
in their morphological differentiation to neurons and Cdc42 and
N-Wasp-dependent signalling was involved [23]. The ability of
C3G to suppress transformation was dependent on its localiza-
tion at the subcortical actin cytoskeleton and its association with
protein phosphatase 2A [45]. It was indicated that C3G could
also directly interact with actin in a yeast two-hybrid assay.
In c-Abl-induced cell death, C3G was phosphorylated select-
ively in actin-rich cellular domains dependent on F-actin-binding
domain of c-Abl [30]. Localized phosphorylation of C3G re-
quired intact actin cytoskeleton, but was not affected by micro-
tubule disruption. Phosphorylation of C3G enhanced its ability
to associate with cytoskeletal structures. Previously it was shown
that C3G phosphorylation on tyrosine in response to adhesion of
NIH 3T3 cells was dependent on an intact cytoskeleton [43]. In
T-cells, it was seen that the actin remodelling protein WAVE-2
was required for C3G phosphorylation on Y504 [29]. In re-
sponse to mechanical signals such as cytoskeletal stretch, C3G
was found associated with Triton-insoluble structures to locally
activate Rap1 [60]. In v-Abl-transformed cells, cytoskeletal re-
arrangement is dependent on the CrkL–C3G complex, Rap1 and
Rac1 [61]. A link between the actin-regulating protein VASP and
C3G has been shown in human polymorphonuclear neutrophils,
with VASP serving to regulate C3G activation [42].
Vesicle traffic which is dependent on actin dynamics is also
regulated by C3G, through its target, TC10. Insulin-stimulated
GLUT4 (glucose transporter type 4) translocation is dependent
on C3G and an intact actin cytoskeleton [17]. TC10 binds COP1
in the Golgi and aids actin polymerization on membrane trans-
port vesicles [62]. Vesicular trafficking of E-cadherin is regulated
by C3G during the formation and breakdown of adherens junc-
tions. Interaction between E-cadherin and C3G is induced on cell
junction disassembly and activation of Rap1 and Rab11 positive
recycling endosomes [63]. In Drosophila, C3G could rescue the
NSF2 (N-ethylmaleimide-sensitive factor 2) phenotype which
shows defects in vesicular trafficking [64].
Targets of C3G also function in actin regulation. Rap1 func-
tions to regulate actin remodelling by engaging diverse effectors
[65]. C3G-Rap1-dependent Rac and Cdc42 activation through
their GEFs, Vav2 and FRG respectively are seen in response to
nectin engagement [66]. C3G-induced morphological changes
associated with neurons are achieved through Cdc42-mediated
signalling to actin [23]. TC10 activity regulates F-actin dynam-
ics and neurite growth [62,67,68]. Membrane protrusion is caused
by interaction between Exo70 and TC10 [69]. R-Ras regulates
cell migration of melanoma cells through association with the
actin-binding scaffold protein Filamin A [70]. R-Ras signals
to cause membrane protrusions through PLC (phospholipase C)
activity [71]. R-Ras also engages Rho and Rac GTPases to cause
morphological changes in epithelial and myeloid cells [72,73].
RgL3, a Ral GDS (guanine nucleotide dissociation stimulator)-
related protein serves to mediate interaction between Rap family
members and profilin, an important activator of actin polymeriz-
ation [74]. Rap2 engages TNIK (TRAF2/Nck-interacting kinase)
to cause changes in the cytoskeleton of cultured mammalian cells
[75]. Rap activation is required for phorbol-ester-induced actin
polymerization and morphological changes in B-cells [65]. Rap1
localizes to cell junctions and is a key regulator of junction form-
ation and disruption [76]. Evidence that C3G signals to actin is
also strengthened by the fact that most of the molecules that in-
teract with C3G such as Crk, Hck, Src, c-Abl etc. are known to
have roles in actin remodelling. Therefore reciprocal regulation
seems to exist between actin dynamics and C3G. On one hand,
polymerized actin serves as a platform for C3G activation and on
the other hand, activated C3G leads to target activation to achieve
changes in actin dynamics. These changes in turn are responsible
for a multitude of cellular functions as described below.
Adhesion and migration
C3G, being a regulator of Rap GTPase, plays an important role
in integrin signalling, adhesion and migration. C3G is phos-
phorylated in response to adhesion to fibronectin and overex-
pression in Ba/F3 haematopoietic cells enhances migration [43].
Expression of membrane-targeted C3G in HeLa cells also induces
extensive cell spreading [77]. Overexpression of C3G in 32D
cells increased adhesion to fibronectin through the activation of
VLA-4 and VLA-5, mediated by R-Ras [78]. Overexpression
of C3G increases adhesion of NIH 3T3 cells to laminin [79]. C3G
localizes to the focal adhesions in v-Crk transformed cells causing
abnormal activation of MAPK and JNK [80]. In TIMP2 (tissue
inhibitor of metalloproteinases 2)-treated human microvascular
endothelial cells, C3G induced RECK expression and reduced
cell migration [81]. SFK-dependent regulation of cell adhesion
also engages C3G.
Cell proliferation
Constitutive activation of C3G by expression of a membrane-
targeted variant in Drosophila resulted in enhanced Ras-MAPK
signalling and overproliferation and cell fate changes [57].
In haematopoietic progenitor cells, expression of membrane-
targeted C3G resulted in expression of double-positive T-cells,
associated with lethal T-cell acute lymphoblastic leukaemia. This
is achieved through enhanced expression of Notch 1 and 3 and
its target genes like Hes1 and c-Myc [82]. SIHA cells expressing
siRNA (small interfering RNA) targeting C3G showed enhanced
proliferation [25]. In NB cells, in addition to causing morpholo-
gical changes of differentiation, C3G induced p21, an inhibitor
of cell proliferation [23]. This is also reflected in vivo in a mouse
model where C3G neuroepithelial cells are retained in the cell
cycle without arresting and differentiating [55].
Differentiation
C3G is induced during neuronal differentiation and regulates
survival and differentiation of human NB cells [23]. Human NB
cells, IMR-32 induced to differentiate by serum starvation or by
treatment with NGF or forskolin showed enhanced C3G pro-
tein levels. Transient overexpression of C3G stimulated neurite
................................................................... ............................................................ .................................................................. ............................................................. ........................................................ ....................................................
238 C The Authors Journal compilation C 2011 Biochemical Society
C3G signals to actin remodelling
growth and also increased responsiveness to NGF and serum
deprivation induced differentiation. Forskolin and NGF treat-
ment resulted in phosphorylation of C3G at Tyr504 predom-
inantly in the Golgi. The activation of the C3G/Rap1 pathway
results in neurite outgrowth of mouse pheochromocytoma cells,
PC12, which is inhibited by either overexpression of Rap1GAP
or siRNA-mediated knockdown of Rap1 or the GEF C3G [83].
Dephosphorylation of Crk and association with C3G was re-
quired for adipocyte differentiation [84]. C3G protein levels also
increased during differentiation of monocytes to macrophage
lineage (Figure 4). The phenotype shown by mice expressing
a hyphomorphic allele was also indicative of a requirement of
C3G for differentiation of a variety of cells [55].
Transformation
C3G expression increases the growth rate, anchorage-
independent growth and JNK activation in v-Crk transformed
NIH 3T3 cells. The catalytic domain of C3G is essential for
this activity. Rap1 does not act as a C3G substrate in this con-
text. Dominant-negative C3G can reverse the transformed phen-
otype suggesting that C3G is essential for v-Crk-induced trans-
formation of NIH 3T3 cells [38]. C3G-dependent Rap1 activ-
ation also contributes to RET/PTC (rearranged during trans-
fection/papillary thyroid carcinomas) oncogene-mediated trans-
formation of thyroid follicular cells [85].
C3G, like Rap1, is capable of down-regulating the trans-
forming ability of Ras and Sis oncogenes [86]. However, the
transformation suppression activity of C3G is higher than that
of Rap1A. Through its ability to activate Rap1, C3G has been
shown to counteract signalling through the Ras/MAPK pathway
and has also been shown to transmit signals through the stress
kinase JNK pathway [15]. Moreover, C3G can also inhibit v-Raf-
and dbl-induced transformation of NIH 3T3 cells. The catalytic
domain of C3G is not required for this transformation suppression
activity, rather the proline-rich motifs of C3G are essential and
sufficient for this. C3G inhibits Ras-induced ERK activation, cyc-
lin A expression and anchorage-independent growth [79]. Farne-
sylated C3G, which localizes to the membrane, causes signific-
antly higher morphological reversion of transformed phenotype
of v-ki-Ras-transformed NIH 3T3 cells than normal C3G.
Apoptosis and cell survival
Co-expression of Hck with C3G induced a high level of apoptosis
in many cell lines and this property was not dependent on Y-504
phosphorylation or the catalytic domain of C3G but required the
catalytic activity of Hck. This indicated that C3G co-expression
could alter Hck activity towards select targets leading to apoptosis
[7]. c-Abl expression-induced cell death was dependent on C3G
and its phosphorylation in distinct actin-rich retracting lamellipo-
dia was associated with apoptosis. Oxidative-stress-induced cell
death mediated through c-Abl activation was dependent on C3G
phosphorylation [30].
By negatively regulating p38α MAPK, C3G plays a dual role
in regulating cell death in MEFs (mouse embryonic fibroblasts)
depending on the stimulus. C3G mediates cell death in response
to oxidative stress, whereas it induces cell survival upon serum
starvation. On serum deprivation, C3G induces survival through
inhibition of p38α MAPK activity, which mediates apoptosis;
whereas, in response to oxidative stress, C3G behaves as a
proapoptotic molecule, as its knockdown or knockout enhances
survival through upregulation of p38α activity, which plays an
antiapoptotic role under these conditions [87]. C3G acts to signal
to apoptosis and cell survival in response to the c-Abl inhibitor,
ST1-571 [88]. Differentiation of NB cells involves activation of
survival pathways along with induction of cell cycle arrest. C3G
is required for cell survival during differentiation as its knock-
down caused enhanced cell death in response to serum starvation
[23].
Filopodia formation and cell junction integrity
Work from our laboratory has shown that C3G plays a role in cyto-
skeletal reorganization and filopodia formation [8]. Knockdown
of C3G inhibited c-Abl-induced filopodia during cell spreading
on fibronectin. C3G expression induces actin cytoskeletal reor-
ganization and promotes filopodia formation independent of its
catalytic activity. It showed enrichment at filopodia tips charac-
teristic of molecules involved in filopodial dynamics (Figure 5).
AJs (adherens junctions) responsible for the integrity of epi-
thelial monolayers are formed by linking actin networks of neigh-
bouring cells. C3G directly interacts with E-cadherin, a primary
component of epithelial AJs, and excludes binding of β-catenin
to E-cadherin [9]. C3G’s function has therefore been implicated
in recruitment of E-cadherin to the junctions. E-cadherin-rich
filopodia extensions function as adhesion zippers to interlock
neighbouring cells before mature junction formation. E-cadherin
internalization on junctional breakdown also depends on C3G
binding to intracellular E-cadherin to activate Rap1 [89]. Nectins
(Ig-like transmembrane molecules) which aid in AJ formation
also signal by recruiting C3G to activate Rap1 [90].
Association with human disease
In malignant transformation associated with human cancers,
changes in C3G expression is tissue-specific. C3G overex-
pression was found in several samples of primary NSCLCs
(non-small-cell lung cancers) compared with corresponding
non-cancerous tissues. Six of seven NSC cell lines also showed
higher levels of C3G [91]. In contrast, cervical squamous cell
carcinomas were associated with decreased C3G levels. This
was attributed to frequent hypermethylation of upstream regu-
latory gene sequence [25]. Gene expression profiling of chronic
lymphocytic leukaemia samples showed downregulation of C3G
during disease progression [92]. Expression of an alternately
spliced form of C3G, p87, lacking N-terminal 305 residues in
CML cell lines and Ph+
[Philadelphia chromosomal transloca-
tion t(9;22)(q34;q11) positive] patients has been suggested to
play a role in the pathogenesis of CML [21,93].
Single-nucleotide polymorphisms in the C3G gene have shown
association with T2D (Type 2 diabetes), but the molecular basis
...................................................................... .......................................................... .................................................................... ........................................................... ........................................................... ................................................
www.bioscirep.org / Volume 31 (4) / Pages 231–244 239
V. Radha and others
is not clear. In a Finnish population, SNP rs4740283, located
4kb downstream of the C3G gene showed positive association
with T2D [94]. The GG phenotype of the polymorphism at
rs11243444, located in intron 13, had a protective effect on the
development of T2D in a Korean population [95]. In an experi-
mental model of glomerular nephritis, C3G and R-Ras-dependent
signalling has been implicated [96]. Disease-associated deletions
are known in the 9q34.3 chromosomal location that harbours the
C3G gene [97]. It is to be determined whether lack of C3G con-
tributes to the disease phenotype. It has also been predicted that
C3G deregulation may be associated with human disorders show-
ing defective leucocyte adhesion to the endothelium [42]. Mice
lacking C3G show cortical neuronal migration defects resulting
in failure to split preplate into marginal zone and subplate [56].
In humans, defective neuronal migration during development
leads to disorders like lissencephaly. It would therefore be in-
teresting to check for defects in C3G in lissencephaly patients.
CONCLUSIONS AND PERSPECTIVES
Multiple lines of evidence exist to show that many of the cellular
functions regulated by C3G involve reorganization of the actin
cytoskeleton. Through its ability to signal to actin reorganization,
C3G is involved in regulation of both structural and functional
processes in the cell. Morphogenesis is primarily dependent on
adhesive and migratory behaviour of cells and these functions of
C3G may be essential during embryonic development. The fact
that C3G is engaged in response to diverse signals indicates its
role in multiple tissue types and also explains the early embryonic
lethality due to defective development of multiple organ systems.
Requirement of C3G for mammalian development leads us to
ask whether C3G mutations could be associated with human
developmental defects. Examining aborted foetuses for mutations
or expression changes in C3G may help in determining whether
it plays a role in human embryonic development.
C3G being a member of a family constituting a large number
of proteins, it was surprising to note that other Rap GEFs do not
compensate C3G function under several situations.
Action of C3G in a spatial and temporal manner appears to be
essential during embryonic development, which may be one of
the reasons as to why its function is not complemented by other
GEFs. There is need to understand much more about the regula-
tion of C3G both in terms of its expression as well as activation.
Isoform-specific functions of C3G need to be elucidated. Iden-
tification of the C3G promoter and the regulatory transcription
factors and their response elements is warranted. There is good
reason to think that transcription factors that regulate differenti-
ation and migratory behaviour of cells may regulate C3G expres-
sion. Other modifications of C3G in addition to phosphorylation
on Y504 need to be investigated and studied. C3G has multiple
proline tracts but it is not clear as to whether it can interact directly
with more than one protein to form a multimolecular complex and
serve as a scaffold. One question that has not been addressed is
whether there is mutual exclusion of interacting partners enabling
the activation of only a subset of downstream effector pathways.
It is also possible that two or more protein binding motifs in C3G
function in a co-operative manner.
Suppression of transformation is an important role played by
C3G, which is a property independent of its catalytic activity.
C3G expression resulted in upregulation of the cell cycle inhibitor
p21 and suppression of cyclin A expression. Understanding how
C3G signals to changes in expression of genes regulating the
cell cycle will be important to understand its role as a tumour
suppressor. In some cell types, C3G also functions to enhance cell
proliferation and therefore its role in enhancing or suppressing
proliferation is context-dependent. At present, it is not clear as
to how C3G activates specific GTPases belonging to either Ras
or Rho family in a stimulus-dependent manner. Further studies
need to be carried out to determine whether C3G can regulate the
activity of other GTPases directly or indirectly.
On the basis of existing evidence, we propose that C3G may
be a master regulator of the differentiated phenotype in multiple
tissues. Differentiation removes cells from the proliferative mode
without affecting their integrity. Differentiation pathways are rel-
evant for tumour suppression in the light of continuous tissue
regeneration and therefore understanding them in various tissue
types has been important. In cells that have defects in apoptotic
pathways, inducing irreversible arrest through differentiation is a
good alternative in cancer therapy. The function of C3G as a reg-
ulator of differentiation in multiple tissues may be an important
property that could be utilized for achieving tumour suppression.
The 3D (three-dimensional) structure of C3G (either of the
whole molecule or its subdomains) has not been elucidated. Ana-
lysis of the 3D structure of C3G will help in understanding its
properties better. A 3D homology model constructed by using
SWISS-MODEL software indicated considerable structural ho-
mology between the catalytic sequence of C3G and the GEF do-
main (Cdc25 homology domain) of Sos, a Ras family GEF whose
crystal structure has been studied [107]. The GEFs interact with
their respective GTPases by using the same overall interface but
different specific interactions provide target specificity [108].
Targeting GEFs for either activation or inhibition for therapy
has been shown to be possible in principle [1]. Small-molecule
inhibitors have been developed for some GEFs and selective ag-
onists used for activation in other instances. C3G being a ubiquit-
ously expressed molecule with a role in pathways triggered by
a variety of signals, any attempt at therapeutic intervention must
aim at achieving selectivity in specific cell types. Some sugges-
ted approaches for activation of C3G are: (1) enabling membrane
targeting; (2) inhibition of tyrosine phosphatases or activation of
kinases that specifically regulate C3G; (3) introduction of pep-
tides that bind negative regulatory sequences; and (4) treatment
with agents that cause increase in C3G levels in specific cell types.
Just as in the case of Rho GEFs, C3G activity can be inhibited
by finding small molecule inhibitors that target its GEF domain.
Since C3G has functions dependent on catalytic activity as well
as protein interaction leading to different cellular functions, it
should be possible to target specific pathways selectively.
Other major questions that remain to be answered are ‘how are
developmental processes co-ordinated by C3G at the molecular
................................................................... ............................................................ .................................................................. ............................................................. ........................................................ ....................................................
240 C The Authors Journal compilation C 2011 Biochemical Society
C3G signals to actin remodelling
level?’ and ‘how does C3G regulate actin dynamics?’ Although
one straight answer would be that these functions are carried out
through activation of GTPases, there appears to be more com-
plexity. Association of C3G directly with actin indicates multiple
mechanisms that could be involved. Although we have high-
lighted a role for C3G in regulating actin dynamics, it is possible
that C3G signals to cytoskeletal changes by also affecting mi-
crotubule dynamics. A detailed knowledge of the regulation and
function of C3G at the cellular and molecular level will hopefully
provide us with means to selectively target it in specific tissues
where its deregulation is associated with pathology.
ACKNOWLEDGEMENTS
We thank Dr Ghanshyam Swarup for a critical reading of the manu-
script prior to submission.
REFERENCES
1 Bos, J. L., Rehmann, H. and Wittinghofer, A. (2007) GEFs and
GAPs: critical elements in the control of small G proteins. Cell
129, 865–877
2 Raaijmakers, J. H. and Bos, J. L. (2009) Specificity in Ras and
Rap signaling. J. Biol. Chem. 284, 10995–10999
3 Quilliam, L. A., Rebhun, J. F. and Castro, A. F. (2002) A growing
family of guanine nucleotide exchange factors is responsible for
activation of Ras-family GTPases. Prog. Nucleic Acid Res. Mol.
Biol. 71, 391–444
4 Tanaka, S., Morishita, T., Hashimoto, Y., Hattori, S., Nakamura,
S., Shibuya, M., Matuoka, K., Takenawa, T., Kurata, T.,
Nagashima, K. et al. (1994) C3G, a guanine nucleotide-releasing
protein expressed ubiquitously, binds to the Src homology 3
domains of CRK and GRB2/ASH proteins. Proc. Natl. Acad. Sci.
U.S.A. 91, 3443–3447
5 Knudsen, B. S., Feller, S. M. and Hanafusa, H. (1994) Four
proline-rich sequences of the guanine-nucleotide exchange factor
C3G bind with unique specificity to the first Src homology 3
domain of Crk. J. Biol. Chem. 269, 32781–32787
6 Kirsch, K. H., Georgescu, M. M. and Hanafusa, H. (1998) Direct
binding of p130(Cas) to the guanine nucleotide exchange factor
C3G. J. Biol. Chem. 273, 25673–25679
7 Shivakrupa, R., Radha, V., Sudhakar, C. and Swarup, G. (2003)
Physical and functional interaction between Hck tyrosine kinase
and guanine nucleotide exchange factor C3G results in
apoptosis, which is independent of C3G catalytic domain. J. Biol.
Chem. 278, 52188–52194
8 Radha, V., Rajanna, A., Mitra, A., Rangaraj, N. and Swarup, G.
(2007) C3G is required for c-Abl-induced filopodia and its
overexpression promotes filopodia formation. Exp. Cell Res. 313,
2476–2492
9 Hogan, C., Serpente, N., Cogram, P., Hosking, C. R., Bialucha, C.
U., Feller, S. M., Braga, V. M., Birchmeier, W. and Fujita, Y. (2004)
Rap1 regulates the formation of E-cadherin-based cell-cell
contacts. Mol. Cell. Biol. 24, 6690–6700
10 Takai, S, Tanaka, M, Sugimura, H, Yamada, K, Naito, Y, Kino, I
and Matsuda, M. (1994) Mapping of the human C3G gene coding
a guanine nucleotide releasing protein for Ras family to 9q34.3
by fluorescence in situ hybridization. Hum. Genet. 94, 549–550
11 Gotoh, T., Hattori, S., Nakamura, S., Kitayama, H., Noda, M.,
Takai, Y., Kaibuchi, K., Matsui, H., Hatase, O., Takahashi, H. et al.
(1995) Identification of Rap1 as a target for the Crk SH3
domain-binding guanine nucleotide-releasing factor C3G. Mol.
