Download as docx, pdf, or txt
Download as docx, pdf, or txt
You are on page 1of 7

Small-molecule modulators of Hedgehog signaling:

identification and characterization of Smoothened


agonists and antagonists

Background
The Hedgehog signaling pathway is vital to animal development and can be
activated to facilitate tissue maintenance and repair. However, little is known about
the communication between the pathway suppressor Patched and the pathway
activator Smoothened.
Results
A synthetic non-peptidyl small molecule, Hh-Ag, promotes cell-type-specific
proliferation and concentration-dependent differentiation in vitro and in utero, and
rescues aspects of the Hh-signaling defect in Sonic hedgehog-null, but not Smo-null,
mouse embryos.
Conclusions
Smo can be modulated by synthetic small molecules, raising the possibility that Hh
signaling may be regulated by endogenous small molecules.

Background
The hedgehog (hh) gene family encodes highly conserved secreted signaling
proteins that are responsible for controlling cell fate determination throughout
development and into adulthood. There are three Hh-family proteins in mammals:
Sonic (Shh), Indian (Ihh), and Desert (Dhh).
In vitro culture systems of neuronal tissues have been used to characterize the
biology of the Hh-signaling pathway. These studies have shown that Hh signals
promote proliferation of granule neuron precursors, which is implicated in the biology
of certain cancers.
The Hh-signaling pathway comprises three main components: the Hh ligand, a
transmembrane receptor circuit, and a cytoplasmic complex. There is positive and
negative feedback at the transcriptional level.
Smo and Ptc are transmembrane proteins that interact with each other and with the
Hh pathway. Ptc inhibits Smo, but it is not clear how Smo communicates with the
Ci/Gli transcription factor complex.
Through a chemical genetic approach, we hoped to uncover some of the
complexities of the Hh-signaling system. We used the plant-derived Hh antagonist
cyclopamine and a synthetic small-molecule Hh-signaling inhibitor, Cur61414.
Although genetic manipulations of Smo have demonstrated that the pathway can be
activated independently of Hh ligand, no small molecules with this capability have
been identified.
A small-molecule pathway activator would need to be capable of interfering with the
inhibitory effect that Ptc exerts on Smo, activating Smo without affecting Ptc, or
activating the pathway downstream of Smo.
A non-peptidyl small-molecule agonist of Hh signaling has been identified that
bypasses the Ptc-regulatory step and interacts directly with Smo. This suggests that
Smo can be activated or inhibited by direct interaction with small-molecule ligands.
Isolation of Hh agonists by high-throughput screening
We established a mammalian-cell-based assay to identify small-molecule agonists of
Hh signaling. We screened 140,000 synthetic compounds at a concentration of 2-5
M and isolated several putative agonists, including Hh-Ag 1.1, which exhibited half-
maximal stimulation at around 3 mM and an activation maximum of approximately
35% compared to the Hh protein control.
Chemical modifications increase potency
Over 300 derivatives of Hh-Ag 1.1 were synthesized and tested in the cell-based
reporter assay. The most potent derivative, Hh-Ag 1.5, had an EC50 of
approximately 1 nM, and was increased over 1000-fold by chemical modification.
Proliferation activity of the agonist
A study showed that primary neonatal cerebellar granule neuron precursors
proliferate in response to Hh stimulation. The study also demonstrated that the Hh
agonists can elicit a biological response in primary cells similar to that produced by
Hh protein.
Morphogenic activity of the agonist
The expression of three transcription factors, Pax7, MNR2 and Nkx2.2, in response
to increasing concentrations of Hh protein in the intermediate region of the chick
neural plate was assayed. This result demonstrated that the Hh agonist mimics the
concentration-dependent inductive activity of Hh protein on neural precursors.
Activity of the agonist in vivo
We developed an in vivo assay for the Hh agonist that would allow us to test its
activity in Shh- and Smo-mutant mouse embryos in utero. The agonist compound
effectively activated Hh signaling in vivo following oral administration.
Agonist site of action in vivo
Pregnant mice from Shh+/- and Smo+/- intercrosses were treated with agonist at 6.5
and 7.5 dpc, and embryos were collected at 6-8 somite stages (E8.5). Agonist
treatment dramatically enhanced and expanded the expression of Ptc1 in these
heterozygous embryos, consistent with what we have observed in wild-type
embryos.
Shh-/- and Smo-/- embryos showed fused ventral lips of the cephalic folds and a
single continuous optic vesicle, indicating lack of a clearly defined midline. Ptc1
