ArticlePDF Available

Dual degradation signals control Gli protein stability and tumor formation

Authors:
  • Independent Researcher

Abstract and Figures

Regulated protein destruction controls many key cellular processes with aberrant regulation increasingly found during carcinogenesis. Gli proteins mediate the transcriptional effects of the Sonic hedgehog pathway, which is implicated in up to 25% of human tumors. Here we show that Gli is rapidly destroyed by the proteasome and that mouse basal cell carcinoma induction correlates with Gli protein accumulation. We identify two independent destruction signals in Gli1, D(N) and D(C), and show that removal of these signals stabilizes Gli1 protein and rapidly accelerates tumor formation in transgenic animals. These data argue that control of Gli protein accumulation underlies tumorigenesis and suggest a new avenue for antitumor therapy.
Onset of BCC formation correlates with Gli protein accumulation. ( a ) Clinical appearance of focal BCCs induced in transgenic animals expressing Gli2 in the skin epithelium with the keratin 5 promoter. ( b ) Bar graph showing representative onset of tumors in K5-Gli2 mice. ( c ) Quantitative PCR of Gli2 RNA levels from skin of wild-type or Gli2 transgenic animals of indicated age. Gli2 RNA levels in each sample were normalized to those of GAPDH. Error bars are standard error of the mean (SEM). ( d ) Immunofluorescence with anti-HA (red) antibody showing protein accumulation only in BCC tumor (arrowhead), not in interfollicular epidermis (arrow). (Green) Anti-laminin 5; (blue) Hoechst. ( e ) Western blot of lysates from explanted K5-Gli2 keratinocytes demonstrating the rapid accumulation of Gli2 protein with the addition of the proteasome inhibitor MG132, but not with DMSO. ( f ) Autoradiogram of 35 S-labeled Gli1 mixed with Xenopus oocyte extract. Gli1 is degraded in a proteasome-dependent manner with a half-life of ∼ 40 min. ( g ) Western blot of HA-Gli1 in NIH 3T3 cells showing rapid, proteasome-dependent destruction. The nonspecific band demonstrates equal protein loading. ( h ) HA-Gli1 protein is rapidly degraded (C) via a process inhibited by proteasome inhibitors (M), but not cathepsin or lysosomal inhibitors E64 (E) or chloroquine (Q), respectively. The efficacy of the E64 and chloroquine used in this experiment was confirmed by their ability to inhibit ligand-dependent lysosomal destruction of EGFR in primary human keratinocytes.
… 
Degron D C (DSGVEM) mediates Gli1 destruction via the ␤ TrCP destruction complex. ( a , left ) Alignment of chordate Gli sequences showing conserved D C sequence. The box details key residues that bind ␤ TrCP. ( Right ) ␤ TrCP-binding sequences from other vertebrate proteins. ( b ) Reciprocal coimmunoprecipitation of HA-Gli1 or HA-Gli1 ⌬ D C with myc- ␤ TrCP. Note the lack of ␤ TrCP binding in the mutant. The characteristic mobility shift of immu- noprecipitated Gli1 is not an artifact of ␤ TrCP overexpression as this shift is observed even in its absence. ( c ) Degradation of trans- fected Gli1 in NIH 3T3 cells. Note the small but significant delay in destruction kinetics of the mutant versus wild-type protein. The densitometry of both assays is shown to the right and is repre- sentative of three independent experiments. Equal sample loading and transfer was confirmed by post-staining of the experimental membranes with Coomassie blue. The 150 – 250-kDa region of the membranes is shown. ( d ) Western blot of Gli1 or Gli1 ⌬ D C and in- creasing amounts of transfected ␤ TrCP. Note the decreased steady- state levels of wild-type, but not mutant Gli1. The difference is quantified below and is representative of three independent experi- ments. Error bars are standard error of the mean (SEM). ( e ) Coim- munoprecipitation assay of 6X-His-tagged ubiquitin and HA- Gli1 ⌬ N398 containing degron D C or HA-Gli1 ⌬ N398 ⌬ D C mutant. Ubiquitinylated Gli ( top panel) is detected in the wild-type Gli1 C terminus, but not the ⌬ D C mutant, in the presence of ␤ TrCP ( bot- tom panel).
… 
Degron D N mediates Gli destruction independent of D C or Sufu function. (a) Alignment showing the conserved N-terminal region containing degron D N. A solid line indicates the most highly conserved region that is deleted in the D N mutant, while the boxed area shows the Sufu-binding site, which is retained in the D N mutant. (b) Destruction assays of HA-Gli1 in NIH 3T3 cells showing the effects of single D C , D N , and double mutants in comparison to wild-type (WT) Gli1 in the presence and absence of MG132. The densitometery of blots is shown to the right and is based on three independent experiments. Note that results are plotted on the base 2 logarithmic scale. Error bars are standard error of the mean (SEM). (c) Coimmunoprecipitation of wild-type and mutant Gli with TrCP. Note that the D N mutation does not affect the binding of TrCP to degron D C. (d) Changes in levels of green fluorescent protein (EGFP) fused to Gli1 N-terminal residues (top), or EGFP (bottom), in the presence of cycloheximide (left) or MG132 (right). The amount of fusion protein is identical at t = 0, but the exposure time for the left and right panels differs to avoid signal saturation. The densitometry is shown to the right with results plotted on a linear scale. The results are representative of three independent experiments. Error bars are SEM. (e) Coprecipitation assays with GST-Sufu and lysates from cells containing wild-type or mutant Gli proteins. Note that the Gli1D N mutation leaves Sufu binding intact. (f) Immunofluorescence of Gli1 shows similar subcellular localization of wild-type and mutant Gli1 proteins in the absence (left) or presence (right) of the Crm1-inhibitor leptomycin B. (g, left) Luciferase transcription assays of wild-type and double-mutant Gli1 protein with increasing amounts of transfected moles of plasmid. Error bars are SEM. Densitometry (middle) of Western blots (right) showing the amount of steady-state protein accumulation corresponding to the increase in luciferase activity. (h, left) Luciferase transcription assays of wild-type, single, and double-mutant Gli1 proteins. Error bars are SEM. Western blot (right) of levels of Gli1 protein in luciferase assay and quantitation (middle) of protein levels normalized for loading and transfer efficiency determined by immunoblot for nuclear pore complex (NPC).
… 
Content may be subject to copyright.
RESEARCH COMMUNICATION
Dual degradation signals
control Gli protein stability
and tumor formation
Erik G. Huntzicker,
1,3
Ivette S. Estay,
1,3
Hanson Zhen,
1
Ludmila A. Lokteva,
1
Peter K. Jackson,
2,3
and Anthony E. Oro
1,3,4
1
Program in Epithelial Biology,
2
Department of Pathology,
School of Medicine, and
3
Cancer Biology Graduate Program,
Stanford University, Stanford, California 94305, USA
Regulated protein destruction controls many key cellu-
lar processes with aberrant regulation increasingly found
during carcinogenesis. Gli proteins mediate the tran-
scriptional effects of the Sonic hedgehog pathway, which
is implicated in up to 25% of human tumors. Here we
show that Gli is rapidly destroyed by the proteasome and
that mouse basal cell carcinoma induction correlates
with Gli protein accumulation. We identify two inde-
pendent destruction signals in Gli1, D
N
and D
C
, and
show that removal of these signals stabilizes Gli1 pro-
tein and rapidly accelerates tumor formation in trans-
genic animals. These data argue that control of Gli pro-
tein accumulation underlies tumorigenesis and suggest a
new avenue for antitumor therapy.
Supplemental material is available at http://www.genesdev.org.
Received October 3, 2005; revised version accepted December
1, 2005.
Factors controlling protein destruction are critical for
the timing of key processes such as the cell cycle, apo-
ptosis, and cell fate decisions, with aberrant regulation
increasingly found during carcinogenesis (Pickart 2004;
Yamasaki and Pagano 2004). Inappropriate Sonic hedge-
hog (Shh) signaling results in a panoply of birth defects
and is implicated in up to 25% of human tumors (Cal-
lahan and Oro 2001; Lum and Beachy 2004). While the
Gli family of proteins mediates the transcriptional ef-
fects of Shh (Methot and Basler 2001; Ruiz i Altaba et al.
2002), the mechanism by which Gli proteins are regu-
lated to achieve changes in pathway output remains
poorly understood. Studies in mice and humans show
that Shh target gene induction is sufficient to induce a
variety of tumors including basal cell carcinomas (BCCs)
(Oro et al. 1997; Nilsson et al. 2000; Hutchin et al. 2005).
However, there is a wide variability in the onset and
severity of phenotypes among patients with mutations
in the Shh pathway (Wicking et al. 1997), and a notice-
ably wide variability of tumor onset in animal models
(Oro and Higgins 2003; Hutchin et al. 2005). This sug-
gests the possibility that additional, previously unchar-
acterized, cellular processes regulate pathway output.
Here we show that Gli protein accumulation correlates
with tumor formation and stabilizing mutations in Gli
protein dramatically accelerate tumor induction.
