ArticlePDF Available

Regulation of Cell Polarity and Protrusion Formation by Targeting RhoA for Degradation

Authors:

Abstract and Figures

The Rho family of small guanosine triphosphatases regulates actin cytoskeleton dynamics that underlie cellular functions such as cell shape changes, migration, and polarity. We found that Smurf1, a HECT domain E3 ubiquitin ligase, regulated cell polarity and protrusive activity and was required to maintain the transformed morphology and motility of a tumor cell. Atypical protein kinase C zeta (PKCζ), an effector of the Cdc42/Rac1-PAR6 polarity complex, recruited Smurf1 to cellular protrusions, where it controlled the local level of RhoA. Smurf1 thus links the polarity complex to degradation of RhoA in lamellipodia and filopodia to prevent RhoA signaling during dynamic membrane movements.
Smurf1 targets RhoA for ubiquitination and degradation through ubiquitin-dependent proteasome pathway. (A) Expression of Smurf1 decreases the steady-state level of RhoA. HEK293T cells were transiently transfected with the indicated combinations of wild-type (WT) or catalytically inactive (CA) Flag-tagged Smurf1 (F/Smurf1) or Flag-tagged Smurf2 (F/Smurf2) and Flag-tagged RhoA (F/RhoA), Cdc42 (F/Cdc42), or Rac1 (F/Rac1). Steady-state protein levels were determined by immunoblotting (IB) total cell lysates with anti-Flag. (B) Proteasome inhibitors decrease RhoA down-regulation by Smurf1. HEK293T cells transfected with hemagglutinin (HA)-tagged RhoA (HA/RhoA) and F/Smurf1 were treated for 4 hours with or without 40 M LLnL, and RhoA steady-state levels determined. (C) Ubiquitination of RhoA in HEK293T cells. After overnight treatment with 20 M LLnL, lysates from cells transfected with HA-tagged ubiquitin (HA/Ub), F/RhoA, or Myc-tagged Smurf1 (M/ Smurf1) (WT or CA) were subjected to anti-Flag immunoprecipitation, eluted by boiling in 1% SDS, and then reprecipitated with anti-Flag (2X IP). Ubiquitin-conjugated RhoA [(Ub) n-RhoA] and free RhoA were detected by immunoblotting with the appropriate antibodies. M/Smurf1 expression was confirmed by anti-Myc immunoblotting samples of total lysates. (D) Interaction of Smurf1 with RhoA. HEK293T cells were transfected with F/Smurf1(CA) and various versions of HA/RhoA (WT, V14, or N19). Cell lysates were subjected to anti-Flag immunoprecipitation followed by immunoblotting (IB) with rat anti-HA to detect associated RhoA. Total protein expression was confirmed by immunoblotting as shown. (E) Interaction between Smurf1 and RhoA in vitro. Samples of bacterially produced RhoA treated with or without 0.25 mM GDP or GTPS, as indicated, were incubated with glutathione beads coupled to glutathione S-transferase (GST ) or GST/Smurf1(CA). Associated RhoA (top) and input RhoA (bottom) were determined by immunoblotting (IB) with the indicated antibody. GST was detected by Ponceau S staining. (F) Direct ubiquitination of RhoA by Smurf1. RhoA and WT or C699A Smurf1 were purified from bacteria and subjected to an in vitro ubiquitination assay.
… 
Content may be subject to copyright.
normal Syn function and its misfunction
in synucleinopathies. We established that
the ability of the protein to inhibit PLD,
promote the accumulation of lipids, influ-
ence the balance of vesicular pools, asso-
ciate with membranes in a highly selective
manner, induce ubiquitin accumulation,
and inhibit the proteasome when misfolded
are all intrinsic and biologically relevant
properties of the protein that can be uncou-
pled from each other by the effects of Syn
mutations. Constructs expressing mutant
Q103 Htt did not produce similar biological
effects (supporting online material), and
unbiased genetic screens confirmed that
distinct pathways are involved in Syn and
Htt toxicities (30). Notably, membrane-
bound Syn is in dynamic equilibrium with
cytoplasmic forms. Just a twofold differ-
ence in expression was sufficient to cause a
catastrophic change in its behavior, induc-
ing nucleated polymerization and recruiting
protein previously associated with mem-
branes to cytoplasmic inclusions. This nu-
cleated polymerization process suggests a
mechanism by which even small changes in
the QC balance of aging neurons could
produce a toxic gain of Syn function con-
comitantly with a loss of normal function.
Thus, two hypotheses [gain of toxic func-
tion or loss of normal function (31)] put
forward to explain PD can be reconciled by
a single molecular mechanism.
Note added in proof: Very recently Sin-
gleton et al.(32) reported that a triplication of
the Syn locus on one chromosome (presum-
ably doubling the expression of wild-type
Syn) causes premature onset of PD, strong-
ly supporting our model that a small change
in the expression of Syn relative to the cell’s
quality-control systems causes disease-relat-
ed toxicity.
References and Notes
1. C. B. Lucking, A. Brice, Cell. Mol. Life Sci. 57, 1894
(2000).
2. T. Gasser, J. Neurol. 248, 833 (2001).
3. T. Kitada et al., Nature 392, 605 (1998).
4. E. Leroy et al., Nature 395, 451 (1998).
5. Y. Liu, L. Fallon, H. A. Lashuel, Z. Liu, P. T. Lansbury Jr.,
Cell 111, 209 (2002).
6. P. J. Muchowski, Neuron 35, 9 (2002).
7. Materials and methods, additional data, and other
supporting materials concerning data not shown are
available on Science Online.
8. P. J. McLean, H. Kawamata, B. T. Hyman, Neuro-
science 104, 901 (2001).
9. T. F. Outeiro, S. Lindquist, data not shown.
10. E. Jo, J. McLaurin, C. M. Yip, P. St George-Hyslop, P. E.
Fraser, J. Biol. Chem. 275, 34328 (2000).
11. M. H. Polymeropoulos et al., Science 276, 2045 (1997).
12. R. Kruger et al., Nature Genet. 18, 106 (1998).
13. E. Jo, N. Fuller, R. P. Rand, P. St George-Hyslop, P. E.
Fraser, J. Mol. Biol. 315, 799 (2002).
14. R. Bussell Jr., D. Eliezer, J. Mol. Biol. 329, 763 (2003).
15. S. J. Berke, H. L. Paulson, Curr. Opin. Genet. Dev. 13,
253 (2003).
16. J. P. Taylor, J. Hardy, K. H. Fischbeck, Science 296,
1991 (2002).
17. K. A. Conway et al., Proc. Natl. Acad. Sci. U.S.A. 97,
571 (2000).
18. A. Hershko, A. Ciechanover, A. Varshavsky, Nature
Med. 6, 1073 (2000).
19. L. Stefanis, K. E. Larsen, H. J. Rideout, D. Sulzer, L. A.
Greene, J. Neurosci. 21, 9549 (2001).
20. D. M. Sampathu, B. I. Giasson, A. C. Pawlyk, J. Q.
Trojanowski, V. M. Lee, Am. J. Pathol. 163, 91 (2003).
21. N. F. Bence, R. M. Sampat, R. R. Kopito, Science 292,
1552 (2001).
22. D. D. Murphy, S. M. Rueter, J. Q. Trojanowski, V. M.
Lee, J. Neurosci. 20, 3214 (2000).
23. N. B. Cole et al., J. Biol. Chem. 277, 6344 (2002).
24. J. M. Jenco, A. Rawlingson, B. Daniels, A. J. Morris,
Biochemistry 37, 4901 (1998).
25. V. A. Bankaitis, J. R. Aitken, A. E. Cleves, W. Dowhan,
Nature 347, 561 (1990).
26. S. A. Rudge, T. R. Pettitt, C. Zhou, M. J. Wakelam, J. A.
Engebrecht, Genetics 158, 1431 (2001).
27. A. Sreenivas, J. L. Patton-Vogt, V. Bruno, P. Griac, S. A.
Henry, J. Biol. Chem. 273, 16635 (1998).