Cell. Biol. 15, 6746–6753
12 Gotoh, T., Niino, Y., Tokuda, M., Hatase, O., Nakamura, S.,
Matsuda, M. and Hattori, S. (1997) Activation of R-Ras by
Ras-guanine nucleotide-releasing factor. J. Biol. Chem. 272,
18602–18607
13 van den Berghe, N., Cool, R. H., Horn, G. and Wittinghofer, A.
(1997) Biochemical characterization of C3G: an exchange factor
that discriminates between Rap1 and Rap2 and is not inhibited
by Rap1A(S17N). Oncogene 15, 845–850
14 York, R. D., Yao, H., Dillon, T., Ellig, C. L., Eckert, S. P., McCleskey,
E. W. and Stork, P. J. (1998) Rap1 mediates sustained MAPK
activation induced by NGF. Nature 392, 622–626
15 Mochizuki, N., Ohba, Y., Kobayashi, S., Otsuka, N., Graybiel, A.
M., Tanaka, S. and Matsuda, M. (2000) Crk activation of JNK via
C3G and R-Ras. J. Biol. Chem. 275, 12667–12671
16 Ohba, Y., Mochizuki, N., Yamashita, S., Chan, A. M., Schrader, J.
W., Hattori, S., Nagashima, K. and Matsuda, M. (2000)
Regulatory proteins of R-Ras, TC21/R-Ras2, and M-Ras/R-Ras3.
J. Biol. Chem. 275, 20020–20026
17 Chiang, S. H., Baumann, C. A., Kanzaki, M., Thurmond, D. C.,
Watson, R. T., Neudauer, C. L., Macara, I. G., Pessin, J. E. and
Saltiel, A. R. (2001) Insulin-stimulated GLUT4 translocation
requires the CAP-dependent activation of TC10. Nature 410,
944–948
18 Ling, L., Zhu, T. and Lobie, P. E. (2003) Src-CrkII-C3G-dependent
activation of Rap1 switches growth hormone-stimulated p44/42
MAP kinase and JNK/SAPK activities. J. Biol. Chem. 278,
27301–27311
19 Wang, Z., Dillon, T. J., Pokala, V., Mishra, S., Labudda, K.,
Hunter, B. and Stork, P. J. (2006) Rap1-mediated activation of
extracellular signal-regulated kinases by cyclic AMP is dependent
on the mode of Rap1 activation. Mol. Cell. Biol. 26, 2130–2145
20 Shivakrupa, R., Singh, R. and Swarup, G. (1999) Identification of
a novel splice variant of C3G which shows tissue-specific
expressionDNA Cell Biol. 18, 701–708
21 Gutierrez-Berzal, J., Castellano, E., Martin-Encabo, S.,
Gutierrez-Cianca, N., Hernandez, J. M., Santos, E. and Guerrero,
C. (2006) Characterization of p87C3G, a novel, truncated C3G
isoform that is overexpressed in chronic myeloid leukemia and
interacts with Bcr-Abl. Exp. Cell Res. 312, 938–948
22 Zhai, B., Huo, H. and Liao, K. (2001) C3G, a guanine nucleotide
exchange factor bound to adapter molecule c-Crk, has two
alternative splicing forms. Biochem. Biophys. Res. Commun.
286, 61–66
23 Radha, V., Rajanna, A., Gupta, R. K., Dayma, K. and Raman, T.
(2008) The guanine nucleotide exchange factor, C3G regulates
differentiation and survival of human neuroblastoma cells.
J. Neurochem. 107, 1424–1435
24 Prince, L. S., Karp, P. H., Moninger, T. O. and Welsh, M. J. (2001)
KGF alters gene expression in human airway epithelia: potential
regulation of the inflammatory response. Physiol. Genom. 6,
81–89
25 Okino, K., Nagai, H., Nakayama, H., Doi, D., Yoneyama, K.,
Konishi, H. and Takeshita, T. (2006) Inactivation of Crk SH3
domain-binding guanine nucleotide-releasing factor (C3G) in
cervical squamous cell carcinoma. Int. J. Gynecol. Cancer 16,
763–771
26 Wang, H., Linghu, H., Wang, J., Che, Y. L., Xiang, T. X., Tang, X.
and Yao, Z. W. (2010) The role of Crk/Dock180/Rac1 pathway in
the malignant behavior of human ovarian cancer cell SKOV3.
Tumour Biol. 31, 59–67
27 Ichiba, T., Hashimoto, Y., Nakaya, M., Kuraishi, Y., Tanaka, S.,
Kurata, T., Mochizuki, N. and Matsuda, M. (1999) Activation of
C3G guanine nucleotide exchange factor for Rap1 by phospho-
rylation of tyrosine 504. J. Biol. Chem. 274, 14376–14381
28 Radha, V., Rajanna, A. and Swarup, G. (2004) Phosphorylated
guanine nucleotide exchange factor C3G, induced by pervanadate
and Src family kinases localizes to the Golgi and subcortical actin
cytoskeleton. BMC Cell Biol. 5, 31
29 Nolz, J. C., Nacusi, L. P., Segovis, C. M., Medeiros, R. B.,
Mitchell, J. S., Shimizu, Y. and Billadeau, D. D. (2008) The WAVE2
complex regulates T cell receptor signaling to integrins via
...................................................................... .......................................................... .................................................................... ........................................................... ........................................................... ................................................
www.bioscirep.org / Volume 31 (4) / Pages 231–244 241
V. Radha and others
Abl- and CrkL-C3G-mediated activation of Rap1. J. Cell Biol. 182,
1231–1244
30 Mitra, A. and Radha, V. (2010) F-actin-binding domain of c-Abl
regulates localized phosphorylation of C3G: role of C3G in
c-Abl-mediated cell death. Oncogene 29, 4528–4542
31 Yokote, K., Hellman, U., Ekman, S., Saito, Y., Ronnstrand, L.,
Heldin, C. H. and Mori, S. (1998) Identification of Tyr-762 in the
platelet-derived growth factor alpha-receptor as the binding site
for Crk proteins. Oncogene 16, 1229–1239
32 Larsson, H., Klint, P., Landgren, E. and Claesson-Welsh, L. (1999)
Fibroblast growth factor receptor-mediated endothelial cell
proliferation is dependent on the Src homology (SH) 2/SH3
domain containing adaptor protein. J. Biol. Chem. 274,
25726–25734
33 Uemura, N. and Griffin, J. D. (1999) The adapter protein Crkl links
Cbl to C3G after integrin ligation and enhances cell migration.
J. Biol. Chem. 274, 37525–37532
34 Sakkab, D., Lewitzky, M., Posern, G., Schaeper, U., Sachs, M.,
Birchmeier, W. and Feller, S. M. (2000) Signaling of hepatocyte
growth factor/scatter factor (HGF) to the small GTPase Rap1 via
the large docking protein Gab1 and the adapter protein CRKL.
J. Biol. Chem. 275, 10772–10778
35 Ichiba, T., Kuraishi, Y., Sakai, O., Nagata, S., Groffen, J.,
Kurata, T., Hattori, S. and Matsuda, M. (1997) Enhancement of
guanine-nucleotide exchange activity of C3G for Rap1 by the
expression of Crk, CrkL, and Grb2. J. Biol. Chem. 272,
22215–22220
36 Okada, S., Matsuda, M., Anafi, M., Pawson, T. and Pessin, J. E.
(1998) Insulin regulates dynamic balance between Ras and Rap1
signaling by coordinating assembly states of Grb2–SOS and
CrkII–C3G complexes. EMBO J. 17, 2554–2565
37 Takino, T., Tamura, M., Miyamori, H., Araki, M., Matsumoto, K.,
Sato, H. and Yamada, K. M. (2003) Tyrosine phosphorylation of
the CrkII adaptor protein modulates cell migration. J. Cell Sci.
116, 3145–3155
38 Tanaka, S., Ouchi, T. and Hanafusa, H. (1997) Downstream of
Crk adaptor signaling pathway: activation of Jun kinase by v-Crk
through the guanine nucleotide exchange protein C3G. Proc. Natl.
Acad. Sci. U.S.A. 94, 2356–2361
39 Buensuceso, C. S. and O’Toole, T. E. (2000) The association of
CRKII with C3G can be regulated by integrins and defines a novel
means to regulate the mitogen-activated protein kinases. J. Biol.
Chem. 275, 13118–13125
40 Zhang, W., Shao, Y., Fang, D., Huang, J., Jeon, M. S. and Liu, Y.
C. (2003) Negative regulation of T-cell antigen receptor mediated
Crk-L-C3G signalling and cell adhesion by cbl-b. J. Biol. Chem.
278, 23978–23983
41 Fecchi, K., Volonte, D., Hezel, M. P., Schmeck, K. and Galbiati, F.
(2006) Spatial and temporal regulation of GLUT4 translocation by
flotillin-1 and caveolin-3 in skeletal muscle cells. FASEB J. 20,
705–707
42 Deevi, R. K., Koney-Dash, M., Kissenpfennig, A., Johnston, J. A.,
Schuh, K., Walter, U. and Dib, K. Vasodilator-stimulated
phosphoprotein regulates inside-out signaling of β2 integrins in
neutrophils. J. Immunol. 184, 6575–6584
43 de Jong, R., van Wijk, A., Heisterkamp, N. and Groffen, J. (1998)
C3G is tyrosine-phosphorylated after integrin-mediated cell
adhesion in normal but not in Bcr/Abl expressing cells. Oncogene
17, 2805–2810
44 Cho, Y. J., Hemmeryckx, B., Groffen, J. and Heisterkamp, N.
(2005) Interaction of Bcr/Abl with C3G, an exchange factor for
the small GTPase Rap1, through the adapter protein Crkl.
Biochem. Biophys. Res. Commun. 333, 1276–1283
45 Martin-Encabo, S., Santos, E. and Guerrero, C. (2007) C3G
mediated suppression of malignant transformation involves
activation of PP2A phosphatases at the subcortical actin
cytoskeleton. Exp. Cell Res. 313, 3881–3891
46 Bivona, T. G., Wiener, H. H., Ahearn, I. M., Silletti, J., Chiu, V. K.
and Philips, M. R. (2004) Rap1 up-regulation and activation on
plasma membrane regulates T cell adhesion. J. Cell Biol. 164,
461–470
47 Okada, S. and Pessin, J. E. (1997) Insulin and epidermal growth
factor stimulate a conformational change in Rap1 and
dissociation of the CrkII–C3G complex. J. Biol. Chem. 272,
28179–28182
48 Kao, S., Jaiswal, R. K., Kolch, W. and Landreth, G. E. (2001)
Identification of the mechanisms regulating differential activation
of the MAPK cascade by EGF and NGF in PC12 cells. J. Biol.
Chem. 276, 18169–18177
49 Li, L., Okura, M. and Imamoto, A. (2002) Focal adhesions require
catalytic activity of Src family kinases to mediate integrin-matrix
adhesion. Mol. Cell. Biol. 22, 1203–1217
50 Kyono, W. T., de Jong, R., Park, R. K., Liy, Y., Heisterkamp, N.,
Groffen, J. and Durden, D. L. (1998) Differential interaction of
Crk1 with Cbl or C3G, Hef-1, and γ subunit immunoreceptor
tyrosine-based activation motif in signaling of myeloid high affinity
Fc receptor for IgG (Fcγ RI). J. Immunol. 161, 5555–5563
51 Uemura, N., Salgia, R., Li, J. L., Pisick, E., Sattler, M. and Griffin,
J. D. (1997) The BCR/ABL oncogene alters interaction of the
adapter proteins CRKL and CRK with cellular proteins. Leukemia
11, 376–385
52 Takahashi, M., Rikitake, Y., Nagamatsu, Y., Hara, T., Ikeda, W.,
Hirata, K. and Takai, Y. (2008) Sequential activation of Rap1 and
Rac1 small G proteins by PDGF locally at leading edges of
NIH3T3 cells. Genes Cells 13, 549–569
53 Ohba, Y., Ikuta, K., Ogura, A., Matsuda, J., Mochizuki, N.,
Nagashima, K., Kurokawa, K., Mayer, B. J., Maki, K., Miyazaki, J.
et al. (2001) Requirement for C3G-dependent Rap1 activation for
cell adhesion and embryogenesis. EMBO J. 20, 3333–3341
54 Voss, A. K., Gruss, P. and Thomas, T. (2003) The guanine
nucleotide exchange factor C3G is necessary for the formation of
focal adhesions and vascular maturation. Development 130,
355–367
55 Voss, A. K., Krebs, D. L. and Thomas, T. (2006) C3G regulates
the size of the cerebral cortex neural precursor population. EMBO
J. 25, 3652–3663
56 Voss, A. K., Britto, J. M., Dixon, M. P., Sheikh, B. N., Collin, C.,
Tan, S. S. and Thomas, T. (2008) C3G regulates cortical neuron
migration, preplate splitting and radial glial cell attachment.
Development 135, 2139–2149
57 Ishimaru, S., Williams, R., Clark, E., Hanafusa, H. and Gaul, U.
(1999) Activation of the Drosophila C3G leads to cell fate
changes and overproliferation during development, mediated by
the RAS–MAPK pathway and RAP1. EMBO J. 18, 145–155
58 Shirinian, M., Grabbe, C., Popovic, M., Varshney, G.,
Hugosson, F., Bos, H., Rehmann, H. and Palmer, R. H. (2010)
The Rap1 guanine nucleotide exchange factor C3G is required for
preservation of larval muscle integrity in Drosophila
melanogaster. PLoS One 5, e9403
59 Luber, B., Candidus, S., Handschuh, G., Mentele, Edith,
Hutzler, P., Feller, S., Voss, J., Hofler, H. and Becke, K. F. (2000)
Tumor-derived mutated E-cadherin influences β-catenin locali-
zation and increases susceptibility to actin cytoskeletal changes
induced by pervanadate. Cell Commun. Adhesion 7, 391–408
60 Tamada, M., Sheetz, M. P. and Sawada, Y. (2004) Activation of a
signaling cascade by cytoskeleton stretch. Dev. Cell 7, 709–718
61 Lee, H., Gaughan, J. P. and Tsygankov, A. Y. (2008) c-Cbl
facilitates cytoskeletal effects in v-Abl transformed fibroblast
through Rac1 and Rap1-mediated signaling. Int. Biochem. Cell
Biol. 40, 1930–1943
62 Kanzaki, M., Watson, R. T., Hou, J. C., Stamnes, M., Saltiel, A. R.
and Pessin, J. E. (2002) Small GTP-binding protein TC10
differentially regulates two distinct populations of filamentous
actin in 3T3L1 adipocytes. Mol. Biol. Cell 13, 2334–2346
63 Balzac, F., Avolio, M., Degani, S., Kaverina, I., Torti, M.,
Silengo, L., Small, J. V. and Retta, S. F. (2005) E-cadherin
endocytosis regulates the activity of Rap1: a traffic light GTPase
at the crossroads between cadherin and integrin function. J. Cell
Sci. 118, 4765–4783
................................................................... ............................................................ .................................................................. ............................................................. ........................................................ ....................................................
242 C The Authors Journal compilation C 2011 Biochemical Society
C3G signals to actin remodelling
64 Laviolette, M. J., Nunes, P., Peyre, J. B., Aigaki, T. and Stewart,
B. A. (2005) A genetic screen for suppressors of Drosophila NSF2
neuromuscular junction overgrowth. Genetics 170, 779–792
65 McLeod, S. J., Shum, A. J., Lee, R. L., Takei, F. and Gold, M. R.
(2004) The Rap GTPases regulate integrin-mediated adhesion,
cell spreading, actin polymerization, and Pyk2 tyrosine phospho-
rylation in B lymphocytes. J. Biol. Chem. 279, 12009–12019
66 Fukuyama, T., Ogita, H., Kawakatsu, T., Fukuhara, T., Yamada, T.,
Sato, T., Shimizu, K., Nakamura, T., Matsuda, M. and Takai, Y.
(2001) Involvement of c-Src-Crk-C3G-C3G-Rap1 signaling in the
nectin induced activation of Cdc42 and formation of adherens
junctions. J. Biol. Chem. 280, 815–825
67 Abe, T., Kato, M., Miki, H., Takenawa, T. and Endo, T. (2003)
Small GTPase Tc10 and its homologue RhoT induce
N-WASP-mediated long process formation and neurite outgrowth.
J. Cell Sci. 116, 155–168
68 Ridley, A. J. (2006) Rho GTPases and actin dynamics in
membrane protrusions and vesicle trafficking. Trends Cell Biol.
16, 522–529
69 Pommereit, D. and Wouters, F. S. (2007) An NGF-induced
Exo70-TC10 complex locally antagonises Cdc42-mediated
activation of N-WASP to modulate neurite outgrowth. J. Cell Sci.
120, 2694–2705
70 Gawecka, J. E., Griffiths, G. S., Ek-Rylander, B., Ramos, J. W. and
Matter, M. L. (2010) R-Ras regulates migration through an
interaction with filamin A in melanoma cells. PLoS One 5, e11269
71 Ada-Nguema, A. S., Xenias, H., Hofman, J. M., Wiggins, C. H.,
Sheetz, M. P. and Keely, P. J. (2006) The small GTPase R-Ras
regulates organization of actin and drives membrane protrusions
through the activity of PLCε. J. Cell Sci. 119, 1307–1319
72 Jeong, H. W., Nam, J. O. and Kim, I. S. (2005) The C-terminal end
of R-Ras alters the motility and morphology of breast epithelial
cells through Rho/Rho kinase. Can. Res. 65, 507–515
73 Holly, S. P., Barson, M. K. and Parise, L. V. (2005) The unique
N-terminus of R-Ras is required for Rac activation and precise
regulation of cell migration. Mol. Biol. Cell 16, 2458–2469
74 Xu, J., Shi, S., Matsumoto, N., Noda, M. and Kitayama, H. (2007)
Identification of Rgl3 as a potential binding partner for Rap-family
small G-proteins and profilin II. Cell Signalling 19, 1575–1582
75 Taira, K., Umikawa, M., Takei, K., Myagmar, B. E., Shinzato, M.,
Machida, N., Uezato, H., Nonaka, S. and Kariya, K. (2004) The
Traf2- and Nck-interacting kinase as a putative effector of Rap2 to
regulate actin cytoskeleton. J. Biol. Chem. 279, 49488–49496
76 Pannekoek, W. J., Kooistra, M. R., Zwartkruis, F. J. and Bos, J. L.
(2009) Cell–cell junction formation: the role of Rap1 and Rap1
guanine nucleotide exchange factors. Biochim. Biophys. Acta
1788, 790–796
77 Tsukamoto, N., Hattori, M., Yang, H., Bos, J. L. and Minato, N.
(1999) Rap1 GTPase-activating protein SPA-1 negatively
regulates cell adhesion. J. Biol. Chem. 274, 18463–18469
78 Arai, A., Nosaka, Y., Kohsaka, H., Miyasaka, N. and Miura, O.
(1999) CrkL activates integrin-mediated hematopoietic cell
adhesion through the guanine nucleotide exchange factor C3G.
Blood 93, 3713–3722
79 Guerrero, C., Martin-Encabo, S., Fernandez-Medarde, A. and
Santos, E. (2004) C3G-mediated suppression of
oncogene-induced focus formation in fibroblasts involves
inhibition of ERK activation, cyclin A expression and alterations of
anchorage-independent growth. Oncogene 23, 4885–4893
80 Nievers, M. G., Birge, R. B., Greulich, H., Verkleij, A. J., Hanafusa,
H. and van Bergen en Henegouwen, P. M. (1997) v-Crk-induced
cell transformation: changes in focal adhesion composition and
signaling. J Cell Sci. 110, 389–399
81 Oh, J., Seo, D. W., Diaz, T., Wei, B., Ward, Y., Ray, J. M., Morioka,
Y., Shi, S., Kitayama, H., Takahashi, C., Noda, M. and
Stetler-Stevenson, W. G. (2004) Tissue inhibitors of
metalloproteinase 2 inhibits endothelial cell migration through
increased expression of RECK. Cancer Res. 64, 9062–9069
82 Wang, S. F., Aoki, M., Nakashima, Y., Shinozuka, Y., Tanaka, H.,
Taniwaki, M., Hattori, M. and Minato, N. (2008) Development of
Notch-dependent T-cell leukemia by deregulated Rap1 signaling.
Blood 111, 2878–2886
83 Schonherr, C., Yang, H. L., Vigny, M., Palmer, R. H. and Hallberg,
B. (2010) Anaplastic lymphoma kinase activates the small
GTPase Rap1 via the Rap1-specific GEF C3G in both
neuroblastoma and PC12 cells. Oncogene 29, 2817–2830
84 Jin, S., Zhai, B., Qiu, Z., Wu, J., Lane, M. D. and Liao, K. (2000)
c-Crk, a substrate of the insulin-like growth factor-1 receptor
tyrosine kinase, functions as an early signal mediator in
adipocyte differentiation process. J. Biol. Chem. 275,
34444–34452
85 De Falco, V., Castellone, M. D., De Vita, G., Cirafici, A. M.,
Hershman, J. M., Guerrero, C., Fusco, A., Melillo, R. M. and
Santoro, M. (2007) RET/papillary thyroid carcinoma oncogenic
signaling through the Rap1 small GTPase. Cancer Res. 67,
381–390
86 Guerrero, C., Fernandez-Medarde, A., Rojas, J. M., Font de Mora,
J., Esteban, L. M. and Santos, E. (1998) Transformation
suppressor activity of C3G is independent of its CDC25-homology
domain. Oncogene 16, 613–624
87 Gutierrez-Uzquiza, A., Arechederra, M., Molina, I., Banos, R.,
Maia, V., Benito, M., Guerrero, C. and Porras, A. (2010) C3G
down-regulates p38 MAPK activity in response to stress by Rap-1
independent mechanisms: involvement in cell death. Cell
Signalling 22, 533–542
88 Maia, V., Sanz, M., Gutierrez-Berzal, J., de Luis, A.,
Gutierrez-Uzquiza, A., Porras, A. and Guerrero, C. (2009) C3G
silencing enhances STI-571-induced apoptosis in CML cells
through p38 MAPK activation, but it antagonizes STI-571
inhibitory effect on survival. Cell Signalling 21, 1229–1235
89 Asuri, S., Yan, J., Paranavitana, N. C. and Quilliam, L. A. (2008)
E-cadherin disengagement activates Rap1 GTPase. J. Cell
Biochem. 105, 1027–1037
90 Sato, T., Fujita, N., Yamada, A., Ooshio, T., Okamoto, R., Irie, K.
and Takai, Y. (2006) Regulation of the assembly and adhesion
activity of E-cadherin by nectin and afadin for the formation of
adherens junctions in Madin-Darby canine kidney cells. J. Biol.
Chem. 281, 5288–5299
91 Hirata, T., Nagai, H., Koizumi, K., Okino, K., Harada, A., Onda, M.,
Nagahata, T., Mikami, I., Hirai, K., Haraguchi, S. et al. (2004)
Amplification, up-regulation and over-expression of C3G (CRK
SH3 domain-binding guanine nucleotide-releasing factor) in
non-small cell lung cancers. J. Hum. Genet. 49, 290–295
92 Fernandez, V., Jares, P., Salaverria, I., Gine, E., Bea, S., Aymerich,
M., Colomer, D., Villamor, N., Bosch, F., Montserrat, E. et al.
(2008) Gene expression profile and genomic changes in disease
progression of early-stage chronic lymphocytic leukemia.
Haematologica 93, 132–136
93 Virgili, A., Brazma, D., Reid, A. G., Howard-Reeves, J., Valga˜n´on,
M., Chanalaris, A., De Melo, V. A., Marin, D., Apperley, J. F. and
Grace, C. (2008) FISH mapping of Philadelphia negative
BCR/ABL1 positive CML. Mol. Cytogenet. 1, 14
94 Gaulton, K. J., Willer, C. J., Li, Y., Scott, L. J., Conneely, K. N.,
Jackson, A. U., Duren, W. L., Chines, P. S., Narisu, N.,
Bonnycastle, L. L. et al. (2008) Comprehensive association study
of type 2 diabetes and related quantitative traits with 222
candidate genes. Diabetes 57, 3136–3144
95 Hong, K. W., Jin, H. S., Lim, J. E., Ryu, H. J., Go, M. J., Lee, J. Y.,
Woo, J. T., Park, H. K. and Oh, B. (2009) RAPGEF1 gene variants
associated with type 2 diabetes in the Korean population.
Diabetes Res. Clin. Pract. 84, 117–122
96 Rufanova, V. A., Lianos, E., Alexanian, A., Sorokina, E.,
Sharma, M., McGinty, A. and Sorokin, A. (2009) C3G
overexpression in glomerular epithelial cells during
anti-GBM-induced glomerulonephritis. Kidney Int. 75, 31–40
97 Nowak, N. J., Sait, S. N., Zeidan, A., Deeb, G., Gaile, D., Liu, S.,
Ford, L., Wallace, P. K., Wang, E. S. and Wetzler, M. (2010)
Recurrent deletion of 9q34 in adult normal karyotype precursor
B-cell ALL. Can. Genet. Cytogenet. 199, 15–20
...................................................................... .......................................................... .................................................................... ........................................................... ........................................................... ................................................
www.bioscirep.org / Volume 31 (4) / Pages 231–244 243
V. Radha and others
98 Arai, A., Nosaka, Y., Kanda, E., Yamamoto, K., Miyasaka, N. and
Miura, O. (2001) Rap1 is activated by erythropoietin or
interleukin-3 and is involved in regulation of β1 integrin-mediated
hematopoietic cell adhesion. J. Biol. Chem. 276, 10453–10462
99 Reedquist, K. A., Fukazawa, T., Panchamoorthy, G., Langdon,
W. Y., Shoelson, S. E., Druker, B. J. and Band, H. (1996)
Stimulation through the T-cell receptor induces Cbl association
with Crk proteins and the guanine nucleotide exchange protein
C3G. J. Biol. Chem. 271, 8435–8442
100 Smit, L., van der Horst, G. and Borst, J. (1996) Sos, Vav, and
C3G participate in B-cell receptor-inducing signaling pathways
and differentially associate with Shc-Grb2, Crk, and Crk-L
adaptors. J. Biol. Chem. 271, 8564–8569
101 Alsayed, Y., Uddin, S., Ahmad, S., Majchrzak, B., Druker, B. J.,
Fish, E. N. and Platimas, L. C. (2000) IFN-γ activates C3G/Rap1
signaling pathway. J. Immunol. 164, 1800–1806
102 Nasaka, Y., Arai, A., Miyasaka, N. and Miura, O. (1999) CrkL
mediates Ras-dependent activation of the Raf/ERK pathway
through the GEF, C3G in hematopoietic cells stimulated with EPO
and IL-3. J. Biol. Chem. 274, 30154–30162
103 Du, J., AlSayed, Y. M., Xin, F., Ackerman, S. J. and Platanias, L. C.
(2000) Engagement of the CrkL adapter in IL-5 signaling in
eosinophils. J. Biol. Chem. 275, 33167–33175
104 Ballif, B. A., Arnaud, L., Arthur, W. T., Guris, D., Imamoto, A. and
Cooper, J. A. (2004) Activation of a Dab1/CrkL/C3G/Rap1
pathway in Reelin-stimulated neurons. Curr. Biol. 14, 606–610
105 Fukuyama, T., Ogita, H., Kawakatsu, T., Inagaki, M. and Takai, Y.
(2006) Activation of Rac by cadherin through the c-Src-Rap1-
phosphatidylinositol 3-kinase-Vav2 pathway. Oncogene 25,
8–19
106 Posern, G., Rapp, U. R. and Feller, S. M. (2000) The Crk signaling
pathway contributes to the bombesin-induced activation of the
small GTPase Rap1 in Swiss 3T3 cells. Oncogene 19,
6361–6368
107 Boriack-Sjodin, P. A., Margarit, S. M., Bar-Sagi, D. and Kuriyan, J.
(1998) The structural basis of the activation of Ras by Sos.
Nature 394, 337–43
108 van den Berghe, N., Cool, R. H. and Wittinghofer, A. (1999)
Discriminatory residues in Ras and Rap for guanine nucleotide
exchange factor recognition. J. Biol. Chem. 274, 11078–11085
Received 17 August 2010/11 October 2010; accepted 13 October 2010
Published on the Internet 2 March 2011, doi 10.1042/BSR20100094
................................................................... ............................................................ .................................................................. ............................................................. ........................................................ ....................................................
244 C The Authors Journal compilation C 2011 Biochemical Society