Results and Discussion
While expression of either Gli1 or Gli2 in the epidermis
of transgenic mice induces BCCs (Fig. 1a), we have ob-
served a considerable delay in the appearance of Gli-de-
pendent tumors. Analysis of transgenic mice expressing
Gli2 revealed an average latency of 7 mo before tumor
appearance (Fig. 1b). We ruled out changes in transcrip-
tion of the transgene with age as a cause of the tumors,
as similar levels of RNA are seen in both age groups as
measured by quantitative PCR (Fig. 1c). This suggested
the existence in keratinocytes of additional processes,
whose loss or dysregulation is required to permit Gli
activity and direct tumor formation. Our previous stud-
ies indicated that differential accumulation of Gli pro-
tein plays an important role in restricting Shh target
gene induction in interfollicular epithelium (Oro and
Higgins 2003). Indeed, we detected no transgenic Gli pro-
tein in normal skin, whereas we found high levels in the
BCC tumors (Fig. 1d). Cultured explants of primary ke-
ratinocytes from normal skin also contained little de-
tectable Gli protein (Fig. 1e). However, treatment of
these cells with the proteasome inhibitor MG132 caused
full-length Gli2 protein to accumulate many fold within
3 h, confirming the presence of an active Gli2 protein
destruction mechanism. These data support the conclu-
sion that proteasome-dependent Gli protein destruction
underlies the latency in Shh target gene response.
To study the molecular mechanisms that govern Gli
protein degradation, we chose to focus our initial studies
on Gli1, which, unlike Gli2 or Gli3, is primarily a tran-
scriptional activator and is not processed to a repressor
form (Dai et al. 1999; von Mering and Basler 1999). In
this way, Gli protein function and degradation could be
examined independently of proteolytic processing and
transcriptional repressor regulation. We tested Gli1 sta-
bility in a variety of in vitro settings and found that Gli1
is degraded by the proteasome. In Xenopus egg extracts,
a system where the ubiquitin–proteasome system (UPS)
is known to be active to control -catenin and IB sta-
bility (Winston et al. 1999; Margottin-Goguet et al.
2003),
35
S-labeled Gli1 protein is destroyed in a protea-
some-dependent manner, with a half-life of 40 min (Fig.
1f). Similar kinetics are seen in a variety of cultured nor-
mal and cancer cells, including the Shh-responsive NIH
3T3 cells (Fig. 1g; Taipale et al. 2000). We ruled out deg-
radation of Gli1 by other mechanisms such as lysosomal
degradation (Dai et al. 2003), as cathepsin and lysosome
inhibitors (E64 and chloroquine, respectively) had no ef-
fect on Gli levels (Fig. 1h). The efficacy of these inhibi-
tors was confirmed in primary human keratinocytes
where they inhibit the EGF-dependent lysosomal de-
struction of EGFR (Fig. 1h). These data provide strong
support for destruction of vertebrate Gli1 by the UPS.
To identify signals that allow Gli1 to interact with the
UPS, we were guided by the previous finding in Dro-
sophila that the TrCP locus is required for Ci process-
ing (Jiang and Struhl 1998). The degron DSGXXS, recog-
nized by TrCP, is present in vertebrate regulatory pro-
[Keywords: Hedgehog; Gli; -TRCP; proteasome; basal cell carcinoma;
hair follicle]
4
Corresponding author.
E-MAIL oro@cmgm.stanford.edu; FAX (650) 723-8762.
Article published online ahead of print. Article and publication date are
at http://www.genesdev.org/cgi/doi/10.1101/gad.1380906.
276 GENES & DEVELOPMENT 20:276–281 © 2006 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/06; www.genesdev.org
teins -catenin, IB/, and Emi1 (Spencer et al. 1999),
although it is absent from Ci. In spite of this, we have
identified a C-terminal motif, DSGVEM, that is con-
served in chordate Gli homologs and vertebrate Gli1 and
Gli2 proteins (Fig. 2a). To determine if the DSGVEM
motif of human Gli1 mediates association with TrCP1,
reciprocal immunoprecipitations were performed from
NIH 3T3 cells transfected with myc-tagged TrCP1 and
HA-tagged Gli1 (Fig. 2b). Gli1 protein lacking the
DSGVEM motif (Gli1D
C
) failed to associate with
TrCP and exhibited delayed degradation kinetics (Fig.
2c). Levels of TrCP appeared to be limiting for Gli1
degradation, as increasing the levels of TrCP protein
significantly decreased steady-state levels of Gli1 protein
(Fig. 2d). Consistent with its role as an E3 ligase, TrCP
association with Gli1 facilitated ubiquitination. In ubiq-
uitin coimmunoprecipitation assays, ubiquitinylated
Gli1 (N398), but not Gli1D
C
(N398), could be de-
tected in the presence of overexpressed TrCP1 (Fig. 2e).
Previous studies have shown that Protein kinase A (PKA)
can enhance TrCP-dependent Ci cleavage in Dro-
sophila (Wang et al. 1999; Jia et al. 2004). We saw similar
effects on Gli1, as inhibition of PKA impeded destruc-
tion (Supplementary Fig. 2a), and Gli1 constructs lacking
consensus PKA sites in the C terminus failed to bind
TrCP and exhibited delayed destruction kinetics
(Supplementary Fig. 2a,b). These data demonstrate that
degron D
C
mediates Gli destruction via the TrCPubiq-
uitin ligase complex.
While TrCP-dependent degradation clearly plays a
role in Gli1 destruction, the Gli1D
C
mutation only par-
tially altered the destruction kinetics of Gli1 protein in
cultured cells. At 3 h after cycloheximide addition, de-
struction of Gli1D
C
was decreased by only 25% rela-
tive to wild-type Gli1 (47.1% ± 6% vs. 21% ± 5%,
Avg. ± SEM) (Fig. 3b). This argued that additional signals
control Gli1 degradation. Through focused mutagenesis,
we found that a small deletion of the N terminus further
stabilized Gli1. As with the D
C
degron, degron D
N
mu-
tations (GliN1116, referred to as Gli1D
N
) alone had
modest effects on Gli destruction kinetics in vitro (3 h:
40.1% ± 6% vs. 21% ± 5%, Avg. ± SEM) (Fig. 3b). How-
ever, Gli1 lacking both degrons (double mutant;
Gli1D
N
D
C
became stable, possessing destruction ki-
netics similar to that with addition of proteasome inhibi-
tor (Figs. 3b, 1g). This argued for an additional degron in
the N terminus. Further mutagenesis narrowed the re-
gion containing the degradation signal to residues 51
116 (Supplementary Fig. 3). This region contains a
stretch of highly conserved residues present in all verte-
brate Gli genes and in Drosophila Ci (Fig. 3a), suggesting
that the destruction signal may be found in many Gli
proteins.
We next determined whether degron D
N
functioned
independently of degron D
C
. We tested whether TrCP
binding depends on D
N
. Consistent with the notion of
distinct signals, coimmunoprecipitation studies showed
that TrCP bound equally well to both wild-type Gli1
and the Gli1D
N
mutant (Fig. 3c). Moreover, we tested
whether degron D
N
could confer instability to a heter-
ologous protein. Green Fluorescent Protein (GFP) is a
stable protein with a long half-life. Addition of amino
acids 1208, a region that encompasses degron D
N
se-
quences, destabilized GFP in a proteasome-dependent
fashion, giving it a half-life of 180 min (Fig. 3d). To-
gether, these data suggest the two destruction signals
function independently.
Degron D
N
is immediately adjacent to the binding site
for Sufu (Fig. 3a), a powerful negative regulator of the Shh
pathway, suggesting that the degron might work in con-
junction with Sufu. Consequently, we tested whether
D
N
mutations affected the known Sufu functions of tran-
scriptional corepression and Gli sequestration in the cy-
tosol (Ding et al. 1999; Kogerman et al. 1999; Cheng and
Bishop 2002). Gli1D
N
bound to Sufu as well as wild-
type Gli1 in GST pull-down (Fig. 3e) assays. Also, Gli1
Figure 1. Onset of BCC formation correlates with Gli protein accumulation. (a) Clinical appearance of focal BCCs induced in transgenic
animals expressing Gli2 in the skin epithelium with the keratin 5 promoter.(b) Bar graph showing representative onset of tumors in K5-Gli2
mice. (c) Quantitative PCR of Gli2 RNA levels from skin of wild-type or Gli2 transgenic animals of indicated age. Gli2 RNA levels in each
sample were normalized to those of GAPDH. Error bars are standard error of the mean (SEM). (d) Immunofluorescence with anti-HA (red)
antibody showing protein accumulation only in BCC tumor (arrowhead), not in interfollicular epidermis (arrow). (Green) Anti-laminin 5; (blue)
Hoechst. (e) Western blot of lysates from explanted K5-Gli2 keratinocytes demonstrating the rapid accumulation of Gli2 protein with the
addition of the proteasome inhibitor MG132, but not with DMSO. (f) Autoradiogram of
35
S-labeled Gli1 mixed with Xenopus oocyte extract.
