You are on page 1of 13

Developmental Cell, Vol.

2, 1–20, May, 2002, Copyright 2002 by Cell Press

Modulation of CREB Activity by the Rho GTPase


Regulates Cell and Organism Size
during Mouse Embryonic Development
Raffaella Sordella,1,4 Marie Classon,1,4 GTPases elicit their biological actions through direct
Kang-Quan Hu,1 Stephen F. Matheson,1 interactions with a variety of so-called effector targets.
Madeleine R. Brouns,1 Barry Fine,1 Le Zhang,1 The biological function of the RhoGAPs is somewhat
Hiroya Takami,2 Yoshihiko Yamada,2 less clear. These negative regulators of Rho GTPases
and Jeffrey Settleman1,3 have been postulated to play a role in precisely modulat-
1
MGH Cancer Center and ing Rho-mediated signaling in vivo, and there are several
Harvard Medical School reports indicating that these proteins, when overex-
Charlestown, Massachusetts 02129 pressed, can inhibit GTPase-mediated signaling path-
2
National Institute of Dental ways (Symons and Settleman, 2000). However, there
and Craniofacial Research has been relatively little analysis of these proteins in a
National Institutes of Health truly in vivo context. Among the various RhoGAPs that
Bethesda, Maryland 20892 have been identified is a family of widely expressed
potent Rho-specific GAPs, referred to as the p190 Rho-
GAPs. These GAPs are unusual in that they contain
an amino-terminal GTPase domain in addition to their
Summary
carboxy-terminal RhoGAP catalytic domain (Burbelo et
al., 1995; Settleman et al., 1992). In mammals, there are
Rho GTPases regulate several aspects of tissue mor-
two p190 RhoGAPs, referred to as p190 RhoGAP and
phogenesis during animal development. We found that
p190-B RhoGAP. Closely related orthologs of these pro-
mice lacking the Rho-inhibitory protein, p190-B Rho-
teins are present in Drosophila and C. elegans as well.
GAP, are 30% reduced in size and exhibit develop-
Recently, RNA interference studies have revealed a role
mental defects strikingly similar to those seen in mice
for the Drosophila p190 RhoGAP in neurite retraction in
lacking the CREB transcription factor. In p190-B Rho-
the developing nervous system (Billuart et al., 2001),
GAP-deficient mice, CREB phosphorylation is sub-
although a loss-of-function mutant of this gene has yet
stantially reduced in embryonic tissues. Embryo-
to be reported. As Rho activation has previously been
derived cells contain abnormally high levels of active
demonstrated to promote neurite retraction in cultured
Rho protein, are reduced in size, and exhibit defects in
neurons (Jalink et al., 1994), this result suggests that
CREB activation upon exposure to insulin or IGF-1.
loss of p190 RhoGAP activity in vivo leads to deregu-
The cell size defect is rescued by expression of consti-
lated Rho GTPase function. Disruption of the p190 Rho-
tutively activated CREB, and in wild-type cells, expres-
GAP gene in mice by gene targeting revealed that it is
sion of activated Rho or dominant-negative CREB re-
an essential protein that mediates an adhesion signaling
sults in reduced cell size. Together, these results
pathway downstream of Src kinases that functions in
suggest that activity of the Rho GTPase modulates a
neural development (Brouns et al., 2000, 2001).
signal from insulin/IGFs to CREB that determines cell
Although the biological function of p190-B RhoGAP
size and animal size during embryogenesis.
is not clear, it has been reported that this RhoGAP ac-
counts for the majority of Rho-inhibitory activity in cul-
Introduction tured fibroblasts, suggesting that it is a major down-
modulator of Rho GTPase activity in vivo (Vincent and
The Rho GTPases mediate signaling pathways that reg- Settleman, 1999). To determine the biological require-
ulate a variety of cellular activities, including cytoskeletal ment for p190-B RhoGAP, we have disrupted the gene
reorganization, gene expression, and cell cycle progres- in mice. Surprisingly, we found that these mice exhibit
sion (Van Aelst and D’Souza-Schorey, 1997). They are phenotypes that are remarkably similar to those seen
also essential regulators of tissue morphogenesis during in mice lacking the CREB transcription factor, including
embryonic development (Settleman, 1999). Like all a substantial reduction in animal size (Rudolph et al.,
GTPases, the Rho proteins cycle between active, GTP- 1998). Additional studies revealed that cell size is re-
bound and inactive, GDP-bound states. This cycling is duced in these animals and that Rho and CREB are
largely regulated by two classes of proteins, the guanine important novel regulators of cell size in a developing
nucleotide exchange factors (GEFs) and the GTPase mouse. The results also suggest a cytoskeleton-inde-
activating proteins (GAPs; Symons and Settleman, pendent role for Rho GTPase signaling in embryonic
2000). The RhoGEFs function to promote the conversion development.
of inactive Rho to active Rho, while the RhoGAPs stimu-
late the intrinsic GTP hydrolysis activity of Rho proteins, Results
thereby converting them to a GDP-bound, inactive form.
Accumulating evidence suggests that the RhoGEFs are Mice Lacking p190-B RhoGAP Closely Resemble
rapidly activated upon ligand-receptor interactions at Mice Lacking CREB
the cell surface, and that the subsequently activated To explore p190-B RhoGAP function in vivo, we dis-
rupted the mouse gene using homologous recombina-
3
Correspondence: settleman@helix.mgh.harvard.edu tion in embryonic stem (ES) cells (Figure 1A). Two lines
4
These authors contributed equally to this work. of properly targeted ES cells were used to establish
Developmental Cell
2

Figure 1. Mice Lacking p190-B RhoGAP Resemble CREB-Deficient Mice


(A) Targeting strategy for disruption of the p190-B RhoGAP gene. Schematic representations of the 5⬘ genomic region of the p190-B RhoGAP
gene (upper line), the targeting vector (middle line), and the mutated allele (lower line) (gray bar depicts exon I). Bar under the upper line
represents the probe used for the identification of heterozygous ES lines. S, StuI; H, HindIII; B, BamHI; N, NotI; NEO, neomycine resistance
gene; TK, thymidine kinase gene.
(B) An immunoblot of embryonic tissue from wild-type (⫹/⫹), heterozygous mutant (⫹/⫺), and homozygous mutant (⫺/⫺) tissue probed with
antisera that specifically recognize either p190-B RhoGAP (top panel) or p190 RhoGAP (bottom panel).
Rho and CREB Regulate Embryonic Cell Size
3

