ArticlePDF Available

Diacylglycerol kinase mediates HGF-induced Rac activation and membrane ruffling by regulating atypical PKC and RhoGDI

Authors:

Abstract and Figures

Diacylglycerol kinases (DGKs) convert diacylglycerol (DAG) into phosphatidic acid (PA), acting as molecular switches between DAG- and PA-mediated signaling. We previously showed that Src-dependent activation and plasma membrane recruitment of DGKα are required for growth-factor-induced cell migration and ruffling, through the control of Rac small-GTPase activation and plasma membrane localization. Herein we unveil a signaling pathway through which DGKα coordinates the localization of Rac. We show that upon hepatocyte growth-factor stimulation, DGKα, by producing PA, provides a key signal to recruit atypical PKCζ/ι (aPKCζ/ι) in complex with RhoGDI and Rac at ruffling sites of colony-growing epithelial cells. Then, DGKα-dependent activation of aPKCζ/ι mediates the release of Rac from the inhibitory complex with RhoGDI, allowing its activation and leading to formation of membrane ruffles, which constitute essential requirements for cell migration. These findings highlight DGKα as the central element of a lipid signaling pathway linking tyrosine kinase growth-factor receptors to regulation of aPKCs and RhoGDI, and providing a positional signal regulating Rac association to the plasma membrane.
Content may be subject to copyright.
Diacylglycerol kinase αmediates HGF-induced Rac
activation and membrane rufing by regulating
atypical PKC and RhoGDI
Federica Chianale
a,1
, Elena Rainero
a,1
, Cristina Cianone
a
, Valentina Bettio
a
, Andrea Pighini
a
, Paolo E. Porporato
a
,
Nicoletta Filigheddu
a
, Guido Serini
b,c
, Fabiola Sinigaglia
a
, Gianluca Baldanzi
a
, and Andrea Graziani
a,2
a
Department of Clinical and Experimental Medicine and Biotecnologie per la Ricerca Medica Applicata, University Amedeo Avogadro of Piemonte Orientale,
28100 Novara, Italy;
b
Department of Oncological Sciences and Division of Molecular Angiogenesis, Istituto per la Ricerca e la Cura del Cancro, Institute for
Cancer Research and Treatment, University of Torino School of Medicine, 10060 Candiolo, Italy; and
c
Center for Complex Systems in Molecular Biology and
Medicine, University of Torino, 10100 Torino, Italy
Edited* by Lewis Clayton Cantley, Harvard Medical School, Boston, MA, and approved December 14, 2009 (received for review July 29, 2009)
Diacylglycerol kinases (DGKs) convert diacylglycerol (DAG) into
phosphatidic acid (PA), acting as molecular switches between
DAG- and PA-mediated signaling. We previously showed that Src-
dependent activation and plasma membrane recruitment of DGKα
are required for growth-factor-induced cell migration and rufing,
through the control of Rac small-GTPase activation and plasma
membrane localization. Herein we unveil a signaling pathway
through which DGKαcoordinates the localization of Rac. We show
that upon hepatocyte growth-factor stimulation, DGKα, by produc-
ing PA, provides a key signal to recruit atypical PKCζ/ι(aPKCζ/ι)in
complex with RhoGDI and Rac at rufing sites of colony-growing
epithelial cells. Then, DGKα-dependent activation of aPKCζ/ιmedi-
ates the release of Rac from the inhibitory complex with RhoGDI,
allowing its activation and leading to formation of membrane ruf-
es, which constitute essential requirements for cell migration.
These ndings highlight DGKαas the central element of a lipid sig-
naling pathway linking tyrosine kinase growth-factor receptors to
regulation of aPKCs and RhoGDI, and providing a positional signal
regulating Rac association to the plasma membrane.
cell migration
|
growth factors
|
phosphatidic acid
Cell migration, central to many biological and pathological
processes such as cancer metastatic progression, is a multistep
cycle involving extension of protrusions and formation of stable
attachments near the leading edge, followed by translocation of the
cell body forward (1). The protrusive activity occurring at the
leading edge depends on the spatial and temporal coordination
between cell substrate adhesion and actin reorganization. Rho-
family small GTPases coordinate the recruitment at the leading
edge of downstream effectors, thereby mediating the formation of
rufes and lamellipodia. Their GTP-bound state is tightly regulated
by both guanine nucleotide exchange factors (GEFs), which stim-
ulate GTP loading, and GTPase activating proteins (GAPs), which
catalyze GTP hydrolysis. Moreover, Rho-family GTPases are re-
gulated by guanine nucleotide dissociation inhibitors (GDIs), which
antagonize both GEFs and GAPs and mediate the cycling of Rho
proteins between the cytosol and the membrane (2, 3).
Atypical protein kinase C ζand ι(aPKCζ/ι), unlike classical
and novel PKCs, feature a C1-like domain which does not bind
to either diacylglycerol or phorbol esters, and have recently been
proposed as key transducers for establishment of cell polarity
and migration (4).
Diacylglycerol kinases (DGKs), which convert diacylglycerol
(DAG) to phosphatidic acid (PA), comprise a family of 10 distinct
enzymes grouped into veclasses, each featuring distinct regulatory
domains and a highly conserved catalytic domain preceded by two
cysteine-rich C1-like domains. An increasing body of evidence
indicates that DGKs, by acting as terminators of diacylglycerol-
triggered signaling, contribute to regulating C1 domain-containing
proteins, such as classical and novel PKCs and the Rac-GAP
β-chimaerin (5). Conversely, by generating PA, DGKs regulate
several signaling proteins, including serine kinases, small-GTPase-
regulating proteins, and lipid-metabolizing enzymes (reviewed in
refs. 6 and 7). Thus, by regulating in a reciprocal manner the level of
both DAG and PA lipid second messengers, DGK enzymes may act
as terminators of DAG-mediated signals as well as activators of
PA-mediated ones. We previously showed that DGKαis activated
by growth factors on recruitment to the plasma membrane through
its Src-mediated phosphorylation on Tyr
335
. Activation of DGKα
mediates growth-factor-induced cell migration and proliferation in
epithelial, endothelial, and lymphoma cells (812). Moreover, we
demonstrated that in epithelial cells, DGKαis required for hep-
atocyte growth factor (HGF) -induced membrane rufing by regu-
lating Rac membrane targeting and activation (13). However, the
molecular mechanisms by which DGKαregulates Rac function still
remain to be elucidated.
Here we unveil a previously undescribed signaling pathway
linking HGF receptor to Rac activation and targeting at the
plasma membrane of epithelial cells, where it drives the formation
of membrane protrusions. Our data highlight that upon HGF-
induced plasma membrane recruitment and activation of DGKα,
PA recruits at the plasma membrane and activates aPKCζ/ι,in
complex with RhoGDI and Rac. Then, Rac is released from the
inhibitory complex with RhoGDI, allowing its activation and for-
mation of membrane rufes.
Results
DGKαRegulates Rac by Directing Its Recruitment to the Plasma
Membrane. MDCK epithelial cells grow in discrete colonies and
on HGF stimulation undergo scatter, involving cell spreading,
dissolution of intercellular adhesions, and migration of cells away
from one another. At early time points from HGF stimulation,
cells at the periphery of a colony reorganize the actin cytoske-
leton and form dynamic protrusions of the outer plasma mem-
brane known as rufes, mediated by membrane targeting and
activation of Rac (14, 15). We previously showed that down-
regulation of DGKαaffected both HGF-induced membrane
rufing and recruitment of Rac to the plasma membrane (13).
Here, by live-cell imaging, we report that HGF-induced rapid
and transient recruitment of EGFP-Rac at sites of intense ruf-
Author contributions: F.C., E.R., F.S., G.B., and A.G. designed research; F.C., E.R., C.C., V.B.,
A.P., G.S., and G.B. performed research; F.C., E.R., C.C., V.B., A.P., P.E.P., N.F., G.S., G.B., and
A.G. analyzed data; G.S. contributed new reagents/analytic tools; and F.C., E.R., and A.G.
wrote the paper.
The authors declare no conict of interest.
*This Direct Submission article had a prearranged editor.
1
F.C. and E.R. contributed equally to this work.
2
To whom correspondence should be addressed. E-mail: graziani@med.unipmn.it.
This article conta ins supporting in formation online at www.pnas.org/cgi/content/full/
0908326107/DCSupplemental.
41824187
|
PNAS
|
March 2, 2010
|
vol. 107
|
no. 9 www.pnas.org/cgi/doi/10.1073/pnas.0908326107
ing was impaired upon DGK inhibition (Fig. S1 and Movies S1
and S2), achieved by cell treatment with 1 μM R59949. R59949 is
a DGK-specic inhibitor interacting with the catalytic domain,
endowed with a strong preference for α,β, and δisoforms (16,
17). We previously showed that overexpression of DGKαre-
verted R59949-mediated inhibition of HGF signaling (8).