Mais conteúdo relacionado

Mais procurados

Structure-Function Analysis of POR Mutants
Structure-Function Analysis of POR MutantsStructure-Function Analysis of POR Mutants
Structure-Function Analysis of POR MutantsAYang999
 
Thant Bio Symposium Poster Spring 2016
Thant Bio Symposium Poster Spring 2016Thant Bio Symposium Poster Spring 2016
Thant Bio Symposium Poster Spring 2016Claire Thant
 
Structure-Function Analysis of POR Mutants
Structure-Function Analysis of POR MutantsStructure-Function Analysis of POR Mutants
Structure-Function Analysis of POR MutantsAYang999
 
Evaluation of the lacZ gene in Escherichia coli mutagenesis using pBluescript...
Evaluation of the lacZ gene in Escherichia coli mutagenesis using pBluescript...Evaluation of the lacZ gene in Escherichia coli mutagenesis using pBluescript...
Evaluation of the lacZ gene in Escherichia coli mutagenesis using pBluescript...Emilio Solomon
 
Molecular biology revision-Part 3 (Regulation of genes expression and Recombi...
Molecular biology revision-Part 3 (Regulation of genes expression and Recombi...Molecular biology revision-Part 3 (Regulation of genes expression and Recombi...
Molecular biology revision-Part 3 (Regulation of genes expression and Recombi...Namrata Chhabra
 
PhD Federica III anno
PhD Federica III annoPhD Federica III anno
PhD Federica III annolab13unisa
 
Study of psymberin's mode of action using forward genetics
Study of psymberin's mode of action using forward geneticsStudy of psymberin's mode of action using forward genetics
Study of psymberin's mode of action using forward geneticsVincent Tsao
 
tuning the pH Response of i-Motif DNA Oligonucleotides_Lannes_et_al-2015-Chem...
tuning the pH Response of i-Motif DNA Oligonucleotides_Lannes_et_al-2015-Chem...tuning the pH Response of i-Motif DNA Oligonucleotides_Lannes_et_al-2015-Chem...
tuning the pH Response of i-Motif DNA Oligonucleotides_Lannes_et_al-2015-Chem...saheli halder
 
Mol. Biol. Cell-2015-Ayache-2579-95
Mol. Biol. Cell-2015-Ayache-2579-95Mol. Biol. Cell-2015-Ayache-2579-95
Mol. Biol. Cell-2015-Ayache-2579-95Jessica Ayache
 
Sigma Xi Presentation
Sigma Xi PresentationSigma Xi Presentation
Sigma Xi PresentationSKU sxi
 
adenylate_cyclase_poster
adenylate_cyclase_posteradenylate_cyclase_poster
adenylate_cyclase_posterKelly Thompson
 
Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human Cancer
Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human CancerSima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human Cancer
Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human CancerSima Lev
 
Small Molecule Interactions with Protein Tyrosine Phosphatase
Small Molecule Interactions with Protein Tyrosine PhosphataseSmall Molecule Interactions with Protein Tyrosine Phosphatase
Small Molecule Interactions with Protein Tyrosine PhosphataseJonathan Paul
 
ShRNA-specific regulation of FMNL2 expression in P19 cells
ShRNA-specific regulation of FMNL2 expression in P19 cellsShRNA-specific regulation of FMNL2 expression in P19 cells
ShRNA-specific regulation of FMNL2 expression in P19 cellsYousefLayyous
 
DNA damage and repair systems
DNA damage and repair systemsDNA damage and repair systems
DNA damage and repair systemsSaira Fatima
 

Mais procurados (20)

Structure-Function Analysis of POR Mutants
Structure-Function Analysis of POR MutantsStructure-Function Analysis of POR Mutants
Structure-Function Analysis of POR Mutants
 
Thant Bio Symposium Poster Spring 2016
Thant Bio Symposium Poster Spring 2016Thant Bio Symposium Poster Spring 2016
Thant Bio Symposium Poster Spring 2016
 
Structure-Function Analysis of POR Mutants
Structure-Function Analysis of POR MutantsStructure-Function Analysis of POR Mutants
Structure-Function Analysis of POR Mutants
 
Evaluation of the lacZ gene in Escherichia coli mutagenesis using pBluescript...
Evaluation of the lacZ gene in Escherichia coli mutagenesis using pBluescript...Evaluation of the lacZ gene in Escherichia coli mutagenesis using pBluescript...
Evaluation of the lacZ gene in Escherichia coli mutagenesis using pBluescript...
 