germline transmission of the targeted allele in mice, and significantly increased in the mutants (Figure 2A). For-
the resulting homozygous mutant mice fail to express ward light scatter FACS analysis, which indirectly mea-
any detectable p190-B RhoGAP (Figure 1B). Homozy- sures changes in cell volume, revealed that this differ-
gous mutant mice die immediately after birth, and, ence reflects an approximately 30% reduction in cell
grossly, they always exhibit a substantial and uniform size in the mutants (Figures 2B and 5D). Thus, they pack
reduction in size (30% reduction in mass) relative to more densely in a monolayer. This result was indepen-
their littermates (more than 100 examined; Figure 1C). dently confirmed by comparing protein:cell number and
Histological analysis revealed several completely pene- protein:DNA ratios between wild-type and mutant cells
trant (30/30 examined) defects in the mutant mice. At (Figures 2C and 2D), as well as by morphometric analysis
birth, they exhibit a severe reduction in thymus size (not shown). Changes in cell size observed by these first
(disproportionate to the reduction in body size; Figure three criteria would not be influenced by changes in the
1D), their lungs fail to inflate properly (not shown), and cytoskeleton, indicating that the reduced cell size in
in brain, they exhibit axon defects that include a severe mutants is not due to Rho-induced effects on actin as-
reduction in the major midline forebrain crossing tracts sembly.
(corpus callosum and anterior commissure), as well as The small size of mutant animals could potentially
a thinner cortex, associated with partial loss of cortical reflect this cell size phenotype seen in 3T3 lines, or
axon tracts (Figures 1E and 1F). The lateral ventricles it could be due to either reduced cell proliferation or
in brain are substantially enlarged in mutants (Figures increased apoptosis in developing tissues. Indeed,
1E and 1F). Aside from a small decrease in overall size, CREB has previously been implicated as a cell survival
all other major organs appear to be unaffected in the factor (Walton and Dragunow, 2000). However, in mutant
mutants. embryonic tissues (E12–E18) analyzed at several levels
The observed spectrum of defects in mutant animals of sectioning, we were not able to detect any significant
strikingly resembles defects previously reported in mice increase in apoptosis by TUNEL staining (data not
lacking the CREB transcription factor (Rudolph et al., shown). Moreover, no evidence of reduced cell prolifera-
1998). CREB-deficient mice also die immediately after tion was seen by BrdU incorporation into embryonic
birth, are 30% reduced in size, fail to inflate their lungs, tissues (Figures 2E and 2F). Then, by comparing pro-
have a small thymus, and exhibit brain defects that affect tein:DNA ratios, we found that in most tissues there is
the corpus callosum, anterior commissure, and lateral a significant reduction in cell size in animals lacking
ventricles (Rudolph et al., 1998). CREB, like its closely p190-B RhoGAP (Figure 2G). Together with the observa-
related family members CREM and the ATFs, undergoes tion that mutant 3T3 cells are reduced in size, these
phosphorylation of serine 133 when activated (Shaywitz results suggest that the small size of the mutant mice
and Greenberg, 1999). Significantly, we determined that reflects a cell-autonomous defect in cell size regulation.
while CREB protein levels are unaffected in mutant tis- Significantly, p190-B RhoGAP is expressed in all tissues
sues, phospho-CREB and phospho-ATF-1 are virtually during embryonic development (Figure 2H).
absent in mutant brain, are substantially reduced in thy- To determine whether the cell size defect that results
mus, and are also reduced in stomach and heart (Fig- from loss of p190-B RhoGAP is due to impaired CREB
ure 1G). activation, we generated p190-B RhoGAP-deficient 3T3
lines that stably express a constitutively activated form
of CREB (CREBFY; Du et al., 2000). These cells are
Loss of p190-B RhoGAP Causes a Cell Size significantly larger than the parental p190-B RhoGAP-
Reduction that Is Rescued by Activated CREB deficient lines, indicating that CREB activation is suffi-
To determine the role of p190-B RhoGAP in CREB regu- cient to rescue the cell size defect in mutants, and that
lation, we established stable fibroblast lines from wild- CREB is probably functioning downstream of p190-B
type and mutant embryos. All results were confirmed RhoGAP (Figures 3A–3C). In addition, we determined
with three independently derived litter-matched wild- that CREBFY rescues the cell size defect in both G1-
type and mutant pairs of spontaneously immortalized and G2-selected cell populations, indicating a cell cycle-
fibroblast (3T3) lines. Morphologically, wild-type and independent role for CREB in cell size regulation (Fig-
mutant cells appear similar, although mutant cells, as ures 3D–3G).
would be expected as a consequence of losing a Rho-
inhibitory protein, contain significantly more polymer-
ized actin (data not shown). By comparing the growth Rho and CREB Activity Modulate Cell Size
properties of these cells, we determined that while they To address the mechanism by which loss of p190-B
exhibit equivalent doubling times, saturation density is RhoGAP affects CREB activation, we considered the

(C) Newborn wild-type (⫹/⫹) and homozygous mutant (⫺/⫺) animals illustrating the size defect seen in animals lacking p190-B RhoGAP.
(D) Thymuses dissected from newborn wild-type (⫹/⫹) and homozygous mutant (⫺/⫺) animals revealing the substantial reduction in the size
of mutant thymus.
(E and F) Coronal forebrain sections from wild-type (E) and mutant (F) embryos at E18.5 stained with Nissl reveals the reduction in the thickness
of the corpus callosum (black arrows) in the mutant as well as the enlargement of lateral ventricles in these animals. Note that the thickness
of the cortex is also reduced in the mutants, and this appears to reflect a reduction of the major forebrain cortical axon tracts (white arrows).
(G) Immunoblots of various tissues from wild-type (⫹/⫹) and mutant (⫺/⫺) E18.5 animals with antibodies that specifically recognize the
phosphorylated form of CREB and ATF-1 (upper panels) or total CREB protein (lower panels).
Developmental Cell
4

Figure 2. Cell Size Is Reduced in Fibroblasts and Tissues from Mice Lacking p190-B RhoGAP
(A) Proliferation curve of wild-type (⫹/⫹) and p190-B RhoGAP mutant (⫺/⫺) 3T3 cells, indicating similar proliferative rates in the two populations
and increased saturation density of the mutant cells.
(B) Cell size distribution of wild-type (⫹/⫹) and p190-B RhoGAP mutant (⫺/⫺) 3T3 cells determined by forward light scatter FACS analysis,
indicating the mean reduction in cell size in a population of G1-selected (2N content) mutant cells.
(C) Ratio of total protein to cell number in a population of confluent wild-type (⫹/⫹) and p190-B RhoGAP mutant (⫺/⫺) cells confirming the
reduced cell size of mutant cells.
(D) Ratio of total protein to total DNA content in a population of confluent wild-type (⫹/⫹) and p190-B RhoGAP mutant cells, again confirming
the reduced cell size of mutant cells. Values in (C) and (D) reflect the average of three independent experiments, and error bars represent
standard error of the mean.
(E and F) BrdU incorporation of intestinal sections of wild-type (E) and p190-B RhoGAP mutant (F) E18.5 embryos (following 4 hr intrauterine
BrdU injection), indicating approximately equivalent levels of cell proliferation. Red, BrdU positivity by anti-BrdU antibody fluorescence; blue,
DAPI staining.
(G) Ratio of total protein to total DNA content in various tissues derived from E18.5 wild-type (⫹/⫹) and p190-B RhoGAP mutant (⫺/⫺) animals,
indicating that tissues from mutant embryos contain abnormally small cells. Values reflect the average of tissue measurements taken from
seven different mice, and error bars represent the standard error of the mean.
(H) Immunoblot of p190-B RhoGAP with the indicated tissues dissected from an E18.5 wild-type embryo.
Rho and CREB Regulate Embryonic Cell Size
5

Figure 3. Activated CREB Rescues the Size Defect in Cells Lacking p190-B RhoGAP
(A) Forward light scatter analysis of G1-selected p190-B RhoGAP mutant (⫺/⫺) cells and a pool of mutant cells stably transfected with an
expression vector expressing activated CREB (CREBFY), indicating that the reduced cell size of mutant cells can be rescued by activated
CREB.
(B) Immunoblot of protein lysates prepared from p190-B RhoGAP mutant 3T3 cells (left lane) and individual clonally derived cell lines of p190-
B RhoGAP mutant cells stably expressing transfected CREBFY (FLAG epitope-tagged; anti-FLAG blot).
(C) Relative cell size of G1-selected wild-type cells (⫹/⫹), p190-B RhoGAP mutant cells (⫺/⫺), and mutant clonal cell lines stably expressing
CREBFY (FY1, FY2, and FY3), indicating the rescue of reduced cell size by CREBFY.
(D and F) FACS gating of G1-selected (D) and G2-selected (F) populations of wild-type 3T3 cells.
(E and G) Forward light scatter profile of wild-type, mutant, and CREBFY-rescued mutant 3T3 cells in both G1 (E) and G2 (G). Note that
CREBFY restores cell size in both G1 and G2 populations.