Thus, we investigated the mechanisms by which DGKαmight
regulate Rac-specic localization. In the absence of growth-fac-
tor stimulation, constitutively active RacV12 mutant localized
mainly in the cytoplasm and at cell-cell adhesions, whereas its
expression was not sufcient to induce rufing at the leading
edge of cells at the periphery of a colony, according to previous
observations (15). HGF cell treatment increased the percentage
of cells featuring concomitant membrane rufes and RacV12 at
the outer plasma membrane in a DGK-dependent manner (Fig.
S2). In addition, we compared endogenous Rac localization in
MDCK cells transiently transfected with either DGKαwild type
(DGKαWT) or a constitutively active membrane-bound DGKα
mutant (myr-DGKα). Indeed, the expression of myr-DGKα, but
not of DGKαWT, promoted a 2.5-fold increase of the percent-
age of cells displaying membrane rufes, along with Rac
recruitment to the plasma membrane at rufing sites (Fig. 1).
Overall, these data indicate that DGKαactivation provides a
crucial signal which is both necessary and sufcient to direct Rac
membrane localization at the leading edge, regardless of its GTP
loading, thereby promoting rufe formation.
DGKαRegulates RhoGDI Membrane Recruitment. An increasing
body of evidence indicates that RhoGDI, which in unstimulated
cells associates with GDP-bound Rac in a cytosolic complex,
regulates Rac targeting to specic adhesion sites at the plasma
membrane through largely unidentied mechanisms (2). Once at
the plasma membrane, Rac is released from the inhibitory com-
plex with RhoGDI and is eventually activated by a Rac-GEF (2,
18). To verify whether growth factors trigger Rac localization at
the plasma membrane through interaction with RhoGDI, we
transiently transfected MDCK cells with a Rac mutant, RacR66E,
unable to bind to RhoGDI (19). Indeed, RacR66E failed to be
recruited at rufing sites upon HGF cell treatment whereas HGF-
induced membrane rufe formation was not impaired, indicating
that endogenous Rac is properly activated (Fig. 2). These data
demonstrate that RhoGDI physical interaction with Rac mediates
growth-factor-induced Rac targeting at rufing sites.
Thus, we investigated whether HGF, although inducing mem-
brane rufes, was able to recruit RhoGDI to the leading edge and
whether this process required DGKαactivity. We show that HGF
increased the percentage of cells displaying RhoGDI at rufing
sites, which was impaired by both pharmacological inhibition (Fig.
3A) and siRNA-mediated silencing of DGKα(Fig. S3A). In
addition, in time-lapse experiments, HGF-induced rapid and
transient recruitment of RhoGDI to rufing sites required DGK
activity (Fig. S4 and Movies S3 and S4).
Moreover, we investigated whether DGKαconstitutive activa-
tion at the plasma membrane was sufcient to recruit RhoGDI.
Thus, the expression of myr-DGKα, but not of DGKαWT, was
sufcient to recruit RhoGDI at rufing sites in almost 80% of
transfected cells (Fig. 3Band Fig. S3B). Altogether, these ndings
demonstrate that stimulation of DGKαenzymatic activity pro-
vides a crucial signal which is necessary and sufcient to direct the
recruitment of RhoGDI, which in turn is required for Rac tar-
geting at protrusion sites and rufe formation.
DGKαRegulates Membrane Rufing by Modulating aPKCζ/ιFunction.
The molecular mechanisms regulating RhoGDI localization are
still largely unknown, as it lacks membrane-interacting domains.
PKCζ, reported to be regulated by direct binding to PA but not to
DAG (20), associates with and phosphorylates RhoGDI, thereby
allowing its dissociation from Rac (21). Indeed, in MDCK cells, a
small amount of the whole cellular aPKCζ/ιwas constitutively
associated with RhoGDI (see below, Fig. 6). Thus, we raised the
hypothesis that DGKαmight control RhoGDI by providing the
lipid-regulating aPKCζ/ι.
To obtain an appreciable down-regulation of cellular aPKCs,
we silenced both PKCζand PKCιby different combinations of
their respective specic siRNAs (Fig. 4A). We observed that
aPKCζ/ιsilencing impaired HGF-induced membrane rufe for-
mation, as well as both RhoGDI and Rac recruitment to the
outer plasma membrane (Fig. 4B). The aPKCζ/ιcatalytic activity
was actually required, as aPKCζ/ιinhibition by cell treatment
with PKCζ/ιpseudosubstrate peptide impaired HGF-induced
membrane rufe formation and Rac plasma membrane trans-
location (Fig. S5A). Similarly, inhibition of PKCζby expression
of PKCζdominant-negative mutant (PKCζKW) impaired HGF-
induced RhoGDI translocation to the outer plasma membrane
Rac
DGK WT
Rac+DGK
+actin
myr-DGK
ZOOM
DGK
0
20
40
60
80
% of cells with Rac
at ruffles
DGKWT
myrDGK
***
Fig. 1. DGKαprovides the signal directing Rac to the nascent rufe. MDCK
cells, transfected with either DGKαWT or myr-DGKα, were cultured over-
night in the absence of serum, xed, and stained for Rac (green), myc tag
(red), and actin (blue). Arrows indicate DGKαWT- or myr-DGKα-transfected
cells. (Scale bar, 10 μm.) C, 30 transfected cells, scored for the presence of Rac
at rufing sites. n= 6, with SEM; ***P= 0.0002.
ZOOM
mCherry-
RacR66E
control HGF
mCherry-
RacR66E
+actin
RacR66E
at pl. membr.
% of cells
*
control
HGF
0
20
40
60
80
ruffles
Fig. 2. Rac plasma membrane targeting requires Rac/RhoGDI interaction.
MDCK cells were transiently transfected with mCherry-RacR66E, treated with
10 ng/mL HGF for 15 min, xed, and stained for actin (green). (Scale bar,
24 μm.) C, 30 transfected cells, scored for the presence of rufes or RacR66E
plasma membrane localization. n= 3, with SEM; *P= 0.0057.
Chianale et al. PNAS
|
March 2, 2010
|
vol. 107
|
no. 9
|
4183
CELL BIOLOGY
(Fig. S5B). Moreover, aPKCζ/ιwas also required for myr-DGKα-
induced membrane rufe formation and Rac and RhoGDI tar-
geting to the outer plasma membrane (Fig. 4Cand Fig. S5C).
Together these data indicate that, upon HGF stimulation or
myr-DGKαexpression, aPKCζ/ιmediates membrane rufe for-
mation and recruitment of the molecular machinery necessary
for polarized extension of plasma membrane protrusions.
We then investigated whether DGKαwas involved in the regu-
lation of aPKCζ/ιdownstream of HGF signaling. Indeed, we ob-
served that HGF induced the recruitment of aPKCζ/ιto membrane
rufes in a DGKα-dependent manner. In fact, both specicdown-
regulation of DGKαby two different siRNAs (Fig. 5Aand Fig.
S6A) and its pharmacological inhibition by R59949 cell treatment
(Fig. S6B) completely abolished HGF-induced recruitment of
aPKCζ/ιto the outer plasma membrane, indicating that enzymatic
activity of DGKa was required.
Moreover, expression of myr-DGKαwas sufcient to recruit
aPKCζ/ιto rufing sites, as the percentage of cells displaying
this localization of aPKCζ/ιincreased 3-fold in myr-DGKα-
expressing cells compared to DGKαWT-expressing ones (Fig. 5B
and Fig. S6C). Accordingly, cell treatment with PA but not with
lysophosphatidic acid (LPA) or DAG was sufcient to promote
concurrent cortical actin rearrangements and plasma membrane
translocation of aPKCζ/ι, Rac, and RhoGDI (Fig. 5Cand Fig.
S7). Moreover, cell treatment with exogenous PA, but not with
DAG, induced aPKCζ/ιactivation, as revealed by the increase in
phosphorylation of the catalytic domain Thr410 (Fig. 5D).
Together, these data strongly support the idea that the gen-
eration of PA by DGKαis a crucial signal driving the recruitment
and activation of the molecular machinery necessary for exten-
sion of membrane protrusions.
As accumulating evidence suggests that the small GTPase Cdc42
acts upstream of PKCζand regulates it through the Par3/Par6
complex, thus driving cell polarity and directional migration (22),
we further veried whether DGKαmight regulate aPKCζ/ιvia
Cdc42. Indeed, the expression of Cdc42N17, although impairing
membrane rufes induced by myr-DGKα, did not affect aPKCζ/ι
plasma membrane recruitment (Fig. S8). Thus, although Cdc42 is
required for rufe formation induced by constitutive generation of
PA at the plasma membrane, it is not required for the recruitment
of aPKCζ/ι. Overall, these data indicate that DGKαacts upstream
of aPKCζ/ι, in a Cdc42-independent manner, by regulating its
recruitment to rufingsitesthroughproductionofPA.
A
control R59949 HGF+R59949
RhoGDI
RhoGDI
+actin
HGF
*
ctr
% of cells with
RhoGDI at the
pl. membr.
