Smoothened (SMO) is a G-protein-coupled receptor-related protein required for the transduction of Hedgehog (HH). The HH gradient leads to graded phosphorylation of SMO, mainly by the PKA and CKI kinases. How thresholds in HH morphogen regulate SMO to promote switch-like transcriptional responses is a central unsolved issue. Using the wing imaginal disc model in Drosophila, we identified new SMO phosphosites that enhance the effects of the PKA/CKI kinases on SMO accumulation, its localization at the plasma membrane and its activity. Surprisingly, phosphorylation at these sites is induced by the kinase Fused (FU), a known downstream effector of SMO. In turn, activation of SMO induces FU to act on its downstream targets. Overall, our data provide evidence for a SMO/FU positive regulatory loop nested within a multikinase phosphorylation cascade. We propose that this complex interplay amplifies signaling above a threshold that allows high HH signaling.

The Hedgehog (HH) signaling pathway is crucial for the growth and patterning of many metazoans (Lee et al., 2016). Its deregulation leads to developmental disorders and cancers (Pak and Segal, 2016). In many cases, HH acts as a morphogen; it spreads as a gradient across a field of receiving cells to promote dose-dependent discrete transcriptional responses (Vervoort, 2000).

HH signaling requires Smoothened (SMO), a member of the G-protein-coupled receptor family (Ayers and Therond, 2010). SMO is regulated by the transmembrane protein Patched (PTC; Nakano et al., 1989), which acts as both an SMO antagonist and an HH co-receptor. Studies in flies show that in the absence of signal, PTC promotes SMO internalization and targeting to the lysosome for degradation (Fan et al., 2013; Li et al., 2012; Xia et al., 2012). These effects are blocked by HH, leading to accumulation of a hyperphosphorylated form of SMO at the plasma membrane. An important contribution to SMO hyperphosphorylation is the sequential action of the cAMP-dependent protein kinase (PKA) and Casein kinase 1 (CKI) on three clusters of serines (S) and threonines (T) located in its C-terminal cytoplasmic tail (cyto-tail; Chen and Jiang, 2013). These phosphorylations are necessary and sufficient for SMO localization to the plasma membrane and activation. They induce SMO clustering and a conformational switch in its cyto-tail. The extent of phosphorylation of the PKA/CKI clusters depends on HH levels and controls in a graded manner SMO clustering, the degree of conformational change in its cyto-tail, its accumulation at the cell surface and its signaling activity (Fan et al., 2012; Su et al., 2011; Zhao et al., 2007). Moreover, CKIγ/Gilgamesh (Gish), G-protein-coupled receptor kinase 2 (GPRK2/GRK2), Casein kinase II and atypical protein kinase C increase the ability of SMO to transduce high levels of HH by phosphorylating residues present in the membrane-proximal and central regions of the SMO cyto-tail (Jia et al., 2010; Jiang et al., 2014; Li et al., 2016; Maier et al., 2014). In vertebrates, SMO phosphorylation has been reported to depend on GRK2 and CKIγ (Chen et al., 2004). However, the role of GRPK2/GRK2 has recently been called into question by a study in zebrafish (Zhao et al., 2016).

The wing imaginal disc has been instrumental in study of the morphogenetic effects of HH. In this epithelium, HH is produced in the posterior (P) compartment and spreads towards the anterior (A) region. The dose of HH controls the fate of Cubitus interruptus (CI), a zinc finger transcription factor of the GLI family (Aza and Kornberg, 1999; Wang and Holmgren, 2000). In the absence of HH, CI undergoes partial degradation that leads to a truncated repressor form (CI-R). Toward the A/P boundary, the progressive suppression of CI processing by increased doses of HH leads to the gradual decrease of CI-R associated with a progressive accumulation of full-length CI (CI-FL). Thus, in the presence of low levels of HH, CI processing is blocked, which promotes decapentaplegic (dpp) expression. In the presence of intermediate levels of HH, the levels of CI-FL are increased, leading to ptc and collier (col; knot) expression. Finally, in the A cells that abut the P compartment HH producing cells, CI-FL is turned into a hyperactive state (CI-A), which enables engrailed (en) expression.

How is the graded phosphorylation of SMO transduced to the intracellular components of the pathway that regulate CI fate? In the fly wing imaginal disc, responses to high levels of HH rely on the protein kinase Fused (FU) and the kinesin Costal2 (COS2; Alves et al., 1998; Ho et al., 2005; Ohlmeyer and Kalderon, 1998). Both proteins are part of a SMO-bound large cytoplasmic complex that controls CI-FL processing, nuclear entry and activation (Jia et al., 2003; Monnier et al., 1998, 2002; Ogden et al., 2003). The relationships between SMO, FU and COS2 are very complex. SMO directly recruits FU to the plasma membrane and promotes its clustering and its trans-autophosphorylation, with each event being sufficient to promote FU activation (Claret et al., 2007; Shi et al., 2011). COS2 and FU control each other's levels and activity (Ruel et al., 2007; Zhou and Kalderon, 2010, 2011) and also feed back on SMO (Claret et al., 2007; Liu et al., 2007; Ranieri et al., 2014). Thus, in the absence of HH, COS2 binding to SMO inhibits its phosphorylation by PKA/CKI and its accumulation at the cell surface, whereas in response to HH, FU activity is essential for the accumulation of SMO at the plasma membrane and full phosphorylation. It has been proposed that positive feedback by FU on SMO could be attributable to FU opposing the negative effect of COS2. However, FU could control the fate of SMO by directly promoting its phosphorylation on new sites.

Here, we report the identification of four new clusters of S/T in the most C-terminal part of the SMO cyto-tail, near to where FU binds. These sites are required for the full hyperphosphorylation of SMO in response to HH. Their phosphorylation depends on SMO binding to FU and on FU kinase activity, arguing that FU might directly phosphorylate SMO. By preventing or mimicking their phosphorylation, we show that they enhance the effects of the phosphorylation of SMO by the PKA/CKI, resulting in high levels of HH signaling. Finally, phosphorylation of these sites is important for FU to phosphorylate its targets COS2 and SU(FU) and to promote high CI-FL activation. We propose that in response to high levels of HH signal, SMO and FU engage in a complex positive feedback loop, in which they mutually enhance each other's activation. This multikinase phosphorylation cascade would result in an amplification of HH signaling that reaches a threshold, promoting ‘high HH’-specific responses.

Phosphorylation at sites near the C-terminal of SMO is required for HH-induced hyperphosphorylation

PKA and CKI act on three clusters of S/T (Fig. 1A). Blocking the phosphorylation of these residues [by replacing the PKA sites by alanines (A), SMOPKA-SA] did not totally prevent SMO phosphorylation in response to HH (Fig. 1B). Four C-terminal (Ct) blocks (GI-GIV) of S or T residues located between amino acid (aa) 916 and 1036 are conserved among Drosophila species (Fig. 1A). To assay their phosphorylation, we transiently expressed wild-type SMO (SMOwt) or SMOCt-SA (all the Ct-sites replaced by A) – both HA tagged – in HH responsive Cl8 cells (Fig. 1B). In the presence of HH, SMOCt-SA-HA phosphorylation levels were significantly reduced compared with that of SMOwt-HA. Mutation of both the PKA/CKI and the Ct-sites (SMOPKA-SA Ct-SA-HA) almost totally suppressed SMO phosphorylation.