known biochemical action of p190-B RhoGAP as a dow- active CREB are in fact reduced in cells lacking p190-
nmodulator of Rho. As would be expected, asynchro- B RhoGAP, we used a CRE-luciferase reporter to deter-
nously growing mutant cells were found to contain ab- mine that in mutant cells, CRE reporter activity is re-
normally high levels of activated, GTP-bound Rho. duced by 6-fold relative to wild-type cells (Figure 4B).
Moreover, these levels are maintained in the CREB “res- Moreover, introduction of constitutively activated Rho
cue” lines, consistent with a role for CREB downstream (RhoV14) into wild-type cells results in a similar degree of
of Rho (Figure 4A). To verify that levels of biologically decreased CRE reporter activity (Figure 4B). Analogous
Developmental Cell
6

Figure 4. Rho Antagonizes CREB-Mediated Transcriptional Activity


(A) Anti-Rho immunoblot of total cell lysates (lys.) and GST-Rhotekin “pull-downs” (GTP-Rho) from wild-type (⫹/⫹), p190-B RhoGAP mutant
(⫺/⫺), and CREBFY-transfected mutant cells, indicating that levels of activated Rho are substantially increased in the mutant cells and that
the proportion of Rho in the active, GTP-bound state is similarly increased in the CREBFY-transfected cells.
(B) CRE-mediated transcriptional activity (CRE-luciferase reporter plasmid) in transiently transfected wild-type (⫹/⫹) and p190-B RhoGAP
mutant (⫺/⫺) 3T3 cells, and in mutant cells cotransfected with activated Rho (RhoV14). The results indicate that the transcriptional activity
of CREB is reduced by approximately 6-fold in mutant cells and is similarly reduced in wild-type cells transfected with RhoV14.
(C) GAL4-CREB activation of a UAS-luciferase reporter confirming the requirement for ser133 phosphorylation and demonstrating the specific
inhibitory effect of Rho on wild-type GAL4-CREB.
(D) Immunoblot of cytochrome c in wild-type, mutant, and two CREBFY-rescued mutant 3T3 lines, indicating reduced expression in mutant
cells that is restored in the CREBFY lines. Normalization is with an anti-actin blot.
(E) Immunoblot of cytochrome c in wild-type and mutant brain (Br.), stomach (St.), and heart (Hrt.), indicating reduced expression in mutant
tissues (from E18.5). Normalization is with an anti-actin blot.
Rho and CREB Regulate Embryonic Cell Size
7

experiments with an E2F-luciferase reporter as a control activation, and that this pathway can be specifically
revealed similar levels of transcriptional activity in wild- downmodulated by increased levels of Rho GTPase ac-
type and mutant cells (data not shown). To address tivity.
specifically the ability of Rho to regulate CREB transcrip- One of the major downstream targets of insulin/IGF
tional activity via phosphorylation of ser133, we used a receptor activation is the insulin receptor substrate IRS,
GAL4-CREB reporter assay. In those experiments, Rho which plays a role in cell size regulation in Drosophila
antagonizes transcriptional activity of wild-type GAL4- (Bohni et al., 1999). Interestingly, one of the major Rho
CREB, but does not affect the residual activity of GAL4- targets, Rho-kinase (ROK; Leung et al., 1995), can re-
CREB containing a ser133 substitution (Figure 4C). We portedly antagonize insulin signaling through an associ-
also determined that expression of one of the well-docu- ation with the major insulin receptor substrate IRS (Be-
mented CREB-regulated genes, cytochrome c (Herzig gum et al., 2001; Farah et al., 1998) and an inhibitory
et al., 2000), is reduced in mutant 3T3s and embryonic phosphorylation (Paz et al., 1997). Therefore, we hypoth-
tissues, and is restored in mutant 3T3s that express esized that increased ROK activity in mutant cells could
activated CREB (Figures 4D and 4E). Together, these account for the observed defects. Indeed, we found that
observations indicate that at least some of the conse- when mutant cells are exposed to a specific pharma-
quences of hyperactivation of Rho in cells lacking p190- cological inhibitor of ROK (Y-27632), levels of IGF-1-
B RhoGAP are likely to be mediated through specific induced CREB phosphorylation are restored to levels
inhibitory effects on CREB-dependent transcriptional seen in wild-type cells (Figure 5C). In addition, when
activity. mutant cells are exposed to Y-27632, cell size is in-
These results implicate both Rho and CREB in cell creased nearly to wild-type size (Figure 5D). Thus, ROK
size regulation. However, in the context of the p190-B inhibition can “rescue” defects in cell size and IGF-1
RhoGAP mutant cells, it is difficult to make this conclu- responsiveness in mutant cells. In addition, exposure
sion definitively because it requires the assumption that of Rat-1 cells to Y-27632, in the absence of other treat-
increased Rho activity is the only significant conse- ments, is sufficient to promote increased cell size (data
quence of losing p190-B RhoGAP. Therefore, to analyze not shown). This suggests that ROK is an important
more directly the role of Rho and CREB in cell size Rho target in regulating cell size, and that the observed
regulation, we examined transiently transfected Rat-1 growth inhibition seen upon Rho activation is not simply
fibroblasts. We observed by FACS analysis that cells a consequence of cell toxicity caused by active Rho,
transfected with RhoV14 are significantly smaller than but rather that this reflects a physiological consequence
nontransfected cells, while transfection with CREBFY of modulating Rho activity.
results in cells that are abnormally large (Figure 5A). In To further establish the role of ROK and IRS in a cell
addition, transfection of wild-type cells with a dominant- size pathway, we examined the IRS status in primary
negative form of CREB (Walton et al., 1992) gives rise MEFs lacking p190-B RhoGAP. After IGF-1 treatment,
to cells that are reduced in size (Figure 5A). As before, levels of tyrosine phosphorylation of the IGF-1 receptor
these effects appear to be independent of potential ef- are normal, as expected; however, in mutant cells, there
fects on cell cycle because the analysis was restricted is a significant decrease in tyrosine phosphorylation of
to cells in G1. These results, which suggest that Rho IRS-1, suggesting that IRS-1 does not interact as effi-
activation inhibits cells growth and that CREB activation ciently with the activated IGF receptor in these cells (Fig-
promotes cell growth, are consistent with the observa- ure 5E). Next, we tested the hypothesis that this reduction
tion that cell size and CREB phosphorylation are both
is associated with an increase in the “inhibitory” phosphor-
reduced in mice lacking p190-B RhoGAP.
ylation of IRS-1 (ser612) in mutant cells. This was found
to be the case (Figure 5F), and additionally, we found
Rho Regulates Insulin Signaling to CREB via
that treatment of mutant cells with Y-27632 “rescues” the
an Inhibitory Effect of Rho-Kinase on IRS
excessive levels of serine phosphorylation of IRS-1, con-
Previous studies in both vertebrate and invertebrate or-
sistent with the postulated role for ROK in antagonizing
ganisms revealed that cell size and animal size are regu-
insulin/IGF signaling through an inhibitory IRS phosphory-
lated by components of the insulin/IGF signaling path-
lation in mutant cells (Figure 5F).
way (Stocker and Hafen, 2000). Significantly, we found
that wild-type fibroblasts undergo a substantial induc-
tion of CREB phosphorylation following exposure to
IGF-1, and that this response is diminished in cells lack- Reduced MAP Kinase Signaling Links Rho-Mediated
ing p190-B RhoGAP (Figure 5B). Similar results were ob- Inhibition of Insulin Signaling to CREB
served with insulin (data not shown). We then determined We next tested the possibility that known downstream
that expression of RhoV14 (Sullivan and Finkel, 2000) in components of insulin/IGF signaling that could poten-
wild-type cells is sufficient to reduce levels of IGF-1- tially provide a link to CREB are affected in cells lacking
induced phospho-CREB (Figure 5B). Significantly, levels p190-B RhoGAP. For these studies, we compared pri-
of IGF-1-induced phospho-ERK and phospho-MEK1 are mary MEFs and embryonic tissues from wild-type and
not reduced in mutant cells, indicating that the observed mutant animals. In MEFs, cell size is also substantially
effect is specific (data not shown). In addition, we found reduced in the mutants (data not shown). In these pri-
that introduction of the activated Rac GTPase into wild- mary cell cultures, we confirmed the defect in CREB
type cells does not inhibit insulin-induced CREB phos- activation in response to IGF-1 in mutants, and addition-
phorylation, and in fact, causes an increase in CREB ally, we found that phosphorylation of several down-
phosphorylation (data not shown). Together, these re- stream pathway components, including two kinases that
sults indicate that insulin/IGF signaling promotes CREB can promote CREB activation (JNK, not shown and p38;
Developmental Cell
8