28. R. Sharon et al., Proc. Natl. Acad. Sci. U.S.A. 98, 9110
(2001).
29. R. Sharon et al., Neuron 37, 583 (2003).
30. S. Willingham, T. F. Outeiro, M. J. DeVit, S. L.
Lindquist, P. J. Muchowski, Science 302, 1769 (2003).
31. T. M. Dawson, V. L. Dawson, J. Clin. Invest. 111, 145
(2003).
32. A.B. Singleton et al., Science 302, 841 (2003).
33. This work was funded by NIH/National Institute of
Neurological Disorders and Stroke (grant NS044829-
01). T.F.O. was partially supported by Programa Prax-
is XXI, Fundacao para a Ciencia e Tecnologia, Portu-
gal. We thank P. Lansbury, R. Esposito, J. Engebrecht,
H. Chang, and S. Henry for plasmids and strains and
members of the Lindquist laboratory for critical read-
ing of this manuscript.
Supporting Online Material
www.sciencemag.org/cgi/content/full/302/5651/1772/
DC1
Materials and Methods
Figs. S1 to S4
Tables S1 and S2
References
14 August 2003; accepted 13 October 2003
Regulation of Cell Polarity and
Protrusion Formation by
Targeting RhoA for Degradation
Hong-Rui Wang,*
1
Yue Zhang,*
1
Barish Ozdamar,
1,2
Abiodun A. Ogunjimi,
1
Evguenia Alexandrova,
3
Gerald H. Thomsen,
3
Jeffrey L. Wrana
1,2
The Rho family of small guanosine triphosphatases regulates actin cytoskeleton
dynamics that underlie cellular functions such as cell shape changes, migration,
and polarity. We found that Smurf1, a HECT domain E3 ubiquitin ligase, reg-
ulated cell polarity and protrusive activity and was required to maintain the
transformed morphology and motility of a tumor cell. Atypical protein kinase
C zeta (PKC), an effector of the Cdc42/Rac1-PAR6 polarity complex, recruited
Smurf1 to cellular protrusions, where it controlled the local level of RhoA.
Smurf1 thus links the polarity complex to degradation of RhoA in lamellipodia
and filopodia to prevent RhoA signaling during dynamic membrane movements.
The Rho family of small guanosine triphos-
phatases (GTPases) cycle between an active
guanosine 5-triphosphate (GTP)– bound and
inactive guanosine 5-diphosphate (GDP)–
bound state to control cell shape, motility,
polarity, and behavior (15). At the leading
edge of motile cells, Cdc42 and Rac1 regulate
the actin cytoskeleton to form fingerlike
filopodia and sheetlike lamellipodia, respec-
tively, whereas in the cell body RhoA induc-
es assembly of focal adhesions and contrac-
tile actin-myosin stress fibers. Active Rho
GTPases signal through effector complexes
(5, 6), one of which is the PAR (for parti-
tioning defective) polarity complex (7).
PAR6 is a key component of this complex
that binds atypical protein kinase C zeta
(PKC)(810), recruits it to active Cdc42
(810), and is important for cell transforma-
tion (10), polarity (11, 12), and epithelial
tight junctions (9, 1315). One effector path-
way of this complex involves GSK-3 and
APC, which links PAR6 to microtubules and
astrocyte polarity (16); another, involving
Lgl, affects asymmetric cell divisions (17)
and polarization of migrating cells (18). A
direct link between the polarity complex and
regulation of actin cytoskeleton dynamics has
not been defined.
Conjugation of polyubiquitin chains to
protein targets triggers their degradation and
is mediated by E3 enzymes, which include
the Smurf family of C2-WW-HECT ubiquitin
ligases (19, 20) that regulate transforming
growth factor– (TGF-) signal transduction
(2123). Ubiquitin ligases are not known to
regulate cell shape, motility, and polarity.
1
Program in Molecular Biology and Cancer, Samuel
Lunenfeld Research Institute, Mount Sinai Hospital,
Toronto M56 15, Canada.
2
Department of Medical
Genetics and Microbiology, University of Toronto,
Toronto M5S 1A8, Canada.
3
Department of Biochem-
istry and Cell Biology, Center for Developmental Ge-
netics, CMM 348, Stony Brook University, Stony
Brook, NY 11794 –5215, USA.
*These authors contributed equally to this work.
To whom correspondence should be addressed. E-
mail: wrana@mshri.on.ca
R EPORTS
www.sciencemag.org SCIENCE VOL 302 5 DECEMBER 2003 1775
However, when Smurf1 or yellow fluorescent
protein (YFP)tagged Smurf1 was over-
expressed in Mv1Lu epithelial cells, we ob-
served large numbers of dynamic protrusions
in 95% of expressing cells (24). These were
not present in the controls, nor in cells ex-
pressing a catalytic mutant of Smurf1, YFP-
Smurf1(C699A) (Movies S1 and S2) (Fig.
1A). Furthermore, movement of YFP-Smurf1
to the tips of processes often preceded active
extension (Fig. 1A), whereas movement to
the cell body presaged retraction (Movie S1).
Smurf1 expression also strongly reduced
stress fiber formation (Movie S1). In contrast,
Smurf2 rarely induced protrusive activity
(15% of Smurf2-expressing cells). Next, we
examined how increased expression of
Smurf1 affected the behavior of NIH3T3 fi-
broblasts in a wounding assay in which cells
are induced to polarize and migrate into a
wound that is created by scratching the
monolayer with a pipette tip. In controls, the
leading-edge cells displayed organized mi-
gration (fig. S1), whereas Smurf1-expressing
cells were highly disorganized and extended
more protrusions at the leading edge (2.4
protrusions per cell in Smurf1-expressing
populations versus 1.3 in the controls) (Fig.
1B). This suggested that Smurf1 disrupted
polarity, which we examined directly by an-
alyzing the localization of pericentrin, a com-
ponent of the microtubule organization center
(MTOC) (25) (fig. S1). In controls, 76 8%
of cells at the wound edge exhibited polarized
MTOC localization, compared with 30 6%
in wild-type Smurf1-expressing cells, indi-
cating nearly random (25%) polarity (Fig.
1B). In contrast, Smurf1(C699A) had only
slight effects on protrusive activity and
MTOC polarity. Thus, elevated Smurf1 ex-
pression induces protrusive activity and dis-
rupts fibroblast polarity in a manner that is
dependent on the catalytic activity of its
HECT domain.
To determine the importance of endoge-
nous Smurf1 on protrusive activity, we de-
signed small interfering RNA (siRNA) to
Smurf1 that reduced Smurf1 protein by 60%
compared with controls (fig. S2A). We used
the human embryonic kidney (HEK) 293T
tumor cell line because these cells are highly
transfectable, express Smurf1, extend many
cellular protrusions, and display a disorga-
nized actin cytoskeleton typical of a trans-
formed phenotype (Fig. 1C). Examination of
the morphology of Smurf1 siRNA-
transfected cells revealed a marked change at
40 hours after transfection. The cells lost their
protrusions, the actin cytoskeleton was re-
arranged into a cortical F-actin staining pat-
tern, and the cells assumed a cuboidal mor-
phology (Fig. 1C). Similar effects were ob-
served with a second siRNA directed to
Smurf1. In addition, HEK293T motility
through modified Boyden chambers was re-
duced sixfold (fig. S3). These data indicate
that Smurf1 plays an important role in regu-
lating protrusive activity and the transformed
phenotype of HEK293T cells.
Elevated expression of the Rho family
GTPases Cdc42 or Rac1 induces high levels
of protrusive activity, disrupts polarity, and
contributes to cell transformation (26, 27).
Previous studies have established that the
PAR6-PKC complex is a key effector of
Cdc42 and Rac1 that controls polarity, is
localized at lamellipodia and filopodia, and is
required for the oncogenic activities of Cdc42
and Rac1 (811). PKC activity also medi-
ates the loss of stress fibers caused by acti-
vated Cdc42 (26), and in HEK293T cells
PKC inhibitors caused morphological
changes that were similar to those in Smurf1
siRNA-treated cells (28). All of this suggest-
ed that Smurf1 might function as an effector
of this complex. In support of this notion, in
Mv1Lu and NIH3T3 cells, Smurf1 was local-
ized to both lamellipodial- and filopodial-like
protrusions (Fig. 2, A and B), where it colo-
calized extensively with PKC (Fig. 2B).