Fig. 1.

Activated FU promotes the phosphorylation of SMO on new sites in its C-terminal intracellular tail. (A) Schematic representation of the Drosophilamelanogaster SMO C-terminal from position 557 to 1036. A multiple sequence alignment of different Drosophilae species (Dm, D. melanogaster; Dps, D. pseudoobscura; Ds, D. simulans; and Dy, D. yakuba). The positions correspond to the first amino acid (aa) of the Dm SMO. Conserved serine (S) and threonine (T) are in red, and the stripes indicate a region with no conserved S or T. PKA/CKI phosphorylation clusters are conserved (dark gray box, with the residues phosphorylated by the PKA underlined). Some conserved S and T are clustered in four groups (GI, II, III and IV) in the C-terminal. Binding to FU was reported to occur between aa 985 and 1036 (Malpel et al., 2007). SMO variants used in this study are shown below, where conserved S or T residues were substituted by alanine (A) or by aspartic acid (D) in the PKA/CKI cluster and/or Ct clusters. (B-F) Immunodetection of various forms of transiently transfected SMO-HA in extracts of Cl8 cells with an antibody against HA (αHA). (B) SMOwt-HA (lanes 1 and 2), SMOCt-SA-HA (lanes 3 and 4), SMOPKA-SA-HA (lanes 5 and 6), SMOPKA-SA Ct-SA-HA (lanes 7 and 8) without (−) or with (+) HH. Here and in the following blots, the bracket indicates the phosphorylated forms of SMO-HA with a slower migration and GMAP is used as loading control. (C) SMOwt-HA (lanes 1 and 2), SMOPKA-SA-HA (lanes 3 and 4) and SMOPKA-SA Ct-SA-HA (lanes 5 and 6) without (−) or with (+) GAP-gFU. Here and in (D-F), the presence of GAP-gFU is verified by immunoblotting with an antibody against GFP (αGFP). (D) SMOwt-HA (lanes 1-3), SMOCt-SA-HA (lanes 4-6) without (−) (lanes 1 and 4) or with GAP-gFU (lanes 2 and 5) or GAP-cFU-DANA (lanes 3 and 6), a kinase dead form of GAP-FU. (E) SMOPKA-SD-HA (lanes 1-4), SMOPKA-SD Ct-SA-HA (lanes 5-8) alone (lanes 1 and 5), with GAP-gFU (lanes 3 and 7) or with GAP-cFU-DANA (lanes 4 and 8); in the absence (lanes 1 and 5) or presence (lanes 2-4 and 6-8) of HH. Note that the shift of SMOPKA-SD was not totally suppressed by GAP-cFU-DANA, probably because of the phosphorylation of sites other than the Ct-sites. (F) SMOwt-HA (lanes 1 and 2), SMOPKA-SD-HA (lanes 3 and 4), SMOPKA-SD Ct-SA-HA (lanes 5 and 6) without (−) or with (+) GAP-gFU. Note that tagging SMO to its N-terminal was shown to preserve its function (Zhu et al., 2003). The asterisk indicates multimeric forms of SMO. (G) Immunodetection of SMO in extracts of wing imaginal disc from MS1096 (expressing GAL4 in the whole wing pouch), MS1096; UAS smowt, MS1096; UAS smoCt-SA and MS1096; UAS smoCt-SD flies, respectively. In MS1096 control discs (lane 1), only phosphorylated forms of SMO (pSMO) are visible. In discs that express the smowt transgene (lane 2), the levels of both forms of SMO (p-SMO and SMO) are increased. In comparison, SMOCt-SA (lane 3) was less phosphorylated than SMOwt, as shown by the reduced pSMO/SMO ratio, especially considering the contribution of endogenous SMO to pSMO. By contrast, SMOCt-SD (lane 4) is mainly present as a phosphorylated form. Red and black arrowheads indicate hyperphosphorylated (pSMO) and non-phosphorylated (SMO) forms of SMO, respectively. TUB, tubulin used as a loading control. All the western blots in all figures have been performed in at least three independent replicates.

Fig. 1.

Activated FU promotes the phosphorylation of SMO on new sites in its C-terminal intracellular tail. (A) Schematic representation of the Drosophilamelanogaster SMO C-terminal from position 557 to 1036. A multiple sequence alignment of different Drosophilae species (Dm, D. melanogaster; Dps, D. pseudoobscura; Ds, D. simulans; and Dy, D. yakuba). The positions correspond to the first amino acid (aa) of the Dm SMO. Conserved serine (S) and threonine (T) are in red, and the stripes indicate a region with no conserved S or T. PKA/CKI phosphorylation clusters are conserved (dark gray box, with the residues phosphorylated by the PKA underlined). Some conserved S and T are clustered in four groups (GI, II, III and IV) in the C-terminal. Binding to FU was reported to occur between aa 985 and 1036 (Malpel et al., 2007). SMO variants used in this study are shown below, where conserved S or T residues were substituted by alanine (A) or by aspartic acid (D) in the PKA/CKI cluster and/or Ct clusters. (B-F) Immunodetection of various forms of transiently transfected SMO-HA in extracts of Cl8 cells with an antibody against HA (αHA). (B) SMOwt-HA (lanes 1 and 2), SMOCt-SA-HA (lanes 3 and 4), SMOPKA-SA-HA (lanes 5 and 6), SMOPKA-SA Ct-SA-HA (lanes 7 and 8) without (−) or with (+) HH. Here and in the following blots, the bracket indicates the phosphorylated forms of SMO-HA with a slower migration and GMAP is used as loading control. (C) SMOwt-HA (lanes 1 and 2), SMOPKA-SA-HA (lanes 3 and 4) and SMOPKA-SA Ct-SA-HA (lanes 5 and 6) without (−) or with (+) GAP-gFU. Here and in (D-F), the presence of GAP-gFU is verified by immunoblotting with an antibody against GFP (αGFP). (D) SMOwt-HA (lanes 1-3), SMOCt-SA-HA (lanes 4-6) without (−) (lanes 1 and 4) or with GAP-gFU (lanes 2 and 5) or GAP-cFU-DANA (lanes 3 and 6), a kinase dead form of GAP-FU. (E) SMOPKA-SD-HA (lanes 1-4), SMOPKA-SD Ct-SA-HA (lanes 5-8) alone (lanes 1 and 5), with GAP-gFU (lanes 3 and 7) or with GAP-cFU-DANA (lanes 4 and 8); in the absence (lanes 1 and 5) or presence (lanes 2-4 and 6-8) of HH. Note that the shift of SMOPKA-SD was not totally suppressed by GAP-cFU-DANA, probably because of the phosphorylation of sites other than the Ct-sites. (F) SMOwt-HA (lanes 1 and 2), SMOPKA-SD-HA (lanes 3 and 4), SMOPKA-SD Ct-SA-HA (lanes 5 and 6) without (−) or with (+) GAP-gFU. Note that tagging SMO to its N-terminal was shown to preserve its function (Zhu et al., 2003). The asterisk indicates multimeric forms of SMO. (G) Immunodetection of SMO in extracts of wing imaginal disc from MS1096 (expressing GAL4 in the whole wing pouch), MS1096; UAS smowt, MS1096; UAS smoCt-SA and MS1096; UAS smoCt-SD flies, respectively. In MS1096 control discs (lane 1), only phosphorylated forms of SMO (pSMO) are visible. In discs that express the smowt transgene (lane 2), the levels of both forms of SMO (p-SMO and SMO) are increased. In comparison, SMOCt-SA (lane 3) was less phosphorylated than SMOwt, as shown by the reduced pSMO/SMO ratio, especially considering the contribution of endogenous SMO to pSMO. By contrast, SMOCt-SD (lane 4) is mainly present as a phosphorylated form. Red and black arrowheads indicate hyperphosphorylated (pSMO) and non-phosphorylated (SMO) forms of SMO, respectively. TUB, tubulin used as a loading control. All the western blots in all figures have been performed in at least three independent replicates.