Gli1 is degraded in a proteasome-dependent manner with a half-life of 40 min. (g) Western blot of HA-Gli1 in NIH 3T3 cells showing rapid,
proteasome-dependent destruction. The nonspecific band demonstrates equal protein loading. (h) HA-Gli1 protein is rapidly degraded (C) via
a process inhibited by proteasome inhibitors (M), but not cathepsin or lysosomal inhibitors E64 (E) or chloroquine (Q), respectively. The efficacy
of the E64 and chloroquine used in this experiment was confirmed by their ability to inhibit ligand-dependent lysosomal destruction of EGFR
in primary human keratinocytes.
Two destruction signals in Gli proteins
GENES & DEVELOPMENT 277
mutants had a similar subcellular distribution to wild-
type Gli1 and accumulated in the nucleus with equal
efficiency in the presence of leptomycin (Fig. 3f). These
data argue that the N-terminal degron regulates Gli1 sta-
bility via a unique Sufu- and degron D
C
-independent
pathway.
Gli1 is known to activate transcription of Shh target
genes via a transactivation domain in its C terminus
(Yoon et al. 1998). To determine the functional signifi-
cance of stabilizing Gli, we assessed the transcriptional
activity of the mutants on Gli-responsive promoters
(Taipale et al. 2000). Gli1D
C
and Gli1D
N
displayed
modest increases in transcription when the same molar
amount of plasmid was transfected into cells, with the
double mutant displaying threefold higher target gene
induction compared with wild-type Gli1 (Fig. 3g,h). This
increase could be due to increased transactivation ability
or increased protein levels. Analysis of protein levels
relative to transcriptional output demonstrated a clear
linear relationship between the amount of Gli1 protein
for each of the mutants and reporter gene output (Fig.
3g,h). These data argue that the greater transcriptional
activity of the mutants is due to increased protein sta-
bility rather than transactivation ability.
Tumor induction in Gli transgenic animals correlates
with Gli protein accumulation. If the degrons identified
in our studies are responsible for restricting Gli1 protein
accumulation in vivo, then expressing Gli1 without
these signals should shorten the latency to tumor induc-
tion. We assayed skin phenotypes of several lines of
transgenic animals expressing different mutants of Gli1
in the basal layer of stratified epithelia. As expected,
transgenic animals expressing wild-type Gli1 were born
normally with no detectable transgenic Gli protein (Fig.
4b,l), and developed the predicted tumor phenotype at
68 wk after birth (Oro and Higgins 2003; data not
shown). In contrast, animals expressing double-mutant
Gli1 (GliD
N
D
C
) exhibited Gli protein accumulation
at the time of birth in tumor and nontumor epithelium
(Fig. 4c; Supplementary Fig. 4). The GliD
N
D
C
-ex-
pressing animals died at birth with shallow skin ulcers
clinically similar to BCCs throughout the body. The tu-
mors demonstrated characteristic features of BCCs (Fig.
4c,m; Supplementary Fig. 4; Oro et al. 1997; Callahan et
al. 2004), including the up-regulation of ptch1 (Fig. 4r).
Moreover, the tumors were rapidly dividing as evidenced
by the significant increase in Ki67 staining and displayed
the BCC marker keratin 17 (Supplementary Fig. 4). This
demonstrates that altered protein accumulation can di-
rectly accelerate tumor induction.
In cultured cells, both degrons were highly active in
restricting Gli1 levels. However, depending on the speci-
ficity and/or capacity of the operative degradation path-
way, one of the degrons may play a more active role in a
given in vivo context. We determined the relative con-
tribution of each degron to Gli1 destruction by compar-
ing the phenotype of single Gli mutant transgenic mice
to those expressing GliD
N
D
C
. In the skin, Gli1 mu-
tants lacking degron D
C
displayed a much stronger phe-
notype than those lacking degron D
N
(Fig. 4v). While
both GliD
C
and GliD
N
transgenic mice were viable
and lacked the ulcerating lesions seen in the double mu-
tant, GliD
C
mutants demonstrated BCC-like lesions at
birth more comparable to those expressing GliD
N
D
C
(depth of invasion, 111 µM vs. 140 µM, respectively) (Fig.
4d). GliD
N
transgenic animals had small BCC-like pro-
liferations that developed slightly after birth and ap-
peared to come directly off the hair follicle (Fig. 4e). Also,
many GliD
N
mutant lesions were benign hair follicle
tumors, indicative of lower Shh target gene induction
(Callahan and Oro 2001; Grachtchouk et al. 2003). In
each of the Gli mutants, the distribution of Gli protein
was both nuclear and cytoplasmic, providing further evi-
dence that the degron sequences do not play a role in
nucleocytoplasmic shuttling of Gli1 (Fig. 4lp). The phe-
notypic differences within each group could not be at-
tributed to transgene expression differences, as only
steady-state protein levels by IHC, not transgene copy
number or RNA expression level, correlated with the
phenotype (Fig. 4; Supplementary Fig. 5). These data
demonstrate the combinatorial action of both D
C
and
D
N
degrons in preventing ectopic Shh target gene induc-
tion and provide in vivo support for the role of Gli de-
struction in controlling tumor formation.
Here we have shown that Gli1 protein contains two
destruction signals that regulate protein stability and tu-
Figure 2. Degron D
C
(DSGVEM) mediates Gli1 destruction via the
TrCP destruction complex. (a, left) Alignment of chordate Gli
sequences showing conserved D
C
sequence. The box details key
residues that bind TrCP. (Right) TrCP-binding sequences from
other vertebrate proteins. (b) Reciprocal coimmunoprecipitation of
HA-Gli1 or HA-Gli1D
C
with myc-TrCP. Note the lack of TrCP
binding in the mutant. The characteristic mobility shift of immu-
noprecipitated Gli1 is not an artifact of TrCP overexpression as
this shift is observed even in its absence. (c) Degradation of trans-
fected Gli1 in NIH 3T3 cells. Note the small but significant delay
in destruction kinetics of the mutant versus wild-type protein.
The densitometry of both assays is shown to the right and is repre-
sentative of three independent experiments. Equal sample loading
and transfer was confirmed by post-staining of the experimental
membranes with Coomassie blue. The 150250-kDa region of the
membranes is shown. (d) Western blot of Gli1 or Gli1D
C
and in-
creasing amounts of transfected TrCP. Note the decreased steady-
state levels of wild-type, but not mutant Gli1. The difference is
quantified below and is representative of three independent experi-
ments. Error bars are standard error of the mean (SEM). (e) Coim-
munoprecipitation assay of 6X-His-tagged ubiquitin and HA-
Gli1N398 containing degron D
C
or HA-Gli1N398D
C
mutant.
Ubiquitinylated Gli (top panel) is detected in the wild-type Gli1 C
terminus, but not the D
C
mutant, in the presence of TrCP (bot-
tom panel).
Huntzicker et al.
278 GENES & DEVELOPMENT
mor formation (Supplementary Fig.
1). As with other key regulatory pro-
teins such as myc, p53, IB, and
-catenin, there appears to be a finely
balanced control of Gli1 protein lev-
els to allow for proper target gene in-
duction while preventing epithelial
tumor formation. Our data suggest
that the BCC tumors observed in the
K5Gli2 transgenic mice likely arise as
a result of secondary changes that
lead to Gli2 stabilization rather than
as a result of gradual saturation of the
destruction machinery. Arguing
against saturation is the lack of in-
creased protein in adjacent normal
tissue or in the explanted cells from
older animals. Furthermore, with the
addition of proteasome inhibitors, we
see rapid accumulation of Gli2 pro-
tein. This suggests that halting the
destruction of Gli proteins is an early
step in the tumor process and that
cellular changes that allow Gli1 pro-
tein accumulation may contribute to
human carcinogenesis (Kinzler et al.
1988). Similarly, targeted therapies
that delay the onset of Gli accumula-
tion may have potent antitumor prop-
erties.
Our study illustrates how two de-
struction signals cooperate to prevent
Gli protein accumulation, target gene
induction, and subsequent tumor for-
mation. While a role for TrCP has
been implicated in Ci processing, the
present study is the first to demon-
strate that it acts by directly binding
Gli to facilitate ubiquitinylation and
destruction. Interestingly, while Ci
and Gli1 are both directed by PKA
and TrCP to interact with the pro-
teasome, the end result differs in that
Gli1 is degraded but not cleaved. This
could be due to either the particular
amino acid sequence of the degron or
to surrounding amino acids that in-
fluence TrCP/UPS function. The
identified Gli degron differs signifi-
cantly from that of -catenin, Emi1,
and IkB in that it lacks a second ser-
ine shown to be important for sequen-
tial phosphorylation and contains a
phosphomimetic glutamic acid residue (Amit et al. 2002;
Moshe et al. 2004). Future studies will focus on whether
these sequence differences are sufficient to account for
the different final disposition of the protein. This study
further identifies a novel degron, D
N
, that shares little
identity with other known degradation signals. The con-
served sequences in this degron are found in both Gli2
and Gli3, and removal of the region containing them has
been associated with activation of Gli2 (Sasaki et al.
1999; Mill et al. 2003). Our data suggest that a portion of
this activation may be due to Gli2 protein stabilization
via degron D
N
rather than simply loss of transcriptional
repressor activity.