Furthermore, both the expression of Smurf1
and the kinase activity of PKC were re-
quired for the localization of both proteins to
Fig. 1. Smurf1 is localized to lamellipodia and filopodia and regulates protrusive activity, polarity,
and motility. (A) Mv1Lu cells were transfected with YFP control or YFP fused to wild-type Smurf1
(WT) or Smurf1(C699A) (CA) and imaged by time lapse fluorescence microscopy. Shown are frames
extracted from the supplemental movies at the indicated times (in minutes). The start position of
the tip of a protrusion in a YFP-Smurf1– expressing cell at the beginning of data collection (time
0) is marked (red bar), and movement of the YFP-Smurf1 puncta is shown highlighted (yellow
arrowhead). Note that extension of the tip coincides with the arrival of Smurf1 at the tip. (B)
Regulation of protrusive activity and polarity in NIH3T3 cells. (Top) Protrusions formed per cell in
a NIH3T3 wounding assay were quantitated (mean SEM) and are plotted for each of the
indicated cell lines. (Bottom) Polarity was measured by counting the percentage (SEM) of cells in
the front row that oriented their MTOC in the forward-facing quadrant. (C) Loss of protrusions and
actin cytoskeleton rearrangements induced by Smurf1 siRNA. HEK293T cells were transfected with
control or Smurf1 siRNA and visualized 40 hours later by phase contrast microscopy (top) or Texas
red–phalloidin staining (red; bottom).
R EPORTS
5 DECEMBER 2003 VOL 302 SCIENCE www.sciencemag.org1776
membrane protrusions (fig. S4, A and B).
This is consistent with the requirement for
atypical PKC activity in the assembly and
function of PAR complexes in Caenorhabdi-
tis elegans and during tight junction forma-
tion in epithelial cells (13, 14, 29, 30). Next,
we demonstrated that endogenous PKC
bound Smurf1 and that this was unaffected by
treatment with PKC kinase inhibitors (Fig.
2C). Similar kinase-independent interactions
were observed between bacterially produced
proteins (fig. S4C) and in transiently trans-
fected HEK293T cells, where the steady-state
level of PKC was unaffected by Smurf1 (fig.
S4D). This indicates that PKC is not a sub-
strate of Smurf1. Thus, PKC binds directly
to Smurf1 independent of its kinase activity,
but both the expression of Smurf1 and PKC
kinase activity are required to localize the
complex to filopodia and lamellipodia.
We hypothesized that Smurf1 might be an
effector of the Cdc42-PAR6-PKC pola-
rity complex that mediates the ubiquitin-
dependent degradation of RhoA. In support
of this, wild-type Smurf1, but neither Smurf2
nor catalytically inactive Smurf1, decreased
the steady-state level of RhoA, but had no
effect on Cdc42 or Rac1 (Fig. 3A); this result
was confirmed with the UPR assay (31) (fig.
S5). Two proteasome inhibitors, LLnL and
lactacystin, reversed this effect (Fig. 3B)
(28). Furthermore, Smurf1 expression mark-
edly increased ubiquitin-conjugated RhoA,
whereas Smurf1(C699A) blocked all detect-
able ubiquitination of RhoA (Fig. 3C). Next,
we examined RhoA interaction with Smurf1,
using catalytically inactive Smurf1 to trap the
ligase substrate (21, 32, 33). Smurf1(C699A)
was found to interact with the dominant in-
active form of RhoA, RhoA
N19
(Fig. 3D),
which binds constitutively to guanine nucle-
otide exchange factors (GEFs) (34). Smurf1
also interacted in vitro with either nucleotide-
free or GDP-bound RhoA, whereas loading
with GTPS inhibited the interaction (Fig.
3E). Because most GDP-bound RhoA in cells
is associated with guanine dissociation inhib-
itor (GDI) (35), these results suggest that GDI
may inhibit binding of Smurf1 to GDP-bound
RhoA in mammalian cells. Finally, Smurf1
directly catalyzed ubiquitination of RhoA in
an in vitro ubiquitination assay (Fig. 3F).
Thus, RhoA is a direct substrate of Smurf1,
and degradation of RhoA in mammalian cells
may require nucleotide exchange and be de-
pendent on a Rho GEF.
The PKC-dependent recruitment of
Smurf1 to active membrane protrusions sug-
gested that at endogenous levels of expres-
sion, Smurf1 activity toward RhoA was re-
stricted to sites where the Smurf1-PKC
complex assembled with Cdc42-PAR6 to in-
duce membrane protrusions. Consistent with
this notion, we did not see marked changes in
total RhoA protein levels upon reducing
Smurf1 expression (28). Thus, we considered
that the morphological change in HEK293T
cells treated with Smurf1 siRNA might be
due to ectopic accumulation of RhoA in pro-
trusions. To test this hypothesis, we exam-
ined endogenous RhoA localization at 18
hours after Smurf1 siRNA treatment. At this
early time point before morphological
changes have occurredwe observed strong
accumulations of RhoA and colocalized F-
actin in the tips of most cellular protrusions.
We also observed the initiation of RhoA ac-
cumulation at cell-cell junctions (Fig. 4A).
Thus, the maintenance of low local levels of
RhoA in protrusions was dependent on en-
dogenous Smurf1. To demonstrate that RhoA
degradation was a key target for Smurf1-
dependent regulation of the transformed phe-
notype of HEK293T cells, we next tested the
epistatic relation between Smurf1 and RhoA
using siRNA. siRNA directed to RhoA
blocked expression of the protein (fig. S2B
and Fig. 4B), but had no effect on the mor-
phology (fig. S6A) or F-actin distribution in
HEK293T cells (Fig. 4B). This result indi-
cates that there was little RhoA-dependent
regulation of the actin cytoskeleton in these
cells, consistent with the absence of a dis-
cernible, well-organized stress fiber network.
Notably, cotransfection with RhoA siRNA
strongly suppressed the effect of Smurf1
siRNA (fig. S6A and Fig. 4B). Thus, reduc-
ing RhoA expression blocks the morpholog-
ical transformation and actin cytoskeleton re-
arrangements caused by reducing Smurf1
levels. We also observed that siRNA to RhoA
severely impaired MTOC polarity in NIH3T3
cells (fig. S6B), supporting the notion that
Smurf1 targeting of RhoA is important for
regulating polarity. Our results point to a key
role for Smurf1 as an effector of PKC that
regulates actin cytoskeleton dynamics during
protrusion formation by targeting the local-
ized degradation of RhoA via a ubiquitin-
dependent mechanism.
Regulation of cell polarity and shape by
RhoGTPase-dependent regulation of the actin
cytoskeleton is a key biological pathway that
governs diverse cell functions such as local-
ization of embryonic determinants, establish-
ment of tissue and organ architecture, and cell
motility. The precise temporal and spatial
coordination of Rho family GTPase activity
is important in a broad range of cellular
activities (25). Our findings strongly sug-
gest that Smurf1 is a key effector of the
Cdc42/Rac1-PAR6-PKC pathway that an-
tagonizes RhoA through ubiquitin-dependent
degradation. In cells in which Smurf1 expres-
sion is knocked down by siRNA, RhoA and
associated F-actin accumulate in cellular pro-
trusions, indicating that the activity of
Fig. 2. PKC colocalizes with and binds
Smurf1. (A) Localization of endogenous
Smurf1 was visualized by immunostaining
Mv1Lu cells with a Smurf1 monoclonal anti-
body followed by fluorescein isothiocyanate–
conjugated secondary antibody (green); the
actin cytoskeleton was visualized with Texas
red–phalloidin staining (red). Smurf1 in protru-
sions is marked (yellow arrowheads). Controls
conducted in the absence of primary antibody
showed no detectable signal (27). (B) Colocal-
ization of Smurf1 and PKC. Endogenous PKC
(i) and Smurf1 CA (ii) were visualized in
NIH3T3-Smurf1 CA cells, and colocalization
(iii) of PKC and Flag-tagged Smurf1 to pro-
trusions and lamellipodial-like structures is in-
dicated (yellow arrowheads). (C) Interaction of
endogenous Smurf1 and PKC in Mv1Lu cells. Lysates prepared from cells incubated in the absence
or presence of the indicated inhibitors were subjected to immunoprecipitation with rabbit antibody
to Smurf1 (anti-Smurf1) followed by immunoblotting with anti-PKC.