We conclude that, in these conditions, the Ct and PKA/CKI sites are the major contributors to the HH-induced SMO hyperphosphorylation.

Activated FU promotes the phosphorylation of the SMO Ct-sites

Downregulation of FU kinase activity reduces the hyperphosphorylation of SMO induced by HH (Liu et al., 2007), and FU is able to induce SMO phosphorylation (Claret et al., 2007). This latter effect depends on FU kinase activity and is enhanced with GAP-gFU, a constitutively active form of FU. GAP-gFU also induces the phosphorylation of SMOPKA-SA-HA (Fig. 1C; Fig. S1A), showing that its target sites are outside the known PKA/CKI targets. The Ct clusters are at or near the FU binding region (59 most C-terminal aa) and, as shown in Fig. 1C,D, GAP-gFU could not induce SMOCt-SA-HA (or SMOPKA-SA Ct-SA) phosphorylation, indicating that the Ct-sites are targeted by activated FU. Note that mutation of each Ct group separately impacted the SMO phosphorylation induced by GAP-cFU to various degrees (Fig. S1B), indicating that they all contribute to it.

Moreover, GAP-gFUDANA, a kinase-dead form of GAP-gFU, which acts in a dominant negative manner (Claret et al., 2007), reduces the HH-induced phosphorylation of SMOPKA-SD, but not of SMOPKA-SD Ct-SA (Fig. 1E). Finally, induction of SMOwt phosphorylation by GAP-gFU was suppressed with a form of SMO lacking its FU binding region (SMOΔ978-HA; Fig. S1C).

These data argue that activated FU can promote the phosphorylation of the Ct-sites involved in the HH-induced hyperphosphorylation of SMO and show that this effect is FU kinase activity and interaction dependent.

Phosphorylation of the PKA/CKI sites favors the phosphorylation of the Ct clusters

We next studied whether the phosphorylation of the PKA/CKI and Ct clusters influenced each other. As shown above, phosphorylation of the PKA sites is not required for that of the Ct-sites induced by GAP-gFU. We also tested SMOPKA-SD-HA, in which the PKA and CKI phosphorylation sites have been replaced by aspartic acids (D) to mimic their phosphorylation. SMOPKA-SD-HA was constitutively phosphorylated in the absence of HH (Fig. 1F; Fig. S1A), probably attributable to the phosphorylation of the Ct clusters as it disappeared with SMOPKA-SD Ct-SA-HA. By contrast, SMOCt-SD-HA (Ct-sites replaced by D to mimick their phosphorylation) displayed no sign of phosphorylation in the absence of HH but was further phosphorylated in the presence of HH, probably on its PKA sites as this was not the case for SMOPKA-SA Ct-SD-HA (Fig. S1D,E).

Finally, we analyzed the effect of Ct-sites on SMO phosphorylation in vivo. The smo transgenes (smowt, smoCt-SA and smoCt-SD) were inserted at the same locus to ensure comparable expression levels and expressed throughout the wing primodium. As seen in Fig. 1G, blocking the phosphorylation of the Ct-sites (smoCt-SA) reduced the levels of SMO phosphorylation, whereas mimicking it (smoCt-SD) led to their increase.

In summary, the phosphorylation of the Ct-sites of SMO is neither necessary for the phosphorylation of the PKA/CKI sites in response to HH nor sufficient to promote it in the absence of HH. By contrast, phosphorylation of the PKA/CKI sites favors the phosphorylation of the Ct-sites, but is not required for their phosphorylation by activated GAP-FU, suggesting that it acts by activating FU.

Phosphorylation of the Ct-sites controls the levels of SMO

Our data indicate that the phosphorylation of the Ct-sites upregulates SMO levels (Fig. 1; Fig. S1). To quantify this, we fused SMO N-terminal to an enzymatic self-labeling tag called SNAP that enables direct detection and quantification of the tagged protein after electrophoresis (Tirat et al., 2006). After checking the functionality of SNAP-SMO in vivo (Fig. S2), we analyzed the effects of HH and the Ct-sites on its levels in Cl8 cells. As shown in Fig. 2A-C (and Fig. S3), HH promoted a more than twofold increase in SNAP-SMO levels. SNAP-SMOCt-SA basal levels were not affected, but its HH-induced accumulation was reduced by ∼30%. Conversely, SNAP-SMOCt-SD) basal levels were significantly upregulated (by threefold), an effect strongly increased (sevenfold compared with SNAP-SMOwt) in response to HH. Moreover, compared with SNAP-SMOPKA-SD, SNAP-SMOPKA-SD Ct-SA levels were downregulated by about twofold both in the presence and in the absence of HH, whereas SNAP-SMOPKA-SD Ct-SD was constitutively accumulated at very high levels, reaching almost fourfold those of SNAP-SMOPKA-SD.

Fig. 2.