Figure 5. A Rho-ROK (Rho-Kinase) Pathway Modulates Cell Size and IGF-Induced CREB Phosphorylation
(A) Relative cell size of G1-selected, GFP-positive Rat-1 fibroblasts transiently transfected with either RhoV14, CREBFY, or CREB-DN, indicating
that expression of activated Rho or dominant-negative CREB results in reduced cell size, and expression of activated CREB promotes
increased cell size.
(B) IGF-1-induced phospho-CREB (after 15 min) as revealed by immunoblotting with a phospho-CREB antibody (upper panel) of protein lysates
prepared from wild-type (⫹/⫹) and p190-B RhoGAP mutant (⫺/⫺) 3T3 cells, and wild-type cells infected with an adenovirus that expresses
RhoV14 (48 hr before IGF-1 treatment). RhoV14 expression blocks approximately 50% of the IGF-induced CREB phosphorylation seen in
wild-type cells. All non-RhoV14-expressing cells were infected with a GFP-expressing adenovirus as a negative control. Middle panel represents
an immunoblot of the same lysates probed with anti-CREB antibody as a normalization control. Lower panel represents an immunoblot of
Rho protein, indicating the size-shifted band corresponding to RhoV14 in the right-most lane.
(C) IGF-induced CREB phosphorylation (after 15 min) similar to that shown in (B), and rescue of IGF-induced CREB phosphorylation in mutant
cells by pretreatment with the Rho-kinase inhibitor Y-27632.
(D) Relative cell size (by forward light scatter) of G1-selected wild-type (⫹/⫹) and p190-B RhoGAP mutant (⫺/⫺) 3T3 cells, and mutant cells
treated for 48 hr with Y-27632, indicating the partial rescue of cell size in mutant cells by inhibition of Rho-kinase. Values in (A) and (D) reflect
the average of three independent experiments, and error bars represent standard error of the mean.
(E) Immunoprecipitation of IGF-1 receptor and IRS-1 from wild-type and mutant 3T3 cells before and after 15 min of IGF-1 treatment, followed
by immunoblot with anti-phosphotyrosine antibodies. Samples were also immunoblotted for total IGF-1 receptor and IRS-1 for normalization.
(F) Immunoprecipitation of IRS-1 from wild-type and mutant 3T3 cells before and after 15 min of IGF-1 treatment, followed by immunoblot with
phospho-IRS-1 (phospho-ser612) antibodies. Y-27632 was included where indicated. Immunoblot of total IRS-1 is included for normalization.

Shaywitz and Greenberg, 1999), as well as Akt, is sub- are consistent with a model wherein increased ROK
stantially reduced in mutant MEFs (Figure 6A). More- activity in mutant cells downmodulates insulin/IGF sig-
over, expression of RhoV14 in wild-type MEFs is suffi- naling to several downstream pathway components, in-
cient to inhibit IGF-1-induced phosphorylation of these cluding MAP kinases that can promote CREB activity
proteins, and treatment of mutant MEFs with Y-27632 (Figure 7A). Consistent with this model, we find that
can restore normal levels IGF-1-induced phosphoryla- levels of phosphorylated p38, JNK, and Akt are substan-
tion of these kinases (Figure 6A). Together, these results tially reduced in embryonic tissues (Figures 6B and 6C),
Rho and CREB Regulate Embryonic Cell Size
9

Figure 6. Downstream Components of the Insulin/IGF Signaling Pathway Are Affected in Mutant Primary MEFs and Tissues
(A) Immunoblots with the indicated antibodies of lysates from wild-type and mutant MEFs upon treatment with IGF-1 (15 min), Y-27632, and/
or adenovirus RhoV14 infection, as indicated.
(B) Immunoblots with the indicated antibodies of tissue lysates from wild-type and mutant embryos (E18.5).
(C) Immunoblot of phospho-Akt in the indicated wild-type and mutant tissues (E18.5).

and, as would be expected, there is a significant in- be uniformly reduced in size, while phospho-CREB lev-
crease in ser612 phosphorylation of IRS (data not els are not substantially reduced in every tissue, raises
shown). The expected reduction in levels of active Rac the possibility that there is additional complexity in-
GTPase in mutant cells was also confirmed (data not volved in determining organism size.
shown). It is possible, for example, that modulation of CREB
is a more significant determinant of cell size at earlier
Discussion stages of development in some tissues (we focused on
E18.5). In addition, there may be both cell-autonomous
Taken together, our observations indicate that activity and -nonautonomous mechanisms involved in vivo. In-
of the Rho GTPase and the CREB transcription factor deed, PI-3 kinase, which appears to play a role in the
are important determinates of cell and animal size during pathway proposed here, has been previously implicated
embryonic development, thereby defining a novel bio- in cell size regulation, and has been reported to have
logical function for both of these widely expressed regu- both cell-autonomous and -nonautonomous functions
latory proteins. In fibroblasts derived from mice lacking in C. elegans (Hsin and Kenyon, 1999). In addition, while
p190-B RhoGAP, it appears that Rho is performing a IRS plays a well-documented cell-autonomous role in
largely cell-autonomous role that can influence respon- regulating cell size in Drosophila (Bohni et al., 1999),
siveness to insulin/IGFs through activation of the Rho studies in mice reveal an additional cell-nonautonomous
target ROK. However, the fact that mutant fibroblasts role in controlling animal size (Withers, 2001). It is also
are not completely defective in insulin/IGF-1 respon- worth noting that both insulin and IGF-1 gene expression
siveness suggests that at least some aspect of insulin- have been found to be CREB responsive (Philippe and
promoted growth is Rho insensitive. Moreover, the ob- Missotten, 1990; Thomas et al., 1996), raising the possi-
servation that mice lacking p190-B RhoGAP appear to bility that impaired CREB activity can also contribute in
Developmental Cell
10