*
RHH+R
0
10
20
30
40
50
B
ZOOM
DGK WT
RhoGDI
RhoGDI+DGK
+actin
myr-DGK
DGK
Fig. 3. DGKαregulatesRhoGDI targeting to the plasma membrane. (A) MDCK
cells were stimulated with 10 ng/mL HGF for 5 min in the presence or absence
of 1 μM R59949, xed, and stained for RhoGDI (green) and actin (blue). Arrows
indicate RhoGDI at membrane rufes. (Scale bar, 24 μm.) C, 70 cells, scored for
RhoGDI localization at rufing sites. n= 3, with SEM; *P<0.05. (B) MDCK cells,
transfected with either DGKαWT or myr-DGKα,were cultured overnight in the
absence of serum, xed, and stained for RhoGDI (green), myc tag (red), and
actin (blue). Arrowheads indicate transfected cells. (Scale bar, 10 μm.)
A
B
C
Fig. 4. aPKCζ/ιmediates both HGF- and myr-DGKα-induced extension of
membrane protrusions. (A) MDCK cells were transfected with three combi-
nations of PKCζ(79, 80) and PKCι(13) specic siRNAs. Whole-cell lysates
were analyzed for levels of aPKCζ/ιexpression by western blot. (B) MDCK
cells were transfected as in A, treated with 10 ng/mL HGF for 15 min, xed,
and stained for Rac or RhoGDI and actin. C, 110 cells, analyzed for the
presence of rufes and Rac or RhoGDI at the plasma membrane. n= 4 (Rac
and RhoGDI), n= 8 (rufes), with SEM; **P<0.005, ***P<0.0005. (C) MDCK
cells were transiently transfected either with myr-DGKαalone or cotrans-
fected with myr-DGKαand PKCζKW, cultured overnight in the absence of
serum, xed, and stained for myc and ag tags, actin, and Rac or RhoGDI. C,
20 transfected cells, scored for the presence of rufes and Rac or RhoGDI at
protrusion sites. n= 4, with SEM; **P<0.001, ***P<0.0001.
4184
|
www.pnas.org/cgi/doi/10.1073/pnas.0908326107 Chianale et al.
We then investigated whether, by regulating the function of
aPKCζ/ι,DGKαmight control the dissociation of Rac/RhoGDI
complex. We immunoprecipitated RhoGDI from MDCK cells sta-
bly expressing either the control vector (MDCK/empty vector) or a
kinase-defective dominant-negative mutant of DGKα(DGKαDN).
Rac coimmunoprecipitated with RhoGDI both in MDCK/empty
vector and in MDCK/DGKαDN unstimulated cells. Upon HGF
stimulation, Rac was released from the complex with RhoGDI in
MDCK/empty vector cells, whereas expression of DGKαDN pre-
vented both dissociation of the complex (Fig. 6 and Fig. S9)andRac
activation (13). Collectively, the data presented above identify
DGKαas an upstream regulator of aPKCζ/ι.DGKα,byproducing
PA, provides the crucial signal for the recruitment at the plasma
membrane and activation of aPKCζ/ι, in complex with RhoGDI and
Rac, which nally dissociates in a DGKα-dependent manner,
thereby allowing Rac activation.
Finally, we tested the hypothesis that DGKαmediates the for-
mation of membrane rufes by regulating aPKCζ/ι.Thus,we
veried whether transient expression of membrane-bound con-
stitutively active myr-PKCζcould overcome DGKαinhibition, as
expected for a downstream effector. Indeed, myr-PKCζ-expressing
cells featured an altered morphology with membrane protrusions
and rufes, even in the absence of stimulation, and in 60% of them
RhoGDI was localized at the outer plasma membrane. Prolonged
(1-h) pharmacological inhibition of DGK activity by R59949 did
not affect either formation of membrane protrusions or RhoGDI
localization to the outer plasma membrane (Fig. 7), conrming that
aPKCζ/ιis responsible, downstream of DGKα, for the recruitment
of RhoGDI at the leading edge and consequent activation of the
molecular machinery necessary for extension of membrane rufes.
Overall, our ndings highlight that DGKα, by producing PA,
provides a crucial signal to recruit aPKCζ/ιand RhoGDI/Rac
complex and to activate aPKCζ/ι, thereby allowing Rac mem-
brane targeting and activation and consequent actin cytoskeleton
remodeling preliminary to cell migration.
Discussion
Epithelial cells at the periphery of a colony initiate migration by
extending membrane protrusions whose formation relies on re-
cruitment and activation of Rac to their tips, to harness and
localize actin polymerization (23). Spatially restricted activation of
Rac at nascent rufes is triggered by coordinated signals provided
by both growth factors and adhesion receptors. Moreover, the
observation that in epithelial cells the expression of a constitutively
active form of Rac is not per se sufcient to promote rufe for-
mation (15, 24) strongly indicates that a further localization signal,
A
B
C
D
Fig. 5. DGKαregulates aPKCζ/ιfunction. (A) MDCK cells were transfected
either with control siRNA or DGKαsiRNA c1 and c2, treated with 10 ng/mL
HGF for 15 min, xed, and stained for aPKCζ/ι(green) and actin (red). (Scale
bar, 10 μm.) (B) MDCK cells, transfected either with DGKαWT or myr-DGKα,
were cultured overnight in the absence of serum, xed, and stained for
aPKCζ/ι(green), myc tag (red), and actin (blue). Arrowheads indicate trans-
fected cells. (Scale bar, 10 μm.) (C) MDCK cells were stimulated with either
250 μM C8-PA, C8-DAG, or C6-LPA or left untreated, and xed and stained
for actin (red) and aPKCζ/ι(green). Arrowheads indicate cortical actin rear-
rangements, while the arrow indicates aPKCζ/ιmembrane localization.
(Scale bar, 19 μm.) (D) MDCK cells were stimulated with either 250 μM C8-PA,
10% FCS medium (as positive control), 250 μM C8-DAG, or left untreated.
Whole-cell lysates were analyzed by western blot and the intensity of
phospho-aPKCζ/ιbands was quantied by densitometry. For each condition,
nine replicate points were performed in four independent experiments. The
densitometry of each band was normalized as the percentage of the den-
sitometry mean of control points in the same experiment, and is shown in
the histogram, with SEM; **P<0.005. A representative picture is shown.
-H
emptyvector
DGKDN
-H
RhoGDI
IB: RhoGDI
IB: Rac
IP:
IB: aPKC /
IB: RhoGDI
IB: aPKC /
IB: Rac
IB: myc
input
-H
emptyvector
DGKDN
-H
Fig. 6. DGKαregulates Rac/RhoGDI complex dissociation. MDCK/empty
vector or MDCK/DGKα-DN cells were stimulated with 50 ng/mL HGF for
15 min. Cell lysates were immunoprecipitated for RhoGDI and analyzed by
western blot.
Chianale et al. PNAS
|
March 2, 2010
|
vol. 107
|
no. 9
|
4185
CELL BIOLOGY
provided by growth factors, is required to generate RacGTP
accumulation at protrusion sites. Starting from our initial obser-
vation that DGKαis required for HGF-induced RacV12 mem-
brane targeting, we unveil a signaling pathway by which HGF
regulates Rac localization to nascent rufes through membrane
recruitment of DGKα, aPKCζ/ι, and RhoGDI.
We previously showed that, upon HGF stimulation, activation
and membrane recruitment of Rac, as well as rufe formation,
are fully dependent on DGKα(13). Our nding that expression
of a myristoylated mutant of DGKαpromoted rufe formation
and Rac recruitment at protrusion sites, in the absence of growth
factor, strongly suggests that activation of DGKαat the plasma
membrane provides a crucial signal to regulate Rac function.
Previous evidence underscored the role of RhoGDI in regu-
lating the dynamics of Rac targeting to the plasma membrane,
where Rac dissociates from the inhibitory complex with RhoGDI
to interact with its downstream effectors (2, 18). Our observation
that in HGF-treated cells RhoGDI is recruited to nascent rufes,
and that a Rac mutant unable to bind to RhoGDI is not, provides
further support to the claim that the interaction between Rac and
RhoGDI is necessary for Rac membrane targeting. Moreover, the
present data demonstrate that growth factors promote Rac
membrane localization by regulating RhoGDI targeting. We show
that DGKαat the plasma membrane provides a key lipid signal
necessary and sufcient to recruit RhoGDI. Furthermore, the
failure to detect DGKαin a complex with RhoGDI and Rac
suggests that DGKα, rather than acting as a scaffolding protein,
may recruit RhoGDI through its enzymatic activity. However,
RhoGDI does not feature any clear domain responsible for
membrane binding, suggesting that the lipid signal generated
by DGKαmay recruit RhoGDI through an interacting lipid-
binding protein.
Recent evidence indicates that PKCζassociates with RhoGDI
and regulates the dissociation of Rac/RhoGDI complex, thereby
allowing Rac activation (21). Moreover, PKCιregulates invasive
behavior by activating Rac in lung cancer cells (25). Interestingly,
PA, the lipid product of DGKαactivity, has been reported to bind
directly to PKCζand to stimulate its enzymatic activity (20),
whereas it was recently shown that DGKαenhances PKCζ-
mediated phosphorylation of p65/Rel (26). Here we show that
upon HGF stimulation, DGKα-generated PA is a necessary and
sufcient signal to recruit aPKCζ/ιat protrusion sites and activate
it, thereby promoting Rac and RhoGDI membrane targeting.