Molecular biology revision-Part 3 (Regulation of genes expression and Recombi...
Molecular biology revision-Part 3 (Regulation of genes expression and Recombi...Molecular biology revision-Part 3 (Regulation of genes expression and Recombi...
Molecular biology revision-Part 3 (Regulation of genes expression and Recombi...
 
PhD Federica III anno
PhD Federica III annoPhD Federica III anno
PhD Federica III anno
 
Study of psymberin's mode of action using forward genetics
Study of psymberin's mode of action using forward geneticsStudy of psymberin's mode of action using forward genetics
Study of psymberin's mode of action using forward genetics
 
tuning the pH Response of i-Motif DNA Oligonucleotides_Lannes_et_al-2015-Chem...
tuning the pH Response of i-Motif DNA Oligonucleotides_Lannes_et_al-2015-Chem...tuning the pH Response of i-Motif DNA Oligonucleotides_Lannes_et_al-2015-Chem...
tuning the pH Response of i-Motif DNA Oligonucleotides_Lannes_et_al-2015-Chem...
 
Mol. Biol. Cell-2015-Ayache-2579-95
Mol. Biol. Cell-2015-Ayache-2579-95Mol. Biol. Cell-2015-Ayache-2579-95
Mol. Biol. Cell-2015-Ayache-2579-95
 
Crispr trap
Crispr trapCrispr trap
Crispr trap
 
Sigma Xi Presentation
Sigma Xi PresentationSigma Xi Presentation
Sigma Xi Presentation
 
Mmp 2
Mmp 2Mmp 2
Mmp 2
 
adenylate_cyclase_poster
adenylate_cyclase_posteradenylate_cyclase_poster
adenylate_cyclase_poster
 
C2H2_research
C2H2_researchC2H2_research
C2H2_research
 
Reprogramming
ReprogrammingReprogramming
Reprogramming
 
Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human Cancer
Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human CancerSima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human Cancer
Sima lev: Lipid Transfer Proteins and Membrane Contact Sites in Human Cancer
 
Small Molecule Interactions with Protein Tyrosine Phosphatase
Small Molecule Interactions with Protein Tyrosine PhosphataseSmall Molecule Interactions with Protein Tyrosine Phosphatase
Small Molecule Interactions with Protein Tyrosine Phosphatase
 
Wang_NSMB2009
Wang_NSMB2009Wang_NSMB2009
Wang_NSMB2009
 
ShRNA-specific regulation of FMNL2 expression in P19 cells
ShRNA-specific regulation of FMNL2 expression in P19 cellsShRNA-specific regulation of FMNL2 expression in P19 cells
ShRNA-specific regulation of FMNL2 expression in P19 cells
 
DNA damage and repair systems
DNA damage and repair systemsDNA damage and repair systems
DNA damage and repair systems
 

Destaque

Presentación de power point oportunidades de exportación - 26 de octubre de...
Presentación de power point   oportunidades de exportación - 26 de octubre de...Presentación de power point   oportunidades de exportación - 26 de octubre de...
Presentación de power point oportunidades de exportación - 26 de octubre de...rodolfo1253
 
Presentation in the British Embassy in Madrid - Threats and opportunities in ...
Presentation in the British Embassy in Madrid - Threats and opportunities in ...Presentation in the British Embassy in Madrid - Threats and opportunities in ...
Presentation in the British Embassy in Madrid - Threats and opportunities in ...col.lab | collaboration laboratory
 
A s nov 2013 full web pdf
A s nov 2013 full web pdfA s nov 2013 full web pdf
A s nov 2013 full web pdfMadhavbaug
 
Textos de magia en papiros griegos
Textos de magia    en papiros griegosTextos de magia    en papiros griegos
Textos de magia en papiros griegosMónica Cano
 
Géneros literarios
Géneros literariosGéneros literarios
Géneros literariosKatalogador
 
Break the Rules - The WOLF & SELL ME THIS PEN
Break the Rules - The WOLF & SELL ME THIS PENBreak the Rules - The WOLF & SELL ME THIS PEN
Break the Rules - The WOLF & SELL ME THIS PENGuillaume Warckol
 
בדיקה בעברית
בדיקה בעבריתבדיקה בעברית
בדיקה בעבריתTom Meir
 
Social justice facilitation pack - introductory letter
Social justice facilitation pack - introductory letterSocial justice facilitation pack - introductory letter
Social justice facilitation pack - introductory letterfairerscotland
 
Lexington Communications: Education & Skills Round-Up
Lexington Communications: Education & Skills Round-Up Lexington Communications: Education & Skills Round-Up
Lexington Communications: Education & Skills Round-Up LexEduSkills
 
What do LinkedIn endorsements mean?
What do LinkedIn endorsements mean?What do LinkedIn endorsements mean?
What do LinkedIn endorsements mean?The Level
 
Eit orginal
Eit orginalEit orginal
Eit orginalanamsini
 
The Habit of Retention
The Habit of RetentionThe Habit of Retention
The Habit of RetentionAmanda Burbage
 
E&y acma presentation_5th_septembr_2012
E&y acma presentation_5th_septembr_2012E&y acma presentation_5th_septembr_2012
E&y acma presentation_5th_septembr_2012keshav_13
 
Five Strategies to Consider as You Seek Capital for Your Business
Five Strategies to Consider as You Seek Capital for Your BusinessFive Strategies to Consider as You Seek Capital for Your Business
Five Strategies to Consider as You Seek Capital for Your BusinessCharlie Stewart
 
Genius Recipe for Homemade Soup
Genius Recipe for Homemade SoupGenius Recipe for Homemade Soup
Genius Recipe for Homemade SoupSuzy Bowler
 
尼斯
尼斯尼斯
尼斯tinaho
 

Destaque (20)

Presentación de power point oportunidades de exportación - 26 de octubre de...
Presentación de power point   oportunidades de exportación - 26 de octubre de...Presentación de power point   oportunidades de exportación - 26 de octubre de...
Presentación de power point oportunidades de exportación - 26 de octubre de...
 
Presentation in the British Embassy in Madrid - Threats and opportunities in ...
Presentation in the British Embassy in Madrid - Threats and opportunities in ...Presentation in the British Embassy in Madrid - Threats and opportunities in ...
Presentation in the British Embassy in Madrid - Threats and opportunities in ...
 
A s nov 2013 full web pdf
A s nov 2013 full web pdfA s nov 2013 full web pdf
A s nov 2013 full web pdf
 
Textos de magia en papiros griegos
Textos de magia    en papiros griegosTextos de magia    en papiros griegos
Textos de magia en papiros griegos
 
Géneros literarios
Géneros literariosGéneros literarios
Géneros literarios
 
Break the Rules - The WOLF & SELL ME THIS PEN
Break the Rules - The WOLF & SELL ME THIS PENBreak the Rules - The WOLF & SELL ME THIS PEN
Break the Rules - The WOLF & SELL ME THIS PEN
 
בדיקה בעברית
בדיקה בעבריתבדיקה בעברית
בדיקה בעברית
 
Social justice facilitation pack - introductory letter
Social justice facilitation pack - introductory letterSocial justice facilitation pack - introductory letter
Social justice facilitation pack - introductory letter
 
Lexington Communications: Education & Skills Round-Up
Lexington Communications: Education & Skills Round-Up Lexington Communications: Education & Skills Round-Up
Lexington Communications: Education & Skills Round-Up
 
What do LinkedIn endorsements mean?
What do LinkedIn endorsements mean?What do LinkedIn endorsements mean?
What do LinkedIn endorsements mean?
 
Eit orginal
Eit orginalEit orginal
Eit orginal
 
投稿用
投稿用投稿用
投稿用
 
transcript
transcripttranscript
transcript
 
The Habit of Retention
The Habit of RetentionThe Habit of Retention
The Habit of Retention
 
E&y acma presentation_5th_septembr_2012
E&y acma presentation_5th_septembr_2012E&y acma presentation_5th_septembr_2012
E&y acma presentation_5th_septembr_2012
 
Apresentação Estilo
Apresentação EstiloApresentação Estilo
Apresentação Estilo
 
Oral Cancer infographic
Oral Cancer infographicOral Cancer infographic
Oral Cancer infographic
 
Five Strategies to Consider as You Seek Capital for Your Business
Five Strategies to Consider as You Seek Capital for Your BusinessFive Strategies to Consider as You Seek Capital for Your Business
Five Strategies to Consider as You Seek Capital for Your Business
 
Genius Recipe for Homemade Soup
Genius Recipe for Homemade SoupGenius Recipe for Homemade Soup
Genius Recipe for Homemade Soup
 
尼斯
尼斯尼斯
尼斯
 

Semelhante a review

PNAS-2013-Arambula-8212-7
PNAS-2013-Arambula-8212-7PNAS-2013-Arambula-8212-7
PNAS-2013-Arambula-8212-7Wenge Wong
 
vulva formation in c.elegans.pptx
vulva formation in c.elegans.pptxvulva formation in c.elegans.pptx
vulva formation in c.elegans.pptxRAHANAMOIDEENKOYAVK
 
vulva formation in c.elegans.pptx
vulva formation in c.elegans.pptxvulva formation in c.elegans.pptx
vulva formation in c.elegans.pptxRAHANAMOIDEENKOYAVK
 
vulva formation in c.elegans.pptx
vulva formation in c.elegans.pptxvulva formation in c.elegans.pptx
vulva formation in c.elegans.pptxRAHANAMOIDEENKOYAVK
 
vulva formation in c.elegans.pptx
vulva formation in c.elegans.pptxvulva formation in c.elegans.pptx
vulva formation in c.elegans.pptxRAHANAMOIDEENKOYAVK
 
transciption powerpoint
transciption powerpointtransciption powerpoint
transciption powerpointNikka Bañez
 
RAGE-Mediated Cell Signaling.pdf
RAGE-Mediated Cell Signaling.pdfRAGE-Mediated Cell Signaling.pdf
RAGE-Mediated Cell Signaling.pdfNazmunNahar479158
 
Molecular cell biology 2 transcription
Molecular cell biology 2 transcriptionMolecular cell biology 2 transcription
Molecular cell biology 2 transcriptionDr. sreeremya S
 
Genetic retargeting of E3 ligases to enhance CAR T therapy
Genetic retargeting of E3 ligases to enhance CAR T therapyGenetic retargeting of E3 ligases to enhance CAR T therapy
Genetic retargeting of E3 ligases to enhance CAR T therapyDrShafi4
 
Expression systems
Expression systemsExpression systems
Expression systemsBruno Mmassy
 
Galectins Potential Targes For Cancer Therapy
Galectins   Potential Targes For Cancer TherapyGalectins   Potential Targes For Cancer Therapy
Galectins Potential Targes For Cancer Therapyguest82d2e9
 
Pells et al [2015] PLoS ONE 10[7] e0131102
Pells et al [2015] PLoS ONE 10[7] e0131102Pells et al [2015] PLoS ONE 10[7] e0131102
Pells et al [2015] PLoS ONE 10[7] e0131102Steve Pells
 
Senior Thesis-Analyzing the interactions between MYOGEF and a component of er...
Senior Thesis-Analyzing the interactions between MYOGEF and a component of er...Senior Thesis-Analyzing the interactions between MYOGEF and a component of er...
Senior Thesis-Analyzing the interactions between MYOGEF and a component of er...Dougan McGrath
 
RNA Sequencing Research
RNA Sequencing ResearchRNA Sequencing Research
RNA Sequencing ResearchTanmay Ghai
 

Semelhante a review (20)

publication 4
publication 4publication 4
publication 4
 
PNAS-2013-Arambula-8212-7
PNAS-2013-Arambula-8212-7PNAS-2013-Arambula-8212-7
PNAS-2013-Arambula-8212-7
 
vulva formation in c.elegans.pptx
vulva formation in c.elegans.pptxvulva formation in c.elegans.pptx
vulva formation in c.elegans.pptx
 
vulva formation in c.elegans.pptx
vulva formation in c.elegans.pptxvulva formation in c.elegans.pptx
vulva formation in c.elegans.pptx
 
vulva formation in c.elegans.pptx
vulva formation in c.elegans.pptxvulva formation in c.elegans.pptx
vulva formation in c.elegans.pptx
 
vulva formation in c.elegans.pptx
vulva formation in c.elegans.pptxvulva formation in c.elegans.pptx
vulva formation in c.elegans.pptx
 
transciption powerpoint
transciption powerpointtransciption powerpoint
transciption powerpoint
 
RAGE-Mediated Cell Signaling.pdf
RAGE-Mediated Cell Signaling.pdfRAGE-Mediated Cell Signaling.pdf
RAGE-Mediated Cell Signaling.pdf
 
Molecular cell biology 2 transcription
Molecular cell biology 2 transcriptionMolecular cell biology 2 transcription
Molecular cell biology 2 transcription
 
Imaginal discs1
Imaginal discs1Imaginal discs1
Imaginal discs1
 
publication 3
publication 3publication 3
publication 3
 
Genetic retargeting of E3 ligases to enhance CAR T therapy
Genetic retargeting of E3 ligases to enhance CAR T therapyGenetic retargeting of E3 ligases to enhance CAR T therapy
Genetic retargeting of E3 ligases to enhance CAR T therapy
 
Expression systems
Expression systemsExpression systems
Expression systems
 
Galectins Potential Targes For Cancer Therapy
Galectins   Potential Targes For Cancer TherapyGalectins   Potential Targes For Cancer Therapy
Galectins Potential Targes For Cancer Therapy
 
769.full
769.full769.full
769.full
 
Pells et al [2015] PLoS ONE 10[7] e0131102
Pells et al [2015] PLoS ONE 10[7] e0131102Pells et al [2015] PLoS ONE 10[7] e0131102
Pells et al [2015] PLoS ONE 10[7] e0131102
 
publication 1
publication 1publication 1
publication 1
 
Senior Thesis-Analyzing the interactions between MYOGEF and a component of er...
Senior Thesis-Analyzing the interactions between MYOGEF and a component of er...Senior Thesis-Analyzing the interactions between MYOGEF and a component of er...
Senior Thesis-Analyzing the interactions between MYOGEF and a component of er...
 