R EPORTS
www.sciencemag.org SCIENCE VOL 302 5 DECEMBER 2003 1777
Fig. 3. Smurf1 targets RhoA for ubiquitination and degradation through
ubiquitin-dependent proteasome pathway. (A) Expression of Smurf1 de-
creases the steady-state level of RhoA. HEK293T cells were transiently
transfected with the indicated combinations of wild-type (WT) or cata-
lytically inactive (CA) Flag-tagged Smurf1 (F/Smurf1) or Flag-tagged
Smurf2 (F/Smurf2) and Flag-tagged RhoA (F/RhoA), Cdc42 (F/Cdc42), or
Rac1 (F/Rac1). Steady-state protein levels were determined by immuno-
blotting (IB) total cell lysates with anti-Flag. (B) Proteasome inhibitors
decrease RhoA down-regulation by Smurf1. HEK293T cells transfected
with hemagglutinin (HA)–tagged RhoA (HA/RhoA) and F/Smurf1 were
treated for 4 hours with or without 40 M LLnL, and RhoA steady-state
levels determined. (C) Ubiquitination of RhoA in HEK293T cells. After
overnight treatment with 20 M LLnL, lysates from cells transfected with
HA-tagged ubiquitin (HA/Ub), F/RhoA, or Myc-tagged Smurf1 (M/
Smurf1) (WT or CA) were subjected to anti-Flag immunoprecipitation,
eluted by boiling in 1% SDS, and then reprecipitated with anti-Flag (2X
IP). Ubiquitin-conjugated RhoA [(Ub)
n
-RhoA] and free RhoA were detected
by immunoblotting with the appropriate antibodies. M/Smurf1 expression
was confirmed by anti-Myc immunoblotting samples of total lysates. (D)
Interaction of Smurf1 with RhoA. HEK293T cells were transfected with
F/Smurf1(CA) and various versions of HA/RhoA (WT, V14, or N19). Cell
lysates were subjected to anti-Flag immunoprecipitation followed by immu-
noblotting (IB) with rat anti-HA to detect associated RhoA. Total protein
expression was confirmed by immunoblotting as shown. (E) Interaction
between Smurf1 and RhoA in vitro. Samples of bacterially produced RhoA
treated with or without 0.25 mM GDP or GTPS, as indicated, were
incubated with glutathione beads coupled to glutathione S-transferase
(GST ) or GST/Smurf1(CA). Associated RhoA (top) and input RhoA (bottom)
were determined by immunoblotting (IB) with the indicated antibody. GST
was detected by Ponceau S staining. (F) Direct ubiquitination of RhoA by
Smurf1. RhoA and WT or C699A Smurf1 were purified from bacteria and
subjected to an in vitro ubiquitination assay.
Fig. 4. Endogenous RhoA is targeted by Smurf1 in protrusions. (A)
Reduction in Smurf1 leads to appearance of endogenous RhoA in pro-
trusions of HEK293T cells. HEK293T cells transfected with luciferase
control or Smurf1 siRNA as indicated were fixed 18 hours after trans-
fection. Endogenous RhoA staining was visualized with an anti-RhoA
primary antibody (green), and actin was detected with Texas red–
conjugated phalloidin (red). Superimposition of the images (merge)
reveals colocalization of RhoA and F-actin in the protrusions of Smurf1
siRNA-treated HEK293T cells (yellow arrowheads). (B) Reduction of
endogenous RhoA by RhoA siRNA rescues the morphological change
caused by Smurf1 siRNA. HEK293T cells were transfected with Smurf1,
RhoA, or control siRNA, as indicated. Cells were fixed 40 hours after
transfection, and RhoA and F actin were visualized by immunofluores-
cence microscopy as described in (A).
R EPORTS
5 DECEMBER 2003 VOL 302 SCIENCE www.sciencemag.org1778
Smurf1 toward RhoA is locally restricted to
sites of active protrusion. This is likely to be
achieved through PKC-dependent recruit-
ment of Smurf1 to filopodia and lamellipo-
dia, which is in agreement with a key role for
this kinase in controlling the activity of the
polarity complex both in C. elegans (29, 30)
and in mammals (13, 14). Furthermore, we
showed that Smurf1 binds RhoA in a GEF-
dependent manner, suggesting that Smurf1
activity is further restricted by a RhoA GEF
that colocalizes with the polarity complex in
filopodia and lamellipodia. Localizing the deg-
radation of RhoA to protrusive regions likely
acts to prevent inappropriate stress fiber forma-
tion during dynamic actin cytoskeletal remod-
eling that is required to drive rapid filopodial
and lamellipodial membrane extensions in re-
sponse to Cdc42 and Rac1 activation. More-
over, our observation that knocking down
Smurf1 expression suppresses the tumorigenic
morphology and motility of HEK293T cells
suggests that this pathway plays a key role in
maintaining the protrusive activity and trans-
formed phenotype of cancer cells.
References and Notes
1. S. Etienne-Manneville, A. Hall, Nature 420, 629
(2002).
2. D. Bar-Sagi, A. Hall, Cell 103, 227 (2000).
3. A. L. Bishop, A. Hall, Biochem. J. 348, 241 (2000).
4. A. Hall, C. D. Nobes, Philos. Trans. R. Soc. London B
Biol. Sci. 355, 965 (2000).
5. L. Van Aelst, M. Symons, Genes Dev. 16, 1032 (2002).
6. A. Hall, Br. J. Cancer 80 (suppl. 1), 25 (1999).
7. J. E. Gomes, B. Bowerman, Curr. Biol. 12, R444
(2002).
8. D. Lin et al., Nature Cell Biol. 2, 540 (2000).
9. G. Joberty, C. Petersen, L. Gao, I. G. Macara, Nature
Cell Biol. 2, 531 (2000).
10. R. G. Qiu, A. Abo, G. Steven Martin, Curr. Biol. 10, 697
(2000).
11. S. Etienne-Manneville, A. Hall, Cell 106, 489 (2001).
12. S.-H. Shi, L. Y. Jan, Y.-N. Jan, Cell 112, 63 (2003).
13. L. Gao, G. Joberty, I. G. Macara, Curr. Biol. 12, 221
(2002).
14. A. Suzuki et al., J. Cell Sci. 115, 3565 (2002).
15. A. Suzuki et al., J. Cell Biol. 152, 1183 (2001).
16. S. Etienne-Manneville, A. Hall, Nature 421,753
(2003).
17. J. Betschinger, K. Mechtler, J. A. Knoblich, Nature 422,
326 (2003).
18. P. J. Plant et al., Nature Cell Biol. 5, 301 (2003).
19. K. F. Harvey, S. Kumar, Trends Cell Biol. 9, 166 (1999).
20. A. Hershko, A. Ciechanover, Annu. Rev. Biochem. 67,
425 (1998).
21. H. Zhu, P. Kavsak, S. Abdollah, J. L. Wrana, G. H.
Thomsen, Nature 400, 687 (1999).
22. Y. Zhang, C. Chang, D. J. Gehling, A. Hemmati-Brivan-
lou, R. Derynck, Proc. Natl. Acad. Sci. U.S.A. 98, 974
(2001).
23. X. Lin, M. Liang, X.-H. Feng, J. Biol. Chem. 275, 36818
(2000).
24. Materials and methods are available as supporting
material on Science Online.