Material and methods
Destruction assays
Xenopus egg extracts. Xenopus egg cytoplasmic extracts were prepared
fresh as previously described (Reimann et al. 2001). Substrate proteins
were in vitro translated in the presence of
35
S-methionine using the TnT
IVT system (Promega). IVT protein was added to egg extract to 10% of
final volume. Destruction assays were conducted in a final volume of
210 µL, and stopped by addition of 2× Sample buffer and snap-freezing in
liquid nitrogen. In some experiments, MG-132 (Calbiochem) was added
to a final concentration of 1 mM.
NIH 3T3. NIH 3T3 cells were transfected as described above. Two days
after transfection cycloheximide was added to final concentration of 20
µg/mL and samples were harvested in 2× Sample buffer at various time
points. Alternatively, cycloheximide was added at various time points
Figure 3. Degron D
N
mediates Gli destruction independent of D
C
or Sufu function. (a) Align-
ment showing the conserved N-terminal region containing degron D
N
. A solid line indicates
the most highly conserved region that is deleted in the D
N
mutant, while the boxed area shows
the Sufu-binding site, which is retained in the D
N
mutant. (b) Destruction assays of HA-Gli1
in NIH 3T3 cells showing the effects of single D
C
,D
N
, and double mutants in comparison to
wild-type (WT) Gli1 in the presence and absence of MG132. The densitometery of blots is
shown to the right and is based on three independent experiments. Note that results are
plotted on the base 2 logarithmic scale. Error bars are standard error of the mean (SEM). (c)
Coimmunoprecipitation of wild-type and mutant Gli with TrCP. Note that the D
N
mutation
does not affect the binding of TrCP to degron D
C
.(d) Changes in levels of green fluorescent
protein (EGFP) fused to Gli1 N-terminal residues (top), or EGFP (bottom), in the presence of
cycloheximide (left) or MG132 (right). The amount of fusion protein is identical at t = 0, but
the exposure time for the left and right panels differs to avoid signal saturation. The densi-
tometry is shown to the right with results plotted on a linear scale. The results are represen-
tative of three independent experiments. Error bars are SEM. (e) Coprecipitation assays with
GST-Sufu and lysates from cells containing wild-type or mutant Gli proteins. Note that the
Gli1D
N
mutation leaves Sufu binding intact. (f) Immunofluorescence of Gli1 shows similar
subcellular localization of wild-type and mutant Gli1 proteins in the absence (left) or presence
(right) of the Crm1-inhibitor leptomycin B. (g, left) Luciferase transcription assays of wild-type
and double-mutant Gli1 protein with increasing amounts of transfected moles of plasmid.
Error bars are SEM. Densitometry (middle) of Western blots (right) showing the amount of
steady-state protein accumulation corresponding to the increase in luciferase activity. (h, left)
Luciferase transcription assays of wild-type, single, and double-mutant Gli1 proteins. Error
bars are SEM. Western blot (right) of levels of Gli1 protein in luciferase assay and quantitation
(middle) of protein levels normalized for loading and transfer efficiency determined by immu-
noblot for nuclear pore complex (NPC).
Two destruction signals in Gli proteins
GENES & DEVELOPMENT 279
prior to lysis of all samples in 2× Sample buffer. Both approaches yielded
similar results. In some experiments MG-132 (Calbiochem) was added to
a final concentration of 30 µM 1 h prior to destruction assay. HA-tagged
Gli1 proteins were detected with a mouse-anti-HA monoclonal antibody
(Covance). Equal transfection was confirmed by blotting with a mouse
antibody for EGFP (Roche), and loading and transfer efficiency were con-
firmed by blotting with a mouse antibody to -actin (Sigma).
Primary human keratinocytes. Primary human foreskin keratinocytes
were passaged in Keratinocyte-SFM medium (Invitrogen) supplemented
with bovine pituitary extract and recombinant human EGF (Invitrogen)
and cultured in unsupplemented Keratinocyte-SFM for 24 h prior to the
destruction assay. For destruction assay, recombinant human EGF (In-
vitrogen) was added to a final concentration of 100 ng/mL with cyclo-
heximide to a concentration of 100 µg/mL. Chloroquine (12.5 g/mL;
Sigma) or E64 (25 µM; Calbiochem) were added 1 h prior to beginning the
destruction assay. Samples were harvested at various time points in 2×
Sample buffer. EGFR protein was detected with a rabbit antibody to
EGFR (Santa Cruz Biotechnology). Equal sample loading was confirmed
by blotting for -actin.
Mice
All mouse studies were performed in accordance with the policies of the
Stanford IUPAC. K5Gli2 animals were generated using full-length mouse
Gli2 (Sasaki et al. 1999) containing a triple HA tag on the N terminus in
pENTR1A (Invitrogen) and then recombined into a transgenic vector
containing the bovine keratin 5 promoter (Ramirez et al. 1994; Callahan
et al. 2004) using Gateway cloning (Invitrogen). Five independent lines
were generated that had similar phenotypes. Line #70 was expanded and
quantified. K5Gli1 wild-type, K5Gli1D
C
; D
N
, K5Gli1D
C
, and
K5Gli1D
N
were constructed as described in the Plasmid section in the
Supplemental Material and then recombined
into the bovine keratin 5 promoter by Gateway
cloning. Transgene copy number was deter-
mined by quantitative real-time PCR (Brilliant
Sybr Green; Stratagene) using DNA isolated
from transgenic mouse tails. We used primers
specific to the 3-region of human Gli1 (F: GC
CGTGCTAAAGCTCCAGTGAACAC; R: AG
AAGTCGAGGTGGTGCTGCTGCCC). These
primers did not amplify mouse Gli1. A 10-fold
dilution series of transgene plasmid diluted into
a constant amount of nontransgenic mouse
DNA was used as a standard to determine trans-
gene copy number in a given amount of tail
DNA. Mouse GAPDH (GAPDH F: TCTTCTT
GTGCAGTGCCAGCCTCGTCC; R: GACT
GTGCCGTTGAATTTGCCGTGAGTG) and
mouse Gli2 primers (F: CCTCCCTGG
GAAGAAGACTTGCGTCTAC; R: TCAAT
GCCTTCAACCTTCCGCTCAAC) were used
as controls for DNA loading and quality. Copy
number results are expressed as copies per dip-
loid genome. Expression analysis of transgene
expression was performed by quantitative real-
time RTPCR (Brilliant Sybr Green; Stratagene)
according to the manufacturers instructions.
RNA was isolated from right hind-limb tissue
using Trizol reagent (Invitrogen). Mouse Keratin
5 primers (F: CTCCAGGAACCATCATGT
CTCGCCAGTC; R: CACCACCGAAGCCA
AAGCCACTACCAG) were used to control for
RNA loading and quality. Template quantity was
determined using the deltadelta CT method ac-
cording to the manufacturers instructions.
Acknowledgments
We thank C.A. Callahan for help in making
transgenic Gli2 animals; Lei Chen and the Stan-
ford Transgenic Facility for help with pronuclear
injections; and Paul Khavari, James Chen,
Howard Chang, and the Oro laboratory for com-
ments on the manuscript. This work is funded
by NIH grants R01ARO46786 (to A.E.O) and R01GM60439 (to P.K.J), a
Stanford Graduate Fellowship (to E.G.H.), and the Cancer Biology gradu-
ate program (to I.S.E.).
References
Amit, S., Hatzubai, A., Birman, Y., Andersen, J.S., Ben-Shushan, E.,
Mann, M., Ben-Neriah, Y., and Alkalay, I. 2002. Axin-mediated CKI
phosphorylation of -catenin at Ser 45: A molecular switch for the
Wnt pathway. Genes & Dev. 16: 10661076.
Callahan, C.A. and Oro, A.E. 2001. Monstrous attempts at adnexogen-
esis: Regulating hair follicle progenitors through Sonic hedgehog sig-
naling. Curr. Opin. Genet. Dev. 11: 541546.
Callahan, C., Ofstad, T., Horng, L., Wang, J., Zhen, H., and Oro, A. 2004.
MIM/BEG4, a Sonic hedgehog-responsive gene that potentiates Gli-
dependent transcription. Genes & Dev. 18: 27242729.
Cheng, S.Y. and Bishop, J.M. 2002. Suppressor of Fused represses Gli-
mediated transcription by recruiting the SAP18mSin3 corepressor
complex. Proc. Natl. Acad. Sci. 99: 54425447.
Dai, P., Akimaru, H., Tanaka, Y., Maekawa, T., Nakafuku, M., and Ishii,
S. 1999. Sonic Hedgehog-induced activation of the Gli1 promoter is
mediated by GLI3. J. Biol. Chem. 274: 81438152.
Dai, P., Akimaru, H., and Ishii, S. 2003. A hedgehog-responsive region in
the Drosophila wing disc is defined by debra-mediated ubiquitination
and lysosomal degradation of Ci. Dev. Cell 4: 917928.