Phosphorylation of the Ct-sites controls the levels of SMO at the plasma membrane. (A) Transfected Cl8 cells expressing the different SNAP-SMO constructs were labeled for 20 min with a permeable fluorescent substrate (TMR STAR) that marked both cell surface and intracellular SNAP-SMO. Extracellular (Ext) in gray and intracellular (Int) in yellow. (B) Representative fluorescent scanning of extracts of cells labeled as indicated in (A) and cotransfected with various forms of SNAP-SMO along with CLIP-GUS, which is a fusion between the Escherichiacoli glucoronidase (GUS) gene and the CLIP-tag, a derivative form of SNAP-tag with different substrate specificity. Here and in (E), CLIP-GUS was used as an internal control for experimental variations including the ones attributable to transfection efficiency variation. (C) Graph showing the ratio between the amounts of SNAP-SMO and those of CLIP-GUS (in arbitrary units). The variants of SMO tested are indicated, and the value for SNAP-SMOwt in the absence of HH was taken as the reference (=1). Here and in (F), yellow is without HH and orange with HH; quantifications are from three biological replicates that were scanned together. Gel images in (B) and (E) are composites of representative results (the dashed lines indicate when the samples were on the same gel and the continuous lines when they came from gels run in parallel and treated in the same conditions). In (C) and (F), the error bars represent the s.d. Statistical analysis by one tailed bivariate Wilcoxon rank tests: black bracket, P=0.05; red bracket, P=0.1. (D) Transfected Cl8 cells expressing the SNAP-SMO constructs were labeled for 10 min with a non-liposoluble 782 nm fluorescent substrate (SNAP surface 782) that could not pass through the membrane and therefore labeled only the population of SMO molecules present at the cell surface. Then, the cells were labeled for 10 min with a membrane permeable fluorescent substrate (SNAP TMR STAR) before being lysed. (E) Representative fluorescent scanning of extracts of transfected Cl8 cells cotransfected with various forms of SNAP-SMO along with CLIP-GUS and labeled as indicated in (D). Fluorescent SNAP surface staining is shown in the upper panel and fluorescent SNAP and CLIP TMR staining in the middle panel. A merged image of the two stainings is shown in the lower panel. Images of corresponding intact cells are shown in Fig. S3. (F) Ratio of the cell surface to the intracellular fluorescence obtained with different forms of SNAP-SMO. The values were normalized to the ratio obtained for SNAP-SMOwt in the presence of HH.

Fig. 2.

Phosphorylation of the Ct-sites controls the levels of SMO at the plasma membrane. (A) Transfected Cl8 cells expressing the different SNAP-SMO constructs were labeled for 20 min with a permeable fluorescent substrate (TMR STAR) that marked both cell surface and intracellular SNAP-SMO. Extracellular (Ext) in gray and intracellular (Int) in yellow. (B) Representative fluorescent scanning of extracts of cells labeled as indicated in (A) and cotransfected with various forms of SNAP-SMO along with CLIP-GUS, which is a fusion between the Escherichiacoli glucoronidase (GUS) gene and the CLIP-tag, a derivative form of SNAP-tag with different substrate specificity. Here and in (E), CLIP-GUS was used as an internal control for experimental variations including the ones attributable to transfection efficiency variation. (C) Graph showing the ratio between the amounts of SNAP-SMO and those of CLIP-GUS (in arbitrary units). The variants of SMO tested are indicated, and the value for SNAP-SMOwt in the absence of HH was taken as the reference (=1). Here and in (F), yellow is without HH and orange with HH; quantifications are from three biological replicates that were scanned together. Gel images in (B) and (E) are composites of representative results (the dashed lines indicate when the samples were on the same gel and the continuous lines when they came from gels run in parallel and treated in the same conditions). In (C) and (F), the error bars represent the s.d. Statistical analysis by one tailed bivariate Wilcoxon rank tests: black bracket, P=0.05; red bracket, P=0.1. (D) Transfected Cl8 cells expressing the SNAP-SMO constructs were labeled for 10 min with a non-liposoluble 782 nm fluorescent substrate (SNAP surface 782) that could not pass through the membrane and therefore labeled only the population of SMO molecules present at the cell surface. Then, the cells were labeled for 10 min with a membrane permeable fluorescent substrate (SNAP TMR STAR) before being lysed. (E) Representative fluorescent scanning of extracts of transfected Cl8 cells cotransfected with various forms of SNAP-SMO along with CLIP-GUS and labeled as indicated in (D). Fluorescent SNAP surface staining is shown in the upper panel and fluorescent SNAP and CLIP TMR staining in the middle panel. A merged image of the two stainings is shown in the lower panel. Images of corresponding intact cells are shown in Fig. S3. (F) Ratio of the cell surface to the intracellular fluorescence obtained with different forms of SNAP-SMO. The values were normalized to the ratio obtained for SNAP-SMOwt in the presence of HH.

We therefore conclude that the phosphorylation of the Ct-sites strongly enhances the stabilizing effects of the phosphorylation of PKA/CKI sites induced by HH.

Phosphorylation of the Ct-sites increases SMO localization at the plasma membrane

We then examined whether the subcellular localization of SMO was regulated by the phosphorylation of its Ct-sites. The cell surface and intracellular pools of SNAP-SMO were differentially marked by sequentially using membrane impermeable and permeable SNAP substrates carrying different fluorophores (Fig. 2D-F; Fig. S3). As expected, HH stimulation led to accumulation of SNAP-SMOwt at the plasma membrane. This initial experiment also revealed two important features. First, all the hyperphosphorylated forms of SMO are at the cell surface and, reciprocally, the intracellular pool of SMO lacks extensive phosphorylation. Second, HH stimulation mainly induced an increase in the levels of SNAP-SMOwt at the cell surface, whereas intracellular levels were almost unaffected. By contrast with SNAP-SMOwt, SNAP-SMOCt-SA stayed intracellular even in the presence of HH. Moreover, in the absence of HH, SNAP-SMOCt-SD displayed a weak but significant increased (twofold) localization to the plasma membrane, which was strongly enhanced by HH (15-fold). As expected, SNAP-SMOPKA-SA remained intracellular in the presence of HH, whereas SNAP-SMOPKA-SD was mostly at the plasma membrane even in the absence of HH. This latter effect was suppressed in SNAP-SMOPKA-SD Ct-SA but was further increased in SNAP-SMOPKA-SD Ct-SD.

We conclude that phosphorylation of the Ct-sites is required for the accumulation of high levels of SMO at the cell surface promoted by the phosphorylation of the PKA/CKI clusters in response to HH.

Phosphorylation of SMO Ct-sites controls its apicobasal distribution

SNAP labeling was then used to analyze the effects of Ct-site phosphorylation on the subcellular distribution of SMO in vivo. Wing imaginal discs expressing SNAP-tagged forms of SMO in the dorsal compartment were labeled with a membrane impermeable SNAP ligand. The ventral compartment served as an internal negative control. Labeled SNAP-SMO thus corresponds to molecules that were – at least transiently – present at the plasma membrane during the labeling time. In agreement with the Cl8 data, labeled SNAP-SMOwt was at higher levels both at the plasma membrane and in intracellular vesicles in HH sending posterior cells and in A/P receiving cells (Fig. 3A). By contrast, SNAP-SMOCt-SA was less labeled and mostly present in intracellular vesicular structures, whereas SNAP-SMOCt-SD was highly labeled, with an enrichment at the plasma membrane of the HH sending and receiving cells (Fig. 3B,C). We also observed distinctions in labeled SNAP-SMOwt distribution along the apicobasal axis of the wing epithelium (Fig. 3D): labeled SNAP-SMOwt was present at the apical side (visualized with aPKC staining) of the P cells and of the A cells that receive HH and also in the most basal region of the P cells and of the A cells abutting A/P in the region where the highest levels of HH are encountered. This basal distribution was lost with labeled SNAP-SMOCt-SA and increased with labeled SNAP-SMOCt-SD, indicating that it is dependent on the phosphorylation of the Ct-sites (Fig. 3E,F). Note that these effects were also seen by immunofluorescence with untagged SMO variants (Fig. S4).