IJBB-10803348
IJBB-10803348IJBB-10803348
IJBB-10803348
 
RNA Sequencing Research
RNA Sequencing ResearchRNA Sequencing Research
RNA Sequencing Research
 

review

  • 1. Biosci. Rep. (2011) / 31 / 231–244 (Printed in Great Britain) / doi 10.1042/BSR20100094 Signalling to actin: role of C3G, a multitasking guanine-nucleotide-exchange factor Vegesna RADHA1 , Aninda MITRA, Kunal DAYMA and Kotagiri SASIKUMAR Centre for Cellular and Molecular Biology (CSIR), Uppal Road, Hyderabad 500 007, India ' & $ % Synopsis C3G (Crk SH3-domain-binding guanine-nucleotide-releasing factor) is a ubiquitously expressed member of a class of molecules called GEFs (guanine-nucleotide-exchange factor) that activate small GTPases and is involved in pathways triggered by a variety of signals. It is essential for mammalian embryonic development and many cellular functions in adult tissues. C3G participates in regulating functions that require cytoskeletal remodelling such as adhesion, migration, maintenance of cell junctions, neurite growth and vesicle traffic. C3G is spatially and temporally regulated to act on Ras family GTPases Rap1, Rap2, R-Ras, TC21 and Rho family member TC10. Increased C3G protein levels are associated with differentiation of various cell types, indicating an important role for C3G in cellular differentiation. In signalling pathways, C3G serves functions dependent on catalytic activity as well as protein interaction and can therefore integrate signals necessary for the execution of more than one cellular function. This review summarizes our current knowledge of the biology of C3G with emphasis on its role as a transducer of signals to the actin cytoskeleton. Deregulated C3G may also contribute to pathogenesis of human disorders and therefore could be a potential therapeutic target. Key words: actin cytoskeleton, C3G (Crk SH3-domain-binding guanine-nucleotide-releasing factor), differentiation, embryonic development, GTPase, signalling INTRODUCTION The ability of small GTPases to switch between active GTP and inactive GDP-bound states enables them to function as hubs in signalling pathways. A small number of GTPases can respond to a multitude of signals and also activate multiple downstream effectors, resulting in diverse and specific responses. GTPases are activated by GEFs (guanine-nucleotide-exchange factors) and inhibited by GTPase-activating proteins that accelarate GTP hy- drolysis [1]. Most upstream signals target GEFs to act on specific GTPases and therefore GEFs serve to link activated receptors to downstream signalling cascades and provide signalling spe- cificity [2]. GEFs are classified on the basis of which family of GTPases they act on, namely Ras GEFs, Rho GEFs etc. A com- mon feature of Ras GEFs is the presence of a CDC25 homology domain that helps in catalysis along with an REM (Ras exchanger motif). In addition, Rap GEFs have multiple modular domains that aid in protein and lipid interactions, and in their regulation [3]. C3G (Crk SH3-domain-binding guanine nucleotide-releasing factor) was the first Rap GEF identified with a domain showing ............................................................................................................................................................................................................................................................................................................ Abbreviations used: 3D, three-dimensional: AJ, adherence junction; C3G, Crk SH3-domain-binding guanine nucleotide releasing factor; CML, chronic myelogenous leukaemia; DC3G, Drosophila C3G; EGF, epidermal growth factor; ERK, extracellular-signal-regulated kinase; GEF, guanine-nucleotide-exchange factor; JNK, c-Jun N-terminal kinase; MAPK, mitogen-activated protein kinase; NB, neuroblastoma; NGF, nerve growth factor; pC3G, Y504 (Tyr504)-phosphorylated C3G; PDGF, platelet-derived growth factor; PV, pervanadate; REM, Ras exchanger motif; SFK, Src family kinase; siRNA, small interfering RNA; T2D, Type 2 diabetes. 1 To whom correspondence should be addressed (e-mail: vradha@ccmb.res.in) homology with the yeast CDC25, and was originally isolated as an interacting partner of CRK (cellular homologue of the v-Crk oncoprotein) [4,5]. Alternate names of C3G are Rap GEF1, GRF2 and DKFZ p781P1719. C3G has the catalytic domain along with REM at the extreme C-terminus and lacks modular protein in- teraction domains found in most other Rap GEFs (Figure 1A). Domains found in Rap GEFs generally are DEP (disheveled- EGL-10-pleckstrin domain), cNB-L (cyclic nucleotide-binding domain-like) and PDZ (PSD-95/Dig/ZO-1). These are shown in Figure 1(B) along with the primary structure of C3G. In humans, the two primary protein products of about 140 kDa are over 1000 residues in length and have multiple proline-rich sequences in the central region through which they interact with proteins con- taining SH3 domains. Crk, Hck, c-Abl and Cas are molecules known to interact directly with the central domain of C3G [4–8]. Residues in the N-terminus are responsible for interaction with E-cadherin, indicating that the N-terminal sequences may also aid in protein interaction [9]. The two isoforms arise due to al- ternate splicing, and primarily differ in the N-terminus where three amino acids of isoform a are replaced by 21 amino acids in isoform b (Figures 2A and 2B). www.bioscirep.org / Volume 31 (4) / Pages 231–244 231 BioscienceReportswww.bioscirep.org
  • 2. V. Radha and others Figure 1 Domain organization of C3G and comparison with other Rap GEFs (A) Schematic diagram showing the domain organization of C3G protein. The C-terminal catalytic domain of C3G is homologous with CDC25 and is responsible for target G protein activation. The N-terminal region has a domain that interacts with E-cadherin. The central protein interaction domain (also known as Crk-binding region, CBR) contains multiple proline-rich sequences that bind SH3 domains of Crk, Cas, c-Abl and Hck. The non-catalytic sequences negatively regulate the catalytic activity of C3G. (B) Domain organization of different Rap GEFs. C3G is a unique Rap GEF member that lacks modular protein interaction domains found in other Rap GEFs. cAMP: cAMP-binding site; EF, EF-hand calcium-binding domain; Ras GEF or CDC25 homology domain; RA, Ras-association domain; Y504, Tyr504 . A single-copy gene at chromosomal location 9q34.3 en- codes C3G [10]. Other proteins encoded from the same region are c-Abl, nucleoporin 214, laminin γ 3, NET39, protein-O- mannosyltransferase, uridine-cytidine kinase 1, mediator com- plex subunit 27 and sarcosine dehydrogenase. The human C3G gene comprises 24 exons spanning 163 kb (Figure 2A). Tran- script size of isoform a is 6085 bp and that of isoform b, 6256 bp. Its homologues have been cloned from several organisms and show a high degree of conservation in the catalytic domain (Figure 3). The proline-rich stretches and E-cadherin-binding domain are conserved among all the vertebrates from which C3G has been cloned. In invertebrates, some putative proline- rich SH3-binding stretches could be identified in the primary sequence, but the E-cadherin-binding domain shows poor con- servation. C3G may therefore have evolved to perform a broader range of functions in the vertebrates. A variety of stimuli such as growth factors, cytokines, integrins, neurotrophins, hormones and mechanical stress have been shown to engage C3G-mediated signalling (Table 1). The GTPases known to be regulated by C3G are Ras family members Rap1, Rap2, R-Ras, TC-21 and the Rho family member TC10 leading to the activation of MAPK (mitogen-activated protein kinase) and other effector pathways [11–19]. Generally, GEFs do not show promiscuity in targeting members of the various G-protein subfamilies and act within their specific family of G-proteins. C3G is an example of a GEF that targets a Rho family member in addition to the Ras family GTPases. Over the past 15 years, several studies have thrown light on the involvement of C3G in multiple signalling pathways and its role in regulating diverse cellular functions. Through the activation of ERK (extracellular-signal- regulated kinase), JNK (c-Jun N-terminal kinase) and Rac signalling, C3G plays a crucial role in integrin-mediated cell adhesion and migration and also regulates cell proliferation, differentiation and apoptosis. These cellular properties are often associated with, or are a consequence of, cytoskeletal reorganiz- ation. In the present review, we highlight these properties of C3G and describe the consequence of its deregulation in cells as well ................................................................... ............................................................ .................................................................. ............................................................. ........................................................ .................................................... 232 C The Authors Journal compilation C 2011 Biochemical Society
  • 3. C3G signals to actin remodelling Figure 2 Isoforms of C3G (A) Schematic diagram showing the genome organization of C3G gene. The human C3G gene comprises 24 exons, spanning 163 kb on chromosome 9q34.3. Human C3G has two predominant isoforms, a and b, which arise due to alternate splicing and differ in their N-termini. Isoform a has 6085 bp of transcript length, whereas isoform b has 6256 bp of transcript length. (B) Characterized mammalian isoforms of C3G. The two isoforms of C3G protein differ such that 3 aa (amino acids) of Isoform a are replaced by 21 aa in isoform b. A truncated isoform is expressed in CML cells (K562), p87C3G which arises from a 4.5 kb transcript. An alternate isoform in rat, which is expressed only in testis and brain, has a 51 aa (153 bp) insertion, just after the proline-rich domain. In mouse an additional isoform is found which has a deletion of 38 aa at the N-terminal. as during embryonic development. The examples of defective signalling due to C3G leading to pathological states are also presented. Isoforms and expression Although C3G is ubiquitously expressed, some tissue-specific differences in expression levels have been seen. C3G transcripts are subject to alternate splicing and variant isoforms have been cloned from different species (Figure 2B). Rat tissues have shown the presence of a major ubiquitously expressed 7 kb transcript and a 4 kb transcript in some tissues [20]. An isoform contain- ing a 153 bp insert after the fifth proline-rich region has also been cloned from rat testis [20]. This isoform is predominantly expressed in the testis and to some extent in the brain unlike all other tissues which show predominant expression of only the isoform without this insert. Specific functions served by these isoforms have not been studied yet. In human tissues, too, C3G shows ubiquitous expression, but levels of a 7.5-kb transcript were high in adult skeletal muscle and placenta, fetal heart and brain and low in the liver [4]. A short 87-kDa isoform encoded by a 4.4-kb transcript is expressed in myeloid leukaemic cells, and lacks N-terminal 305 amino acids of the full-length C3G [21]. This lacks the first two polyproline regions and interacts with Bcr-Abl through the third proline-rich sequence. The p87 isoform showed differences in expression levels depending on disease remission during treatment, suggest- ing a role for C3G in CML (chronic myelogenous leukaemia) ...................................................................... .......................................................... .................................................................... ........................................................... ........................................................... ................................................ www.bioscirep.org / Volume 31 (4) / Pages 231–244 233
  • 4. V. Radha and others Figure 3 C3G protein from various species A comparative analysis of C3G protein from various species is shown with human (H. sapiens) C3G. Human C3G (isoform b) shows E-cadherin-binding domain (red), proline-rich regions (light blue), REM (dark blue) and Ras GEF domain (black). Amino acid residues belonging to each domain are mentioned and the position of amino acid residue at the start of each proline-rich region is also mentioned. The longest isoform of C3G from each species is depicted along with all the domains and the regions showing significant homology with human C3G with percentage identity. Corresponding regions are indicated (in red). Analysis reveals the presence of putative proline-rich SH3 binding sequences in C3G protein from various species. The N-terminal region in C3G protein from rat (R. norvegicus), mouse (M. musculus), Xenopus (X. laevis) and zebrafish (D. rerio) showing high identity with E-cadherin-binding domain of human C3G is also depicted. pathogenesis. In mouse tissues, two transcripts with and without a 114 bp insertion in the N-terminal were expressed in most tis- sues. C3G expression was high in brain, heart, liver and muscle and low in adipose tissue, kidney and spleen [22]. Regulation Currently, very little information is available on regulation of C3G expression. Difference in relative expression of the two mouse isoforms was seen during adipocyte differentiation [22]. ................................................................... ............................................................ .................................................................. ............................................................. ........................................................ .................................................... 234 C The Authors Journal compilation C 2011 Biochemical Society
  • 5. C3G signals to actin remodelling Table 1 Stimuli that engage C3G JAK, Janus kinase; STAT, signal transducer and activator of transcription; TIMP2 tissue inhibitor of metallo- proteinases 2. Stimulus Molecules involved in the pathway Reference(s) Integrin binding VLA-4/VLA-5/R-Ras [33,78,98] T-cell receptor c-Cbl/CrkL [99] B-cell receptor c-Cbl/CrkL [100] Insulin Cbl/Crk/C3G/TC10 [17,36,47] EGF EGF/Crk/C3G/Rap1/B-Raf [47,48] NGF FRS2/Crk/C3G /Rap1/B-Raf [14,48] Interferon γ c-Cbl/CrkL [101] Erythropoetin, interleukin-3 and interleukin-5 CrkL/STAT5 [102,103] Hepatocyte growth factor Gab1-CrkL [34] Growth hormone JAK2 and c-Src [18] Reelin stimulation via Dab1/CrkL [104] Mechanical force CrkII/Cas [60] Nectin c-Src/Crk [66] Cadherins c-Src/Vav2/Crk [105] Bombesin Crk/CrkL [106] TIMP2 Crk [81] Figure 4 Differentiated human monocytes express higher levels of C3G protein Two human monocytic cell lines U937 and HL-60 were induced to dif- ferentiate to a macrophage lineage by treating with 10 ng of PMA for 48 h or 1 % DMSO for 24 and 48 h respectively. Whole-cell lysates were prepared along with UT (untreated) cells and subjected to Western blotting by using indicated antibodies. Hck was used as a marker of differentiation and Cdk2 as a protein loading control. C3G protein levels also increase on differentiation of NB (neur- oblastoma) cells [23]. Similarly, enhancement in C3G protein was seen on differentiation of human monocytic cells to a mac- rophage lineage (Figure 4). A several-fold increase in C3G gene expression was observed on keratinocyte growth factor treatment of human airway epithelia, indicating that C3G expression may be regulated transcriptionally [24]. No information is available on the promoter of C3G. In silico analysis carried out by us using web-based software, Promo 3.0 and BKL TRANSFAC, has shown binding sites for multiple transcription factors in the upstream regulatory region, but they require experimental valida- tion. Decreased C3G expression was found in cervical squamous cell carcinomas due to hypermethylation of upstream regulat- ory sequences [25]. There appears to be an inter-relationship in the expression of some GEFs. Knocking down of DOCK-180, a GEF for Rac resulted in an increase in C3G levels leading to changes in many cellular properties such as reduced proliferation and attenuated migration in ovarian carcinoma cells [26]. Most of the Rap GEFs are multidomain proteins and their activation is regulated by protein–lipid interaction, binding of second messengers, post-translational modification and subcel- lular localization. C3G activation has been shown to be regulated by tyrosine phosphorylation at Y504 and membrane targeting, en- abled through its interaction with the adaptor protein Crk [27]. c- Src, Hck, Fyn and c-Abl are kinases known to phosphorylate C3G at Y504 [7,28–30]. The sequence surrounding Y504 of human C3G is not totally conserved in rat and mouse, indicating species- specific differences in C3G regulation. In addition to Y504, C3G is phosphorylated on other tyrosine residues, but their contri- bution to C3G regulation has not been studied [30]. The SH2 domain in Crk enables translocation of the Crk–C3G complex to tyrosine-phosphorylated molecules [such as receptor tyrosine kinases, p130Cas, Cbl, ARMS (ankyrin repeat-rich membrane spanning), IRS-1 (insulin receptor substrate-1) and paxillin] in response to extracellular stimuli [31–34]. Complex formation between Crk and C3G is influenced by Crk phosphorylation and the tyrosine phosphatase PTP1B regulates this modification [6,35–37]. C3G regulation to activate specific GTPases may be complex. C3G shows constitutive membrane binding upon v-Crk trans- formation [38]. C3G expression enhances JNK activation and transformation in v-Crk NIH 3T3 cells. In this case, localization to the plasma membrane was not sufficient for JNK activation. The catalytic domain was required but was independent of Rap1 indicating that, under these conditions, C3G targeted other GT- Pases. Constitutive association of C3G with Crk has been de- scribed. This interaction seems to vary in an adhesion-dependent ...................................................................... .......................................................... .................................................................... ........................................................... ........................................................... ................................................ www.bioscirep.org / Volume 31 (4) / Pages 231–244 235