Ding, Q., Fukami, S., Meng, X., Nishizaki, Y., Zhang, X., Sasaki, H.,
Dlugosz, A., Nakafuku, M., and Hui, C. 1999. Mouse suppressor of
fused is a negative regulator of Sonic hedgehog signaling and alters
the subcellular distribution of Gli1. Curr. Biol. 9: 11191122.
Grachtchouk, V., Grachtchouk, M., Lowe, L., Johnson, T., Wei, L., Wang,
Figure 4. Removal of two destruction signals rapidly accelerates tumor induction. (a) Dia-
gram of Gli1 wild-type (b,g,l,q), Gli1D
C
D
N
(c,h,m,r), Gli1D
C
(d,i,n,s), Gli1D
N
(e,j,o,t), or
nontransgenic (f,k,p,u) mice analyzed in this study. The number of independent founders is
shown in parentheses. (bf) Representative H&E sections from each founder line. Note BCC-
like lesions from interfollicular epithelium in c and d, and BCC-like tumors from hair follicle
in e. Bar, 50 µm. (gk) In situ hybridization of transgene expression using transgene-specific
gli1 probe. Bar, 25 µm. (lp) Immunohistochemistry with anti-HA antibody for Gli1 protein.
Note the absence of Gli1 protein in wild-type Gli1 transgenics and nuclear and cytoplasmic
distribution in mutant Gli animals. Bar, 10 µm. (qu) In situ hybridization with ptch1 probe
showing Shh target gene induction in tumors. Bar, 10 µm. (v) Table of representative features
of each group of Gli1 transgenic mice.
Huntzicker et al.
280 GENES & DEVELOPMENT
A., de Sauvage, F., and Dlugosz, A.A. 2003. The magnitude of Hedge-
hog signaling activity defines skin tumor phenotype. EMBO J. 22:
27412751.
Hutchin, M.E., Kariapper, M.S., Grachtchouk, M., Wang, A., Wei, L.,
Cummings, D., Liu, J., Michael, L.E., Glick, A., and Dlugosz, A.A.
2005. Sustained Hedgehog signaling is required for basal cell carci-
noma proliferation and survival: Conditional skin tumorigenesis re-
capitulates the hair growth cycle. Genes & Dev. 19: 214223.
Jia, J., Tong, C., Wang, B., Luo, L., and Jiang, J. 2004. Hedgehog signalling
activity of Smoothened requires phosphorylation by protein kinase A
and casein kinase I. Nature 432: 10451050.
Jiang, J. and Struhl, G. 1998. Regulation of the Hedgehog and Wingless
signalling pathways by the F-box/WD40-repeat protein Slimb. Nature
391: 493496.
Kinzler, K.W., Ruppert, J.M., Bigner, S.H., and Vogelstein, B. 1988. The
GLI gene is a member of the Kruppel family of zinc finger proteins.
Nature 332: 371374.
Kogerman, P., Grimm, T., Kogerman, L., Krause, D., Undén, A.B., Sand-
stedt, B., Toftgård, R., and Zaphiropoulos, P.G. 1999. Mammalian
Suppressor-of-fused modulates nuclear-cytoplasmic shuttling of Gli-
1. Nat. Cell Biol. 1: 312319.
Lum, L. and Beachy, P.A. 2004. The Hedgehog response network: Sen-
sors, switches, and routers. Science 304: 17551759.
Margottin-Goguet, F., Hsu, J.Y., Loktev, A., Hsieh, H.M., Reimann, J.D.,
and Jackson, P.K. 2003. Prophase destruction of Emi1 by the
SCF(TrCP/Slimb) ubiquitin ligase activates the anaphase promoting
complex to allow progression beyond prometaphase. Dev. Cell 4:
813826.
Methot, N. and Basler, K. 2001. An absolute requirement for Cubitus
interruptus in Hedgehog signaling. Development 128: 733742.
Mill, P., Mo, R., Fu, H., Grachtchouk, M., Kim, P.C., Dlugosz, A.A., and
Hui, C.C. 2003. Sonic hedgehog-dependent activation of Gli2 is es-
sential for embryonic hair follicle development. Genes & Dev. 17:
282294.
Moshe, Y., Boulaire, J., Pagano, M., and Hershko, A. 2004. Role of Polo-
like kinase in the degradation of early mitotic inhibitor 1, a regulator
of the anaphase promoting complex/cyclosome. Proc. Natl. Acad.
Sci. 101: 79377942.
Nilsson, M., Unden, A., Krause, D., Malmqwist, U., Raza, K., Zaphiro-
poulos, P., and Toftgard, R. 2000. Induction of basal cell carcinomas
and trichoepitheliomas in mice overexpressing Gli-1. Proc. Natl.
Acad. Sci. 97: 34383443.
Oro, A.E. and Higgins, K.M. 2003. Hair cycle regulation of Hedgehog
signal reception. Dev. Biol. 255: 238248.
Oro, A.E., Higgins, K.M., Hu, Z., Bonifas, J.M., Epstein Jr., E.H., and
Scott, M.P. 1997. Basal cell carcinomas in mice overexpressing Sonic
hedgehog. Science 276: 817821.
Pickart, C.M. 2004. Back to the future with ubiquitin. Cell 116: 181190.
Ramirez, A., Bravo, A., Jorcano, J.L., and Vidal, M. 1994. Sequences 5 of
the bovine keratin 5 gene direct tissue- and cell-type-specific expres-
sion of a lacZ gene in the adult and during development. Differen-
tiation 58: 5364.
Reimann, J.D., Freed, E., Hsu, J.Y., Kramer, E.R., Peters, J.M., and Jack-
son, P.K. 2001. Emi1 is a mitotic regulator that interacts with Cdc20
and inhibits the anaphase promoting complex. Cell 105: 645655.
Ruiz i Altaba, A., Sanchez, P., and Dahmane, N. 2002. Gli and Hedgehog
in cancer: Tumours, embryos and stem cells. Nat. Rev. Cancer 2:
361372.
Sasaki, H., Nishizaki, Y., Hui, C., Nakafuku, M., and Kondoh, H. 1999.
Regulation of Gli2 and Gli3 activities by an amino-terminal repres-
sion domain: Implication of Gli2 and Gli3 as primary mediators of
Shh signaling. Development 126: 39153924.
Spencer, E., Jiang, J., and Chen, Z.J. 1999. Signal-induced ubiquitination
of IB by the F-box protein Slimb/-TrCP. Genes & Dev. 13: 284
294.
Taipale, J., Chen, J.K., Cooper, M.K., Wang, B., Mann, R.K., Milenkovic,
L., Scott, M.P., and Beachy, P.A. 2000. Effects of oncogenic mutations
in Smoothened and Patched can be reversed by cyclopamine. Nature
406: 10051009.
von Mering, C. and Basler, K. 1999. Distinct and regulated activities of
human Gli proteins in Drosophila. Curr. Biol. 9: 13191322.
Wang, G., Wang, B., and Jiang, J. 1999. Protein kinase A antagonizes
Hedgehog signaling by regulating both the activator and repressor
forms of Cubitus interruptus. Genes & Dev. 13: 28282837.
Wicking, C., Shanley, S., Smyth, I., Gillies, S., Negus, K., Graham, S.,
Suthers, G., Haites, N., Edwards, M., Wainwright, B., et al. 1997.
Most germ-line mutations in the nevoid basal cell carcinoma syn-
drome lead to a premature termination of the PATCHED protein, and
no genotype-phenotype correlations are evident. Am. J. Hum. Genet.
60: 2126.
Winston, J.T., Strack, P., Beer-Romero, P., Chu, C.Y., Elledge, S.J., and
Harper, J.W. 1999. The SCFTRCPubiquitin ligase complex asso-
ciates specifically with phosphorylated destruction motifs in IB
and -catenin and stimulates IB ubiquitination in vitro. Genes &
Dev. 13: 270283.
Yamasaki, L. and Pagano, M. 2004. Cell cycle, proteolysis and cancer.
Curr. Opin. Cell Biol. 16: 623628.
Yoon, J.W., Liu, C.Z., Yang, J.T., Swart, R., Iannaccone, P., and Walter-
house, D. 1998. Gli activates transcription through a herpes simplex
viral protein 16-like activation domain. J. Biol. Chem. 273: 3496
3501.
Two destruction signals in Gli proteins
GENES & DEVELOPMENT 281
... GLI1 is an unstable protein whose protein abundance is regulated in a proteasome-dependent manner. Consistently, it has been reported that mutations causing the removal of the protein destruction signals of GLI1 result in its accumulation and tumor formation (38). Previously, the E3 ubiquitin-ligase beta-transducin repeat containing E3 ubiquitin (b-TrCP) has been shown to interact with and induce the degradation of GLI1 (38). ...
... Consistently, it has been reported that mutations causing the removal of the protein destruction signals of GLI1 result in its accumulation and tumor formation (38). Previously, the E3 ubiquitin-ligase beta-transducin repeat containing E3 ubiquitin (b-TrCP) has been shown to interact with and induce the degradation of GLI1 (38). Another E3 ubiquitin ligase, Cullin 3 (CUL3), has also been reported to cause the degradation of another GLI family protein, GLI2, in glioblastoma cells (39). ...