Fig. 3.

Phosphorylation of the Ct-sites controls SMO localization and activity in vivo. (A-F) XY confocal images (A-C) or XZ confocal images (D-F) of wing discs expressing UAS snap-smowt (A,D), UAS snap-smoCt-SA (B,E) and UAS snap-smoCt-SD (C,F) driven by apGal4 and labeled with a non-liposoluble fluorescent substrate (A-F) and stained with aPKC (green; D2-F2). The XZ sections are perpendicular to the A/P axis through the dorsal compartment. The images in (D3-F3) show the distribution of SNAP-SMO forms along the apicobasal axis in three-dimensional false color. The rectangle in (A1-C1) corresponds to the region enlarged in (A2-C2). The A/P boundary (determined by CI-FL staining, not shown) is indicated either by two arrowheads or by a dotted line. Ap, apical; Ba, basal. Note that the imaging conditions used optimized the acquisition of SMO for each genotype. The XZ images correspond to maximal intensity projection from 13 sections. Here and in the following figures, XY disc images are oriented dorsal up and anterior left. (G-I) Representative XY confocal images of wing discs of apGAL4 flies expressing UAS smowt (G), UAS smoCt-SA (H) and UAS smoCt-SD (I) and stained with antibodies against CI-FL (G1-I1) or PTC (G2-I2). The white arrowhead indicates the position of the dorsoventral boundary. (J,K) For each labeling, the discs were classified in a blind experiment according to four phenotypic criteria that reflect the strength of HH activation: decreased accumulation at A/P (down arrow), no effect on accumulation at A/P (=), increased accumulation at A/P (up arrow) and ectopic accumulation (ectopic). The percentage of the different phenotypes observed for each genotype is presented on the radar diagram for CI-FL (J) and for PTC (K) staining. The number of discs analyzed is indicated in brackets. Scale bars: 50 µm in A1,B1,C1,G,H,I; 20 µm in the other panels.

Fig. 3.

Phosphorylation of the Ct-sites controls SMO localization and activity in vivo. (A-F) XY confocal images (A-C) or XZ confocal images (D-F) of wing discs expressing UAS snap-smowt (A,D), UAS snap-smoCt-SA (B,E) and UAS snap-smoCt-SD (C,F) driven by apGal4 and labeled with a non-liposoluble fluorescent substrate (A-F) and stained with aPKC (green; D2-F2). The XZ sections are perpendicular to the A/P axis through the dorsal compartment. The images in (D3-F3) show the distribution of SNAP-SMO forms along the apicobasal axis in three-dimensional false color. The rectangle in (A1-C1) corresponds to the region enlarged in (A2-C2). The A/P boundary (determined by CI-FL staining, not shown) is indicated either by two arrowheads or by a dotted line. Ap, apical; Ba, basal. Note that the imaging conditions used optimized the acquisition of SMO for each genotype. The XZ images correspond to maximal intensity projection from 13 sections. Here and in the following figures, XY disc images are oriented dorsal up and anterior left. (G-I) Representative XY confocal images of wing discs of apGAL4 flies expressing UAS smowt (G), UAS smoCt-SA (H) and UAS smoCt-SD (I) and stained with antibodies against CI-FL (G1-I1) or PTC (G2-I2). The white arrowhead indicates the position of the dorsoventral boundary. (J,K) For each labeling, the discs were classified in a blind experiment according to four phenotypic criteria that reflect the strength of HH activation: decreased accumulation at A/P (down arrow), no effect on accumulation at A/P (=), increased accumulation at A/P (up arrow) and ectopic accumulation (ectopic). The percentage of the different phenotypes observed for each genotype is presented on the radar diagram for CI-FL (J) and for PTC (K) staining. The number of discs analyzed is indicated in brackets. Scale bars: 50 µm in A1,B1,C1,G,H,I; 20 µm in the other panels.

In summary, the phosphorylation of the Ct-sites regulates the subcellular localization of SMO in response to HH by two means: (i) it increases its accumulation at the plasma membrane; and (ii) it promotes its localization in the basal region of the polarized epithelial cells of the wing imaginal disc.

Phosphorylation of the SMO Ct-sites upregulates its activity in vivo

We then assayed whether the phosphorylation of the Ct-sites regulates SMO activity. SMOwt, SMOCt-SA and SMOCt-SD were expressed in the D compartment, and HH signaling was monitored by following CI-FL and PTC accumulation. Despite some variation, clear signatures specific to the different forms of SMO were observed (Fig. 3G-K). SMOwt overexpression led to a mild upregulation of HH signaling as indicated by: (1) an upregulation of CI-FL levels at the A/P boundary (in >50% of discs) and its ectopic accumulation in the A part of the wing discs (in >65% of discs; Fig. 3G1,J); and (2) an increase in PTC levels at the A/P border (in >60% of discs) and its occasional ectopic expression towards the anterior (20% of discs; Fig. 3G2,K). By contrast, SMOCt-SA tended to decrease HH signaling (Fig. 3H,J,K). It never led to ectopic PTC and led to ectopic CI-FL accumulation about four times less often than SMOwt. Moreover, it downregulated PTC levels and/or CI-FL in almost 30% of discs, two events rarely seen with SMOwt expression. Conversely, expression of SMOCt-SD led to a stronger activation of HH signaling than SMOwt, with an enhanced accumulation of CI-FL and PTC in the whole A compartment in 80 and 72% of the discs, respectively (Fig. 3I-K).

Together, these results indicate that the phosphorylation of the Ct-sites of SMO upregulates its activity.

Phosphorylation of the SMO Ct-sites enhances the activating effects of phosphorylation at the PKA/CKI sites

For better assessment of the roles of the phosphorylation of Ct-sites, we analyzed the impact of mutating them on SMOPKA-SD activity. SMOPKA-SD is known to be active constitutively (Jia et al., 2004), and its expression in the wing pouch led to an increased expression of all known HH targets (Fig. 4; Fig. S5). This was associated with an increase in CI-FL levels throughout the A compartment, with an enlargement of the region near the A/P boundary that normally displays low levels of CI-FL (corresponding to CI-A and therefore called the ‘CI-A region’) owing to the presence of high levels of HH. All these effects were stronger with SMOPKA-SD Ct-SD, with a very strong ectopic en expression and an important enlargement of the CI-A region. By contrast, SMOPKA-SD Ct-SA led to CI-FL accumulation in the A region but it was unable to promote ectopic target gene expression. Moreover, en expression at the A/P boundary was suppressed along with the CI-A region, and col and ptc expression were strongly downregulated. However, the expression domains of the ‘low HH’ responsive genes dpp and iroquois (iro) were almost normal. Notably, these negative effects of SMOPKA-SD Ct-SA are also seen in the absence of endogeneous SMO protein (see Fig. S6).