Kuribayashi et al. showed that PKCζ, associated in a complex
with RhoGDI, mediates its phosphorylation on threonine, thereby
allowing Rac release (21). Based on this observation and on our
data, we expected that Rac release from the complex with RhoGDI
depended on DGKαactivity. Consistently, in thepresent report, we
show that DGKαenzymatic activity mediated HGF-induced Rac
release from RhoGDI, although we could not detect any threonine
phosphorylation of endogenous RhoGDI. Finally, these results
suggest the following working model: Upon HGF stimulation, Src-
mediated activation of DGKα, once targeted to the plasma mem-
brane, results in accumulation of PA, whichdirects the recruitment
of aPKC/RhoGDI/Rac complex at protrusion sites, likely through
the interaction of DGKα-produced PA with the C1-like domain of
aPKCζ/ι, which is nally activated. Thus, aPKCζ/ιis identied as
the direct downstream effector of DGKα. Once at the plasma
membrane, aPKCζ/ιmay allow Rac release from RhoGDI, likely
through RhoGDI phosphorylation. This model is conrmed by the
nding that expression of myr-PKCζ, in the absence of growth
factors, recapitulates both RhoGDI recruitment and protrusion
formation in a DGKα-independent manner, providing further
support to the tenet that DGKαregulates RhoGDI and Rac by
acting upstream of aPKCζ/ι(Fig. 8).
aPKC is recruited to the plasma membrane through binding to
active Cdc42 (27) or by directed interaction with ceramide in a
Cdc42-independent manner (28). Our nding that Cdc42 is dis-
pensable for aPKCζ/ιrecruitment induced by myr-DGKαsuggests
that Cdc42 function is not required for lipid-mediated aPKCζ/ι
membrane targeting, albeit necessary for rufe formation down-
stream of DGKα.
As RhoGDI also controls Cdc42, we may speculate that acti-
vation of DGKαprovides a localization signal driving plasma
membrane recruitment of aPKCs in complex with RhoGDI and
Rac or Cdc42, eventually leading to the activation of Cdc42 and
Rac, which may further recruit aPKCs in a Cdc42-dependent
manner. This might establish a feed-forward mechanism that
allows the full activation of the molecular machinery driving
actin polymerization and consequent formation of membrane
protrusions.
PAK1-mediated serine phosphorylation of RhoGDI promotes
the selective release of Rac downstream of Cdc42 activation
(29). Recently, DGKζwas shown to regulate PAK1-mediated
phosphorylation of RhoGDI, leading to selective activation of
Rac but not of Cdc42 (30). Conversely, PKCζ-dependent
phosphorylation of RhoGDI promotes the release of both Rac
and Cdc42 (21). Thus, it seems that DGKαand DGKζ, by reg-
ulating, respectively, aPKCζ/ιand PAK1, are involved in two
myr-aPKC /RhoGDI
RhoGDI
ZOOM
Myr-aPKC /+
RhoGDI+actin
control
R59949
0
20
40
60
80
100
protrusions
Pl.membr.
RhoGDI
% of cells
vehicle
R59949
Fig. 7. DGKαdoes not affect myr-PKCζ-induced events. MDCK cells were
transiently transfected with myr-PKCζ, grown overnight in the absence of
serum, and treated with 1 μM R59949 for 1 h. Cells were xed and stained for
ag tag (red), RhoGDI (green), and actin (blue). Arrows indicate RhoGDI
staining at protrusion sites. (Scale bar, 24 μm.) C, 30 transfected cells, scored for
the presence of protrusions and RhoGDI at protrusion sites. n= 3, with SEM.
HGF
actin
polymerization
ruffling
migration
Src
Src
P
RhoGDI
Rac
GDP
aPKC/
PA
PA
DGK
DGK
DAG
RhoGDI
aPKC/
Rac
GTP
P
PA
Fig. 8. Model proposed for Rac-localized activation at the leading edge
upon growth-factor stimulation. Upon growth-factor stimulation, DGKαis
activated in a Src-dependent manner and recruited to the plasma mem-
brane. The production of PA is the crucial signal to direct the recruitment of
aPKCζ/ι, in complex with RhoGDI and Rac. PKCζ/ι, in turn, mediates the dis-
sociation of Rac from the inhibitory complex with RhoGDI, which may
become prone to activation by a RacGEF.
4186
|
www.pnas.org/cgi/doi/10.1073/pnas.0908326107 Chianale et al.
distinct molecular mechanisms leading to the control of small-
GTPase function.
In conclusion, our ndings constitute a coherent demon-
stration that DGKα, upon Src-mediated activation, acts as a
positive transducer of growth-factor signaling by producing PA
rather than removing DAG. Here we unveil a PA-mediated
signaling pathway linking tyrosine kinase receptors to Rac acti-
vation, membrane rufing, and cell migration. In particular, we
highlight a pivotal role for a DGKα-aPKCζ/ι-RhoGDI axis in the
regulation of the initial events leading to activation of Rac at the
leading edge of migrating cells.
Materials and Methods
Cell Stimulation. Before any treatment, cells were cultured overnight in the
absence of serum. In the case of R59949 cell treatment, a 15-min pretreat-
ment with R59949, or vehicle alone, was performed.
Immunouorescence. Culturing, xing, and staining detailed procedures are
described in SI Materials and Methods and ref. 13. Representative pictures
are shown in the gures.
Statistical Analysis. In experiments involving counting of cells displaying
rufes, membrane protrusions, or localization of proteins at the outer plasma
membrane (not at cell-cell contacts), only colony-edge cells were considered.
The average number of cells scored for each condition in each experiment is
indicated in gure legends by C.Statistical analysis was performed con-
sidering nindependent experiments (indicated in the gure legends). The P
values were calculated by one-tailed Standard ttest.
Further details are provided in SI Materials and Methods.
ACKNOWLEDGMENTS. This work was supported by Associazione Italiana per
la Ricerca sul Cancro, Ministero dellIstruzione, dellUniversità e della Ricerca,
PRIN Program 2007, and Regione Piemonte (Ricerca Sanitaria Finalizzata).
F.C. is funded by Regione Piemonte (L.R. 4/2006 for brain drain containment,
D.R. 392-2007). We thank Paola Chiarugi, Celine DerMardirossian, Louise
Hodgson, Giorgio Scita, and Alex Toker for providing reagents.
1. Webb D-J, Parsons J-T, Horwitz A-F (2002) Adhesion assembly, disassembly and
turnover in migrating cells: over and over and over again. Nat Cell Biol 4:97100.
2. Del Pozo M-A, et al. (2002) Integrins regulate GTP-Rac localized effector interactions
through dissociation of Rho-GDI. Nat Cell Biol 4:232239.
3. Del Pozo M-A, Schwartz M-A (2007) Rac, membrane heterogeneity, caveolin and
regulation of growth by integrins. Trends Cell Biol 17:246250.
4. Nishimura T, Yamaguchi T, Kato K, Yoshizawa M, Nabeshima Y, Ohno S, Hoshino M,
Kaibuchi K (2005) PAR-6-PAR-3 mediates Cdc42-induced Rac activation through the
Rac GEFs STEF/Tiam1. Nat Cell Biol 7:27027.
5. Carrasco S, Merida I (2006) Diacylglycerol, when simplicity becomes complex. Trends
Biochem Sci 32:2736.
6. Sakane F, Imai S, Kai M, Yasuda S, Kanoh H (2007) Diacylglycerol kinases: why so many
of them? Biochim. Biophys Acta 1771:793806.
7. Mérida I, Avila-Flores A, Merino E (2008) Diacylglycerol kinases: at the hub of cell
signalling. Biochem J 409:118.
8. Cutrupi S, et al. (2000) Src-mediated activation of alpha-diacylglycerol kinase is
required for hepatocyte growth factor-induced cell motility. EMBO J 19:46144622.
9. Baldanzi G, et al. (2004) Activation of diacylglycerol kinase alpha is required for VEGF-
induced angiogenic signaling in vitro. Oncogene 23:48284838.
10. Bacchiocchi R, et al. (2005) Activation of alpha-diacylglycerol kinase is critical for the
mitogenic properties of anaplastic lymphoma kinase. Blood 106:21752182.
11. Baldanzi G, et al. (2008) Diacylglycerol kinase-alpha phosphorylation by Src on Y335 is
required for activation, membrane recruitment and HGF-induced cell motility.
Oncogene 27:942956.
12. Filigheddu N, et al. (2007) Diacylglycerol kinase is required for HGF-induced
invasiveness and anchorage-independent growth of MDA-MB-231 breast cancer cells.
Anticancer Research 27:14891492.
13. Chianale F, et al. (2007) Diacylglycerol kinase-alpha mediates hepatocyte growth
factor-induced epithelial cell scatter by regulating Rac activation and membrane
rufing. Mol Biol Cell 18:48594871.