expression was only seen in lateral plate mesoderm and weakly in somites.
Following agonist treatment, Shh-/- embryos had greater neural tube and somite
expression of Ptc1, and the midline defects were at least partly rescued by agonist
treatment. However, Smo-/- embryos had no detectable effect on either morphology
or Ptc1 expression.
Chemical epistasis studies
We sought to determine the level at which the agonist acts in the Hh pathway, by
conducting competition experiments between the agonist and known Hh- signaling
antagonists that block the pathway at different levels.
The Hh-blocking antibody 5E1 had no effect on pathway activation by the agonist,
while forskolin, cyclopamine and Cur61414 were all inhibitory. The agonist appears
to act downstream of the Hh-Ptc interaction, while cyclopamine and Cur61414 may
act at a similar level in the Hh-signaling cascade.
Regulation of Ptc and Smo by Hh protein and Hh agonist
Recent work in Drosophila tissue culture has shown that endogenous Ptc and Smo
proteins are differentially affected by the addition of Hh to the growth medium. We
generated stable cell lines expressing two epitope-tagged proteins, Ptc coupled to
green fluorescent protein, and Smo coupled to a fragment of influenza
hemagglutinin, HA-Smo.
Figure 5b shows that both Hh protein and the small-molecule agonist cause the
accumulation of HA-Smo protein in cells, but only Hh protein can destabilize Ptc. The
agonist is fully capable of activating the full signaling pathway.
Binding in whole cells
The agonist modulates Smo levels and may activate Hh signaling by directly binding
Smo. A tritiated form of the agonist analog Hh-Ag 1.5 could form a complex with
Smo in 293T cells.
Incubation of cells with Hh-Ag 1.5 or an Hh-Ag 1.1 derivative agonist resulted in the
complete absence of counts in the immunocomplex, suggesting that a stable,
specific interaction can form between Smo and the Hh agonist.
The Hh-pathway antagonists cyclopamine and Cur61414 block signaling in a Ptc-
independent manner and therefore may act directly on Smo. The Hh-signaling
inhibitors, but not structurally related inactive compounds, can significantly compete
out binding of [3H]-Hh-Ag 1.5 to Smo-expressing cells.
We synthesized a derivative of Hh-Ag 1.2 with a photoactivatable crosslinker and
subsequently UV-irradiated 293T cells to initiate crosslinking. The resulting
immunocomplexes showed crosslinking exclusively to HA-Smo, but with an
efficiency of less than 1%.
Cell-free membrane-binding assays
To test whether the Hh agonist could interact with Smo in vitro, we transiently
overexpressed Smo, Ptc, rat 2-adrenergic receptor and GFP in 293T cells, harvested
membranes and performed a filtration membrane-binding assay.
To assess the specificity of binding, we added a 1000-fold excess of unlabeled
agonist to the binding assay, and the addition of an unlabeled, inactive Hh agonist
did not compete out the binding of [3H]-Hh-Ag 1.5 to the Smo-containing
membranes.
We tested whether Ptc-independent Hh antagonists could selectively compete with
the interaction of the Hh agonist to Smo membranes. The data show that the Hh-
signaling inhibitors, but not structurally related inactive compounds, can significantly
compete with the interaction.
Kinetics, saturation and competition binding analysis
We generated association, dissociation and saturation-binding curves for the
interaction of the Hh agonist and Smo, and used control membranes to define the
non-specific level.
We performed a kinetic analysis to establish the reversibility of the binding reaction
and the approximate incubation time required for equilibrium binding studies. The
results demonstrate that equilibrium binding will require binding reaction times of
approximately 50 hours.
We performed a saturation binding experiment with [3H]-Hh-Ag 1.5 to establish total,
nonspecific, and specific binding curves. The data best fit a simple one-site binding
model with a predicted Kd of 0.37 nM for Hh-Ag 1.5.
We performed a competition assay using several agonist derivatives across a range
of concentrations (0.01 nM to 1 M). The results show that the binding of Hh-Ag 1.5,
Hh-Ag 1.3, Hh-Ag 1.2, Hh-Ag 1.1, and the signaling-inactive Hh-Ag 1.1 derivative to
Smo-containing membranes is competitive.
We compared binding of KAAD-cyclopamine, Cur61414 and Hh-Ag 1.5 to a
constitutively active mutant of Smo (Smoact) or to wild-type Smo (Smowt) to
determine if the Hh agonist binds Smoact with a higher affinity.
We performed competition binding studies using Hh-Ag 1.5, KAAD-cyclopamine and
Cur61414 on Smoact- and Smowt-containing membranes. The results strongly
support the model that the reduced potency of Hh antagonists on Smoact-expressing
cells is directly due to a reduced affinity of the antagonist for the mutated Smo
protein.