Article
Small cell lung cancer (SCLC) is among the most aggressive and lethal human malignancies. Most patients with SCLC who initially respond to chemotherapy develop disease relapse. Therefore, there is a pressing need to identify novel driver mechanisms of SCLC progression to unlock treatment strategies to improve patient prognosis. SCLC cells comprise subsets of cells possessing progenitor or stem cell properties, while the underlying regulatory pathways remain elusive. Here, we identified the isoform 1 of the neurogenesis-associated protein ASPM (ASPM-I1) as a prominently upregulated stemness-associated gene during the self-renewal of SCLC cells. The expression of ASPM-I1 was found to be upregulated in SCLC cells and tissues, correlated with poor patient prognosis, and indispensable for SCLC stemness and tumorigenesis. A reporter array screening identified multiple developmental signaling pathways, including Hedgehog (Hh) and Wnt pathways, whose activity in SCLC cells depended upon ASPM-I1 expression. Mechanistically, ASPM-I1 stabilized the Hh transcriptional factor GLI1 at the protein level through a unique exon-18–encoded region by competing with the E3 ligases β-TrCP and CUL3. In parallel, ASPM-I1 sustains the transcription of the Hh pathway transmembrane regulator SMO through the Wnt−DVL3−β-catenin signaling axis. Functional studies verified that the ASPM-I1–regulated Hh and Wnt activities significantly contributed to SCLC aggressiveness in vivo. Consistently, the expression of ASPM-I1 positively correlated with GLI1 and stemness markers in SCLC tissues. This study illuminates an ASPM-I1–mediated regulatory module that drives tumor stemness and progression in SCLC, providing an exploitable diagnostic and therapeutic target. Significance ASPM promotes SCLC stemness and aggressiveness by stabilizing the expression of GLI1, DVL3, and SMO, representing a novel regulatory hub of Hh and Wnt signaling and targetable vulnerability.
... The secreted Shh binds to the Patched1 receptor and relieves repression of the 12-transmembrane protein Smoothened (Smo), resulting in activation of the Gli family of transcription factors. Since Gli1 could be degraded via CRLsmediated UPS [25], we guessed that MLN4924 might act through upregulating Gli1 and thus activating Shh pathway. But contrary to our hypothesis, both the transcription and expression of Gli1 were decreased in PSCs when treated with MLN4924 (Fig. 2a). ...
... Although Gli1 is regulated by CRLs-mediated degradation [39], the experimental results are contrary to our original hypothesis that Gli1 is elevated after MLN4924 treatment, and it is speculated that other regulation mechanisms might be involved in the reduction of Gli1, and some further researches are need. It is reported that Shh overexpression or Gli1 protein accumulation promotes tumor growth and contributes to the invasiveness of pancreatic cancer [25,40], but Simon et al. [41] also reported that Gli1 inhibition promotes epithelial-to-mesenchymal transition in pancreatic cancer cells, so the overall effects of Gli1 inhibition in PDAC by MLN4924 should be evaluated more comprehensively. ...
Article
Full-text available
PurposePancreatic cancer is characterized by a dense desmoplasia stroma, which hinders efficient drug delivery and plays a critical role in tumor progression and metastasis. MLN4924 is a first-in-class NEDD8-activating enzyme inhibitor that exhibits anti-tumor activities toward pancreatic cancer, and given the comprehensive effects that MLN4924 could have, we ask what impact MLN4924 would have on the stroma of pancreatic cancer and its underlying mechanisms.Methods Primary pancreatic stellate cells (PSCs) and human HMEC-1 cells were treated with MLN4924 in vitro. The proliferation and extracellular matrix protein levels of PSCs were tested, and their relationship with transcription factor Gli1 in PSCs was investigated. The angiogenic phenotypes of HMEC-1 cells were evaluated using capillary-like tube formation assay, and their relationship with REDD1 in HMEC-1 cells was investigated.ResultsIn this study, we found that MLN4924 inhibited the proliferation of pancreatic stellate cells and their secretion of collagen and CXCL-1, and the collagen secretion inhibiting effect of MLN4924 was related with transcription factor Gli1. MLN4924 inhibited multiple angiogenic phenotypes of HMEC-1 cells, and mTOR agonist partially relieved the inhibition of MLN4924 on HEMCs. MLN4924 increased the expression of REDD1 and REDD1 knockdown promoted the angiogenic phenotypes of HMEC-1 cells.Conclusions Our study suggests that MLN4924 inhibits both the tumor stroma and angiogenesis in pancreatic cancer, and the inhibition effect is related with Gli1 in pancreatic stellate cells and REDD1 in vascular endothelial cells, respectively.
... Ubiquitylation processes are crucial events by which GLI1 activity is finely regulated [38,[51][52][53][54][55][56][57][58][59][60]. Of note, we previously reported that the CRL3 REN E3 ligase complex finely suppresses GLI1 functions by promoting ubiquitylation and degradation of HDAC1, a strong activator of the SHH pathway [6]. ...
Article
Full-text available
BACKGROUND Medulloblastoma (MB) is the most common malignant pediatric brain tumor arising from alterations in cerebellum development. The Sonic Hedgehog variant (SHH MB) is the best genetically characterized, however the molecular mechanisms responsible for its pathogenesis are not fully understood and therapeutic benefits are still limited. Mutations in key components of the SHH pathway and cytogenetic alterations (such as chromosome 17p deletion) lay the pathogenetic foundation for SHH MB subgroup. RENKCTD11 (here REN) is a tumor suppressor and substrate-receptor subunit of the Cullin3 Ring E3 ubiquitin ligase (CRL3) complex mapping on chromosome 17p and frequently deleted in SHH MB. We wished to expand the knowledge of CRL3REN activity to unveil molecular circuitries whose deregulation can contribute to SHH MB onset and to uncover novel therapeutic targets. METHODS Affinity purification coupled to mass spectrometry was performed to identify new REN interactors. Co-immunoprecipitation and ubiquitylation assays validated proteomic data. The proliferation of primary murine SHH MB cells and Patient Derived Xenografts (PDXs) models was evaluated in vitro by using the IncuCyte imaging system. Heterotopic and orthotopic allograft in vivo models of SHH MB were established in immunodeficient mice. RESULTS Proteomic analysis revealed that the pro-oncogenic stemness Spalt-like transcriptional factor 4 (SALL4) interacts with REN. We found that SALL4 is re-expressed in mouse SHH MB models and its high levels correlate with worse overall survival in SHH MB patients. We demonstrate that CRL3REN induces polyubiquitylation and degradation of wild type SALL4, but not of a SALL4 mutant lacking zinc finger cluster 1 domain (ΔZFC1). Interestingly, SALL4 binds GLI1 and cooperates with HDAC1 to potentiate GLI1 deacetylation and transcriptional activity. Notably, inhibition of SALL4 suppress SHH MB growth both in murine and PDX models. CONCLUSIONS Our findings identify SALL4 as a CRL3REN substrate and a promising therapeutic target in SHH-dependent cancers.
... A C-terminal motif of Gli1, DSGVEM, has been identified to stabilize the Gli1 protein and to rapidly accelerate the tumor formation 29 . AMP-activated protein kinase (AMPK) phosphorylates Ser102-, Ser408-, and Thr1074-GLI1, leading to its ubiquitinated degradation via β-TrCP 30,31 , and the proteasome co-activator TRIM16 regulates GLI1 intracellular levels by directly mediating GLI1 ubiquitination degradation 32 . ...
Article
Full-text available
Aberrant activation of sonic hedgehog (SHH) signaling and its effector transcriptional factor GLI1 are essential for oncogenesis of SHH-dependent medulloblastoma (MBSHH) and basal cell carcinoma (BCC). Here, we show that SHH inactivates p38α (MAPK14) in a smoothened-dependent manner, conversely, p38α directly phosphorylates GLI1 on Ser937/Ser941 (human/mouse) to induce GLI1’s proteasomal degradation and negates the transcription of SHH signaling. As a result, Gli1S941E loss-of-function knock-in significantly reduces the incidence and severity of smoothened-M2 transgene-induced spontaneous MBSHH, whereas Gli1S941A gain-of-function knock-in phenocopies Gli1 transgene in causing BCC-like proliferation in skin. Correspondingly, phospho-Ser937-GLI1, a destabilized form of GLI1, positively correlates to the overall survival rate of children with MBSHH. Together, these findings indicate that SHH-induced p38α inactivation and subsequent GLI1 dephosphorylation and stabilization in controlling SHH signaling and may provide avenues for future interventions of MBSHH and BCC.
... Ubiquitylation processes are crucial events by which GLI1 activity is finely regulated [38,[51][52][53][54][55][56][57][58][59][60]. Of note, we previously reported that the CRL3 REN E3 ligase complex finely suppresses GLI1 functions by promoting ubiquitylation and degradation of HDAC1, a strong activator of the SHH pathway [6]. ...