25. C. D. Nobes, A. Hall, J. Cell Biol. 144, 1235 (1999).
26. M. P. Coghlan, M. M. Chou, C. L. Carpenter, Mol. Cell.
Biol. 20, 2880 (2000).
27. R. G. Qiu, A. Abo, F. McCormick, M. Symons, Mol.
Cell. Biol. 17, 3449 (1997).
28. H.-R. Wang, Y. Zhang, B. Ozdamar, J. L. Wrana, un-
published data.
29. T. J. Hung, K. J. Kemphues, Development 126, 127
(1999).
30. Y. Tabuse et al., Development 125, 3607 (1998).
31. F. Levy, N. Johnsson, T. Rumenapf, A. Varshavsky,
Proc. Natl. Acad. Sci. U.S.A. 93, 4907 (1996).
32. P. Kavsak et al., Mol. Cell 6, 1365 (2000).
33. S. Bonni et al., Nature Cell Biol. 3, 587 (2001).
34. A. Schmidt, A. Hall, Genes Dev. 16, 1587 (2002).
35. K. Kaibuchi, S. Kuroda, M. Amano, Annu. Rev. Bio-
chem. 68, 459 (1999).
36. We thank L. Attisano and members of the Wrana
laboratory for discussion and comments on the
manuscript; S. Elowe, D. Lin, T. Pawson, I. B. Wein-
stein, D. Bohmann, J. Sheng, and A. Varshavsky for
providing reagents; S. Kulkarni for help with decon-
volution microscopy and time-lapse imaging; and L.
Locke for advice on the motility assay. We are in-
debted to J. Trimmer and L. Buckwalder for help in
preparation of monoclonal antibodies. This work was
supported by grants from the Canadian Cancer Soci-
ety and the Canadian Institutes of Health Research
(J.L.W.) and NIH grant HD32429 and Carol M. Bald-
win Breast Cancer Foundation (G.H.T.). Y.Z. and B.O.
are supported by a Canadian Institutes of Health
Research (CIHR) Postdoctoral Fellowship and Doctor-
al Studentship awards, respectively, and E.A. is the
recipient of an Institute for Cell and Developmental
Biology Predoctoral Fellowship from Stony Brook
University. J.L.W. is a CIHR Investigator and an Inter-
national Scholar of the Howard Hughes Medical
Institute.
Supporting Online Material
www.sciencemag.org/cgi/content/full/302/5651/1775/
DC1
Materials and Methods
Figs. S1 to S6
Movies S1 and S2
25 August 2003; accepted 16 October 2003
Redox Regulation of Germline
and Vulval Development in
Caenorhabditis elegans
Yukimasa Shibata,* Robyn Branicky, Irene Oviedo Landaverde,
Siegfried Hekimi
In vitro studies have indicated that reactive oxygen species (ROS) and the
oxidation of signaling molecules are important mediators of signal transduc-
tion. We have identified two pathways by which the altered redox chemistry
of the clk-1 mutants of Caenorhabditis elegans acts in vivo on germline de-
velopment. One pathway depends on the oxidation of an analog of vertebrate
low density lipoprotein (LDL) and acts on the germline through the Ack-related
tyrosine kinase (ARK-1) kinase and inositol trisphosphate (IP
3
) signaling. The
other pathway is the oncogenic ras signaling pathway, whose action on germ-
line as well as vulval development appears to be modulated by cytoplasmic ROS.
Reactive oxygen species (ROS) are short-
lived reactive molecules that can modify
cellular components including nucleic ac-
ids, proteins, and lipids. For example, the
oxidation of LDLs by ROS is one of the
causative factors of atherosclerosis (1).
ROS are toxic but the oxidation of macro-
molecules can also serve as a signaling
device (2). Moreover, in vitro studies have
shown that ROS act as intracellular mes-
sengers in signal transduction pathways,
such as ras signaling (3, 4). However, little
is known about the effect of ROS on signal
transduction in intact animals (5).
Ubiquinone (UQ or coenzyme Q) is a
redox-active lipid that has numerous bio-
chemical roles and is involved in the
production of ROS. However, UQ is also an
antioxidant that prevents the initiation and/
or propagation of lipid peroxidation in cellu-
lar membranes (6). The Caenorhabditis el-
egans clk-1 gene encodes a conserved en-
zyme that is necessary for UQ biosynthesis
(7). In the absence of CLK-1, mutants accu-
mulate demethoxyubiquinone (DMQ) (8, 9),
which can partially replace UQ as an electron
carrier (8). However, clk-1 mutants require
dietary UQ for their survival (9, 10).
clk-1 mutants show a highly pleiotropic
phenotype that includes an average slowing
down and deregulation of a number of phys-
iological processes, including aging (11).
Presumably, given that clk-1 mutants obtain
significant amounts of UQ from their diet
(12), these phenotypes result from the pres-
ence of DMQ, which might be a better anti-
oxidant than UQ (13). Thus, the clk-1 mutant
phenotypes could be the consequence of al-
tered redox signaling.
In addition to the previously described
phenotypes, we find that somatic and germ-
line development are desynchronized in clk-1
mutants. In wild-type hermaphrodites, prima-
ry spermatocytes and sperm are observed at
the late fourth larval stage (L4), and oogen-
esis commences shortly after the adult molt
(Fig. 1). However, the majority of clk-1 mu-
tants are either before or in the process of
spermatogenesis at the adult molt (fig. S1A),
and only 3% of the anterior gonads have
initiated oogenesis at 6 hours after the adult
molt (Fig. 1, B and C). Although the devel-
Department of Biology, McGill University, 1205 Ave-
nue Docteur Penfield, Montre´al, Que´bec, Canada, H3A
1B1.
*Present address: RIKEN Center for Developmental
Biology, 2-2-3 Minatojima Minamimachi, Chuo-ku,
Kobe 650-0047, Japan.
To whom correspondence may be addressed. E-mail:
siegfried.hekimi@mcgill.ca
R EPORTS
www.sciencemag.org SCIENCE VOL 302 5 DECEMBER 2003 1779
... CXCL12 binds to its receptor CXCR4 and activates RhoA through activation of the small G proteins, Gi and Gα13, which leads to directional cell migration [34,35]. Western blot analysis of tissues on Day 7 confirmed that protein expression of CXCR4 and RhoA was significantly higher in the + Mig group than in the PBS group (Fig. 5I-K), indicating that migrasomes activate CXCR4/RhoA signaling. ...
... Cell migration requires actin cytoskeletal reorganization, which is mediated by Rho signaling. RhoA stimulates myosin-based retraction of the cell rear and generates the force needed for retraction of the trailing edge during migration [34]. The CXCL12/CXCR4 chemokine signaling activated RhoA has been a well-documented driver for cell migration. ...
Article
Full-text available
Background Adipose-derived stem cells (ASCs) represent the most advantageous choice for soft tissue regeneration. Studies proved the recruitment of ASCs post tissue injury was mediated by chemokine CXCL12, but the mechanism by which CXCL12 is generated after tissue injury remains unclear. Migrasomes are newly discovered membrane-bound organelles that could deliver CXCL12 spatially and temporally in vivo. In this study, we sought to investigate whether migrasomes participate ASC-mediated tissue regeneration. Methods Discrepant and asymmetrical soft tissue regeneration mice model were established, in which HE staining, immunofluorescent staining, western blot and qPCR were conducted to confirm the role of CXCL12 and migrasomes in ASC-mediated tissue regeneration. Characterization of ASC-derived migrasomes were carried out by confocal microscopy, scanning electron microscopy, transmission electron microscopy as well as western blot analysis. The function and mechanism of migrasomes were further testified by assisting tissue regeneration with isolated migrasomes in vivo and by in vitro transwell combined with co-culture system. Results Here, we show for the first time that migrasomes participate in soft tissue regeneration. ASCs generate migrasomes enriched with CXCL12 to mediate tissue regeneration. Migrasomes from ASCs could promote stem cells migration by activating CXCR4/RhoA signaling in vivo and in vitro. Chemoattracted ASCs facilitate regeneration, as demonstrated by the upregulation of an adipogenesis-associated protein. This positive feed-back-loop creates a favorable microenvironment for soft tissue regeneration. Thus, migrasomes represent a new therapeutic target for ASC-mediated tissue regeneration. Conclusions Our findings reveal a previously unknown function of ASCs in mediating tissue regeneration by generating migrasomes. The ASC-derived migrasomes can restore tissue regeneration by recruiting stem cells, which highlighting the potential application of ASC-derived migrasomes in regenerative medicine.