Fig. 4.

Phosphorylation of the Ct-sites increases the effects of PKA/CKI phosphorylation. (A-D) Wing discs expressing UAS smowt (A), UAS smoPKA-SD (B), UAS smoPKA-SD Ct-SD (C) and UAS smoPKA-SD Ct-SA (D) in the dorsal compartment using apGAL4 and stained with antibodies against EN (A1-D1), β-galactosidase (ptc-Z) to follow ptc-Z (A2-D2) or dpp-Z transcriptional fusion reporters (a4-d4) and CI-FL (A3-D3). A slight expansion of the dorsal expression domain of dpp is observed when smoPKA-SD Ct-SA is expressed. This is probably attributable to further spreading of HH resulting from the strong reduction of PTC at the A/P boundary. The brackets in A3-C3 indicate the reduced accumulation of CI-FL near the A/P boundary associated with its high activation state. Scale bars: 50 µm. At least 20 discs per phenotype were analyzed.

Fig. 4.

Phosphorylation of the Ct-sites increases the effects of PKA/CKI phosphorylation. (A-D) Wing discs expressing UAS smowt (A), UAS smoPKA-SD (B), UAS smoPKA-SD Ct-SD (C) and UAS smoPKA-SD Ct-SA (D) in the dorsal compartment using apGAL4 and stained with antibodies against EN (A1-D1), β-galactosidase (ptc-Z) to follow ptc-Z (A2-D2) or dpp-Z transcriptional fusion reporters (a4-d4) and CI-FL (A3-D3). A slight expansion of the dorsal expression domain of dpp is observed when smoPKA-SD Ct-SA is expressed. This is probably attributable to further spreading of HH resulting from the strong reduction of PTC at the A/P boundary. The brackets in A3-C3 indicate the reduced accumulation of CI-FL near the A/P boundary associated with its high activation state. Scale bars: 50 µm. At least 20 discs per phenotype were analyzed.

In conclusion, phosphorylation of the Ct clusters is required for the constitutive activity of SMOPKA-SD and enhances its effects. By contrast, preventing Ct cluster phosphorylation in SMOPKA-SD blocks the response to high levels of HH but has almost no effect on the target genes that respond to low HH levels.

The negative effects of SMOPKA-SD Ct-SA are suppressed by activated FU

Strikingly, the expression of SMOPKA-SD Ct-SA phenocopies the effects of mutations disrupting FU kinase activity. In fu mutants altered in the kinase domain, en and col expression are strongly reduced, inactive CI-FL accumulates and the CI-A region disappears, whereas dpp is almost unaffected (Alves et al., 1998). It might thus be the case that SMOPKA-SD Ct-SA traps or freezes endogenous FU in an inactive state. This prompted us to examine the consequences of SMOPKA-SD Ct-SA expression on the three proteins whose phosphorylation depends on FU activity and are likely targets of FU: COS2, FU itself and its antagonist, SU(FU). Myc-tagged versions of these proteins were transiently coexpressed with SMOwt-HA in Cl8 cells (Fig. 5A). All three displayed low levels of phosphorylation (visualized by a shift in their electrophoretic migration) in the presence of SMOwt and HH. Consistent with their signaling activity, SMOCt-SA-HA and SMOCt-SD-HA led to a slight (but reproducibly) reduced and increased phosphorylation, respectively. As expected, SMOPKA-SD also upregulated their phosphorylation in the absence of HH. By contrast, SMOPKA-SD Ct-SA expression prevented (COS2 and FU) or reduced [SU(FU)] phosphorylation, both in the absence and in the presence of HH. This indicates that SMOPKA-SD Ct-SA negatively affects the kinase activity of endogenous FU.

Fig. 5.

The negative effects of SMOPKA-SD Ct-SA are suppressed by coexpression of constitutively active FU. (A) Immunodetection of myc-COS2, myc-FU and SU(FU)-myc in extracts of Cl8 cells coexpressing SMOwt-HA (lanes 1 and 2), SMOCt-SA-HA (lanes 3 and 4), SMOCt-SD-HA (lanes 5 and 6), SMOPKA-SD-HA (lanes 7 and 8) and SMOPKA-SD Ct-SA-HA (lanes 9 and 10) without (−) or with (+) HH. Arrowheads indicate the most phosphorylated forms of myc-COS2, myc-FU and SU(FU)-myc. (B-E) Wing discs of apGAL4 flies (B) or expressing UAS smoPKA-SD Ct-SA (C), UAS GAP-cFU (D) and UAS GAP-cFU with UAS smoPKA-SD Ct-SA (E) and stained with antibodies against EN (B1-E1), COL (B2-E2) and CI-FL (B3-E3). Expression of GAP-cFU led to anterior expansion of the expression of col and en and to an ectopic accumulation of CI-FL. Coexpression of GAP-cFU and smoPKA-SD Ct-SA led to ectopic expression of all HH targets. Note that the effects of GAP-cFU overexpression were further enhanced in the presence of SMOPKA-SD Ct-SA and that GAP-cFU has a mild effect compared with previous published data (Claret et al., 2007) owing to lower expression levels. Scale bars: 50 µm. Note that SMOPKA-SD also increases the effect of GAP-cFU (Fig. S8).

Fig. 5.

The negative effects of SMOPKA-SD Ct-SA are suppressed by coexpression of constitutively active FU. (A) Immunodetection of myc-COS2, myc-FU and SU(FU)-myc in extracts of Cl8 cells coexpressing SMOwt-HA (lanes 1 and 2), SMOCt-SA-HA (lanes 3 and 4), SMOCt-SD-HA (lanes 5 and 6), SMOPKA-SD-HA (lanes 7 and 8) and SMOPKA-SD Ct-SA-HA (lanes 9 and 10) without (−) or with (+) HH. Arrowheads indicate the most phosphorylated forms of myc-COS2, myc-FU and SU(FU)-myc. (B-E) Wing discs of apGAL4 flies (B) or expressing UAS smoPKA-SD Ct-SA (C), UAS GAP-cFU (D) and UAS GAP-cFU with UAS smoPKA-SD Ct-SA (E) and stained with antibodies against EN (B1-E1), COL (B2-E2) and CI-FL (B3-E3). Expression of GAP-cFU led to anterior expansion of the expression of col and en and to an ectopic accumulation of CI-FL. Coexpression of GAP-cFU and smoPKA-SD Ct-SA led to ectopic expression of all HH targets. Note that the effects of GAP-cFU overexpression were further enhanced in the presence of SMOPKA-SD Ct-SA and that GAP-cFU has a mild effect compared with previous published data (Claret et al., 2007) owing to lower expression levels. Scale bars: 50 µm. Note that SMOPKA-SD also increases the effect of GAP-cFU (Fig. S8).