Article
Full-text available
The Sonic Hedgehog (SHH) pathway is crucial regulator of embryonic development and stemness. Its alteration leads to medulloblastoma (MB), the most common malignant pediatric brain tumor. The SHH-MB subgroup is the best genetically characterized, however the molecular mechanisms responsible for its pathogenesis are not fully understood and therapeutic benefits are still limited. Here, we show that the pro-oncogenic stemness regulator Spalt-like transcriptional factor 4 (SALL4) is re-expressed in mouse SHH-MB models, and its high levels correlate with worse overall survival in SHH-MB patients. Proteomic analysis revealed that SALL4 interacts with REN/KCTD11 (here REN), a substrate receptor subunit of the Cullin3-RING ubiquitin ligase complex (CRL3 REN ) and a tumor suppressor lost in ~30% of human SHH-MBs. We demonstrate that CRL3 REN induces polyubiquitylation and degradation of wild type SALL4, but not of a SALL4 mutant lacking zinc finger cluster 1 domain (ΔZFC1). Interestingly, SALL4 binds GLI1 and cooperates with HDAC1 to potentiate GLI1 deacetylation and transcriptional activity. Notably, inhibition of SALL4 suppresses SHH-MB growth both in murine and patient-derived xenograft models. Our findings identify SALL4 as a CRL3 REN substrate and a promising therapeutic target in SHH-dependent cancers.
... In addition, we found three tumor-derived mutations in this region-R100C, S105F, and L124P-that also disrupted SUFU binding. Interestingly, several other important functional motifs are also found in this region, including a degradation signal that spans the entire region (Huntzicker et al, 2006) and a ciliary localization signal that partially overlaps with the D-site and with the S70 and S86 MAPK target sites (Han et al, 2017). Other kinases also phosphorylate GLI1 in this region (Wang et al, 2012;Li et al, 2015;Schneider et al, 2015). ...
Article
Full-text available
Crosstalk between the Hedgehog and MAPK signaling pathways occurs in several types of cancer and contributes to clinical resistance to Hedgehog pathway inhibitors. Here we show that MAP kinase-mediated phosphorylation weakens the binding of the GLI1 transcription factor to its negative regulator SUFU. ERK2 phosphorylates GLI1 on three evolutionarily conserved target sites (S102, S116, and S130) located near the high-affinity binding site for SUFU; these phosphorylations cooperate to weaken the affinity of GLI1–SUFU binding by over 25-fold. Phosphorylation of any one, or even any two, of the three sites does not result in the level of SUFU release seen when all three sites are phosphorylated. Tumor-derived mutations in R100 and S105, residues bordering S102, also diminish SUFU binding, collectively defining a novel evolutionarily conserved SUFU affinity–modulating region. In cultured mammalian cells, GLI1 variants containing phosphomimetic substitutions of S102, S116, and S130 displayed an increased ability to drive transcription. We conclude that multisite phosphorylation of GLI1 by ERK2 or other MAP kinases weakens GLI1-SUFU binding, thereby facilitating GLI1 activation and contributing to both physiological and pathological crosstalk.
Article
Full-text available
The Sonic Hedgehog (Shh) signalling pathway plays a critical role in normal development and tissue homeostasis, guiding cell differentiation, proliferation, and survival. Aberrant activation of this pathway, however, has been implicated in the pathogenesis of various cancers, largely due to its role in regulating cancer stem cells (CSCs). CSCs are a subpopulation of cancer cells with the ability to self-renew, differentiate, and initiate tumour growth, contributing significantly to tumorigenesis, recurrence, and resistance to therapy. This review focuses on the intricate activity of the Shh pathway within the context of CSCs, detailing the molecular mechanisms through which Shh signalling influences CSC properties, including self-renewal, differentiation, and survival. It further explores the regulatory crosstalk between the Shh pathway and other signalling pathways in CSCs, highlighting the complexity of this regulatory network. Here, we delve into the upstream regulators and downstream effectors that modulate Shh pathway activity in CSCs. This review aims to cast a specific focus on the role of the Shh pathway in CSCs, provide a detailed exploration of molecular mechanisms and regulatory crosstalk, and discuss current and developing inhibitors. By summarising key findings and insights gained, we wish to emphasise the importance of further elucidating the interplay between the Shh pathway and CSCs to develop more effective cancer therapies.
Article
Cashmere, a highly valuable animal product derived from cashmere goats, holds significant economic importance. MiRNAs serve as crucial regulators in the developmental processes of mammalian hair follicles. Understanding the regulation of miRNAs during the hair follicle cycle is essential for enhancing cashmere quality. In this investigation, we employed high-throughput sequencing technology to analyze the expression profiles of miRNAs in the secondary hair follicles of Jiangnan cashmere goats at different stages. Through bioinformatics analysis, we identified differentially expressed miRNAs (DE miRNAs). The regulatory relationships between miRNAs and their target genes were verified using multiple techniques, including RT-qPCR, Western Blot, Dual-Luciferase Reporter, and CKK-8 assays. Our findings revealed the presence of 193 DE miRNAs during various stages of the hair follicle cycle in Jiangnan cashmere goats. Based on the previously obtained mRNA data, the target genes of DE miRNA were predicted, and 1472 negative regulatory relationships between DE miRNAs and target genes were obtained. Notably, the expression of chi-miR-877-3p was down-regulated during the telogen (Tn) phase compared to the anagen (An) and catagen (Cn) phases, while the IGFBP5 gene exhibited up-regulation. Further validation experiments confirmed that overexpression of chi-miR-877-3p in dermal papilla cells (DPCs) suppressed IGFBP5 gene expression and facilitated cell proliferation. The results of this study provide novel insights for analyzing the hair follicle cycle.
Article
Gastric cancer (GC) is a highly prevalent and aggressive malignancy with a poor prognosis. Recent evidence suggested that metallothionein 1 M (MT1M) may play a critical role in cancer development, progression, and drug resistance; however, its role in GC remains largely unknown. In this study, we investigated the expression and function of MT1M in GC both in vitro and in vivo. We found that MT1M expression was significantly downregulated in GC tissues and cell lines. Decreased expression of MT1M was associated with worse clinical prognosis, particularly in patients treated with 5-fluorouracil. Low expression of MT1M was indicative of poor overall survival (OS, HR 0.56 [95% CI 0.37-0.84], P < 0.005), first progression survival (FP, HR 0.54 [95% CI 0.36-0.79], P < 0.005), and post-progression survival (PPS, HR 0.65 [95% CI 0.45-0.94], P < 0.05). We also demonstrated that overexpression of MT1M inhibited cell proliferation and induced apoptosis in GC cells and in tumor xenografts, and it improved chemosensitivity to 5-fluorouracil. Furthermore, we found that MT1M overexpression could inhibit stem cell characteristics by targeting GLI1 and affecting GLI1 ubiquitination. Collectively, these findings indicated that MT1M may act as a tumor suppressor in GC and could serve as a potential therapeutic target to attenuate stemness and chemotherapy resistance of GC.
Article
Full-text available
Basal cell carcinoma, medulloblastoma, rhabdomyosarcoma and other human tumours are associated with mutations that activate the proto-oncogene Smoothened (SMO) or that inactivate the tumour suppressor Patched (PTCH). Smoothened and Patched mediate the cellular response to the Hedgehog (Hh) secreted protein signal, and oncogenic mutations affecting these proteins cause excess activity of the Hh response pathway1, 2. Here we show that the plant-derived teratogen cyclopamine, which inhibits the Hh response3, 4, is a potential 'mechanism-based' therapeutic agent for treatment of these tumours. We show that cyclopamine or synthetic derivatives with improved potency block activation of the Hh response pathway and abnormal cell growth associated with both types of oncogenic mutation. Our results also indicate that cyclopamine may act by influencing the balance between active and inactive forms of Smoothened.
Article
Full-text available
Mutations in the tumor suppressor gene PATCHED (PTC) are found in human patients with the basal cell nevus syndrome, a disease causing developmental defects and tumors, including basal cell carcinomas. Gene regulatory relationships defined in the fruit fly Drosophila suggest that overproduction of Sonic hedgehog (SHH), the ligand for PTC, will mimic loss of ptc function. It is shown here that transgenic mice overexpressing SHH in the skin develop many features of basal cell nevus syndrome, demonstrating that SHH is sufficient to induce basal cell carcinomas in mice. These data suggest that SHH may have a role in human tumorigenesis.
Article
Full-text available
Three proteins have been identified in mammals, GLI, GLI2, and GLI3, which share a highly conserved zinc finger domain with Drosophila Cubitus interruptus and are believed to function as transcription factors in the vertebrate Sonic hedgehog-Patched signaling pathway. To understand the role GLI plays in the Sonic hedgehog-Patched pathway and mechanisms of GLI-induced transcriptional regulation, we have characterized its transcriptional regulatory properties and contributions of specific domains to transcriptional regulation. We have demonstrated that GLI activates expression of reporter constructs in HeLa cells in a concentration-dependent manner through the GLI consensus binding motif and that a GAL4 binding domain-GLI fusion protein activates reporter expression through the GAL4 DNA binding site. GLI-induced transcriptional activation requires the carboxyl-terminal amino acids 1020–1091, which includes an 18-amino acid region highly similar to the α-helical herpes simplex viral protein 16 activation domain, including the consensus recognition element for the human TFIID TATA box-binding protein-associated factor TAFII31 and conservation of all three amino acid residues believed to contact directly chemically complementary residues in TAFII31. The presence of this region in the GLI activation domain provides a mechanism for GLI-induced transcriptional regulation.