14. Royal I, Lamarche-Vane N, Lamorte L, Kaibuchi K, Park M (2000) Activation of cdc42,
rac, PAK, and rho-kinase in response to hepatocyte growth factor differentially
regulates epithelial cell colony spreading and dissociation. Mol Biol Cell 11:
17091725.
15. Ridley A-J, Comoglio P-M, Hall A (1995) Regulation of scatter factor/hepatocyte
growth factor responses by Ras, Rac, and Rho in MDCK cells. Mol Cell Biol 15:
11101122.
16. Jiang Y, Sakane F, Kanoh H, Walsh JP (2000) Selectivity of the diacylglycerol kinase
inhibitor 3-[2-(4-[bis-(4-uorophenyl)methylene]-1-piperidinyl)ethyl]-2,3-dihydro-2-
thioxo-4(1H)quinazolinone (R59949) among diacylglycerol kinase subtypes. Biochem
Pharmacol 59:763772.
17. Miele C, et al. (2007) Glucose regulates diacylglycerol intracellular levels and protein
kinase C activity by modulating diacylglycerol kinase subcellular localization. J Biol
Chem 282:3183531843.
18. Moissoglu K, Slepchenko B-M, Meller N, Horwitz A-F, Schwartz M-A (2006) In vivo
dynamics of Rac-membrane interactions. Mol Biol Cell 17:27702779.
19. Gandhi P-N, et al. (2004) An activating mutant of Rac1 that fails to interact with Rho
GDP-dissociation inhibitor stimulates membrane rufing in mammalian cells.
Biochem J 378:409419.
20. Limatola C, Schaap D, Moolenaar W-H, van Blitterswijk W-J (1994) Phosphatidic acid
activation of protein kinase C-zeta overexpressed in COS cells: comparison with other
protein kinase C isotypes and other acidic lipids. Biochem J 304:10011008.
21. Kuribayashi K, et al. (2007) Essential role of protein kinase C zeta in transducing a
motility signal induced by superoxide and a chemotactic peptide, fMLP. J Cell Biol
176:10491060.
22. Henrique D, Schweisguth F (2003) Cell polarity: the ups and downs of the Par6/aPKC
complex. Curr Opin Genet Dev 13:341350.
23. Small J-V, Stradal T, Vignal E, Rottner K (2002) The lamellipodium: where motility
begins. Trends Cell Biol 12:112120.
24. Kurokawa K, Itoh R-E, Yoshizaki H, Nakamura Y-O, Matsuda M (2004) Coactivation of
Rac1 and Cdc42 at lamellipodia and membrane rufes induced by epidermal growth
factor. Mol Biol Cell 15:10031010.
25. Frederick, et al. (2008) Matrix metalloproteinase-10 is a critical effector of protein
kinase Ciota-Par6alpha-mediated lung cancer. Oncogene 27:48414853.
26. Kai M, et al. (2009) Diacylglycerol kinase alpha enhances protein kinase Czeta-
dependent phosphorylation at Ser311 of p65/RelA subunit of nuclear factor-kappaB.
FEBS Lett 583:32653268.
27. Etienne-Manneville S (2008) Polarity proteins in migration and invasion. Oncogene
27:69706980.
28. Krishnamurthy K, Wang G, Silva J, Condie B-G, Bieberich E (2007) Ceramide regulates
atypical PKCzeta/lambda-mediated cell polarity in primitive ectoderm cells. A novel
function of sphingolipids in morphogenesis. J Biol Chem 282:33793390.
29. DerMardirossian C, Schnelzer A, Bokoch G-M (2004) Phosphorylation of RhoGDI by
Pak1 mediates dissociation of Rac GTPase. Mol Cell 15:117127.
30. Abramovici H, et al. (2009) Diacylglycerol Kinase {zeta} Regulates Actin Cytoskeleton
Reorganization through Dissociation of Rac1 from RhoGDI. Mol Biol Cell, 10.1091/
mbc.E07-12-1248.
Chianale et al. PNAS
|
March 2, 2010
|
vol. 107
|
no. 9
|
4187
CELL BIOLOGY
... In this process, it has been shown that DGKα plays a role in the formation and elongation of invasive protrusions. Upon growth factor or chemokine stimulation, DGKα binds to the PM leading to the production of PA at the tips of invasive pseudopods [39,79,80,[108][109][110]. The DGKα-produced PA, then, acts to dock atypical PKCζ/ι, Rab11 family of interacting protein 1 (Rab11-FIP1), and integrin α5β1, which triggers actin polymerization, elongation of invasive protrusions, and polarize vesicular trafficking as well as promote directional migration [80,108,109]. ...
... In this process, it has been shown that DGKα plays a role in the formation and elongation of invasive protrusions. Upon growth factor or chemokine stimulation, DGKα binds to the PM leading to the production of PA at the tips of invasive pseudopods [39,79,80,[108][109][110]. The DGKα-produced PA, then, acts to dock atypical PKCζ/ι, Rab11 family of interacting protein 1 (Rab11-FIP1), and integrin α5β1, which triggers actin polymerization, elongation of invasive protrusions, and polarize vesicular trafficking as well as promote directional migration [80,108,109]. In invasive pseudopods, actin polymerization at the cell leading edge leads to an outward directed force that creates membrane tension and, therefore, cause membrane bending [111]. ...
Article
Full-text available
Cancer immunotherapy has revolutionized the oncology field. Despite the success, new molecular targets are needed to increase the percentage of patients that benefits from this therapy. Diacylglycerol kinase α (DGKα) has gathered great attention as a potential molecular target in immunotherapy because of its role in cancer proliferation and immunosuppression. DGKα catalyzes the ATP-dependent phosphorylation of diacylglycerol (DAG) to produce phosphatidic acid (PA). Since both lipids are potent signaling messengers, DGKα acts as a switch between different signaling pathways. Its role in cancer and immunosuppression has long been ascribed to the regulation of DAG/PA levels. However, this paradigm has been challenged with the identification of DGKα substrate acyl chain specificity, which suggests its role in signaling could be specific to DAG/PA molecular species. In several biological processes where DGKα plays a role, large membrane morphological changes take place. DGKα substrate specificity depends on the shape of the membrane that the enzyme binds to. Hence, DGKα can act as a bridge between large membrane morphological changes and the regulation of specific molecular species of DAG/PA. Bearing in mind the potential therapeutic benefits of targeting DGKα, here, the role of DGKα in cancer and T cell biology with a focus on the modulation of its enzymatic properties by membrane shape is reviewed. The goal is to contribute to a global understanding of the molecular mechanisms governing DGKα biology. This will pave the way for future experimentation and, consequently, the design of better, more potent therapeutic strategies aiming at improving the health outcomes of cancer patients.
... These subclasses of lipids have been identified as potential molecular activators of downstream signaling, most importantly, PKC activation. Correlation analysis has shown interconnections between DGK and PKC isoforms in distinct signaling nodes (Chianale et al., 2010;Kai et al., 2009;Rainero et al., 2014). For example, DGKα act as a central element of a lipid signaling pathway to regulate PKC that allows spatiotemporal signaling in epithelial cells (Chianale et al., 2010) and matrix invasion of carcinoma cells (Rainero et al., 2014). ...
... Correlation analysis has shown interconnections between DGK and PKC isoforms in distinct signaling nodes (Chianale et al., 2010;Kai et al., 2009;Rainero et al., 2014). For example, DGKα act as a central element of a lipid signaling pathway to regulate PKC that allows spatiotemporal signaling in epithelial cells (Chianale et al., 2010) and matrix invasion of carcinoma cells (Rainero et al., 2014). Therefore, cellular DAG and PA equilibrium could be a tool to fine-tune DGK and PKC downstream signaling (Mochly-Rosen et al., 2012). ...
Article
Full-text available
Diacylglycerol kinase (DGK), a lipid kinase, catalyzes the conversion of diacylglycerol (DAG) to phosphatidic acid, thereby terminating DAG-mediated signaling by Gq-coupled receptors that regulate contraction of airway smooth muscle (ASM). A previous study from our laboratory demonstrated that DGK inhibition or genetic ablation leads to reduced ASM contraction and provides protection for allergen-induced airway hyperresponsiveness. However, the mechanism by which DGK regulates contractile signaling in ASM is not well established. Herein, we investigated the role of prorelaxant cAMP-protein kinase A (PKA) signaling in DGK-mediated regulation of ASM contraction. Pretreatment of human ASM cells with DGK inhibitor I activated PKA as demonstrated by the phosphorylation of PKA substrates, VASP, Hsp20, and CREB, which was abrogated when PKA was inhibited pharmacologically or molecularly using overexpression of the PKA inhibitor peptide, PKI. Furthermore, inhibition of DGK resulted in induction of cyclooxygenase (COX) and generation of prostaglandin E2 (PGE2) with concomitant activation of Gs-cAMP-PKA signaling in ASM cells in an autocrine/paracrine fashion. Inhibition of protein kinase C (PKC) or extracellular-signal-regulated kinase (ERK) attenuated DGK-mediated production of PGE2 and activation of cAMP-PKA signaling in human ASM cells, suggesting that inhibition of DGK activates the COX-PGE2 pathway in a PKC-ERK-dependent manner. Finally, DGK inhibition-mediated attenuation of contractile agonist-induced phosphorylation of myosin light chain 20 (MLC-20), a marker of ASM contraction, involves COX-mediated cAMP production and PKA activation in ASM cells. Collectively these findings establish a novel mechanism by which DGK regulates ASM contraction and further advances DGK as a potential therapeutic target to provide effective bronchoprotection in asthma.