To test this possibility further, we examined whether the negative effects of SMOPKA-SD Ct-SA were rescued by the expression of GAP-CFP-FU (GAP-cFU; Fig. 5B-E; Fig. S7). As previously reported, GAP-cFU expression led to an activation of the pathway, with ectopic ptc, col and dpp expression and ectopic accumulation of CI-FL. When GAP-cFU was coexpressed with SMOPKA-SD Ct-SA, all HH targets were ectopically expressed. Surprisingly, these effects were stronger than those of GAP-cFU or SMOPKA-SD Ct-SA alone as indicated by high levels of ectopic en expression throughout the entire A compartment. Notably, CI-FL levels were strongly reduced, presumably because of the generation of the CI-A form throughout the A compartment.

These data show that the expression of an active form of FU can totally suppress the negative effects of SMOPKA-SD Ct-SA on the ‘high HH’ targets and that, reciprocally, its effects on all HH targets are increased by SMOPKA-SD Ct-SA.

We identified new phosphorylations of the fly SMO cyto-tail and showed that they are promoted by the protein kinase FU. This phosphorylation is part of a multisite, multikinase phosphorylation cascade, which controls both SMO accumulation at the plasma membrane and its distribution along the apicobasal axis. These phosphorylation events participate in amplification feedback loops, which promote high levels of SMO-FU activation that allow responses to high doses of the HH signal.

Multisite, multikinase phosphorylation of SMO

Several kinases are involved in SMO cyto-tail hyperphosphorylation, including PKA/CKI, CKII, aPKC, GPRK2/GRK2 and CK2γ/Gish. Here, we identify four new phosphorylation clusters in its most C-terminal part, near the FU binding site, that are required for the full phosphorylation of SMO in response to HH. FU might act on SMO phosphorylation directly or indirectly through the recruitment, increased expression (via CI activation) or activation of another kinase. Although definite proof of a direct effect is lacking in the absence of an in vitro FU kinase assay, several lines of evidence support that possibility, as follows: (i) blocking endogenous FU activity by a dominant negative form reduces the HH induced phosphorylation of SMO; (ii) expressing a form of FU that is not anchored to the plasma membrane also promotes SMO phosphorylation (although less efficiently than GAP-FU); (iii) both GAP-gFU-kinase activity and its interaction with SMO are required for GAP-gFU to promote the phosphorylation of Ct-sites; and (iv) the induction of SMO phosphorylation by FU or GAP-FU on SMO is also seen with S2 cells that fail to express CI. Note also that FU might act on only a few of these sites, which would prime the phosphorylation of adjacent sites by another kinase, as shown for its own autophosphorylation (Zhou and Kalderon, 2011).

Our data also suggest that sequential phosphorylation of SMO initiated at the PKA/CKI sites leads to the phosphorylation of the Ct-sites. This could be attributable to the conformational changes induced by the PKA/CKI phosphorylation, which could increase the accessibility of the Ct-sites. Alternatively, the phosphorylation of the PKA/CKI sites could act indirectly by promoting FU activation. We favor this latter possibility for the following reasons: (i) FU can be directly activated by SMO (Claret et al., 2007; Zhou and Kalderon, 2011); and (ii) GAP-gFU can promote the phosphorylation of SMOwt in the absence of phosphorylation of the PKA/CKI sites of SMO (Claret et al., 2007; and present study).

SMO localization and levels

SMO phosphorylation by PKA/CKI is necessary and sufficient to promote an increase in SMO levels and its localization at the plasma membrane, but the temporal, spatial and functional connections between these events remain elusive. Several scenarios can be considered. For instance, SMO phosphorylation could block SMO endocytosis once it has reached the cell surface or it could upregulate the targeting of newly synthesized SMO to the plasma membrane. Our data, in agreement with (Kupinski et al., 2013), strongly support the first hypothesis, as follows: (i) the increase of SMO induced by HH, by the phosphorylation of its PKA/CKI sites, or both, is almost entirely attributable to an increase of its cell surface pool, with only very modest effects on intracellular SMO levels; and (ii) the phosphorylated SMO molecules are entirely located at the cell surface and, reciprocally, all of the cell surface fraction is phosphorylated.

Moreover, the phosphorylation of the Ct-sites seems to act by increasing the effects of the PKA/CKI sites on the stabilization of SMO at the plasma membrane. Mimicking the phosphorylation of the Ct-sites leads to a strong increase of the fraction of SMO present at the plasma membrane, and this effect is most visible either in the presence of HH – when the PKA/CKI sites are phosphorylated – or when their phosphorylation is mimicked.

SMO activation

Phosphorylation of the PKA/CKI sites increases SMO activity by antagonizing a region called the SMO auto-inhibitory domain (SAID), which shuts down SMO activity in the absence of HH, owing to its intermolecular electrostatic interaction with acidic aa of the most C-terminal region of SMO (Zhao et al., 2007). As a result, the SMO cyto-tail dimerizes. This effect is increased by GPRK2/GRK2 phosphorylation (Maier et al., 2012). By contrast, FU acts on sites located in or near the acidic region, and their phosphorylation further increases the negative charges in this region, which suggests that they might activate SMO by a new (so far, not understood) mechanism. Nonetheless, the effects of phosphorylation of Ct-sites strengthen the tight correlation between SMO signaling activity and its accumulation at the cell surface, and they further support the notion that the concentration or number of SMO molecules present at the cell surface is crucial for HH signaling. Finally, the phosphorylation of the Ct-sites of SMO appears also to be involved in the apicobasal localization of SMO, leading to increased basal localization in the HH receiving cells. Although the significance of this differential localization awaits further experimentation, it is interesting to consider it in the light of the double HH gradient that has been described: apical for the long range, ‘low HH’ responses and basal for the short range, ‘high HH’ responses (for review, see Guerrero and Kornberg, 2014).

Function and regulation of the kinase FU

Both phosphorylation of SMO Ct-sites and FU kinase activity are necessary for high levels of HH signaling and for the increase in SMO levels and cell surface localization induced by HH (Claret et al., 2007; Liu et al., 2007). So far, these effects have been attributed to the ability of FU to phosphorylate COS2 and SU(FU). Our results provide a new output for FU as they show that it is likely to upregulate the levels of SMO directly at the cell surface and its signaling activity by phosphorylation. It is noteworthy that the FU-dependent phosphorylation of COS2 has been recently shown not to be essential for its activity as SMO and COS2 interact (Zadorozny et al., 2015); it is thus possible that the effect of FU on COS2 activity could in fact occur mainly via SMO. Our work also reveals that the FU downstream activity [i.e. the phosphorylation of SU(FU) and COS2] requires the phosphorylation of SMO Ct-sites. It indicates that SMO activates FU in a two-step mechanism: (i) an initial activation of FU by recruitment to the plasma membrane and that promotes its action on SMO; and (ii) a second step that requires the phosphorylation of SMO Ct-sites, which is necessary for FU effects on its cytosolic targets and the activation of CI-FL.