Article
Ubiquitin-mediated proteolysis has a central role in controlling the intracellular levels of several important regulatory molecules such as cyclins, CKIs, p53, and IκBα. Many diverse proinflammatory signals lead to the specific phosphorylation and subsequent ubiquitin-mediated destruction of the NF-κB inhibitor protein IκBα. Substrate specificity in ubiquitination reactions is, in large part, mediated by the specific association of the E3- ubiquitin ligases with their substrates. One class of E3 ligases is defined by the recently described SCF complexes, the archetype of which was first described in budding yeast and contains Skp1, Cdc53, and the F-box protein Cdc4. These complexes recognize their substrates through modular F-box proteins in a phosphorylation-dependent manner. Here we describe a biochemical dissection of a novel mammalian SCF complex, SCF(β-TRCP), that specifically recognizes a 19-amino-acid destruction motif in IκBα (residues 21-41) in a phosphorylation-dependent manner. This SCF complex also recognizes a conserved destruction motif in β-catenin, a protein with levels also regulated by phosphorylation-dependent ubiquitination. Endogenous IκBα-ubiquitin ligase activity cofractionates with SCF(β-TRCP). Furthermore, recombinant SCF(β-TRCP) assembled in mammalian cells contains phospho-IκBα-specific ubiquitin ligase activity. Our results suggest that an SCF(β-TRCP) complex functions in multiple transcriptional programs by activating the NF-κB pathway and inhibiting the β-catenin pathway.
Article
Many studies have established that a select subset of normal cellular genes are altered in cancer by point mutations, translocations or gene amplification. However, the vast majority of genetic changes that occur in neoplastic cells have not yet been identified. In an attempt to identify some of these other genetic changes, we have recently isolated a gene, GLI, by virtue of its amplification in a human glioblastoma. Subsequently, GLI was found to be amplified in other human glioblastomas (ref. 3 and unpublished data). To understand better the role of GLI in human neoplasia, we have now cloned the GLI complementary DNA (cDNA) and determined its nucleotide sequence. Analysis of the predicted translation product reveals that it contains five repeats of a DNA binding consensus sequence (zinc finger) originally described in Xenopus Transcription Factor III A (TFIIIA). Furthermore, these zinc fingers contain sequence elements that suggest the GLI gene product is a member of the recently described Kruppel family of zinc finger proteins. Additional experiments demonstrate that GLI is an evolutionarily conserved gene that is expressed in embryonal carcinoma cells but not in most adult tissues. The link between the developmentally important Kruppel family of genes and GLI is interesting considering the similarities between developing embryonic and neoplastic tissue.
Article
Expression of keratin K5 (and K14) in multilayered epithelia occurs predominantly in the basal layer of proliferating keratinocytes. When a keratinocyte becomes committed to terminal differentiation, it moves out of the basal layer towards the epithelial surface. As part of this program of terminal differentiation, the expression of K5 (and K14) is downregulated in suprabasal cells, and new pairs of differentiation-specific keratins are expressed. To define the cis-acting DNA sequences required for K5 cell-type- and differentiation-specific expression, chimeric gene fusions between portions of the bovine keratin K5 locus and the Escherichia coli lacZ gene were used to generate transgenic mice. In the genomic fragment consisting of 5.3 kb of 5' flanking sequences, 6.1 kb corresponding to the body of the gene and 4.5 kb of 3' flanking sequences, the subfragment extending from -5300 bp to +138 bp was the smaller region that directed lacZ expression to stratified epithelia in a manner analogous to the endogenous keratin K5. Proximal sequences from -1300 bp to +138 bp were inactive. We also determined the expression pattern of keratin K5 during mouse development using an antiserum specific for mouse keratin K5. Expression was first detected in ectodermal cells of 11.5 days postcoitum embryos, and from day 13.5 postcoitum onwards K5 was detected in the precursors of most epithelia and organs which express K5 at adult stages. This pattern was reproduced, with few differences, by the construct with sequences from -5300 bp to +138 bp fused to the lacZ gene. These findings identify sequences between -5.3 kb and -1.3 kb of the bovine K5 gene as being important for cell-type- and differentiation-specific gene expression both during mouse development and in the adult.
Article
The human homologue of the Drosophila segment polarity gene patched is implicated in the development of nevoid basal cell carcinoma syndrome (NBCCS) and in the genesis of sporadic basal cell carcinomas. In order to examine the phenotypic variability in NBCCS and to highlight functionally important domains of the PTCH protein, we have now screened 71 unrelated NBCCS individuals for mutations in the PTCH exons. We identified 28 mutations that are distributed throughout the entire gene, and most (86%) cause protein truncation. As part of this analysis, we demonstrate that failure of one NBCCS family to show clear linkage to chromosome 9q22.3-31 is most likely due to germinal mosaicism. We have identified three families bearing identical mutations with variable phenotypes, suggesting phenotypic variability in NBCCS is a complex genetic event. No phenotype genotype correlation between the position of truncation mutations and major clinical features was evident. Two missense mutations have been identified, and their location within transmembrane domains supports the notion that PTCH may have a transport function. The preponderance of truncation mutants in the germ line of NBCCS patients suggests that the developmental defects associated with the disorder are most likely due to haploinsufficiency.
Article
Members of the Hedgehog (Hh) and Wnt/Wingless (Wg) families of secreted proteins control many aspects of growth and patterning during animal development. Hh signal transduction leads to increased stability of a transcription factor, Cubitus interruptus (Ci), whereas Wg signal transduction causes increased stability of Armadillo (Arm/beta-catenin), a possible co-factor for the transcriptional regulator Lef1/TCF. Here we describe a new gene, slimb (for supernumerary limbs), which negatively regulates both of these signal transduction pathways. Loss of function of slimb results in a cell-autonomous accumulation of high levels of both Ci and Arm, and the ectopic expression of both Hh- and Wg- responsive genes. The slimb gene encodes a conserved F-box/WD40-repeat protein related to Cdc4p, a protein in budding yeast that targets cell-cycle regulators for degradation by the ubiquitin/proteasome pathway. We propose that Slimb protein normally targets Ci and Arm for processing or degradation by the ubiquitin/proteasome pathway, and that Hh and Wg regulate gene expression at least in part by inducing changes in Ci and Arm, which protect them from Slimb-mediated proteolysis.
Article
Signal-induced phosphorylation of IkappaBalpha targets this inhibitor of NF-kappaB for ubiquitination and subsequent degradation, thus allowing NF-kappaB to enter the nucleus to turn on its target genes. We report here the identification of an IkappaB-ubiquitin (Ub) ligase complex containing the F-box/WD40-repeat protein, beta-TrCP, a vertebrate homolog of Drosophila Slimb. beta-TrCP binds to IkappaBalpha only when the latter is specifically phosphorylated by an IkappaB kinase complex. Moreover, immunopurified beta-TrCP ubiquitinates phosphorylated IkappaBalpha at specific lysines in the presence of Ub-activating (E1) and -conjugating (Ubch5) enzymes. A beta-TrCP mutant lacking the F-box inhibits the signal-induced degradation of IkappaBalpha and subsequent activation of NF-kappaB-dependent transcription. Furthermore, Drosophila embryos deficient in slimb fail to activate twist and snail, two genes known to be regulated by the NF-kappaB homolog, Dorsal. These biochemical and genetic data strongly suggest that Slimb/beta-TrCP is the specificity determinant for the signal-induced ubiquitination of IkappaBalpha.
Article
Ubiquitin-mediated proteolysis has a central role in controlling the intracellular levels of several important regulatory molecules such as cyclins, CKIs, p53, and IkappaBalpha. Many diverse proinflammatory signals lead to the specific phosphorylation and subsequent ubiquitin-mediated destruction of the NF-kappaB inhibitor protein IkappaBalpha. Substrate specificity in ubiquitination reactions is, in large part, mediated by the specific association of the E3-ubiquitin ligases with their substrates. One class of E3 ligases is defined by the recently described SCF complexes, the archetype of which was first described in budding yeast and contains Skp1, Cdc53, and the F-box protein Cdc4. These complexes recognize their substrates through modular F-box proteins in a phosphorylation-dependent manner. Here we describe a biochemical dissection of a novel mammalian SCF complex, SCFbeta-TRCP, that specifically recognizes a 19-amino-acid destruction motif in IkappaBalpha (residues 21-41) in a phosphorylation-dependent manner. This SCF complex also recognizes a conserved destruction motif in beta-catenin, a protein with levels also regulated by phosphorylation-dependent ubiquitination. Endogenous IkappaBalpha-ubiquitin ligase activity cofractionates with SCFbeta-TRCP. Furthermore, recombinant SCFbeta-TRCP assembled in mammalian cells contains phospho-IkappaBalpha-specific ubiquitin ligase activity. Our results suggest that an SCFbeta-TRCP complex functions in multiple transcriptional programs by activating the NF-kappaB pathway and inhibiting the beta-catenin pathway.