Figure 7. Model Depicting the Proposed


Role of p190-B RhoGAP in Modulating Insu-
lin/IGF-1 Signaling to CREB

a cell-nonautonomous manner to growth regulation by CREB-related protein provides some redundant func-
affecting levels of secreted and circulating insulin/IGF. tion for cell survival in this setting, or that there is a
Such findings point to a complex regulatory system for phosphorylation-independent function for CREB in pro-
the control of cell, organ, and animal size that involves moting survival. Interestingly, it has been reported that
both cell-autonomous and -nonautonomous mecha- while mutational loss of one of the CREB homologs in
nisms that may require an overlapping set of proteins. Drosophila, dCREB2, results in a lethal phenotype, the
In fact, there may be additional compensatory mecha- few “escapers” that survive to adulthood are substan-
nisms that serve to uniformly adjust the size of organs tially reduced in size (Belvin et al., 1999). This suggests
to accommodate changes that may be limited to a sub- that a role for CREB in determining animal size has been
set of organs in the mutant animals. evolutionarily conserved. It is also notable that CREB is
Rho GTPase signaling has not previously been directly a direct phosphorylation target of PKA (Colbran et al.,
implicated in cell size regulation. In fact, the vast majority 1992), which, as described above, regulates animal size
of biological functions associated with the Rho GTPase in Drosophila. Possibly, crosstalk between Rho- and
in vivo appear to reflect the ability of Rho proteins to CREB-mediated pathways occurs at multiple levels.
regulate the actin cytoskeleton (Settleman, 2000). How- The vast majority of studies of cell size regulation in
ever, there is indirect evidence to suggest a potential both vertebrates and invertebrates have focused on the
role for Rho in cell size regulation. The human tumor insulin/IGF cell size pathway, and accumulating evi-
suppressor TSC1 encodes a protein that reportedly pro- dence suggests that an important “endpoint” of this
motes Rho activation (Lamb et al., 2000) and which, pathway is the upregulation of components of the pro-
when disrupted in Drosophila, results in tissue out- tein translational machinery (Stocker and Hafen, 2000).
growth due to a substantial increase in cell size (Tapon However, there is also evidence that transcriptional reg-
et al., 2001). Significantly, overexpression of TSC1, like ulation plays a role in regulating cell size, and it appears
Rho activation, reportedly causes reduced cell growth
that one of the relevant transcription factors is Myc,
by inhibiting insulin signaling (Gao and Pan, 2001; Potter
whose expression can promote cell growth in a cell
et al., 2001). In addition, the PKA kinase, which, when
cycle-independent manner (Iritani and Eisenman, 1999).
disrupted in Drosophila, results in a small fly phenotype
Our results indicate that CREB also plays a significant
(Lane and Kalderon, 1993), can directly phosphorylate
role in regulating cell growth, presumably through its
Rho, resulting in Rho inactivation (Dong et al., 1998).
ability to regulate transcriptionally the expression of one
Both of these results are consistent with the role of Rho
or more target genes. Thus far, the relevant transcrip-
in cell size regulation that has been proposed here, and
tional target(s) have not been identified, although, as
significantly, this role for Rho appears to be independent
discussed, insulin and IGF-1 may be important CREB
of its ability to regulate the cytoskeleton.
Previous studies to address the in vivo function of targets in this context. Significantly, many of the identi-
CREB have highlighted its role in cellular differentiation fied CREB targets are regulators of metabolism (Mayr
and protection form apoptosis (survival), as well as in and Montminy, 2001), raising the possibility that CREB-
learning and memory (Mayr and Montminy, 2001). Con- induced cell growth is due to increased expression of
sistent with this, we also see evidence of differentiation several essential metabolic enzymes. Thus, these col-
defects in brain and thymus of mice lacking p190-B lective results suggest a model wherein cellular growth
RhoGAP (S.F.M., R.S., and J.S., unpublished observa- requires increased protein synthesis together with in-
tions). However, we do not see evidence of any signifi- creased metabolism. Significantly, CREBFY-rescued
cant increase in the level of apoptosis in our mutant mutant cells are resistant to the ability of the p70 S6
animals, despite extensive analysis of embryos at sev- kinase inhibitor rapamycin to cause cell size reduction,
eral developmental stages. Thus, it is possible that a suggesting that CREB may be additionally required
Rho and CREB Regulate Embryonic Cell Size
11

downstream of the translational machinery for cell size TCTTCACTTAGAACGG-3⬘), and primer 3, complementary to se-
regulation (M.C. and J.S., unpublished observations). quences at the 5⬘ end of the PGK-neo promoter (5⬘-dCGGTGGATGT
GGAATGTG-3⬘). Primers 1 and 2 were used to identify a 392 bp
Rho and CREB have also both been previously impli-
fragment of the wild-type allele, whereas primers 2 and 3 were used
cated in the differentiation of thymocytes and neurons to amplify a 521 bp fragment of the mutated allele. PCR products
(Henning et al., 1997; Luo, 2000), and, as described were generated after a 5 min hot start at 94⬚C by 30 cycles at 94⬚C
above, we found that the brain and thymus defects in (1 min), 60⬚C (1 min), and 72⬚C (1 min) using Taq polymerase in the
p190-B RhoGAP-deficient mice are associated with a presence of 1.25 mM MgCl2.
failure in cell differentiation, as revealed by immuno-
staining with specific markers of differentiation. Signifi- Immunodetection of Proteins
To detect p190 RhoGAP proteins, lysates from embryo-derived cells
cantly, in CREB-deficient mice, the small thymus is simi-
were prepared in ice-cold lysis buffer (50 mM HEPES [pH 7.4], 150
larly associated with an excess of undifferentiated cells mM NaCl, 1.5 mM MgCl2, 5 mM EGTA, 10% glycerol, 1% Triton
(Rudolph et al., 1998). Thus, our results are consistent X-100) containing protease inhibitors. Debris was removed by cen-
with a role for Rho in regulating CREB function both for trifugation in a microfuge at 12,000 ⫻ g for 10 min at 4⬚C. Clarified
differentiation as well as for determining cell size and lysates were boiled in gel loading buffer and separated by 7.5%
animal size. It is therefore interesting to note that in SDS-PAGE. Proteins were electrotransferred to nitrocellulose and
detected with monoclonal antibodies directed against p190 Rho-
a cell culture model of neuronal differentiation (serum
GAP or p190-B RhoGAP (hybridoma supernate diluted 1:5) followed
withdrawal from neuro-2a neuroblastoma cells), we ob- by incubation with horseradish peroxidase-conjugated secondary
serve, by FACS analysis, a substantial increase in cell goat anti-mouse antibody (Bio-Rad; 1:5000) and development with
size during differentiation (M.C. and J.S., unpublished enhanced chemiluminescence (DuPont NEN) and autoradiography.
observations). Such cell growth is particularly notable For immunoprecipitation, cells that were treated or not treated with
when considering that it takes place in the absence IGF-1 for 15 min were extracted with RIPA buffer, clarified by centrif-
ugation, and subjected to immunoprecipitation with IGF-1 or IRS-1
of any serum-containing factors. In similar cell culture
antibodies and protein A agarose for 2 hr. After washing the beads
models of neuronal differentiation, CREB has been several times with RIPA buffer, samples were subjected to immu-
found to play an important role in promoting differentia- noblotting. For immunoblotting of CREB, JNK, p38, Akt, IRS-1, IGF-1
tion, and it will certainly be of interest in the future to receptor, cytochrome c, actin, and the various phosphoproteins,
determine whether CREB utilizes a distinct or overlap- extracts were prepared from cells and tissues in RIPA buffer and
ping set of transcriptional targets for regulating cell size lysates were clarified by centrifugation. Fifteen micrograms of pro-
tein of each sample were resolved by 12.5% SDS-PAGE, and the
and differentiation.
proteins were then electrotransferred to an Immobilon P membrane.
Immunoblotting was performed as described for p190-B RhoGAP.
Experimental Procedures All antibodies were obtained from New England Biolabs.