... Among others, the hepatocyte growth factor (HGF) receptor (MET) has emerged as central regulator of actin cytoskeleton remodeling in the context of cell migration and scattering modulation. It has been accepted that MET activation induces a Rac1-mediated signaling cascade with actin remodeling and membrane ruffling, both by recruiting Rac1 to the plasma membrane 12 and by endosome signaling 13 . Interestingly, MET is able to activate Rac1 from different endosomes, boosting an acute Rac1 signaling when localized in peripheral endosomes (PEs) and a sustained PI3K/Vav2-mediated Rac1 activation when trafficking to a perinuclear endosome (PNE) 13 . ...
Article
Full-text available
Little is known about the signaling network responsible for the organization of the perinuclear actin cap, a recently identified structure holding unique roles in the regulation of nuclear shape and cell directionality. In cancer cells expressing a constitutively active MET, we show a rearrangement of the actin cap filaments, which crash into perinuclear patches associated with spherical nuclei, meandering cell motility and inactivation of the mechano-transducer YAP1. MET ablation is sufficient to reactivate YAP1 and restore the cap, leading to enhanced directionality and flattened nuclei. Consistently, the introduction of a hyperactive MET in normal epithelial cells, enhances nuclear height and alters the cap organization, as also confirmed by TEM analysis. Finally, the constitutively active YAP1 mutant YAP5SA is able to overcome the effects of oncogenic MET. Overall, our work describes a signaling axis empowering MET-mediated YAP1 dampening and actin cap misalignment, with implications for nuclear shape and cell motility.
... PKC activation is likely upstream in the context of DGK inhibition considering PKC has a DAG-binding domain and move closer to membrane upon stimulation of cells by an external signal. Additionally, DGK enzyme activity is regulated by (i) second messengers (Ca 2+ ) (ii) protein-protein interactions (PKC MARCKS domain) or (iii) cellular sub-localization [39,44,45]. It is probable that the proximity of DGK and PKC to the cell membrane allow for feed-forward signaling inducing a stronger, more vigorous signal inducing COXII production. ...
Article
Full-text available
Background Diacylglycerol kinase (DGK) regulates intracellular signaling and functions by converting diacylglycerol (DAG) into phosphatidic acid. We previously demonstrated that DGK inhibition attenuates airway smooth muscle (ASM) cell proliferation, however, the mechanisms mediating this effect are not well established. Given the capacity of protein kinase A (PKA) to effect inhibition of ASM cells growth in response to mitogens, we employed multiple molecular and pharmacological approaches to examine the putative role of PKA in the inhibition of mitogen-induced ASM cell proliferation by the small molecular DGK inhibitor I (DGK I). Methods We assayed cell proliferation using CyQUANT™ NF assay, protein expression and phosphorylation using immunoblotting, and prostaglandin E2 (PGE2) secretion by ELISA. ASM cells stably expressing GFP or PKI-GFP (PKA inhibitory peptide-GFP chimera) were stimulated with platelet-derived growth factor (PDGF), or PDGF + DGK I, and cell proliferation was assessed. Results DGK inhibition reduced ASM cell proliferation in cells expressing GFP, but not in cells expressing PKI-GFP. DGK inhibition increased cyclooxygenase II (COXII) expression and PGE2 secretion over time to promote PKA activation as demonstrated by increased phosphorylation of (PKA substrates) VASP and CREB. COXII expression and PKA activation were significantly decreased in cells pre-treated with pan-PKC (Bis I), MEK (U0126), or ERK2 (Vx11e) inhibitors suggesting a role for PKC and ERK in the COXII-PGE2-mediated activation of PKA signaling by DGK inhibition. Conclusions Our study provides insight into the molecular pathway (DAG-PKC/ERK-COXII-PGE2-PKA) regulated by DGK in ASM cells and identifies DGK as a potential therapeutic target for mitigating ASM cell proliferation that contributes to airway remodeling in asthma.
... In cancer cells, macropinocytosis is in part driven by activation of phospholipid synthesis enzymes including, PI3K, and phospholipase C (53)(54)(55). Diacylglycerol kinases are also known to be recruited to the plasma membrane where they initiate membrane ruffling (the first step in macropinocytosis) via activation of protein kinase c and Rac1 (56). Interestingly, our lipidomic analysis revealed that ritanserin inhibits DGKA-mediated metabolism of diacylglycerol, which results in depletion of phosphatidic acid and inhibition of macropinocytosis in TSC2-deficient cells (Figs. 3 and 5). ...
Article
Full-text available
Lymphangioleiomyomatosis is a rare destructive lung disease affecting primarily women and is the primary lung manifestation of tuberous sclerosis complex (TSC). In lymphangioleiomyomatosis, biallelic loss of TSC1/2 leads to hyperactivation of mTORC1 and inhibition of autophagy. To determine how the metabolic vulnerabilities of TSC2-deficient cells can be targeted, we performed a high-throughput screen utilizing the “Repurposing” library at the Broad Institute of MIT and Harvard (Cambridge, MA), with or without the autophagy inhibitor chloroquine. Ritanserin, an inhibitor of diacylglycerol kinase alpha (DGKA), was identified as a selective inhibitor of proliferation of Tsc2−/− mouse embryonic fibroblasts (MEF), with no impact on Tsc2+/+ MEFs. DGKA is a lipid kinase that metabolizes diacylglycerol to phosphatidic acid, a key component of plasma membranes. Phosphatidic acid levels were increased 5-fold in Tsc2−/− MEFs compared with Tsc2+/+ MEFs, and treatment of Tsc2−/− MEFs with ritanserin led to depletion of phosphatidic acid as well as rewiring of phospholipid metabolism. Macropinocytosis is known to be upregulated in TSC2-deficient cells. Ritanserin decreased macropinocytic uptake of albumin, limited the number of lysosomes, and reduced lysosomal activity in Tsc2−/− MEFs. In a mouse model of TSC, ritanserin treatment decreased cyst frequency and volume, and in a mouse model of lymphangioleiomyomatosis, genetic downregulation of DGKA prevented alveolar destruction and airspace enlargement. Collectively, these data indicate that DGKA supports macropinocytosis in TSC2-deficient cells to maintain phospholipid homeostasis and promote proliferation. Targeting macropinocytosis with ritanserin may represent a novel therapeutic approach for the treatment of TSC and lymphangioleiomyomatosis. Significance: This study identifies macropinocytosis and phospholipid metabolism as novel mechanisms of metabolic homeostasis in mTORC1-hyperactive cells and suggest ritanserin as a novel therapeutic strategy for use in mTORC1-hyperactive tumors, including pancreatic cancer. Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/81/8/2086/F1.large.jpg.
... Those findings are in line with previous results obtained with a lipid motility shift assay [19]. Previously, our group demonstrated that DGKα-produced PA is required to localise the aPKCs to the plasma membrane, where their activity leads to cytoskeletal remodelling and membrane ruffles formation, two essential processes required for cell migration [14,15,21]. These findings indicate that PA provides a key signal to recruit and activate aPKCs at specific membrane compartments. ...
Article
Full-text available
PKCζ and PKCι/λ form the atypical protein kinase C subgroup, characterized by lack of regulation by calcium and the neutral lipid diacylglycerol. To better understand the regulation of these kinases, we systematically explored their interaction with various purified phospholipids using the lipid overlay assays, followed by kinase activity assays to evaluate lipid effects on their enzymatic activity. We observed that both PKCζ and PKCι interact with phosphatidic acid and phosphatidylserine. Conversely, PKCι is unique in binding also to phosphatidylinositol-monophosphates (e.g. phosphatidylinositol 3-phosphate, 4-phosphate, and 5-phosphate). Moreover, we observed that phosphatidylinositol 4-phosphate specifically activates PKCι, while both isoforms are responsive to phosphatidic acid and phosphatidylserine. Overall, our results suggest that atypical PKCs localization and activity are regulated by membrane lipids distinct from those involved in conventional PKCs and unveil a specific regulation of PKCι by phosphatidylinositol-monophosphates.
... PKCs are classified into "classical/conventional" or calcium-dependent cPKCs (α, βI, βII, and γ), "novel" or calciumindependent nPKCs (δ, ε, η, and θ), and "atypical" aPKCs (ζ and ι) [12,13]. A lot of metastatic factors including PDGF, EGF, and HGF can activate PKC to release the second messenger diacylglycerol (DAG) in a PLC-dependent manner [14,15]. Specifically, TPA can potently activate PKC mimicking the action of DAG. ...