  • 6. V. Radha and others Figure 5 C3G localizes to filopodia tips Left panel: HeLa cells transfected with C3G expression vector were stained for C3G and F-actin. C3G expressing cells show stable filopodial extensions with C3G localized to their tips. Right panel: HeLa cells treated with 50 mM PV, for 20 min show filopodia extensions. Staining for p-C3G and F-actin showed pC3G localized predominantly at the Golgi and filopodia tips. Images were captured using a confocal microscope. manner and in response to other stimuli [36,39]. The ability of molecules such as Cbl to alter CrkL–C3G interaction affects C3G activation [40]. Cbl-b plays a negative role since Cbl− / − T-cells show better interaction and higher Rap1 activation. In response to insulin receptor signalling in skeletal muscle cells, translocation of C3G to lipid rafts regulates its activation, and disruption of flotillin-based membrane domains prevents C3G activation [41]. In neutrophils, the bacterial chemoattractant pro- tein fMLP (fMet-Leu-Phe) causes membrane targeting of C3G dependent on function of the cytoskeletal regulator protein VASP (vasodilator-stimulated phosphoprotein) [42]. Expression of pro- teins like Bcr-Abl reduces the interaction of C3G and CrkL and inhibits tyrosine phosphorylation of C3G upon cell spreading and attachment of NIH 3T3 cells [43]. Bcr-Abl has been found in a complex containing C3G dependent on CrkL [44]. C3G is also subject to autoregulation. It is known that C3G enzyme activity is regulated negatively by its non-catalytic se- quence since deletion of non-catalytic residues results in con- stitutive catalytic activity [27]. The activation of C3G in the cells may also be regulated through targeting to specific intracellu- lar domains [45,46]. All studies so far have shown that C3G localizes to the cytoplasmic compartment. In epithelial cells, overexpressed C3G induces filopodia and localizes to filopodia tips (Figure 5). PV (pervanadate)-induced filopodia show pC3G (Y504-phosphorylated C3G) localized to their tips indicating a role for C3G in filopodia functions (Figure 5). C3G, upon being phosphorylated by SFKs (Src family kinases) or c-Abl, has been shown to localize to the subcortical actin cytoskeleton, Golgi and retracting lamellipodia of cells undergoing apoptosis [23,28,30]. Multimolecular complex formation involving C3G in response to stimuli is a major means of activating C3G. Several proteins that are capable of interacting with C3G directly or indirectly have been identified and their involvement in pathways leading to specific functions are shown in Figure 6. Components of mul- timolecular complexes containing C3G also vary depending on the stimulus [39,47]. Stimulation of PC12 cells by EGF (epi- dermal growth factor), results in the formation of a short-lived complex containing Crk, C3G, Rap1 and B-Raf. NGF (nerve growth factor) stimulation causes formation of a stable complex containing FRS2 (fibroblast growth factor receptor substrate 2), Crk, C3G, Rap1 and B-Raf leading to prolonged MAPK activa- tion [48]. In response to cell adhesion, Cas association with C3G brings it into proximity of Src and focal adhesion kinase at focal adhesions leading to the activation of JNK by integrins in fibro- blasts [49]. In response to the activation of Fcγ R1 of myeloid cells, complex formation is seen with the cytoskeletal protein Hef-1, Crk, Cbl and C3G [50]. In Ba/F3 haematopoietic cells, CrkL was found in a complex with C3G, Sos (Son of seven- less) and c-Abl, but upon Bcr-Abl expression this complex is disrupted [51]. In NIH 3T3 cells, PDGF (platelet-derived growth factor) induces formation of complexes containing Necl-5, Integ- rin α1βIII, PDGF-R (PDGF receptor), Rap1, Crk, C3G and Ral GDS that enable cell movement [52]. FUNCTIONS Role in embryonic development The in vivo function of mammalian C3G has been studied by de- veloping mice lacking C3G expression (knockout) or having very low expression from a hypomorphic allele. C3G− / − homozyg- ous mice died before embryonic day 7.5, suggesting a significant role for C3G during mammalian development [53]. The lethality was rescued by expression of the human C3G transgene. Em- bryonic fibroblasts from C3G knockout mouse embryos showed impaired cell adhesion, delayed cell spreading and accelerated cell migration. These effects were suppressed by expression of active Rap1, Rap2 or R-Ras. This suggested the requirement of C3G-dependent activation of GTPase targets for adhesion and spreading of embryonic fibroblasts and for early embryogenesis [53]. The fact that other Rap GEFs do not compensate for em- bryonic lethality indicated that spatial and temporal functions of C3G other than Rap1 activation may be required during em- bryonic development. To help study the role of C3G in other tissues and at later devel- opmental stages, a mouse strain carrying a hypomorphic C3G al- lele, C3Ggt , was developed. Lysates of primary embryonic fibro- blasts from C3Ggt/gt mice showed less than 5% protein seen in cells from wild-type animals, but they survived up to embryonic day 14.5 [54]. C3Ggt/gt mutant embryos die due to a blood ves- sel maturation defect caused by inappropriate development of vascular supporting cells. C3G-deficient fibroblasts responded to PDGF-BB abnormally, exhibited cell adhesion defects and lacked paxillin and integrin-β1-positive cell adhesions. This study elu- cidated the requirement of C3G for vascular myogenesis, cell adhesion and response to PDGF, necessary for vascular myogen- esis [54]. C3Ggt/gt mice also showed over proliferation of the cortical neuroepithelium [55]. Neuroepithelial cells from these animals failed to exit the cell cycle in vivo. C3G mutant neural pre- cursor cells failed to activate Rap1, exhibited Akt/PKB activation, ................................................................... ............................................................ .................................................................. ............................................................. ........................................................ .................................................... 236 C The Authors Journal compilation C 2011 Biochemical Society
  • 7. C3G signals to actin remodelling Figure 6 Interacting partners of C3G and their involvement in pathways leading to specific functions These members interact with the proline-rich Crk-binding region of C3G through their SH3 domain, except for some members such as E-cadherin. A direct interaction has been characterized only in case of some members like Crk, Cas, Hck and Abl. IL3, interleukin 3. Gsk3β inhibition and β-catenin accumulation, when exposed to growth factors, in vitro. These findings indicated that the size of the cortical neural precursor population is controlled by C3G- mediated inhibition of the Ras signalling pathway [55]. Mutant embryos also exhibited a cortical neuron migration defect leading to a failure of preplate splitting into marginal zone and subplate and a failure to form a cortical plate. The basement membrane was disrupted and radial glial processes were disorganized indic- ating the requirement of C3G in neuronal migration and radial glial attachment during cerebral cortex development [56]. A role for C3G in the development of invertebrates is also known. During Drosophila eye and wing development, overex- pression of membrane targeted full-length C3G phenotypically mimics activation of the Ras-MAPK pathway, suggesting that DC3G (Drosophila C3G) is involved in MAPK activation in vivo [57]. The effects of C3G overactivity can be suppressed by re- ducing the gene dose of components of the Ras-MAPK pathway and of Rap1. DC3G is likely to stimulate both Ras1 and Rap1 dir- ectly, which in turn leads to a convergent activation of the MAPK pathway [57]. Deletion of C3G caused semi-lethality [58]. It is an accessory component of the Drosophila musculature, essen- tial for the proper localization of integrins at muscle–muscle and muscle–epidermis attachment sites and important for maintain- ing muscle integrity during larval stages. Cellular functions Various cellular functions regulated by C3G are mediated either through changes in gene expression or through signalling to actin cytoskeletal reorganization. Expression of constitutively active C3G, or knocking down endogenous C3G have been used to understand these functions. Changes in gene expression have been seen under conditions of C3G overexpression as well as repression [25]. Actin remodelling Initial evidence that C3G is involved in signalling pathways lead- ing to actin rearrangement came from studies which showed that C3G expression resulted in filopodia formation in epithelial cell lines dependent on an intact actin cytoskeleton [8]. C3G was also required for c-Abl-induced filopodia formation. It was shown that C3G could signal to actin by engaging N-Wasp, but independent of Cdc42, a Rho family GTPase whose activation has generally been associated with filopodia formation. C3G expressing cells showed loss of stress fibres suggesting that C3G can alter actin dynamics in these cells. In response to PV treatment, which is known to cause filopodia formation [59], pC3G localized to the subcortical actin cytoskeleton and to the tips of filopodia (Fig- ure 5). The unique morphology of neuronal cells is achieved and ...................................................................... .......................................................... .................................................................... ........................................................... ........................................................... ................................................ www.bioscirep.org / Volume 31 (4) / Pages 231–244 237
  • 8. V. Radha and others maintained through extensive changes in microfilaments and mi- crotubules. Neurite extension is also dependent on filopodia at the growth cone. C3G expression in human NB cells resulted in their morphological differentiation to neurons and Cdc42 and N-Wasp-dependent signalling was involved [23]. The ability of C3G to suppress transformation was dependent on its localiza- tion at the subcortical actin cytoskeleton and its association with protein phosphatase 2A [45]. It was indicated that C3G could also directly interact with actin in a yeast two-hybrid assay. In c-Abl-induced cell death, C3G was phosphorylated select- ively in actin-rich cellular domains dependent on F-actin-binding domain of c-Abl [30]. Localized phosphorylation of C3G re- quired intact actin cytoskeleton, but was not affected by micro- tubule disruption. Phosphorylation of C3G enhanced its ability to associate with cytoskeletal structures. Previously it was shown that C3G phosphorylation on tyrosine in response to adhesion of NIH 3T3 cells was dependent on an intact cytoskeleton [43]. In T-cells, it was seen that the actin remodelling protein WAVE-2 was required for C3G phosphorylation on Y504 [29]. In re- sponse to mechanical signals such as cytoskeletal stretch, C3G was found associated with Triton-insoluble structures to locally activate Rap1 [60]. In v-Abl-transformed cells, cytoskeletal re- arrangement is dependent on the CrkL–C3G complex, Rap1 and Rac1 [61]. A link between the actin-regulating protein VASP and C3G has been shown in human polymorphonuclear neutrophils, with VASP serving to regulate C3G activation [42]. Vesicle traffic which is dependent on actin dynamics is also regulated by C3G, through its target, TC10. Insulin-stimulated GLUT4 (glucose transporter type 4) translocation is dependent on C3G and an intact actin cytoskeleton [17]. TC10 binds COP1 in the Golgi and aids actin polymerization on membrane trans- port vesicles [62]. Vesicular trafficking of E-cadherin is regulated by C3G during the formation and breakdown of adherens junc- tions. Interaction between E-cadherin and C3G is induced on cell junction disassembly and activation of Rap1 and Rab11 positive recycling endosomes [63]. In Drosophila, C3G could rescue the NSF2 (N-ethylmaleimide-sensitive factor 2) phenotype which shows defects in vesicular trafficking [64]. Targets of C3G also function in actin regulation. Rap1 func- tions to regulate actin remodelling by engaging diverse effectors [65]. C3G-Rap1-dependent Rac and Cdc42 activation through their GEFs, Vav2 and FRG respectively are seen in response to nectin engagement [66]. C3G-induced morphological changes associated with neurons are achieved through Cdc42-mediated signalling to actin [23]. TC10 activity regulates F-actin dynam- ics and neurite growth [62,67,68]. Membrane protrusion is caused by interaction between Exo70 and TC10 [69]. R-Ras regulates cell migration of melanoma cells through association with the actin-binding scaffold protein Filamin A [70]. R-Ras signals to cause membrane protrusions through PLC (phospholipase C) activity [71]. R-Ras also engages Rho and Rac GTPases to cause morphological changes in epithelial and myeloid cells [72,73]. RgL3, a Ral GDS (guanine nucleotide dissociation stimulator)- related protein serves to mediate interaction between Rap family members and profilin, an important activator of actin polymeriz- ation [74]. Rap2 engages TNIK (TRAF2/Nck-interacting kinase) to cause changes in the cytoskeleton of cultured mammalian cells [75]. Rap activation is required for phorbol-ester-induced actin polymerization and morphological changes in B-cells [65]. Rap1 localizes to cell junctions and is a key regulator of junction form- ation and disruption [76]. Evidence that C3G signals to actin is also strengthened by the fact that most of the molecules that in- teract with C3G such as Crk, Hck, Src, c-Abl etc. are known to have roles in actin remodelling. Therefore reciprocal regulation seems to exist between actin dynamics and C3G. On one hand, polymerized actin serves as a platform for C3G activation and on the other hand, activated C3G leads to target activation to achieve changes in actin dynamics. These changes in turn are responsible for a multitude of cellular functions as described below. Adhesion and migration C3G, being a regulator of Rap GTPase, plays an important role in integrin signalling, adhesion and migration. C3G is phos- phorylated in response to adhesion to fibronectin and overex- pression in Ba/F3 haematopoietic cells enhances migration [43]. Expression of membrane-targeted C3G in HeLa cells also induces extensive cell spreading [77]. Overexpression of C3G in 32D cells increased adhesion to fibronectin through the activation of VLA-4 and VLA-5, mediated by R-Ras [78]. Overexpression of C3G increases adhesion of NIH 3T3 cells to laminin [79]. C3G localizes to the focal adhesions in v-Crk transformed cells causing abnormal activation of MAPK and JNK [80]. In TIMP2 (tissue inhibitor of metalloproteinases 2)-treated human microvascular endothelial cells, C3G induced RECK expression and reduced cell migration [81]. SFK-dependent regulation of cell adhesion also engages C3G. Cell proliferation Constitutive activation of C3G by expression of a membrane- targeted variant in Drosophila resulted in enhanced Ras-MAPK signalling and overproliferation and cell fate changes [57]. In haematopoietic progenitor cells, expression of membrane- targeted C3G resulted in expression of double-positive T-cells, associated with lethal T-cell acute lymphoblastic leukaemia. This is achieved through enhanced expression of Notch 1 and 3 and its target genes like Hes1 and c-Myc [82]. SIHA cells expressing siRNA (small interfering RNA) targeting C3G showed enhanced proliferation [25]. In NB cells, in addition to causing morpholo- gical changes of differentiation, C3G induced p21, an inhibitor of cell proliferation [23]. This is also reflected in vivo in a mouse model where C3G neuroepithelial cells are retained in the cell cycle without arresting and differentiating [55]. Differentiation C3G is induced during neuronal differentiation and regulates survival and differentiation of human NB cells [23]. Human NB cells, IMR-32 induced to differentiate by serum starvation or by treatment with NGF or forskolin showed enhanced C3G pro- tein levels. Transient overexpression of C3G stimulated neurite ................................................................... ............................................................ .................................................................. ............................................................. ........................................................ .................................................... 238 C The Authors Journal compilation C 2011 Biochemical Society
  • 9. C3G signals to actin remodelling growth and also increased responsiveness to NGF and serum deprivation induced differentiation. Forskolin and NGF treat- ment resulted in phosphorylation of C3G at Tyr504 predom- inantly in the Golgi. The activation of the C3G/Rap1 pathway results in neurite outgrowth of mouse pheochromocytoma cells, PC12, which is inhibited by either overexpression of Rap1GAP or siRNA-mediated knockdown of Rap1 or the GEF C3G [83]. Dephosphorylation of Crk and association with C3G was re- quired for adipocyte differentiation [84]. C3G protein levels also increased during differentiation of monocytes to macrophage lineage (Figure 4). The phenotype shown by mice expressing a hyphomorphic allele was also indicative of a requirement of C3G for differentiation of a variety of cells [55]. Transformation C3G expression increases the growth rate, anchorage- independent growth and JNK activation in v-Crk transformed NIH 3T3 cells. The catalytic domain of C3G is essential for this activity. Rap1 does not act as a C3G substrate in this con- text. Dominant-negative C3G can reverse the transformed phen- otype suggesting that C3G is essential for v-Crk-induced trans- formation of NIH 3T3 cells [38]. C3G-dependent Rap1 activ- ation also contributes to RET/PTC (rearranged during trans- fection/papillary thyroid carcinomas) oncogene-mediated trans- formation of thyroid follicular cells [85]. C3G, like Rap1, is capable of down-regulating the trans- forming ability of Ras and Sis oncogenes [86]. However, the transformation suppression activity of C3G is higher than that of Rap1A. Through its ability to activate Rap1, C3G has been shown to counteract signalling through the Ras/MAPK pathway and has also been shown to transmit signals through the stress kinase JNK pathway [15]. Moreover, C3G can also inhibit v-Raf- and dbl-induced transformation of NIH 3T3 cells. The catalytic domain of C3G is not required for this transformation suppression activity, rather the proline-rich motifs of C3G are essential and sufficient for this. C3G inhibits Ras-induced ERK activation, cyc- lin A expression and anchorage-independent growth [79]. Farne- sylated C3G, which localizes to the membrane, causes signific- antly higher morphological reversion of transformed phenotype of v-ki-Ras-transformed NIH 3T3 cells than normal C3G. Apoptosis and cell survival Co-expression of Hck with C3G induced a high level of apoptosis in many cell lines and this property was not dependent on Y-504 phosphorylation or the catalytic domain of C3G but required the catalytic activity of Hck. This indicated that C3G co-expression could alter Hck activity towards select targets leading to apoptosis [7]. c-Abl expression-induced cell death was dependent on C3G and its phosphorylation in distinct actin-rich retracting lamellipo- dia was associated with apoptosis. Oxidative-stress-induced cell death mediated through c-Abl activation was dependent on