Generation of p190-B RhoGAP Mutant Mice Histology


To construct the p190-B targeting vector, a PGK-neo cassette was Newborn mice were sacrificed by CO2 asphyxiation and either dis-
inserted in reverse transcriptional orientation relative to the p190- sected immediately or fixed by immersion in Bouin’s fixative. For
B coding sequence in a BamHI site (bp 1196) of a 4.5 kb p190-B Nissl staining, E18.5 wild-type and mutant forebrains were fixed by
SV129 5⬘ genomic clone, disrupting the gene immediately upstream immersion (4% paraformaldehyde) and then cryoprotected in PBS/
of the GTPase domain (Burbelo et al., 1998). The complete insert 20% sucrose. Coronal cryostat sections (10–14 ␮M) were stained
of the resulting plasmid was subcloned as a KpnI-NotI fragment with cresyl violet acetate (0.5% in H2O) for 10 min.
into a PGK-neo-free pPNT vector (Tybulewicz et al., 1991), resulting
in the addition of the PGK-tk gene at the 5⬘ end of the p190-B
Cell Size and Cell Cycle Analysis
genomic DNA. NotI-linearized targeting vector (25 ␮g/ 8 ⫻ 106 cells)
For determination of protein:cell number and protein:DNA ratios,
was electroporated (400 V, 25 ␮F) into D3 ES cells of the mouse
cells were counted and lysed in RIPA buffer, and DNA and protein
SV129 strain. ES cells were maintained on a feeder layer of neor
content were determined as previously described (Franch et al.,
embryonic fibroblasts in DMEM supplemented with 15% fetal bovine
1995). For FACS analysis (forward light scatter), 3T3 cells were la-
serum (FBS) in the presence of 500 U/ml LIF (GIBCO). Transfected
beled with 0.1 mg/ml BrdU (cell labeling reagent; Amersham) for
ES cells were selected for growth in G418 at 300 ␮g/ml for 10
30 min, harvested, and analyzed according to the manufacturer’s
days and gancyclovir at 2 mM the first 5 days (initiated 36 hr after
instructions (Becton Dickinson). Briefly, cells were fixed in 80%
electroporation). Single colonies were expanded for DNA isolation
ethanol, and the DNA was denatured by treatment with 2 N HCl/
and Southern blot analysis was performed using standard methods,
0.5% Triton X-100 at room temperature for 30 min followed by a
to identify correctly targeted p190-B heterozygous ES clones (14
neutralization step in 0.1 M Na2B4O7 ⫻ 10H2O (pH 8.5). For immuno-
out of 130 screened). A 500 bp HindIII fragment (bp 1088–1586) was
fluorescence staining, anti-BrdU antibodies (Becton Dickinson) and
used to distinguish the mutated allele from the wild-type allele based
FITC-conjugated horse anti-mouse antibodies (Vector) were used.
on an altered StuI restriction pattern after homologous recombina-
The immunofluorescence step was followed by incubation in propid-
tion. 5⬘ and 3⬘ external probes were used to confirm the absence
ium iodide and RNase prior to two-dimensional FACS analysis using
of rearrangements 5⬘ and 3⬘ of the targeted region. ES cells from
Cell Quest software (Becton Dickinson). To measure the relative cell
three p190-B heterozygous clones were injected into the blastocoel
size of asynchronously growing cells corresponding to the wild-
cavity of 3.5-day-old C57BL/6J blastocysts, which were then trans-
type and mutant 3T3 lines, single cells with a DNA content of 2N
planted into the uteri of pseudopregnant females to generate chime-
(the G1 population) were gated and forward scatter height (FSC-H)
ric mice. Germline transmission was determined by agouti color
histograms were plotted. The mean FSC-H was calculated using
offspring of chimeric males mated with C57BL/6J females and con-
Cell Quest software. The analysis was performed with similar results
firmed by Southern blot analysis of StuI-digested tail DNA.
on three independently derived pairs of wild-type and mutant 3T3
cell lines.
Genotyping For FACS analysis of transiently transfected Rat-1 cells, cells (60
Genotype analysis was performed by PCR using a three-primer reac- hr posttransfection) were fixed in 1% formaldehyde in PBS for 30
tion containing primer 1, complementary to p190-B sequences adja- min, and permeabilized with 0.1% Triton X-100 in PBS followed by
cent to the PGK-neo gene insertion site (bp 1185–1202; 5⬘-dTAATG propidium iodide staining in the presence of RNase A, as described
ATAGGCGGATCCC-3⬘), primer 2, complementary to p190-B coding above. The forward scatter height of the GFP-positive transfected
sequence 3⬘ of the region of gene disruption (bp 1559–1577; 5⬘-dGGT cells with a 2N DNA content was analyzed as described above.
Developmental Cell
12