Article
Full-text available
Both protein kinase C (PKC) and reactive oxygen species (ROS) are well-known signaling messengers cross-talking with each other to activate mitogen-activated protein kinases (MAPKs) for progression of hepatocellular carcinoma (HCC). However, the underlying mechanisms are not well elucidated. Especially, whether mitochondrial ROS (mtROS) is involved and how it triggers MAPK signaling are intriguing. In this study, we found mtROS generation and phosphorylation of MAPKs were mediated by PKCδ in HCCs treated with the tumor promoter 12-O-tetradecanoyl-phorbol-13-acetate (TPA). Heat shock protein 60 (HSP60), one of the chaperones in mitochondria was the major protein oxidized in TPA-treated HCCs. Moreover, depletion of HSP60 or expression of HSP60 cysteine mutant prevented TPA-induced phosphorylation of MAPKs. To delineate how HSP60 mediated MAPK activation, the role of Raf kinase inhibitor protein (RKIP), a negative regulator of MAPK, was investigated. TPA dissociated RKIP from HSP60 in both mitochondria and cytosol, concurrently with translocation of HSP60 and MAPK from mitochondria to cytosol, which was associated with robust phosphorylation of MAPKs in the cytosol. Moreover, TPA induced opposite phenotypical changes of HCCs, G1 cell cycle arrest, and cell migration, which were prevented by mtROS scavengers and depletion of PKC and HSP60. Consistently, TPA increased the migration-related genes, hydrogen peroxide inducible clone5, matrix metalloproteinase-1/3, laminin𝛾2, and suppressed the cell cycle regulator cyclin E1 (CCNE1) via PKC/mtROS/HSP60/MAPK-axis. Finally, c-jun and c-fos were required for TPA-induced expression of the migration-related genes and a novel microRNA, miR-6134, was responsible for TPA-induced suppression of CCNE1. In conclusion, PKC cross-talked with mtROS to trigger HSP60 oxidation for release of RKIP to activate MAPK, regulating gene expression for migration, and G1 cell cycle arrest in HCC. Targeted therapy aiming at key players like PKCδ, RKIP, and HSP60 is promising for preventing HCC progression. Keywords: HSP60; PKCδ; RKIP; MAPK; ERK; JNK; TPA; ROS; mtROS; HCC
... Moreover, DGKζ-deficient CTL show an impairment in MTOC docking to the IS, which correlates with a reduced translocation of phosphorylated PKCζ [85]. PKCζ is known to be regulated by DGK-derived PA in non-T cells [86], and it was shown to promote MTOC polarization in primary CD4 + and CD8 + T cells [87,88], suggesting a role for the DGKζ/PA/PKCζ axis in this process. Remarkably, the impact of this kinase is not limited to T cells, as DGKζ-mediated PA production was shown to play a role in the assembly of the B-cell IS, regulating actin remodeling, MTOC translocation, force generation, and antigen-uptake related processes [89]. ...
Article
Full-text available
Recognition of antigens displayed on the surface of an antigen-presenting cell (APC) by T-cell receptors (TCR) of a T lymphocyte leads to the formation of a specialized contact between both cells named the immune synapse (IS). This highly organized structure ensures cell–cell communication and sustained T-cell activation. An essential lipid regulating T-cell activation is diacylglycerol (DAG), which accumulates at the cell–cell interface and mediates recruitment and activation of proteins involved in signaling and polarization. Formation of the IS requires rearrangement of the cytoskeleton, translocation of the microtubule-organizing center (MTOC) and vesicular compartments, and reorganization of signaling and adhesion molecules within the cell–cell junction. Among the multiple players involved in this polarized intracellular trafficking, we find sorting nexin 27 (SNX27). This protein translocates to the T cell–APC interface upon TCR activation, and it is suggested to facilitate the transport of cargoes toward this structure. Furthermore, its interaction with diacylglycerol kinase ζ (DGKζ), a negative regulator of DAG, sustains the precise modulation of this lipid and, thus, facilitates IS organization and signaling. Here, we review the role of SNX27, DAG metabolism, and their interplay in the control of T-cell activation and establishment of the IS.
Article
Rho subfamily of G proteins (e.g., Rac1) have been implicated in glucose-stimulated insulin secretion from the pancreatic β-cell. Interestingly, metabolic stress (e.g., chronic exposure to high glucose) results in sustained activation of Rac1 leading to increased oxidative stress, impaired insulin secretion and β-cell dysfunction. Activation-deactivation of Rho G proteins is mediated by three classes of regulatory proteins, namely the guanine nucleotide exchange factors (GEFs), which facilitate the conversion of inactive G proteins to their active conformations; the GTPase-activating proteins (GAPs), which convert the active G proteins to their inactive forms); and the GDP-dissociation inhibitors (GDIs), which prevent the dissociation of GDP from G proteins. Contrary to a large number of GEFs (82 members) and GAPs (69 members), only three members of RhoGDIs (RhoGDIα, RhoGDIβ and RhoGDIγ) are expressed in mammalian cells. Even though relatively smaller in number, the GDIs appear to play essential roles in G protein function (e.g., subcellular targeting) for effector activation and cell regulation. Emerging evidence also suggests that the GDIs are functionally regulated via post-translational modification (e.g., phosphorylation) and by lipid second messengers, lipid kinases and lipid phosphatases. We highlight the underappreciated regulatory roles of RhoGDI-Rho G protein signalome in islet β-cell function in health and metabolic stress. Potential knowledge gaps in the field, and directions for future research for the identification of novel therapeutic targets to loss of functional β-cell mass under the duress of metabolic stress are highlighted.
Article
Diacylglycerol kinase (DGK) phosphorylates diacylglycerol to produce phosphatidic acid (PtdOH) and regulates the balance between two lipid second messengers: diacylglycerol and PtdOH. Several lines of evidence suggest that the η isozyme of DGK is involved in the pathogenesis of bipolar disorder. However, the detailed molecular mechanisms regulating the pathophysiological functions remain unclear. One reason is that it is difficult to detect the cellular activity of DGKη. To overcome this difficulty, we utilized protein myristoylation and a cellular PtdOH sensor, the N‐terminal region of α‐synuclein (α‐Syn‐N). Although DGKη expressed in COS‐7 cells was broadly distributed in the cytoplasm, myristoylated (Myr)‐AcGFP‐DGKη and Myr‐AcGFP‐DGKη‐KD (inactive (kinase‐dead) mutant) were substantially localized in the plasma membrane. Moreover, DsRed monomer‐α‐Syn‐N significantly colocalized with Myr‐AcGFP‐DGKη but not Myr‐AcGFP‐DGKη‐KD at the plasma membrane. When COS‐7 cells were osmotically shocked, all DGKη constructs were exclusively translocated to osmotic shock‐responsive granules (OSRG). DsRed monomer‐α‐Syn‐N markedly colocalized with only Myr‐AcGFP‐DGKη at OSRG and exhibited a higher signal/background ratio (3.4) than Myr‐AcGFP‐DGKη at the plasma membrane in unstimulated COS‐7 cells (2.5), indicating that α‐Syn‐N more effectively detects Myr‐AcGFP‐DGKη activity in OSRG. Therefore, these results demonstrated that the combination of myristoylation and the PtdOH sensor effectively detects DGKη activity in cells and that this method is convenient to examine the molecular functions of DGKη. Moreover, this method will be useful for the development of drugs targeting DGKη. Furthermore, the combination of myristoylation (intensive accumulation in membranes) and α‐Syn‐N can be applicable to assays for various cytosolic PtdOH‐generating enzymes.
Article
Full-text available
Activation of Rac1 GTPase signaling is stimulated by phosphorylation and release of RhoGDI by the effector p21-activated kinase 1 (PAK1), but it is unclear what initiates this potential feed-forward mechanism for regulation of Rac activity. Phosphatidic acid (PA), which is produced from the lipid second messenger diacylglycerol (DAG) by the action of DAG kinases (DGKs), is known to activate PAK1. Here, we investigated whether PA produced by DGKzeta initiates RhoGDI release and Rac1 activation. In DGKzeta-deficient fibroblasts PAK1 phosphorylation and Rac1-RhoGDI dissociation were attenuated, leading to reduced Rac1 activation after platelet-derived growth factor stimulation. The cells were defective in Rac1-regulated behaviors, including lamellipodia formation, membrane ruffling, migration, and spreading. Wild-type DGKzeta, but not a kinase-dead mutant, or addition of exogenous PA rescued Rac activation. DGKzeta stably associated with PAK1 and RhoGDI, suggesting these proteins form a complex that functions as a Rac1-selective RhoGDI dissociation factor. These results define a pathway that links diacylglycerol, DGKzeta, and PA to the activation of Rac1: the PA generated by DGKzeta activates PAK1, which dissociates RhoGDI from Rac1 leading to changes in actin dynamics that facilitate the changes necessary for cell motility.