C3G phosphorylation [30]. By negatively regulating p38α MAPK, C3G plays a dual role in regulating cell death in MEFs (mouse embryonic fibroblasts) depending on the stimulus. C3G mediates cell death in response to oxidative stress, whereas it induces cell survival upon serum starvation. On serum deprivation, C3G induces survival through inhibition of p38α MAPK activity, which mediates apoptosis; whereas, in response to oxidative stress, C3G behaves as a proapoptotic molecule, as its knockdown or knockout enhances survival through upregulation of p38α activity, which plays an antiapoptotic role under these conditions [87]. C3G acts to signal to apoptosis and cell survival in response to the c-Abl inhibitor, ST1-571 [88]. Differentiation of NB cells involves activation of survival pathways along with induction of cell cycle arrest. C3G is required for cell survival during differentiation as its knock- down caused enhanced cell death in response to serum starvation [23]. Filopodia formation and cell junction integrity Work from our laboratory has shown that C3G plays a role in cyto- skeletal reorganization and filopodia formation [8]. Knockdown of C3G inhibited c-Abl-induced filopodia during cell spreading on fibronectin. C3G expression induces actin cytoskeletal reor- ganization and promotes filopodia formation independent of its catalytic activity. It showed enrichment at filopodia tips charac- teristic of molecules involved in filopodial dynamics (Figure 5). AJs (adherens junctions) responsible for the integrity of epi- thelial monolayers are formed by linking actin networks of neigh- bouring cells. C3G directly interacts with E-cadherin, a primary component of epithelial AJs, and excludes binding of β-catenin to E-cadherin [9]. C3G’s function has therefore been implicated in recruitment of E-cadherin to the junctions. E-cadherin-rich filopodia extensions function as adhesion zippers to interlock neighbouring cells before mature junction formation. E-cadherin internalization on junctional breakdown also depends on C3G binding to intracellular E-cadherin to activate Rap1 [89]. Nectins (Ig-like transmembrane molecules) which aid in AJ formation also signal by recruiting C3G to activate Rap1 [90]. Association with human disease In malignant transformation associated with human cancers, changes in C3G expression is tissue-specific. C3G overex- pression was found in several samples of primary NSCLCs (non-small-cell lung cancers) compared with corresponding non-cancerous tissues. Six of seven NSC cell lines also showed higher levels of C3G [91]. In contrast, cervical squamous cell carcinomas were associated with decreased C3G levels. This was attributed to frequent hypermethylation of upstream regu- latory gene sequence [25]. Gene expression profiling of chronic lymphocytic leukaemia samples showed downregulation of C3G during disease progression [92]. Expression of an alternately spliced form of C3G, p87, lacking N-terminal 305 residues in CML cell lines and Ph+ [Philadelphia chromosomal transloca- tion t(9;22)(q34;q11) positive] patients has been suggested to play a role in the pathogenesis of CML [21,93]. Single-nucleotide polymorphisms in the C3G gene have shown association with T2D (Type 2 diabetes), but the molecular basis ...................................................................... .......................................................... .................................................................... ........................................................... ........................................................... ................................................ www.bioscirep.org / Volume 31 (4) / Pages 231–244 239
  • 10. V. Radha and others is not clear. In a Finnish population, SNP rs4740283, located 4kb downstream of the C3G gene showed positive association with T2D [94]. The GG phenotype of the polymorphism at rs11243444, located in intron 13, had a protective effect on the development of T2D in a Korean population [95]. In an experi- mental model of glomerular nephritis, C3G and R-Ras-dependent signalling has been implicated [96]. Disease-associated deletions are known in the 9q34.3 chromosomal location that harbours the C3G gene [97]. It is to be determined whether lack of C3G con- tributes to the disease phenotype. It has also been predicted that C3G deregulation may be associated with human disorders show- ing defective leucocyte adhesion to the endothelium [42]. Mice lacking C3G show cortical neuronal migration defects resulting in failure to split preplate into marginal zone and subplate [56]. In humans, defective neuronal migration during development leads to disorders like lissencephaly. It would therefore be in- teresting to check for defects in C3G in lissencephaly patients. CONCLUSIONS AND PERSPECTIVES Multiple lines of evidence exist to show that many of the cellular functions regulated by C3G involve reorganization of the actin cytoskeleton. Through its ability to signal to actin reorganization, C3G is involved in regulation of both structural and functional processes in the cell. Morphogenesis is primarily dependent on adhesive and migratory behaviour of cells and these functions of C3G may be essential during embryonic development. The fact that C3G is engaged in response to diverse signals indicates its role in multiple tissue types and also explains the early embryonic lethality due to defective development of multiple organ systems. Requirement of C3G for mammalian development leads us to ask whether C3G mutations could be associated with human developmental defects. Examining aborted foetuses for mutations or expression changes in C3G may help in determining whether it plays a role in human embryonic development. C3G being a member of a family constituting a large number of proteins, it was surprising to note that other Rap GEFs do not compensate C3G function under several situations. Action of C3G in a spatial and temporal manner appears to be essential during embryonic development, which may be one of the reasons as to why its function is not complemented by other GEFs. There is need to understand much more about the regula- tion of C3G both in terms of its expression as well as activation. Isoform-specific functions of C3G need to be elucidated. Iden- tification of the C3G promoter and the regulatory transcription factors and their response elements is warranted. There is good reason to think that transcription factors that regulate differenti- ation and migratory behaviour of cells may regulate C3G expres- sion. Other modifications of C3G in addition to phosphorylation on Y504 need to be investigated and studied. C3G has multiple proline tracts but it is not clear as to whether it can interact directly with more than one protein to form a multimolecular complex and serve as a scaffold. One question that has not been addressed is whether there is mutual exclusion of interacting partners enabling the activation of only a subset of downstream effector pathways. It is also possible that two or more protein binding motifs in C3G function in a co-operative manner. Suppression of transformation is an important role played by C3G, which is a property independent of its catalytic activity. C3G expression resulted in upregulation of the cell cycle inhibitor p21 and suppression of cyclin A expression. Understanding how C3G signals to changes in expression of genes regulating the cell cycle will be important to understand its role as a tumour suppressor. In some cell types, C3G also functions to enhance cell proliferation and therefore its role in enhancing or suppressing proliferation is context-dependent. At present, it is not clear as to how C3G activates specific GTPases belonging to either Ras or Rho family in a stimulus-dependent manner. Further studies need to be carried out to determine whether C3G can regulate the activity of other GTPases directly or indirectly. On the basis of existing evidence, we propose that C3G may be a master regulator of the differentiated phenotype in multiple tissues. Differentiation removes cells from the proliferative mode without affecting their integrity. Differentiation pathways are rel- evant for tumour suppression in the light of continuous tissue regeneration and therefore understanding them in various tissue types has been important. In cells that have defects in apoptotic pathways, inducing irreversible arrest through differentiation is a good alternative in cancer therapy. The function of C3G as a reg- ulator of differentiation in multiple tissues may be an important property that could be utilized for achieving tumour suppression. The 3D (three-dimensional) structure of C3G (either of the whole molecule or its subdomains) has not been elucidated. Ana- lysis of the 3D structure of C3G will help in understanding its properties better. A 3D homology model constructed by using SWISS-MODEL software indicated considerable structural ho- mology between the catalytic sequence of C3G and the GEF do- main (Cdc25 homology domain) of Sos, a Ras family GEF whose crystal structure has been studied [107]. The GEFs interact with their respective GTPases by using the same overall interface but different specific interactions provide target specificity [108]. Targeting GEFs for either activation or inhibition for therapy has been shown to be possible in principle [1]. Small-molecule inhibitors have been developed for some GEFs and selective ag- onists used for activation in other instances. C3G being a ubiquit- ously expressed molecule with a role in pathways triggered by a variety of signals, any attempt at therapeutic intervention must aim at achieving selectivity in specific cell types. Some sugges- ted approaches for activation of C3G are: (1) enabling membrane targeting; (2) inhibition of tyrosine phosphatases or activation of kinases that specifically regulate C3G; (3) introduction of pep- tides that bind negative regulatory sequences; and (4) treatment with agents that cause increase in C3G levels in specific cell types. Just as in the case of Rho GEFs, C3G activity can be inhibited by finding small molecule inhibitors that target its GEF domain. Since C3G has functions dependent on catalytic activity as well as protein interaction leading to different cellular functions, it should be possible to target specific pathways selectively. Other major questions that remain to be answered are ‘how are developmental processes co-ordinated by C3G at the molecular ................................................................... ............................................................ .................................................................. ............................................................. ........................................................ .................................................... 240 C The Authors Journal compilation C 2011 Biochemical Society
  • 11. C3G signals to actin remodelling level?’ and ‘how does C3G regulate actin dynamics?’ Although one straight answer would be that these functions are carried out through activation of GTPases, there appears to be more com- plexity. Association of C3G directly with actin indicates multiple mechanisms that could be involved. Although we have high- lighted a role for C3G in regulating actin dynamics, it is possible that C3G signals to cytoskeletal changes by also affecting mi- crotubule dynamics. A detailed knowledge of the regulation and function of C3G at the cellular and molecular level will hopefully provide us with means to selectively target it in specific tissues where its deregulation is associated with pathology. ACKNOWLEDGEMENTS We thank Dr Ghanshyam Swarup for a critical reading of the manu- script prior to submission. REFERENCES 1 Bos, J. L., Rehmann, H. and Wittinghofer, A. (2007) GEFs and GAPs: critical elements in the control of small G proteins. Cell 129, 865–877 2 Raaijmakers, J. H. and Bos, J. L. (2009) Specificity in Ras and Rap signaling. J. Biol. Chem. 284, 10995–10999 3 Quilliam, L. A., Rebhun, J. F. and Castro, A. F. (2002) A growing family of guanine nucleotide exchange factors is responsible for activation of Ras-family GTPases. Prog. Nucleic Acid Res. Mol. Biol. 71, 391–444 4 Tanaka, S., Morishita, T., Hashimoto, Y., Hattori, S., Nakamura, S., Shibuya, M., Matuoka, K., Takenawa, T., Kurata, T., Nagashima, K. et al. (1994) C3G, a guanine nucleotide-releasing protein expressed ubiquitously, binds to the Src homology 3 domains of CRK and GRB2/ASH proteins. Proc. Natl. Acad. Sci. U.S.A. 91, 3443–3447 5 Knudsen, B. S., Feller, S. M. and Hanafusa, H. (1994) Four proline-rich sequences of the guanine-nucleotide exchange factor C3G bind with unique specificity to the first Src homology 3 domain of Crk. J. Biol. Chem. 269, 32781–32787 6 Kirsch, K. H., Georgescu, M. M. and Hanafusa, H. (1998) Direct binding of p130(Cas) to the guanine nucleotide exchange factor C3G. J. Biol. Chem. 273, 25673–25679 7 Shivakrupa, R., Radha, V., Sudhakar, C. and Swarup, G. (2003) Physical and functional interaction between Hck tyrosine kinase and guanine nucleotide exchange factor C3G results in apoptosis, which is independent of C3G catalytic domain. J. Biol. Chem. 278, 52188–52194 8 Radha, V., Rajanna, A., Mitra, A., Rangaraj, N. and Swarup, G. (2007) C3G is required for c-Abl-induced filopodia and its overexpression promotes filopodia formation. Exp. Cell Res. 313, 2476–2492 9 Hogan, C., Serpente, N., Cogram, P., Hosking, C. R., Bialucha, C. U., Feller, S. M., Braga, V. M., Birchmeier, W. and Fujita, Y. (2004) Rap1 regulates the formation of E-cadherin-based cell-cell contacts. Mol. Cell. Biol. 24, 6690–6700 10 Takai, S, Tanaka, M, Sugimura, H, Yamada, K, Naito, Y, Kino, I and Matsuda, M. (1994) Mapping of the human C3G gene coding a guanine nucleotide releasing protein for Ras family to 9q34.3 by fluorescence in situ hybridization. Hum. Genet. 94, 549–550 11 Gotoh, T., Hattori, S., Nakamura, S., Kitayama, H., Noda, M., Takai, Y., Kaibuchi, K., Matsui, H., Hatase, O., Takahashi, H. et al. (1995) Identification of Rap1 as a target for the Crk SH3 domain-binding guanine nucleotide-releasing factor C3G. Mol. Cell. Biol. 15, 6746–6753 12 Gotoh, T., Niino, Y., Tokuda, M., Hatase, O., Nakamura, S., Matsuda, M. and Hattori, S. (1997) Activation of R-Ras by Ras-guanine nucleotide-releasing factor. J. Biol. Chem. 272, 18602–18607 13 van den Berghe, N., Cool, R. H., Horn, G. and Wittinghofer, A. (1997) Biochemical characterization of C3G: an exchange factor that discriminates between Rap1 and Rap2 and is not inhibited by Rap1A(S17N). Oncogene 15, 845–850 14 York, R. D., Yao, H., Dillon, T., Ellig, C. L., Eckert, S. P., McCleskey, E. W. and Stork, P. J. (1998) Rap1 mediates sustained MAPK activation induced by NGF. Nature 392, 622–626 15 Mochizuki, N., Ohba, Y., Kobayashi, S., Otsuka, N., Graybiel, A. M., Tanaka, S. and Matsuda, M. (2000) Crk activation of JNK via C3G and R-Ras. J. Biol. Chem. 275, 12667–12671 16 Ohba, Y., Mochizuki, N., Yamashita, S., Chan, A. M., Schrader, J. W., Hattori, S., Nagashima, K. and Matsuda, M. (2000) Regulatory proteins of R-Ras, TC21/R-Ras2, and M-Ras/R-Ras3. J. Biol. Chem. 275, 20020–20026 17 Chiang, S. H., Baumann, C. A., Kanzaki, M., Thurmond, D. C., Watson, R. T., Neudauer, C. L., Macara, I. G., Pessin, J. E. and Saltiel, A. R. (2001) Insulin-stimulated GLUT4 translocation requires the CAP-dependent activation of TC10. Nature 410, 944–948 18 Ling, L., Zhu, T. and Lobie, P. E. (2003) Src-CrkII-C3G-dependent activation of Rap1 switches growth hormone-stimulated p44/42 MAP kinase and JNK/SAPK activities. J. Biol. Chem. 278, 27301–27311 19 Wang, Z., Dillon, T. J., Pokala, V., Mishra, S., Labudda, K., Hunter, B. and Stork, P. J. (2006) Rap1-mediated activation of extracellular signal-regulated kinases by cyclic AMP is dependent on the mode of Rap1 activation. Mol. Cell. Biol. 26, 2130–2145 20 Shivakrupa, R., Singh, R. and Swarup, G. (1999) Identification of a novel splice variant of C3G which shows tissue-specific expressionDNA Cell Biol. 18, 701–708 21 Gutierrez-Berzal, J., Castellano, E., Martin-Encabo, S., Gutierrez-Cianca, N., Hernandez, J. M., Santos, E. and Guerrero, C. (2006) Characterization of p87C3G, a novel, truncated C3G isoform that is overexpressed in chronic myeloid leukemia and interacts with Bcr-Abl. Exp. Cell Res. 312, 938–948 22 Zhai, B., Huo, H. and Liao, K. (2001) C3G, a guanine nucleotide exchange factor bound to adapter molecule c-Crk, has two alternative splicing forms. Biochem. Biophys. Res. Commun. 286, 61–66 23 Radha, V., Rajanna, A., Gupta, R. K., Dayma, K. and Raman, T. (2008) The guanine nucleotide exchange factor, C3G regulates differentiation and survival of human neuroblastoma cells. J. Neurochem. 107, 1424–1435 24 Prince, L. S., Karp, P. H., Moninger, T. O. and Welsh, M. J. (2001) KGF alters gene expression in human airway epithelia: potential regulation of the inflammatory response. Physiol. Genom. 6, 81–89 25 Okino, K., Nagai, H., Nakayama, H., Doi, D., Yoneyama, K., Konishi, H. and Takeshita, T. (2006) Inactivation of Crk SH3 domain-binding guanine nucleotide-releasing factor (C3G) in cervical squamous cell carcinoma. Int. J. Gynecol. Cancer 16, 763–771 26 Wang, H., Linghu, H., Wang, J., Che, Y. L., Xiang, T. X., Tang, X. and Yao, Z. W. (2010) The role of Crk/Dock180/Rac1 pathway in the malignant behavior of human ovarian cancer cell SKOV3. Tumour Biol. 31, 59–67 27 Ichiba, T., Hashimoto, Y., Nakaya, M., Kuraishi, Y., Tanaka, S., Kurata, T., Mochizuki, N. and Matsuda, M. (1999) Activation of C3G guanine nucleotide exchange factor for Rap1 by phospho- rylation of tyrosine 504. J. Biol. Chem. 274, 14376–14381 28 Radha, V., Rajanna, A. and Swarup, G. (2004) Phosphorylated guanine nucleotide exchange factor C3G, induced by pervanadate and Src family kinases localizes to the Golgi and subcortical actin cytoskeleton. BMC Cell Biol. 5, 31 29 Nolz, J. C., Nacusi, L. P., Segovis, C. M., Medeiros, R. B., Mitchell, J. S., Shimizu, Y. and Billadeau, D. D. (2008) The WAVE2 complex regulates T cell receptor signaling to integrins via ...................................................................... .......................................................... .................................................................... ........................................................... ........................................................... ................................................ www.bioscirep.org / Volume 31 (4) / Pages 231–244 241