Fibroblast Cell Culture Studies Reporter Assays


To prepare embryo-derived fibroblasts, E13.5 embryos were ob- Reporter assays were performed following transfection (Fugene;
tained from timed matings of p190-B RhoGAP heterozygotes. DNA Boehringer Mannheim) of 3T3 cells with the CRE-luciferase reporter
was extracted from tail tissue for genotype analysis. After removal plasmid (Stratagene; 100 ng) or the UAS-luciferase reporter plasmid
of the head and internal organs, embryos were washed in PBS under (Stratagene; 250 ng). Cotransfections were with plasmids express-
sterile conditions, followed by trituration and 30 min trypsinization ing either RhoV14 (100 ng), wild-type GAL4-CREB, or GAL4-CREB
(using 5⫻ trypsin) at 37⬚C. Trypsin was inactivated by reconstituting containing an ala133 substitution (100 ng). GAL4-CREB fusions con-
the cells in DMEM containing 15% fetal calf serum (FCS) followed tain the entire CREB coding sequence. Twenty hours after transfec-
by centrifugation at 1500 rpm for 5 min at room temperature. Mouse tion, the cells were switched to media containing 1% serum and 40
embryonic fibroblasts (MEFs) were then resuspended in DMEM (plus hr later luciferase assays were performed (according to the manu-
15% fetal calf serum) and plated at high density (cells derived from facturer’s instructions; Promega). RSV-LacZ (200 ng) was used as
one embryo in one 10 cm dish). 3T3 cell lines were established using an internal control for transfection efficiency. CRE-luciferase activ-
standard procedures (Todaro and Green, 1963). Briefly, cells were ity, as indicated in graph form, represents activity normalized to
trypsinized, counted, and reseeded in DMEM supplemented with ␤-galactosidase activity from the same extracts. Results were quan-
10% calf serum at a density of 106 cells/10 cm dish every 3 days tified using a luminometer from EG&G Berthold for luciferase and
with a media change the subsequent day. After 30 to 50 passages, an ELISA plate reader for ␤-galactosidase.
stable lines of immortalized cells were established. Saturation den-
sity was determined by plating 104 wild-type or mutant cells in a 60
mm dish and then counting in triplicate the number of cells per dish Acknowledgments
every day for 6 consecutive days following replating. 3T3 and Rat-1
fibroblast lines were maintained in DMEM containing 10% calf serum We thank Drs. Daniel Haber, Iswar Hariharan, and Sofie Salama for
or 10% fetal bovine serum, respectively, and antibiotics. helpful comments on the manuscript. We appreciate the generous
To generate “CREB rescue” 3T3 cells, homozygous mutant 3T3 assistance of Dr. Nadia Carlesso with GFP FACS analysis, Dr. Fred
cells were cotransfected (Fugene; Boehringer Mannheim) with plas- Preffer with interpretation of FACS data, Dr. En Li of the MGH mouse
mids encoding constitutively active CREB (CREBFY) and the puro- transgenic/knockout facility with the production of mutant mice, Dr.
mycin resistance protein (pBabe-puro), then selected with puromy- Thaddeus Stappenbeck with the analysis of lung tissue, and Ms.
cin (Sigma; 2 ␮g/ml) for 2 weeks, and pooled. The mean FSC-H of Tracy Huang with assistance with immunoblotting. We are grateful
the G1 population was compared to that of the homozygous mutant to members of the Settleman laboratory for advice throughout the
parental 3T3 cells. Cells were plated at 1 ⫻ 106 cells/10 cm dish course of these studies. The CREB expression plasmids were kindly
and the G1 and G2 populations of cells were analyzed for forward provided by Dr. Marc Montminy (Salk Institute), Dr. Linda Kobierski
light scatter FACS analysis as described above. In addition, three (MGH), and Drs. Jon Kornhauser and Michael Greenberg (Harvard
individual clonal lines of stably transfected CREBFY-expressing Medical School), and the RhoV14 adenovirus was a generous gift
cells were established, and their relative cell sizes were also deter- of Drs. Enzo Calautti and Paolo Dotto (MGH). These studies were
mined by forward light scatter FACS analysis. supported by NIH and ACS awards to J.S.
For experiments with Rat-1 cells, 106 asynchronously growing
cells were plated per 10 cm dish in DMEM containing 10% FBS the
day before transfection. The next day, the cells were transfected Received: February 15, 2002
with 10 ␮g of DNA (5 ␮g vector control, CMV-RhoV14, CMV-CREBDN Revised: March 19, 2002
[dominant-negative], or CMV-CREBFY [activated], together with 5
␮g CMV-GFP vector) and 10 ␮l of Lipofectamine 2000 transfection References
reagent (Life Technologies). Twelve hours posttransfection, the me-
dium was replaced with serum-free DMEM. After an additional 12
Begum, N., Sandu, O.A., Ito, M., Lohmann, S.M., and Smolenski,
hr, the medium was replaced with DMEM containing 2.5% FBS and
A. (2002). Active Rho kinase (ROK-alpha) associates with insulin
2 ␮g/ml aphidicolin (to enrich for a G1 population of cells; Urbani
receptor substrate-1 and inhibits insulin signaling in vascular
et al., 1995). Thirty-six hours later, the cell size of the G1 cell popula-
smooth muscle cells. J. Biol. Chem. 277, 6214–6222.
tion was determined by FACS as described above.
For experiments with IGF-1 treatment, 106 asynchronously grow- Belvin, M.P., Zhou, H., and Yin, J.C. (1999). The Drosophila dCREB2
ing 3T3 cells were plated in 10 cm dishes in DMEM with 10% calf gene affects the circadian clock. Neuron 22, 777–787.
serum. After 3 days, the cells reached confluency and the medium Billuart, P., Winter, C.G., Maresh, A., Zhao, X., and Luo, L. (2001).
was replaced with serum-free DMEM. After an additional 24 hr, cells Regulating axon branch stability: the role of p190 RhoGAP in re-
were incubated with medium containing DMEM alone or DMEM and pressing a retraction signaling pathway. Cell 107, 195–207.
80 ng/ml of IGF-1 (Life Technologies). After 15 min, cells were lysed
in RIPA buffer and 15 ␮g of protein was analyzed by immunoblotting Bohni, R., Riesgo-Escovar, J., Oldham, S., Brogiolo, W., Stocker, H.,
for the indicated proteins as described above. Adenoviral infections Andruss, B.F., Beckingham, K., and Hafen, E. (1999). Autonomous
with virus expressing both RhoV14 (myc epitope-tagged) and GFP control of cell and organ size by CHICO, a Drosophila homolog of
or GFP alone (negative control) were performed 48 hr before IGF-1 vertebrate IRS1–4. Cell 97, 865–875.
treatment as previously described (Sullivan and Finkel, 2000). For Brouns, M.R., Matheson, S.F., Hu, K., Delalle, I., Caviness, V.S.,
treatments with the Y-27632 compound (Welfide), the drug (10 ␮M) Silver, J., Bronson, R.T., and Settleman, J. (2000). The adhesion
was added to culture media 15 min prior to IGF-1 treatment in signaling molecule p190 RhoGAP is required for morphogenetic
phospho-CREB/p38/Akt experiments, and for cell size studies, it processes in neural development. Development 127, 4891–4903.
was added once per day on 2 consecutive days in the presence of
2.5% FBS and 2 ␮g/ml aphidicolin prior to collecting G1 cells for Brouns, M.R., Matheson, S.F., and Settleman, J. (2001). p190 Rho-
forward light scatter FACS analysis as described above. GAP is the principal Src substrate in brain and regulates axon out-
growth, guidance and fasciculation. Nat. Cell Biol. 3, 361–367.
Measuring Rho Activation Burbelo, P.D., Miyamoto, S., Utani, A., Brill, S., Yamada, K.M., Hall,
Levels of activated, GTP-bound RhoA were determined using the A., and Yamada, Y. (1995). p190-B, a new member of the Rho GAP
Rhotekin “pull-down” assay as previously described (Ren and family, and Rho are induced to cluster after integrin cross-linking.
Schwartz, 2000). Briefly, asynchronously growing wild-type and mu- J. Biol. Chem. 270, 30919–30926.
tant cells were collected, lysed, and following clarification, lysates
Burbelo, P.D., Finegold, A.A., Kozak, C.A., Yamada, Y., and Takami,
were incubated with GST-Rhotekin on beads to capture GTP-bound
H. (1998). Cloning, genomic organization and chromosomal assign-
Rho proteins. Bound proteins were washed several times and eluted
ment of the mouse p190-B gene. Biochim. Biophys. Acta 1443,
with sample buffer before being subjected to SDS-PAGE and immu-
203–210.
noblotting with an anti-RhoA antibody (Santa Cruz). Whole-cell ly-
sates (prior to incubation with GST-Rhotekin) were analyzed in paral- Colbran, J.L., Roach, P.J., Fiol, C.J., Dixon, J.E., Andrisani, O.M.,
lel. The amount of whole-cell lysate loaded in each case represents and Corbin, J.D. (1992). cAMP-dependent protein kinase, but not
approximately 5% of the input material in binding assays. the cGMP-dependent enzyme, rapidly phosphorylates delta-CREB,
Rho and CREB Regulate Embryonic Cell Size
13