... SMAD-specific E3 ubiquitin protein ligase 1 colocalizes with CCM2 and associated proteins (CCM complex), 33 where it degrades RhoA in a CCM2-dependent manner. 33,34 Localized degradation of RhoA may have physiologic relevance at sites of CCM complex localization and function. 34 Ste-20 oxidant stress response kinase 1 (SOK-1, STK25), a GCK-III serine/threonine kinase, associates with CCM3 to phosphorylate moesin, which reduces RhoA activity. ...
... 33,34 Localized degradation of RhoA may have physiologic relevance at sites of CCM complex localization and function. 34 Ste-20 oxidant stress response kinase 1 (SOK-1, STK25), a GCK-III serine/threonine kinase, associates with CCM3 to phosphorylate moesin, which reduces RhoA activity. 9,35 Loss of CCM3 and/or SOK-1 attenuates the moesin inhibitory action on RhoA, leading to RhoA activation. ...
Article
Full-text available
Cerebral cavernous malformations (CCMs) are vascular lesions characterized by a porous endothelium. The lack of a sufficient endothelial barrier can result in microbleeds and frank intracerebral hemorrhage. A primary mechanism for lesion development is a sequence variant in at least 1 of the 3 CCM genes (CCM1, CCM2, and CCM3), which influence various signaling pathways that lead to the CCM phenotype. A common downstream process associated with CCM gene loss of function involves overactivation of RhoA and its effector Rho-associated kinase (ROCK). In this study, we review RhoA/ROCK–related mechanisms involved in CCM pathophysiology as potential therapeutic targets. Literature searches were conducted in PubMed using combinations of search terms related to RhoA/ROCK and CCMs. In endothelial cells, CCM1, CCM2, and CCM3 proteins normally associate to form the CCM protein complex, which regulates the functions of a wide variety of protein targets (e.g., MAP3K3, SMURF1, SOK-1, and ICAP-1) that directly or indirectly increase RhoA/ROCK activity. Loss of CCM complex function and increased RhoA/ROCK activity can lead to the formation of stress fibers that contribute to endothelial junction instability. Other RhoA/ROCK–mediated pathophysiologic outcomes include a shift to a senescence-associated secretory phenotype (primarily mediated by ROCK2), which is characterized by endothelial cell migration, cell cycle arrest, extracellular matrix degradation, leukocyte chemotaxis, and inflammation. ROCK represents a potential therapeutic target, and direct (fasudil, NRL-1049) and indirect (statins) ROCK inhibitors have demonstrated various levels of efficacy in reducing lesion burden in preclinical models of CCM. Current (atorvastatin) and planned (NRL-1049) clinical studies will determine the efficacy of ROCK inhibitors for CCM in humans, for which no US Food and Drug Administration–approved or EU-approved pharmacologic treatment exists.
... Proteasomes are essential for proper neuronal function. Using cell-permeable inhibitors that block the catalytic activity of all proteasomes, previous work has revealed proteasome-dependent changes in neuronal functions that occur over hours to days, such as regulation of synaptic plasticity and synaptic vesicle release (Ehlers, 2003;Wang et al., 2003;Rinetti and Schweizer, 2010). Interestingly, proteasome inhibitors have also been shown to have short-term effects in modulating acute neuronal signaling (Bingol and Schuman, 2006;Fonseca et al., 2006;Karpova et al., 2006;Dong et al., 2008;Cai et al., 2010). ...
Article
Full-text available
The neuronal membrane proteasome (NMP) degrades intracellular proteins into peptides that are released directly into the extracellular space, whereby they stimulate neurons to promote signaling mechanisms that remain unknown. Here, we demonstrate that neuronal stimulation promotes NMP activity and, subsequently, enhanced production of NMP peptides. We show that these neuronal activity-dependent NMP peptides can rapidly promote N-methyl-D-aspartate receptor (NMDAR)-dependent calcium influx in neurons. This leads to sustained phosphorylation of the well-defined stimulus-induced transcription factor, cyclic AMP response element (CRE)-binding protein (CREB). Downstream of these events, we identified changes to neuronal target genes which included increased expression of immediate early genes (e.g., Fos, Npas4, Egr4) and other genes known to have critical neuroregulatory roles. Further observations led to the discovery that NMP peptide-induced changes in gene expression is dependent on NMDARs and independent of AMPA receptors or voltage-gated sodium channels. These data demonstrate that NMP peptides are endogenous and selective activators of NMDA receptors and act as sufficient and novel stimuli within the context of neuronal activity-dependent signaling. This novel pathway is parallel to classic neuronal activity-dependent programs and points to NMP and its resulting peptides as potential modulators of neuronal function. [Media: see text] [Media: see text] [Media: see text]
... In present study, we demonstrate that Talin-1 modulates Stat3 protein level in β-cells at least partially through Smurf1, a member of the E3 ubiquitin ligase family, which plays crucial roles in modulating cell differentiation, proliferation, apoptosis, etc. by modulating protein ubiquitination and degradation [66][67][68][69][70]. No physical interaction between Talin-1 and Stat3 is detected in this study, indicating the indirect regulation of Stat3 by Talin-1. ...
Article
Full-text available
Insufficient pancreatic β-cell mass and reduced insulin expression are key events in the pathogenesis of diabetes mellitus (DM). Here we demonstrate the high expression of Talin-1 in β-cells and that deficiency of Talin-1 reduces β-cell proliferation, which leads to reduced β-cell mass and insulin expression, thus causing glucose intolerance without affecting peripheral insulin sensitivity in mice. High-fat diet fed exerbates these phenotypes. Mechanistically, Talin-1 interacts with the E3 ligase smad ubiquitination regulatory factor 1 (Smurf1), which prohibits ubiquitination of the signal transducer and activator of transcription 3 (Stat3) mediated by Smurf1, and ablation of Talin-1 enhances Smurf1-mediated ubiquitination of Stat3, leading to decreased β-cell proliferation and mass. Furthermore, haploinsufficiency of Talin-1 and Stat3 genes, but not that of either gene, in β-cell in mice significantly impairs glucose tolerance and insulin expression, indicating that both factors indeed function in the same genetic pathway. Finally, inducible deletion Talin-1 in β-cell causes glucose intolerance in adult mice. Collectively, our findings reveal that Talin-1 functions as a crucial regulator of β-cell mass, and highlight its potential as a therapeutic target for DM patients.
... Smurf1 and Smurf2 are E3 ubiquitin ligases that modulate PCP signaling to control convergent extension, cell protrusion and motility (Wang et al, 2003;Sahai et al, 2007;Narimatsu et al, 2009;Luga et al., 2012) and have also been shown to play a key role in regulating neuronal morphology (Vohra et al, 2007;Cheng et al, 2011;Kannan et al, 2012). Since cortical neurons express both Smurf1 and Smurf2 as determined by RNA sequencing analysis (Fig. S5A), we next explored whether Smurfs might have a role in exosome-induced axon outgrowth. ...
Preprint
In neurons, the acquisition of a polarized morphology is achieved upon the outgrowth of a single axon from one of several neurites. Exosomes from diverse sources are known to promote the neurite outgrowth, however, the role of fibroblast-derived exosomes on axon elongation in neurons of the central nervous system under growth permissive conditions remains unclear. Here, we show that fibroblast-derived exosomes promote axon outgrowth and a polarized neuronal morphology in mouse primary embryonic cortical neurons. Mechanistically, we demonstrate that the exosome-induced increase in axon outgrowth requires endogenous Wnts and core PCP components including Prickle, Vangl, Frizzled and Dishevelled. We demonstrate that exosomes are internalized by neurons, colocalize with Wnt7b and induce relocalization of Vangl2 to the distal axon during axon outgrowth. In contrast, exosomes derived from neurons or astrocytes do not promote axon outgrowth, while exosomes from activated astrocytes inhibit elongation. Thus, our data reveals that fibroblast-derived exosomes promote axon elongation through the Wnt-PCP pathway in a manner that is dependent on endogenous Wnts.