Article
Full-text available
Cancer is the result of the deregulation of cell proliferation and cell migration. In advanced tumors, cells invade the surrounding tissue and eventually form metastases. This is particularly evident in carcinomas in which epithelial cells have undergone epithelial-mesenchymal transition. Increased cell migration often correlates with a weakening of intercellular interactions. Junctions between neighboring epithelial cells are required to establish and maintain baso-apical polarity, suggesting that not only loss of cell-cell adhesion but also alteration of cell polarity is involved during invasion. Accordingly, perturbation of cell polarity is an important hallmark of advanced invasive tumors. Cell polarity is also essential for cell migration. Indeed, a front-rear polarity axis has first to be generated to allow a cell to migrate. Because cells migrate during invasion, cell polarity is not completely lost. Instead, polarity is modified. From a nonmigrating baso-apically polarized epithelial phenotype, cells acquire a polarized migrating mesenchymal phenotype. The aim of this review is to highlight the molecular relationship between the control of cell polarity and the regulation of cell motility during oncogenesis.
Article
Full-text available
Hepatocyte growth factor (HGF), the ligand for the Met receptor tyrosine kinase, is a potent modulator of epithelial-mesenchymal transition and dispersal of epithelial cells, processes that play crucial roles in tumor development, invasion, and metastasis. Little is known about the Met-dependent proximal signals that regulate these events. We show that HGF stimulation of epithelial cells leads to activation of the Rho GTPases, Cdc42 and Rac, concomitant with the formation of filopodia and lamellipodia. Notably, HGF-dependent activation of Rac but not Cdc42 is dependent on phosphatidylinositol 3-kinase. Moreover, HGF-induced lamellipodia formation and cell spreading require phosphatidylinositol 3-kinase and are inhibited by dominant negative Cdc42 or Rac. HGF induces activation of the Cdc42/Rac-regulated p21-activated kinase (PAK) and c-Jun N-terminal kinase, and translocation of Rac, PAK, and Rho-dependent Rho-kinase to membrane ruffles. Use of dominant negative and activated mutants reveals an essential role for PAK but not Rho-kinase in HGF-induced epithelial cell spreading, whereas Rho-kinase activity is required for the formation of focal adhesions and stress fibers in response to HGF. We conclude that PAK and Rho-kinase play opposing roles in epithelial-mesenchymal transition induced by HGF, and provide new insight regarding the role of Cdc42 in these events.
Article
Full-text available
Diacylglycerol kinases are involved in cell signaling, either as regulators of diacylglycerol levels or as intracellular signal-generating enzymes. However, neither their role in signal transduction nor their biochemical regulation has been elucidated. Hepatocyte growth factor (HGF), upon binding to its tyrosine kinase receptor, activates multiple signaling pathways stimulating cell motility, scattering, proliferation and branching morphogenesis. Herein we demonstrate that: (i) the enzymatic activity of alpha-diacylglycerol kinase (alphaDgk) is stimulated by HGF in epithelial, endothelial and alphaDgk-transfected COS cells; (ii) cellular expression of an alphaDgk kinase-defective mutant inhibits activation of endogenous alphaDgk acting as dominant negative; (iii) specific inhibition of alphaDgk prevents HGF-induced cell movement of endothelial cells; (iv) HGF induces the association of alphaDgk in a complex with Src, whose tyrosine kinase activity is required for alphaDgk activation by HGF; (v) Src wild type stimulates alphaDgk activity in vitro; and (vi) alphaDgk can be tyrosine phosphorylated in intact cells.
Article
We recently reported that diacylglycerol kinase (DGK) alpha enhanced tumor necrosis factor-alpha (TNF-alpha)-induced activation of nuclear factor-kappaB (NF-kappaB). However, the signaling pathway between DGKalpha and NF-kappaB remains unclear. Here, we found that small interfering RNA-mediated knockdown of DGKalpha strongly attenuated protein kinase C (PKC) zeta-dependent phosphorylation of a large subunit of NF-kappaB, p65/RelA, at Ser311 but not PKCzeta-independent phosphorylation at Ser468 or Ser536. Moreover, knockdown and overexpression of PKCzeta suppressed and synergistically enhanced DGKalpha-mediated NF-kappaB activation, respectively. These results strongly suggest that DGKalpha positively regulates TNF-alpha-dependent NF-kappaB activation via the PKCzeta-mediated Ser311 phosphorylation of p65/RelA.
Article
Phosphatidic acid (PA) is produced rapidly in agonist-stimulated cells, but the physiological function of this PA is unknown. We have examined the effects of PA on distinct isoforms of protein kinase C (PKC) using a new cell-free assay system. Addition of PA to cytosol from COS cells overexpressing PKC-alpha, -epsilon or -zeta differentially-activated all three isotypes, as shown by PKC autophosphorylation, and prominent phosphorylation of multiple endogenous substrates. In the absence of Ca2+, the diacylglycerol-insensitive zeta-isotype of PKC was most strongly activated by both PA and bisPA, a newly identified product of activated phospholipase D, with each lipid inducing its own profile of protein phosphorylation. BisPA was also a strong activator of PKC-epsilon, but a weak activator of PKC-alpha. Ca2+, at > or = 0.1 microM, inhibited PA and bisPA activation of PKC-zeta, but did not affect PKC-epsilon activation. In contrast, PKC-alpha was strongly activated by PA only in the presence of Ca2+. BisPA-induced phosphorylations mediated by PKC-zeta could be mimicked in part by other acidic phospholipids and unsaturated fatty acids. PA activation of PKC-zeta was unique in that PA not only stimulated PKC-zeta-mediated phosphorylation of distinctive substrates, but also caused an upward shift in electrophoretic mobility of PKC-zeta, which was not observed with other acidic lipids or with PKC-alpha or -epsilon. We have presented evidence that this mobility shift is not caused by PKC-zeta autophosphorylation, but it coincides with physical binding of PA to PKC-zeta. These results suggest that in cells stimulated under conditions where intracellular Ca2+ is at (or has returned to) basal level, PA may be a physiological activator of PKC-zeta.
Article
Scatter factor/hepatocyte growth factor (SF/HGF) stimulates the motility of epithelial cells, initially inducing centrifugal spreading of cell colonies followed by disruption of cell-cell junctions and subsequent cell scattering. These responses are accompanied by changes in the actin cytoskeleton, including increased membrane ruffling and lamellipodium extension, disappearance of peripheral actin bundles at the edges of colonies, and an overall decrease in stress fibers. The roles of the small GTP-binding proteins Ras, Rac, and Rho in regulating responses to SF/HGF were investigated by microinjection. Inhibition of endogenous Ras proteins prevented SF/HGF-induced actin reorganization, spreading, and scattering, whereas microinjection of activated H-Ras protein stimulated spreading and actin reorganization but not scattering. When a dominant inhibitor of Rac was injected, SF/HGF- and Ras-induced spreading and actin reorganization were prevented, although activated Rac alone did not stimulate either response. Microinjection of activated Rho inhibited spreading and scattering, while inhibition of Rho function led to the disappearance of stress fibers and peripheral bundles but did not prevent SF/HGF-induced motility. We conclude that Ras and Rac act downstream of the SF/HGF receptor p190Met to mediate cell spreading but that an additional signal is required to induce scattering.
Article
Diacylglycerol kinases (DGKs) attenuate diacylglycerol-induced protein kinase C activation during stimulated phosphatidylinositol turnover. This reaction also initiates phosphatidylinositol resynthesis. Two agents, 3-(2-(4-[bis-(4-fluorophenyl)methylene]-1-piperidinyl)ethyl)-2,3-dihydro -2-thioxo-4(1H)quinazolinone (R59949) and 6-(2-(4-[(4-fluorophenyl)phenylmethylene]-1-piperidinyl)ethyl)-7-m ethyl-5H-thiazolo(3,2-a)pyrimidin-5-one (R59022), inhibit diacylglycerol phosphorylation in several systems. To examine the mechanism of this effect, we developed a mixed micelle method suitable for in vitro study of DGK inhibition. Animal cells express multiple DGK isoforms. In a survey of DGK isotypes, these agents selectively inhibited Ca2+-activated DGKs. R59949 was the more selective of the two. To map the site of interaction with the enzyme, a series of DGKalpha deletion mutants were prepared and examined. Deletion of the Ca2+-binding EF hand motif, which is shared by Ca2+-activated DGKs, had no effect on inhibition. Consistent with this observation, inhibition kinetics were noncompetitive with Ca2+. A construct expressing only the catalytic domain was also inhibited by R59949. Studies of substrate kinetics demonstrated that MgATP potentiated R59949 inhibition, indicating synergy of inhibitor and MgATP binding. These results indicate that R59949 inhibits DGKalpha by binding to its catalytic domain.
Article
Lamellipodia, filopodia and membrane ruffles are essential for cell motility, the organization of membrane domains, phagocytosis and the development of substrate adhesions. Their formation relies on the regulated recruitment of molecular scaffolds to their tips (to harness and localize actin polymerization), coupled to the coordinated organization of actin filaments into lamella networks and bundled arrays. Their turnover requires further molecular complexes for the disassembly and recycling of lamellipodium components. Here, we give a spatial inventory of the many molecular players in this dynamic domain of the actin cytoskeleton in order to highlight the open questions and the challenges ahead.