and a synthetic delta-CREB peptide. Biochem. Cell Biol. 70, 1277– lethality in mice lacking the cAMP response element binding protein.
1282. Proc. Natl. Acad. Sci. USA 95, 4481–4486.
Dong, J.M., Leung, T., Manser, E., and Lim, L. (1998). cAMP-induced Settleman, J. (1999). Rho GTPases in development. Prog. Mol. Sub-
morphological changes are counteracted by the activated RhoA cell. Biol. 22, 201–229.
small GTPase and the Rho kinase ROKalpha. J. Biol. Chem. 273, Settleman, J. (2000). Getting in shape with Rho. Nat. Cell Biol. 2,
22554–22562. E7–E9.
Du, K., Asahara, H., Jhala, U.S., Wagner, B.L., and Montminy, M. Settleman, J., Narasimhan, V., Foster, L.C., and Weinberg, R.A.
(2000). Characterization of a CREB gain-of-function mutant with (1992). Molecular cloning of cDNAs encoding the GAP-associated
constitutive transcriptional activity in vivo. Mol. Cell. Biol. 20, 4320– protein p190; implications for a signaling pathway from ras to the
4327. nucleus. Cell 63, 539–549.
Farah, S., Agazie, Y., Ohan, N., Ngsee, J.K., and Liu, X.J. (1998). Shaywitz, A.J., and Greenberg, M.E. (1999). CREB: a stimulus-
A rho-associated protein kinase, ROKalpha, binds insulin receptor induced transcription factor activated by a diverse array of extracel-
substrate-1 and modulates insulin signaling. J. Biol. Chem. 273, lular signals. Annu. Rev. Biochem. 68, 821–861.
4740–4746.
Stocker, H., and Hafen, E. (2000). Genetic control of cell size. Curr.
Franch, H.A., Shay, J.W., Alpern, R.J., and Preisig, P.A. (1995). Opin. Genet. Dev. 10, 529–535.
Involvement of pRB family in TGF beta-dependent epithelial cell
Sullivan, D.M., and Finkel, T. (2000). Expression of Rho GTPases
hypertrophy. J. Cell Biol. 129, 245–254.
using adenovirus vectors. Methods Enzymol. 325, 303–314.
Gao, X., and Pan, D. (2001). TSC1 and TSC2 tumor suppressors
Symons, M., and Settleman, J. (2000). Rho family GTPases: more
antagonize insulin signaling in cell growth. Genes Dev. 15, 1383–
than simple switches. Trends Cell Biol. 10, 415–419.
1392.
Tapon, N., Ito, N., Dickson, B.J., Treisman, J.E., and Hariharan, I.K.
Henning, S.W., Galandrini, R., Hall, A., and Cantrell, D.A. (1997). The (2001). The Drosophila tuberous sclerosis complex gene homologs
GTPase Rho has a critical regulatory role in thymus development. restrict cell growth and cell proliferation. Cell 105, 345–355.
EMBO J. 16, 2397–2407.
Thomas, M.J., Umayahara, Y., Shu, H., Centrella, M., Rotwein, P.,
Herzig, R.P., Scacco, S., and Scarpulla, R.C. (2000). Sequential se- and McCarthy, T.L. (1996). Identification of the cAMP response ele-
rum-dependent activation of CREB and NRF-1 leads to enhanced ment that controls transcriptional activation of the insulin-like
mitochondrial respiration through the induction of cytochrome c. J. growth factor-I gene by prostaglandin E2 in osteoblasts. J. Biol.
Biol. Chem. 275, 13134–13141. Chem. 271, 21835–21841.
Hsin, H., and Kenyon, C. (1999). Signals from the reproductive sys- Todaro, G.J., and Green, H. (1963). Quantitative studies of the growth
tem regulate the lifespan of C. elegans. Nature 399, 362–366. of mouse embryo cells in culture and their development into estab-
Iritani, B.M., and Eisenman, R.N. (1999). c-Myc enhances protein lished lines. J. Cell Biol. 17, 299–313.
synthesis and cell size during B lymphocyte development. Proc. Tybulewicz, V.L.J., Crawford, C.E., Jackson, P.K., Bronson, R.T.,
Natl. Acad. Sci. USA 96, 13180–13185. and Mulligan, R.C. (1991). Neonatal lethality and lymphopenia in
Jalink, K., van Corven, E.J., Hengeveld, T., Morii, N., Narumiya, S., mice with a homozygous disruption of the c-abl proto-oncogene.
and Moolenaar, W.H. (1994). Inhibition of lysophosphatidate- and Cell 65, 1153–1163.
thrombin-induced neurite retraction and neuronal cell rounding by Urbani, L., Sherwood, S.W., and Schimke, R.T. (1995). Dissociation
ADP ribosylation of the small GTP-binding protein Rho. J. Cell Biol. of nuclear and cytoplasmic cell cycle progression by drugs em-
126, 801–810. ployed in cell synchronization. Exp. Cell Res. 219, 159–168.
Lamb, R.F., Roy, C., Diefenbach, T.J., Vinters, H.V., Johnson, M.W., Van Aelst, L., and D’Souza-Schorey, C. (1997). Rho GTPases and
Jay, D.G., and Hall, A. (2000). The TSC1 tumour suppressor hamartin signaling networks. Genes Dev. 11, 2295–2322.
regulates cell adhesion through ERM proteins and the GTPase Rho.
Vincent, S., and Settleman, J. (1999). Inhibition of RhoGAP activity
Nat. Cell Biol. 2, 281–287.
is sufficient for the induction of Rho-mediated actin reorganization.
Lane, M.E., and Kalderon, D. (1993). Genetic investigation of cAMP- Eur. J. Cell Biol. 78, 539–548.
dependent protein kinase function in Drosophila development.
Walton, M.R., and Dragunow, I. (2000). Is CREB a key to neuronal
Genes Dev. 7, 1229–1243.
survival? Trends Neurosci. 23, 48–53.
Leung, T., Manser, E., Tan, L., and Lim, L. (1995). A novel serine/
Walton, K.M., Rehfuss, R.P., Chrivia, J.C., Lochner, J.E., and Goodman,
threonine kinase binding the Ras-related RhoA GTPase which trans-
R.H. (1992). A dominant repressor of cyclic adenosine 3⬘,5⬘-mono-
locates the kinase to peripheral membranes. J. Biol. Chem. 270,
phosphate (cAMP)-regulated enhancer-binding protein activity in-
29051–29054.
hibits the cAMP-mediated induction of the somatostatin promoter
Luo, L. (2000). Rho GTPases in neuronal morphogenesis. Nat. Rev. in vivo. Mol. Endocrinol. 6, 647–655.
Neurosci. 1, 173–180.
Withers, D.J. (2001). Insulin receptor substrate proteins and neuro-
Mayr, B., and Montminy, M. (2001). Transcriptional regulation by the endocrine function. Biochem. Soc. Trans. 29, 525–529.
phosphorylation-dependent factor CREB. Nat. Rev. Mol. Cell. Biol.
2, 599–609.
Paz, K., Hemi, R., LeRoith, D., Karasik, A., Elhanany, E., Kanety, H.,
and Zick, Y. (1997). A molecular basis for insulin resistance. Elevated
serine/threonine phosphorylation of IRS-1 and IRS-2 inhibits their
binding to the juxtamembrane region of the insulin receptor and
impairs their ability to undergo insulin-induced tyrosine phosphory-
lation. J. Biol. Chem. 272, 29911–29918.
Philippe, J., and Missotten, M. (1990). Functional characterization
of a cAMP-responsive element of the rat insulin I gene. J. Biol.
Chem. 265, 1465–1469.
Potter, C.J., Huang, H., and Xu, T. (2001). Drosophila Tsc1 functions
with Tsc2 to antagonize insulin signaling in regulating cell growth,
cell proliferation, and organ size. Cell 105, 357–368.
Ren, X.D., and Schwartz, M.A. (2000). Determination of GTP loading
on Rho. Methods Enzymol. 325, 264–272.
Rudolph, D., Tafuri, A., Gass, P., Hammerling, G.J., Arnold, B., and
Schutz, G. (1998). Impaired fetal T cell development and perinatal

You might also like