... WW domain-containing protein 1 (WWP1) induces the ubiquitination and degradation of KLF2 [78]. Smurf1 plays important roles in regulating cell polarity and tumor progression via mediating BMP-Smad, RhoA signaling pathways [79,80]. WWP1 and Smurf1/2 have been also demonstrated to mediate Smads degradation in TGF-b signaling pathway [81]. ...
Article
Full-text available
Surgery and chemo-radiotherapy are used as the common first-line treatment options in many cancers. However, tumor relapse is observed in many cancer patients following such first-line treatments. Therefore, targeted therapy according to the molecular cancer biology can be very important in reducing tumor recurrence. In this regard, a wide range of monoclonal antibodies against the growth factors and their receptors can offer more targeted treatment in cancer patients. However, due to the importance of growth factors in the normal biology of body cells, side effects can also be observed following the application of growth factor inhibitors. Therefore, more specific factors should be introduced as therapeutic targets with less side effects. Krüppel-like factors 2 (KLF2) belongs to the KLF family of transcription factors that are involved in the regulation of many cellular processes. KLF2 deregulations have been also reported during the progression of many tumors. In the present review we discussed the molecular mechanisms of KLF2 during tumor growth and invasion. It has been shown that the KLF2 as a tumor suppressor is mainly inhibited by the non-coding RNAs (ncRNAs) through the polycomb repressive complex 2 (PRC2) recruitment. This review is an effective step towards introducing the KLF2 as a suitable diagnostic and therapeutic target in cancer patients.
Preprint
Full-text available
The extracellular matrix (ECM) supports blood vessel architecture and functionality and undergoes active remodelling during vascular repair and atherogenesis. Vascular smooth muscle cells (VSMCs) are essential for vessel repair and, via their secretome, are able to invade from the vessel media into the intima to mediate ECM remodelling. Accumulation of fibronectin (FN) is a hallmark of early vascular repair and atherosclerosis and here we show that FN stimulates VSMCs to secrete small extracellular vesicles (sEVs) by activating the β1 integrin/FAK/Src pathway as well as Arp2/3-dependent branching of the actin cytoskeleton. Spatially, sEV were secreted via filopodia-like cellular protrusions at the leading edge of migrating cells. We found that sEVs are trapped by the ECM in vitro and colocalise with FN in symptomatic atherosclerotic plaques in vivo. Functionally, ECM-trapped sEVs induced the formation of focal adhesions (FA) with enhanced pulling forces at the cellular periphery. Proteomic and GO pathway analysis revealed that VSMC-derived sEVs display a cell adhesion signature and are specifically enriched with collagen VI. In vitro assays identified collagen VI as playing the key role in cell adhesion and invasion. Taken together our data suggests that the accumulation of FN is a key early event in vessel repair acting to promote secretion of collage VI enriched sEVs by VSMCs. These sEVs stimulate migration and invasion by triggering peripheral focal adhesion formation and actomyosin contraction to exert sufficient traction forces to enable VSMC movement within the complex vascular ECM network. Vascular smooth muscle cells sense fibronectin via β1 integrin and secrete small extracellular vesicles loaded with collagen VI via filopodia-like protrusions. These extracellular vesicles are entrapped in the extracellular matrix and induce formation of peripheral focal adhesions. Focal adhesions anchor extracellular matrix to the actin fibrils in the cell. Contraction of the actin fibrils generates the mechanical force for cell locomotion and invasion through the matrix. This figure was created with BioRender(https://biorender.com/).
Preprint
Full-text available
The extracellular matrix (ECM) supports blood vessel architecture and functionality and undergoes active remodelling during vascular repair and atherogenesis. Vascular smooth muscle cells (VSMCs) are essential for vessel repair and, via their secretome, are able to invade from the vessel media into the intima to mediate ECM remodelling. Accumulation of fibronectin (FN) is a hallmark of early vascular repair and atherosclerosis and here we show that FN stimulates VSMCs to secrete small extracellular vesicles (sEVs) by activating the β1 integrin/FAK/Src pathway as well as Arp2/3-dependent branching of the actin cytoskeleton. Spatially, sEV were secreted via filopodia-like cellular protrusions at the leading edge of migrating cells. We found that sEVs are trapped by the ECM in vitro and colocalise with FN in symptomatic atherosclerotic plaques in vivo . Functionally, ECM-trapped sEVs induced the formation of focal adhesions (FA) with enhanced pulling forces at the cellular periphery. Proteomic and GO pathway analysis revealed that VSMC-derived sEVs display a cell adhesion signature and are specifically enriched with collagen VI. In vitro assays identified collagen VI as playing the key role in cell adhesion and invasion. Taken together our data suggests that the accumulation of FN is a key early event in vessel repair acting to promote secretion of collage VI enriched sEVs by VSMCs. These sEVs stimulate migration and invasion by triggering peripheral focal adhesion formation and actomyosin contraction to exert sufficient traction forces to enable VSMC movement within the complex vascular ECM network.
Article
Full-text available
Cellular asymmetry is critical for the development of multicellular organisms. Here we show that homologues of proteins necessary for asymmetric cell division in Caenorhabditis elegans associate with each other in mammalian cells and tissues. mPAR-3 and mPAR-6 exhibit similar expression patterns and subcellular distributions in the CNS and associate through their PDZ (PSD-95/Dlg/ZO-1) domains. mPAR-6 binds to Cdc42/Rac1 GTPases, and mPAR-3 and mPAR-6 bind independently to atypical protein kinase C (aPKC) isoforms. In vitro, mPAR-3 acts as a substrate and an inhibitor of aPKC. We conclude that mPAR-3 and mPAR-6 have a scaffolding function, coordinating the activities of several signalling proteins that are implicated in mammalian cell polarity.
Article
Full-text available
The evolutionarily conserved proteins Par-6, atypical protein kinase C (aPKC), Cdc42 and Par-3 associate to regulate cell polarity and asymmetric cell division, but the downstream targets of this complex are largely unknown. Here we identify direct physiological interactions between mammalian aPKC, murine Par-6C (mPar-6C) and Mlgl, the mammalian orthologue of the Drosophila melanogaster tumour suppressor Lethal (2) giant larvae. In cultured cell lines and in mouse brain, aPKC, mPar-6C and Mlgl form a multiprotein complex in which Mlgl is targeted for phosphorylation on conserved serine residues. These phosphorylation sites are important for embryonic fibroblasts to polarize correctly in response to wounding and may regulate the ability of Mlgl to direct protein trafficking. Our data provide a direct physical and regulatory link between proteins of distinct polarity complexes, identify Mlgl as a functional substrate for aPKC in cell polarization and indicate that aPKC is directed to cell polarity substrates through a network of protein–protein interactions.