  • 12. V. Radha and others Abl- and CrkL-C3G-mediated activation of Rap1. J. Cell Biol. 182, 1231–1244 30 Mitra, A. and Radha, V. (2010) F-actin-binding domain of c-Abl regulates localized phosphorylation of C3G: role of C3G in c-Abl-mediated cell death. Oncogene 29, 4528–4542 31 Yokote, K., Hellman, U., Ekman, S., Saito, Y., Ronnstrand, L., Heldin, C. H. and Mori, S. (1998) Identification of Tyr-762 in the platelet-derived growth factor alpha-receptor as the binding site for Crk proteins. Oncogene 16, 1229–1239 32 Larsson, H., Klint, P., Landgren, E. and Claesson-Welsh, L. (1999) Fibroblast growth factor receptor-mediated endothelial cell proliferation is dependent on the Src homology (SH) 2/SH3 domain containing adaptor protein. J. Biol. Chem. 274, 25726–25734 33 Uemura, N. and Griffin, J. D. (1999) The adapter protein Crkl links Cbl to C3G after integrin ligation and enhances cell migration. J. Biol. Chem. 274, 37525–37532 34 Sakkab, D., Lewitzky, M., Posern, G., Schaeper, U., Sachs, M., Birchmeier, W. and Feller, S. M. (2000) Signaling of hepatocyte growth factor/scatter factor (HGF) to the small GTPase Rap1 via the large docking protein Gab1 and the adapter protein CRKL. J. Biol. Chem. 275, 10772–10778 35 Ichiba, T., Kuraishi, Y., Sakai, O., Nagata, S., Groffen, J., Kurata, T., Hattori, S. and Matsuda, M. (1997) Enhancement of guanine-nucleotide exchange activity of C3G for Rap1 by the expression of Crk, CrkL, and Grb2. J. Biol. Chem. 272, 22215–22220 36 Okada, S., Matsuda, M., Anafi, M., Pawson, T. and Pessin, J. E. (1998) Insulin regulates dynamic balance between Ras and Rap1 signaling by coordinating assembly states of Grb2–SOS and CrkII–C3G complexes. EMBO J. 17, 2554–2565 37 Takino, T., Tamura, M., Miyamori, H., Araki, M., Matsumoto, K., Sato, H. and Yamada, K. M. (2003) Tyrosine phosphorylation of the CrkII adaptor protein modulates cell migration. J. Cell Sci. 116, 3145–3155 38 Tanaka, S., Ouchi, T. and Hanafusa, H. (1997) Downstream of Crk adaptor signaling pathway: activation of Jun kinase by v-Crk through the guanine nucleotide exchange protein C3G. Proc. Natl. Acad. Sci. U.S.A. 94, 2356–2361 39 Buensuceso, C. S. and O’Toole, T. E. (2000) The association of CRKII with C3G can be regulated by integrins and defines a novel means to regulate the mitogen-activated protein kinases. J. Biol. Chem. 275, 13118–13125 40 Zhang, W., Shao, Y., Fang, D., Huang, J., Jeon, M. S. and Liu, Y. C. (2003) Negative regulation of T-cell antigen receptor mediated Crk-L-C3G signalling and cell adhesion by cbl-b. J. Biol. Chem. 278, 23978–23983 41 Fecchi, K., Volonte, D., Hezel, M. P., Schmeck, K. and Galbiati, F. (2006) Spatial and temporal regulation of GLUT4 translocation by flotillin-1 and caveolin-3 in skeletal muscle cells. FASEB J. 20, 705–707 42 Deevi, R. K., Koney-Dash, M., Kissenpfennig, A., Johnston, J. A., Schuh, K., Walter, U. and Dib, K. Vasodilator-stimulated phosphoprotein regulates inside-out signaling of β2 integrins in neutrophils. J. Immunol. 184, 6575–6584 43 de Jong, R., van Wijk, A., Heisterkamp, N. and Groffen, J. (1998) C3G is tyrosine-phosphorylated after integrin-mediated cell adhesion in normal but not in Bcr/Abl expressing cells. Oncogene 17, 2805–2810 44 Cho, Y. J., Hemmeryckx, B., Groffen, J. and Heisterkamp, N. (2005) Interaction of Bcr/Abl with C3G, an exchange factor for the small GTPase Rap1, through the adapter protein Crkl. Biochem. Biophys. Res. Commun. 333, 1276–1283 45 Martin-Encabo, S., Santos, E. and Guerrero, C. (2007) C3G mediated suppression of malignant transformation involves activation of PP2A phosphatases at the subcortical actin cytoskeleton. Exp. Cell Res. 313, 3881–3891 46 Bivona, T. G., Wiener, H. H., Ahearn, I. M., Silletti, J., Chiu, V. K. and Philips, M. R. (2004) Rap1 up-regulation and activation on plasma membrane regulates T cell adhesion. J. Cell Biol. 164, 461–470 47 Okada, S. and Pessin, J. E. (1997) Insulin and epidermal growth factor stimulate a conformational change in Rap1 and dissociation of the CrkII–C3G complex. J. Biol. Chem. 272, 28179–28182 48 Kao, S., Jaiswal, R. K., Kolch, W. and Landreth, G. E. (2001) Identification of the mechanisms regulating differential activation of the MAPK cascade by EGF and NGF in PC12 cells. J. Biol. Chem. 276, 18169–18177 49 Li, L., Okura, M. and Imamoto, A. (2002) Focal adhesions require catalytic activity of Src family kinases to mediate integrin-matrix adhesion. Mol. Cell. Biol. 22, 1203–1217 50 Kyono, W. T., de Jong, R., Park, R. K., Liy, Y., Heisterkamp, N., Groffen, J. and Durden, D. L. (1998) Differential interaction of Crk1 with Cbl or C3G, Hef-1, and γ subunit immunoreceptor tyrosine-based activation motif in signaling of myeloid high affinity Fc receptor for IgG (Fcγ RI). J. Immunol. 161, 5555–5563 51 Uemura, N., Salgia, R., Li, J. L., Pisick, E., Sattler, M. and Griffin, J. D. (1997) The BCR/ABL oncogene alters interaction of the adapter proteins CRKL and CRK with cellular proteins. Leukemia 11, 376–385 52 Takahashi, M., Rikitake, Y., Nagamatsu, Y., Hara, T., Ikeda, W., Hirata, K. and Takai, Y. (2008) Sequential activation of Rap1 and Rac1 small G proteins by PDGF locally at leading edges of NIH3T3 cells. Genes Cells 13, 549–569 53 Ohba, Y., Ikuta, K., Ogura, A., Matsuda, J., Mochizuki, N., Nagashima, K., Kurokawa, K., Mayer, B. J., Maki, K., Miyazaki, J. et al. (2001) Requirement for C3G-dependent Rap1 activation for cell adhesion and embryogenesis. EMBO J. 20, 3333–3341 54 Voss, A. K., Gruss, P. and Thomas, T. (2003) The guanine nucleotide exchange factor C3G is necessary for the formation of focal adhesions and vascular maturation. Development 130, 355–367 55 Voss, A. K., Krebs, D. L. and Thomas, T. (2006) C3G regulates the size of the cerebral cortex neural precursor population. EMBO J. 25, 3652–3663 56 Voss, A. K., Britto, J. M., Dixon, M. P., Sheikh, B. N., Collin, C., Tan, S. S. and Thomas, T. (2008) C3G regulates cortical neuron migration, preplate splitting and radial glial cell attachment. Development 135, 2139–2149 57 Ishimaru, S., Williams, R., Clark, E., Hanafusa, H. and Gaul, U. (1999) Activation of the Drosophila C3G leads to cell fate changes and overproliferation during development, mediated by the RAS–MAPK pathway and RAP1. EMBO J. 18, 145–155 58 Shirinian, M., Grabbe, C., Popovic, M., Varshney, G., Hugosson, F., Bos, H., Rehmann, H. and Palmer, R. H. (2010) The Rap1 guanine nucleotide exchange factor C3G is required for preservation of larval muscle integrity in Drosophila melanogaster. PLoS One 5, e9403 59 Luber, B., Candidus, S., Handschuh, G., Mentele, Edith, Hutzler, P., Feller, S., Voss, J., Hofler, H. and Becke, K. F. (2000) Tumor-derived mutated E-cadherin influences β-catenin locali- zation and increases susceptibility to actin cytoskeletal changes induced by pervanadate. Cell Commun. Adhesion 7, 391–408 60 Tamada, M., Sheetz, M. P. and Sawada, Y. (2004) Activation of a signaling cascade by cytoskeleton stretch. Dev. Cell 7, 709–718 61 Lee, H., Gaughan, J. P. and Tsygankov, A. Y. (2008) c-Cbl facilitates cytoskeletal effects in v-Abl transformed fibroblast through Rac1 and Rap1-mediated signaling. Int. Biochem. Cell Biol. 40, 1930–1943 62 Kanzaki, M., Watson, R. T., Hou, J. C., Stamnes, M., Saltiel, A. R. and Pessin, J. E. (2002) Small GTP-binding protein TC10 differentially regulates two distinct populations of filamentous actin in 3T3L1 adipocytes. Mol. Biol. Cell 13, 2334–2346 63 Balzac, F., Avolio, M., Degani, S., Kaverina, I., Torti, M., Silengo, L., Small, J. V. and Retta, S. F. (2005) E-cadherin endocytosis regulates the activity of Rap1: a traffic light GTPase at the crossroads between cadherin and integrin function. J. Cell Sci. 118, 4765–4783 ................................................................... ............................................................ .................................................................. ............................................................. ........................................................ .................................................... 242 C The Authors Journal compilation C 2011 Biochemical Society
  • 13. C3G signals to actin remodelling 64 Laviolette, M. J., Nunes, P., Peyre, J. B., Aigaki, T. and Stewart, B. A. (2005) A genetic screen for suppressors of Drosophila NSF2 neuromuscular junction overgrowth. Genetics 170, 779–792 65 McLeod, S. J., Shum, A. J., Lee, R. L., Takei, F. and Gold, M. R. (2004) The Rap GTPases regulate integrin-mediated adhesion, cell spreading, actin polymerization, and Pyk2 tyrosine phospho- rylation in B lymphocytes. J. Biol. Chem. 279, 12009–12019 66 Fukuyama, T., Ogita, H., Kawakatsu, T., Fukuhara, T., Yamada, T., Sato, T., Shimizu, K., Nakamura, T., Matsuda, M. and Takai, Y. (2001) Involvement of c-Src-Crk-C3G-C3G-Rap1 signaling in the nectin induced activation of Cdc42 and formation of adherens junctions. J. Biol. Chem. 280, 815–825 67 Abe, T., Kato, M., Miki, H., Takenawa, T. and Endo, T. (2003) Small GTPase Tc10 and its homologue RhoT induce N-WASP-mediated long process formation and neurite outgrowth. J. Cell Sci. 116, 155–168 68 Ridley, A. J. (2006) Rho GTPases and actin dynamics in membrane protrusions and vesicle trafficking. Trends Cell Biol. 16, 522–529 69 Pommereit, D. and Wouters, F. S. (2007) An NGF-induced Exo70-TC10 complex locally antagonises Cdc42-mediated activation of N-WASP to modulate neurite outgrowth. J. Cell Sci. 120, 2694–2705 70 Gawecka, J. E., Griffiths, G. S., Ek-Rylander, B., Ramos, J. W. and Matter, M. L. (2010) R-Ras regulates migration through an interaction with filamin A in melanoma cells. PLoS One 5, e11269 71 Ada-Nguema, A. S., Xenias, H., Hofman, J. M., Wiggins, C. H., Sheetz, M. P. and Keely, P. J. (2006) The small GTPase R-Ras regulates organization of actin and drives membrane protrusions through the activity of PLCε. J. Cell Sci. 119, 1307–1319 72 Jeong, H. W., Nam, J. O. and Kim, I. S. (2005) The C-terminal end of R-Ras alters the motility and morphology of breast epithelial cells through Rho/Rho kinase. Can. Res. 65, 507–515 73 Holly, S. P., Barson, M. K. and Parise, L. V. (2005) The unique N-terminus of R-Ras is required for Rac activation and precise regulation of cell migration. Mol. Biol. Cell 16, 2458–2469 74 Xu, J., Shi, S., Matsumoto, N., Noda, M. and Kitayama, H. (2007) Identification of Rgl3 as a potential binding partner for Rap-family small G-proteins and profilin II. Cell Signalling 19, 1575–1582 75 Taira, K., Umikawa, M., Takei, K., Myagmar, B. E., Shinzato, M., Machida, N., Uezato, H., Nonaka, S. and Kariya, K. (2004) The Traf2- and Nck-interacting kinase as a putative effector of Rap2 to regulate actin cytoskeleton. J. Biol. Chem. 279, 49488–49496 76 Pannekoek, W. J., Kooistra, M. R., Zwartkruis, F. J. and Bos, J. L. (2009) Cell–cell junction formation: the role of Rap1 and Rap1 guanine nucleotide exchange factors. Biochim. Biophys. Acta 1788, 790–796 77 Tsukamoto, N., Hattori, M., Yang, H., Bos, J. L. and Minato, N. (1999) Rap1 GTPase-activating protein SPA-1 negatively regulates cell adhesion. J. Biol. Chem. 274, 18463–18469 78 Arai, A., Nosaka, Y., Kohsaka, H., Miyasaka, N. and Miura, O. (1999) CrkL activates integrin-mediated hematopoietic cell adhesion through the guanine nucleotide exchange factor C3G. Blood 93, 3713–3722 79 Guerrero, C., Martin-Encabo, S., Fernandez-Medarde, A. and Santos, E. (2004) C3G-mediated suppression of oncogene-induced focus formation in fibroblasts involves inhibition of ERK activation, cyclin A expression and alterations of anchorage-independent growth. Oncogene 23, 4885–4893 80 Nievers, M. G., Birge, R. B., Greulich, H., Verkleij, A. J., Hanafusa, H. and van Bergen en Henegouwen, P. M. (1997) v-Crk-induced cell transformation: changes in focal adhesion composition and signaling. J Cell Sci. 110, 389–399 81 Oh, J., Seo, D. W., Diaz, T., Wei, B., Ward, Y., Ray, J. M., Morioka, Y., Shi, S., Kitayama, H., Takahashi, C., Noda, M. and Stetler-Stevenson, W. G. (2004) Tissue inhibitors of metalloproteinase 2 inhibits endothelial cell migration through increased expression of RECK. Cancer Res. 64, 9062–9069 82 Wang, S. F., Aoki, M., Nakashima, Y., Shinozuka, Y., Tanaka, H., Taniwaki, M., Hattori, M. and Minato, N. (2008) Development of Notch-dependent T-cell leukemia by deregulated Rap1 signaling. Blood 111, 2878–2886 83 Schonherr, C., Yang, H. L., Vigny, M., Palmer, R. H. and Hallberg, B. (2010) Anaplastic lymphoma kinase activates the small GTPase Rap1 via the Rap1-specific GEF C3G in both neuroblastoma and PC12 cells. Oncogene 29, 2817–2830 84 Jin, S., Zhai, B., Qiu, Z., Wu, J., Lane, M. D. and Liao, K. (2000) c-Crk, a substrate of the insulin-like growth factor-1 receptor tyrosine kinase, functions as an early signal mediator in adipocyte differentiation process. J. Biol. Chem. 275, 34444–34452 85 De Falco, V., Castellone, M. D., De Vita, G., Cirafici, A. M., Hershman, J. M., Guerrero, C., Fusco, A., Melillo, R. M. and Santoro, M. (2007) RET/papillary thyroid carcinoma oncogenic signaling through the Rap1 small GTPase. Cancer Res. 67, 381–390 86 Guerrero, C., Fernandez-Medarde, A., Rojas, J. M., Font de Mora, J., Esteban, L. M. and Santos, E. (1998) Transformation suppressor activity of C3G is independent of its CDC25-homology domain. Oncogene 16, 613–624 87 Gutierrez-Uzquiza, A., Arechederra, M., Molina, I., Banos, R., Maia, V., Benito, M., Guerrero, C. and Porras, A. (2010) C3G down-regulates p38 MAPK activity in response to stress by Rap-1 independent mechanisms: involvement in cell death. Cell Signalling 22, 533–542 88 Maia, V., Sanz, M., Gutierrez-Berzal, J., de Luis, A., Gutierrez-Uzquiza, A., Porras, A. and Guerrero, C. (2009) C3G silencing enhances STI-571-induced apoptosis in CML cells through p38 MAPK activation, but it antagonizes STI-571 inhibitory effect on survival. Cell Signalling 21, 1229–1235 89 Asuri, S., Yan, J., Paranavitana, N. C. and Quilliam, L. A. (2008) E-cadherin disengagement activates Rap1 GTPase. J. Cell Biochem. 105, 1027–1037 90 Sato, T., Fujita, N., Yamada, A., Ooshio, T., Okamoto, R., Irie, K. and Takai, Y. (2006) Regulation of the assembly and adhesion activity of E-cadherin by nectin and afadin for the formation of adherens junctions in Madin-Darby canine kidney cells. J. Biol. Chem. 281, 5288–5299 91 Hirata, T., Nagai, H., Koizumi, K., Okino, K., Harada, A., Onda, M., Nagahata, T., Mikami, I., Hirai, K., Haraguchi, S. et al. (2004) Amplification, up-regulation and over-expression of C3G (CRK SH3 domain-binding guanine nucleotide-releasing factor) in non-small cell lung cancers. J. Hum. Genet. 49, 290–295 92 Fernandez, V., Jares, P., Salaverria, I., Gine, E., Bea, S., Aymerich, M., Colomer, D., Villamor, N., Bosch, F., Montserrat, E. et al. (2008) Gene expression profile and genomic changes in disease progression of early-stage chronic lymphocytic leukemia. Haematologica 93, 132–136 93 Virgili, A., Brazma, D., Reid, A. G., Howard-Reeves, J., Valga˜n´on, M., Chanalaris, A., De Melo, V. A., Marin, D., Apperley, J. F. and Grace, C. (2008) FISH mapping of Philadelphia negative BCR/ABL1 positive CML. Mol. Cytogenet. 1, 14 94 Gaulton, K. J., Willer, C. J., Li, Y., Scott, L. J., Conneely, K. N., Jackson, A. U., Duren, W. L., Chines, P. S., Narisu, N., Bonnycastle, L. L. et al. (2008) Comprehensive association study of type 2 diabetes and related quantitative traits with 222 candidate genes. Diabetes 57, 3136–3144 95 Hong, K. W., Jin, H. S., Lim, J. E., Ryu, H. J., Go, M. J., Lee, J. Y., Woo, J. T., Park, H. K. and Oh, B. (2009) RAPGEF1 gene variants associated with type 2 diabetes in the Korean population. Diabetes Res. Clin. Pract. 84, 117–122 96 Rufanova, V. A., Lianos, E., Alexanian, A., Sorokina, E., Sharma, M., McGinty, A. and Sorokin, A. (2009) C3G overexpression in glomerular epithelial cells during anti-GBM-induced glomerulonephritis. Kidney Int. 75, 31–40 97 Nowak, N. J., Sait, S. N., Zeidan, A., Deeb, G., Gaile, D., Liu, S., Ford, L., Wallace, P. K., Wang, E. S. and Wetzler, M. (2010) Recurrent deletion of 9q34 in adult normal karyotype precursor B-cell ALL. Can. Genet. Cytogenet. 199, 15–20 ...................................................................... .......................................................... .................................................................... ........................................................... ........................................................... ................................................ www.bioscirep.org / Volume 31 (4) / Pages 231–244 243
  • 14. V. Radha and others 98 Arai, A., Nosaka, Y., Kanda, E., Yamamoto, K., Miyasaka, N. and Miura, O. (2001) Rap1 is activated by erythropoietin or interleukin-3 and is involved in regulation of β1 integrin-mediated hematopoietic cell adhesion. J. Biol. Chem. 276, 10453–10462 99 Reedquist, K. A., Fukazawa, T., Panchamoorthy, G., Langdon, W. Y., Shoelson, S. E., Druker, B. J. and Band, H. (1996) Stimulation through the T-cell receptor induces Cbl association with Crk proteins and the guanine nucleotide exchange protein C3G. J. Biol. Chem. 271, 8435–8442 100 Smit, L., van der Horst, G. and Borst, J. (1996) Sos, Vav, and C3G participate in B-cell receptor-inducing signaling pathways and differentially associate with Shc-Grb2, Crk, and Crk-L adaptors. J. Biol. Chem. 271, 8564–8569 101 Alsayed, Y., Uddin, S., Ahmad, S., Majchrzak, B., Druker, B. J., Fish, E. N. and Platimas, L. C. (2000) IFN-γ activates C3G/Rap1 signaling pathway. J. Immunol. 164, 1800–1806 102 Nasaka, Y., Arai, A., Miyasaka, N. and Miura, O. (1999) CrkL mediates Ras-dependent activation of the Raf/ERK pathway through the GEF, C3G in hematopoietic cells stimulated with EPO and IL-3. J. Biol. Chem. 274, 30154–30162 103 Du, J., AlSayed, Y. M., Xin, F., Ackerman, S. J. and Platanias, L. C. (2000) Engagement of the CrkL adapter in IL-5 signaling in eosinophils. J. Biol. Chem. 275, 33167–33175 104 Ballif, B. A., Arnaud, L., Arthur, W. T., Guris, D., Imamoto, A. and Cooper, J. A. (2004) Activation of a Dab1/CrkL/C3G/Rap1 pathway in Reelin-stimulated neurons. Curr. Biol. 14, 606–610 105 Fukuyama, T., Ogita, H., Kawakatsu, T., Inagaki, M. and Takai, Y. (2006) Activation of Rac by cadherin through the c-Src-Rap1- phosphatidylinositol 3-kinase-Vav2 pathway. Oncogene 25, 8–19 106 Posern, G., Rapp, U. R. and Feller, S. M. (2000) The Crk signaling pathway contributes to the bombesin-induced activation of the small GTPase Rap1 in Swiss 3T3 cells. Oncogene 19, 6361–6368 107 Boriack-Sjodin, P. A., Margarit, S. M., Bar-Sagi, D. and Kuriyan, J. (1998) The structural basis of the activation of Ras by Sos. Nature 394, 337–43 108 van den Berghe, N., Cool, R. H. and Wittinghofer, A. (1999) Discriminatory residues in Ras and Rap for guanine nucleotide exchange factor recognition. J. Biol. Chem. 274, 11078–11085 Received 17 August 2010/11 October 2010; accepted 13 October 2010 Published on the Internet 2 March 2011, doi 10.1042/BSR20100094 ................................................................... ............................................................ .................................................................. ............................................................. ........................................................ .................................................... 244 C The Authors Journal compilation C 2011 Biochemical Society