Discussion
Hh signal transduction has been the focus of intense research over the past decade
due to its central role in development and its emerging biomedical relevance. We
identified potent small molecule agonists of Hh signaling and two Ptc-independent
inhibitors of Hh signaling.
Models of Smo-ligand interaction
The mutation in Smoact indirectly influences ligand binding by creating a change in
the normal equilibrium between the different conformations of Smo. A simple two-
state model predicts that the agonist and antagonist compete for the same site on
Smo to activate or inactivate Hh-pathway signaling. A ternary complex model
suggests that there are two independent binding sites on Smowt, one specific for the
agonist and another specific for antagonists. A signaling pathway coupler, or
effector, is proposed to bind activated Smowt and trigger signal transduction. The
coupler/effector complex resides in a stable conformation in the absence of agonist.
We propose a variation of the classic 'ternary complex model' to explain the results
of our experiments, in which the agonist binds and stabilizes an active signaling state
of Smo while the antagonists bind and stabilize an inactive form.
This model proposes that Hh signaling involves Smo, an undefined coupler/effector,
and a gain-of-function mutant form of Smo that adopts an abnormal conformation.
Activating Hh signaling through the GPCR-like Smo receptor
GPCRs are considered excellent drug targets because they are often regulated
through interactions with small natural ligands. A number of compound classes have
been isolated based on their ability to compete for the binding of GPCRs by their
natural ligands.
Many GPCRs have allosteric sites that can bind small molecules in addition to the
orthosteric sites. These allosteric sites can be used to modulate activity of a receptor
without directly mimicking the interaction of ligands to the orthosteric sites.
Smo is a GPCR with clear structural homology to other GPCRs, but there are no
endogenous ligands known. Furthermore, there is no compelling data that directly
links classic G-protein activation to the canonical Hh pathway involving Ci/Gli
stimulation.
Our studies indicate that Smo behaves like a classic GPCR in many regards, and
that the models used to describe this large family of receptors can be applied to Hh
signaling.
GPCR models for Smo and potential mechanisms of Ptc function
A recent study in Drosophila suggested that Hh stimulates the pathway via Ptc
degradation and that Smo is stabilized through extensive phosphorylation at the
plasma membrane where it initiates signaling. However, in our mammalian cell
experiments we did not detect Smo phosphorylation or translocation to the plasma
membrane.
Ptc may regulate Smo-coupler/effector complex formation by affecting the levels of
Smo or its subcellular localization, or by limiting access of Smo to its effector through
targeted vesicle trafficking. Alternatively, Ptc may more directly maintain Smo in an
inactive state.
Therapeutic potential of a Hh-pathway agonist
Studies in mammals have shown that Hh genes are expressed in discrete areas of
the adult organism, and that the Hh pathway may represent a point of intervention for
treating certain degenerative disorders. A Hh agonist may represent an attractive
alternative to an expensive Hh-protein therapeutic.
Chemical libraries and medicinal chemistry
The compound libraries used in our screens were generated by combinatorial
chemistry approaches, and the Hh-agonist class was isolated by standard
procedures.
Cultured cell line assays
TM3 and C3H10T1/2 cells were maintained according to the instructions of ATCC,
and stable Hh-signaling reporter cell lines were established by G418 selection
following transfection with a luciferase reporter plasmid containing the neomycin-
resistance gene.
Retroviral cell lines
Mouse Smo and Ptc1 genes were introduced by a retroviral approach using the
pLPCX vector to limit the copy number per cell. Stable HA-Smo and Ptc-GFP lines
were established by puromycin selection following infection of TM3 cells with the
respective retroviruses.
In uteroHh-signaling assays
Generation of Ptc1lacZ, Shh and Smo mutant mice was described previously [25,
26]. Embryos were collected 24 hours later and hybridized with Ptc1 probe and
stained with X-gal for whole-mount -galactosidase detection.
Primary cerebellar cultures
Cerebellar neurons were dissected out of postnatal rat brains and placed into
primary cell culture. Treatment agents were added once, on the first day of culture (0
DIV).
Neural plate explant assay
Stage 10-11 chick embryos were dissected and embedded in collagen gel.
Intermediate regions of the open neural tube were cultured in Ham-F12
supplemented with 3 g/l D-glucose, Mito Serum Extender, penicillin/streptomycin, 2
mM L-glutamine and Hh-Ag 1.3.
Whole cell/immunocomplex binding assay
After 48 hours, cultured cells were either left untransfected or transfected using
Fugene6 with a pCDNA3.1 construct containing HA-tagged Smo or v5-epitope
tagged 2AR. The cells were then incubated with [3H]-Hh-Ag 1.5 for 2 hours, and
then lysed in lysis/wash buffer for 20 minutes.
Membrane binding assays
Membranes were prepared by transfecting cells with pcDNA 3.1 constructs,
cavitating for 10 minutes at 230 psi, and centrifuging for 20 minutes at 4°C. Filtration
binding assays were performed according to previously described protocols .
In association and dissociation studies, membranes were incubated with [3H]-Hh-Ag
1.5 in the presence or absence of 2 M competitor in binding buffer plus EDTA-free
protease inhibitor cocktail (Roche) for 1-26 hours at 37°C. For saturation and
competition binding analysis, membranes were incubated with various
concentrations of the [3H]-Hh-Ag 1.5 for 45 hours. Binding-kinetics experiments were
performed similarly to the saturation and competition studies. The Kd for Hh-Ag 1.5
was determined to be 0.37 nM.

Acknowledgments
Douglas Melton, Andrew McMahon, Thomas Jessell and Phillip Beachy provided
comments on the manuscript. Andrew McMahon and Matthew Scott provided Shh-,
Smo- and Ptc-transgenic lines, and James Chen provided KAAD-cyclopamine.

You might also like