Article
Full-text available
We have previously shown that during early Caenorhabditis elegans embryogenesis PKC-3, a C. elegans atypical PKC (aPKC), plays critical roles in the establishment of cell polarity required for subsequent asymmetric cleavage by interacting with PAR-3 [Tabuse, Y., Y. Izumi, F. Piano, K.J. Kemphues, J. Miwa, and S. Ohno. 1998. Development (Camb.). 125:3607–3614]. Together with the fact that aPKC and a mammalian PAR-3 homologue, aPKC-specific interacting protein (ASIP), colocalize at the tight junctions of polarized epithelial cells (Izumi, Y., H. Hirose, Y. Tamai, S.-I. Hirai, Y. Nagashima, T. Fujimoto, Y. Tabuse, K.J. Kemphues, and S. Ohno. 1998. J. Cell Biol. 143:95–106), this suggests a ubiquitous role for aPKC in establishing cell polarity in multicellular organisms. Here, we show that the overexpression of a dominant-negative mutant of aPKC (aPKCkn) in MDCK II cells causes mislocalization of ASIP/PAR-3. Immunocytochemical analyses, as well as measurements of paracellular diffusion of ions or nonionic solutes, demonstrate that the biogenesis of the tight junction structure itself is severely affected in aPKCkn-expressing cells. Furthermore, these cells show increased interdomain diffusion of fluorescent lipid and disruption of the polarized distribution of Na+,K+-ATPase, suggesting that epithelial cell surface polarity is severely impaired in these cells. On the other hand, we also found that aPKC associates not only with ASIP/PAR-3, but also with a mammalian homologue of C. elegans PAR-6 (mPAR-6), and thereby mediates the formation of an aPKC-ASIP/PAR-3–PAR-6 ternary complex that localizes to the apical junctional region of MDCK cells. These results indicate that aPKC is involved in the evolutionarily conserved PAR protein complex, and plays critical roles in the development of the junctional structures and apico-basal polarization of mammalian epithelial cells.
Article
Full-text available
We describe a method that can be used to produce equimolar amounts of two or more specific proteins in a cell. In this approach, termed the ubiquitin/protein/reference (UPR) technique, a reference protein and a protein of interest are synthesized as a polyprotein separated by a ubiquitin moiety. This tripartite fusion is cleaved, cotranslationally or nearly so, by ubiquitin-specific processing proteases after the last residue of ubiquitin, producing equimolar amounts of the protein of interest and the reference protein bearing a C-terminal ubiquitin moiety. In applications such as pulse-chase analysis, the UPR technique can compensate for the scatter of immunoprecipitation yields, sample volumes, and other sources of sample-to-sample variation. In particular, this method allows a direct comparison of proteins' metabolic stabilities from the pulse data alone. We used UPR to examine the N-end rule (a relation between the in vivo half-life of a protein and the identity of its N-terminal residue) in L cells, a mouse cell line. The increased accuracy afforded by the UPR technique underscores insufficiency of the current "half-life" terminology, because in vivo degradation of many proteins deviates from first-order kinetics. We consider this problem and discuss other applications of UPR.
Article
Full-text available
Asymmetric cell divisions, critically important to specify cell types in the development of multicellular organisms, require polarized distribution of cytoplasmic components and the proper alignment of the mitotic apparatus. In Caenorhabditis elegans, the maternally expressed protein, PAR-3, is localized to one pole of asymmetrically dividing blastomeres and is required for these asymmetric divisions. In this paper, we report that an atypical protein kinase C (PKC-3) is essential for proper asymmetric cell divisions and co-localizes with PAR-3. Embryos depleted of PKC-3 by RNA interference die showing Par-like phenotypes including defects in early asymmetric divisions and mislocalized germline-specific granules (P granules). The defective phenotypes of PKC-3-depleted embryos are similar to those exhibited by mutants for par-3 and another par gene, par-6. Direct interaction of PKC-3 with PAR-3 is shown by in vitro binding analysis. This result is reinforced by the observation that PKC-3 and PAR-3 co-localize in vivo. Furthermore, PKC-3 and PAR-3 show mutual dependence on each other and on three of the other par genes for their localization. We conclude that PKC-3 plays an indispensable role in establishing embryonic polarity through interaction with PAR-3.
Article
Full-text available
Cell movement is essential during embryogenesis to establish tissue patterns and to drive morphogenetic pathways and in the adult for tissue repair and to direct cells to sites of infection. Animal cells move by crawling and the driving force is derived primarily from the coordinated assembly and disassembly of actin filaments. The small GTPases, Rho, Rac, and Cdc42, regulate the organization of actin filaments and we have analyzed their contributions to the movement of primary embryo fibroblasts in an in vitro wound healing assay. Rac is essential for the protrusion of lamellipodia and for forward movement. Cdc42 is required to maintain cell polarity, which includes the localization of lamellipodial activity to the leading edge and the reorientation of the Golgi apparatus in the direction of movement. Rho is required to maintain cell adhesion during movement, but stress fibers and focal adhesions are not required. Finally, Ras regulates focal adhesion and stress fiber turnover and this is essential for cell movement. We conclude that the signal transduction pathways controlled by the four small GTPases, Rho, Rac, Cdc42, and Ras, cooperate to promote cell movement.
Article
The Rho family members Cdc42, Rac, and Rho play a central role in the organization of the actin cytoskeleton and regulate transcription. Whereas Rac and Rho have been implicated in transformation by oncogenic Ras, the role of Cdc42 in this process remains unknown. In this study, we found that Rat1 fibroblasts expressing constitutively active V12-Cdc42 were anchorage independent and proliferated in nude mice but failed to show enhanced growth in low serum. Similar to V12-Rac1-expressing Rat1 fibroblasts, V12-Cdc42 lines displayed a high frequency of multinucleated cells. Interestingly, coexpression of dominant negative N17-Rac1 blocked the V12-Cdc42-induced multinucleated phenotype but not growth in soft agar, indicating that Cdc42 controls anchorage independence in a Rac-independent fashion. We also showed that dominant negative N17-Cdc42 inhibited Ras focus formation and anchorage-independent growth and caused reversion of the transformed morphology, indicating that Cdc42 is necessary for Ras transformation. N17-Cdc42 caused only partial inhibition of Ras-induced low-serum growth, however. In contrast, whereas N17-Rac1 also effectively inhibited Ras-induced anchorage independence, it did not revert the morphology of Ras-transformed cells. N17-Rac1 strongly inhibited low-serum growth of Ras-transformed cells, however. Together, these data provide a novel function for Cdc42 in cell proliferation and indicate that Cdc42 and Rac play distinct roles in growth control and Ras transformation.
Article
The selective degradation of many short-lived proteins in eukaryotic cells is carried out by the ubiquitin system. In this pathway, proteins are targeted for degradation by covalent ligation to ubiquitin, a highly conserved small protein. Ubiquitin-mediated degradation of regulatory proteins plays important roles in the control of numerous processes, including cell-cycle progression, signal transduction, transcriptional regulation, receptor down-regulation, and endocytosis. The ubiquitin system has been implicated in the immune response, development, and programmed cell death. Abnormalities in ubiquitin-mediated processes have been shown to cause pathological conditions, including malignant transformation. In this review we discuss recent information on functions and mechanisms of the ubiquitin system. Since the selectivity of protein degradation is determined mainly at the stage of ligation to ubiquitin, special attention is focused on what we know, and would like to know, about the mode of action of ubiquitin-protein ligation systems and about signals in proteins recognized by these systems.
Article
The par genes are required to establish polarity in the Caenorhabditis elegans embryo. Mutations in two of these genes, par-3 and par-6, exhibit similar phenotypes. A third gene, pkc-3, gives a similar phenotype when the protein is depleted by RNA interference. PAR-3 and PKC-3 protein are colocalized to the anterior periphery of asymmetrically dividing cells of the germline lineage and the peripheral localizations of both proteins depends upon the activity of par-6. Here we report the molecular cloning of par-6 and the immunolocalization of PAR-6 protein. We found that par-6 encodes a PDZ-domain-containing protein and has homologues in mammals and flies. Moreover, we discovered that PAR-6 colocalizes with PAR-3 and that par-3 and pkc-3 activity are required for the peripheral localization of PAR-6. The localization of both PAR-3 and PAR-6 proteins is affected identically by mutations in the par-2, par-4 and par-5 genes. The co-dependence of PAR-3, PAR-6 and PKC-3 for peripheral localization and the overlap in their distributions lead us to propose that they act in a protein complex.
Article
The members of an emerging family of proteins similar to Nedd4 have a unique modular structure consisting of a Ca2+/lipid-binding domain, multiple protein-protein interaction modules and a ubiquitin-protein ligase domain. Although little is known about the physiological roles of these proteins, studies in both mammals and yeast are providing evidence that members of this family might be involved in diverse cellular functions, such as regulation of membrane channels and permeases, the cell cycle and transcription. This article attempts to bring together what is currently known about these evolutionarily conserved ubiquitin-protein ligases.