FU/SMO mutual activation controls high levels of HH signaling

These results provide evidence of the interplay between SMO and FU which, by increasing SMO phosphorylation, leads to high SMO/FU activation. We propose a model whereby SMO/FU activation is based on the progressive multisite phosphorylation of SMO and on a series of activation loops that take place between SMO and FU in the presence of high levels of HH (Fig. 6). This amplifying loop finally leads to a high accumulation of the hyperactivated SMO/FU couple at the cell surface, reaching a threshold of activation sufficient to promote high levels of CI activation and the ‘high HH’ specific responses. Interestingly, multisite phosphorylation can lead to ultrasensitivity under some circumstances and to bistability when combined with a positive feedback loop (for review, see Ferrell and Ha, 2014a,b; Lander, 2007). In this context, the crosstalk between SMO and FU might provide both robustness and switch-like behavior to the HH pathway that could account in part for its morphogenetic effects. Although SMO has been conserved from flies to mammals, significant differences have appeared between human and fly SMO and its regulation (Ingham et al., 2011). This has probably led to some changes in the molecular mechanism, but the underlying principles of the regulation might well have been conserved.

Fig. 6.

Model: SMO/FU act as a molecular switch that participates in high levels of HH signaling. HH gradient promotes graded phosphorylation of the PKA/CKI sites of SMO, leading to a progressive increase of SMO levels and an increase in CI-FL. At low concentrations of HH, PKA/CKI leads to a low activity of SMO (called SMO 1) sufficient to block CI processing, leading to the expression of ‘low HH’ targets, such as dpp and iro. Higher levels of HH lead to increased SMO phosphorylation, and SMO (called SMO 2) reaches a threshold of activation sufficient to promote the activation of FU (FU 1; step 1). In turn, activated FU phosphorylates (directly or indirectly) SMO on its Ct-sites (step 2), leading to higher levels of SMO activity (SMO 3) that allow further FU activation (FU 2; step 3) and action on its downstream targets. As a result, high levels of the hyperactive SMO/FU (SMO 3/FU 2) pair are accumulated at the plasma membrane and allow very high levels of HH signaling associated with the accumulation of low levels of CI-A, which controls ‘high HH’ targets, such as en. SMO/FU 0, 1 and 2 (and 3 for SMO) represent increasing levels of SMO or FU activation, respectively.

Fig. 6.

Model: SMO/FU act as a molecular switch that participates in high levels of HH signaling. HH gradient promotes graded phosphorylation of the PKA/CKI sites of SMO, leading to a progressive increase of SMO levels and an increase in CI-FL. At low concentrations of HH, PKA/CKI leads to a low activity of SMO (called SMO 1) sufficient to block CI processing, leading to the expression of ‘low HH’ targets, such as dpp and iro. Higher levels of HH lead to increased SMO phosphorylation, and SMO (called SMO 2) reaches a threshold of activation sufficient to promote the activation of FU (FU 1; step 1). In turn, activated FU phosphorylates (directly or indirectly) SMO on its Ct-sites (step 2), leading to higher levels of SMO activity (SMO 3) that allow further FU activation (FU 2; step 3) and action on its downstream targets. As a result, high levels of the hyperactive SMO/FU (SMO 3/FU 2) pair are accumulated at the plasma membrane and allow very high levels of HH signaling associated with the accumulation of low levels of CI-A, which controls ‘high HH’ targets, such as en. SMO/FU 0, 1 and 2 (and 3 for SMO) represent increasing levels of SMO or FU activation, respectively.

Drosophila strains and genetics

For transgenesis, wild-type and mutants smo cDNAs were introduced into the same landing site (9738) on 3R (at 99F8) by BestGene, using the PhiC31 integration system. For MARCM clones, the larvae were heat-shocked 3-4 days after egg laying for 2 hours at 37°C. Further details are given in the supplementary Materials and Methods.

Immunostaining of imaginal discs

Discs were dissected and treated as described by Claret et al. (2007) with minor modifications (see supplementary Materials and Methods). The primary antibodies used were: mouse anti-PTC, 1:1000 [Apa 1, from Developmental Studies Hybridoma Bank (DHSB)] (Martin et al., 2001); mouse anti-SMO, 1:100 (20C6, from DSHB) (Lum et al., 2003); mouse anti-COL, 1:100 (a gift from M. Crozatier, Center for Integrative Biology, Toulouse, France) (Crozatier et al., 2003); rat anti-full length CI, 1:5 [2A1 from R. Holmgren, Northwestern University, Evanston, IL, USA (Motzny and Holmgren, 1995)]; and rabbit polyclonal anti-β-galactosidase 1/100 (ABR, Affinity BioReagents).

Plasmids

All expression vectors were constructed by the Gateway recombination method (Invitrogen) and checked by sequencing the mutated regions. Details of the constructs used here are presented in the supplementary Materials and Methods.

Cl8 cell culture, transfection and western blotting

Cl-8 cells were cultured as described previously (Claret et al., 2007). Transient transfections were then carried out using Transit Insect Reagent (Mirus) and analyzed 48 h post transfection. Note that to visualize SMO protein, the samples were not heated. The samples were run on 10% Anderson gels. See supplementary Materials and Methods for more details.

Cl8 SNAP labeling experiments

Forty-eight hours after transfection, cells were washed in PBS then either labeled for 20 min in complete cell medium with 0.75 µM SNAP TMR-star and 0.75 µM CLIP TMR-star (NEB) for total SMO accumulation or labeled for 10 min with 0.75 µM SNAP-Surface 782 in complete cell medium, before adding 0.75 µM SNAP TMR-star and 0.75 µM CLIP TMR-star for 10 min for surface and intracellular staining. In both cases, cells were then washed twice and lysed in 1% NP-40, 150 mM NaCl, 50 mM Tris, 1 mM dithiothreitol with complete EDTA-free antiprotease mix (Roche) and Phosphatase Inhibitor Cocktail Set II (Calbiochem). The lysate was centrifuged (12,000 g) for 10 min at 4°C and mixed with Laemmli sample buffer (Bio-Rad) and 0.1 M dithiothreitol. Fifteen microliters of each sample was loaded on precast 4-20% Mini-Protean TGX gels (Bio-Rad) and run as for western blots (see supplementary Materials and Methods). Gels were scanned either on a Typhoon Trio imager (GE Healthcare; excitation 532 nm, emission 580 nm, PMT 700 V) for total accumulation or sequentially (for surface and intracellular staining) on a Lycor Odyssey at 800 nm before Typhoon Trio. The merged image was obtained by overlapping the two images. Three independent experiments (with all the constructs together) were performed. All gels were run and scanned together. Images were analyzed and quantified using ImageJ software. One tailed bivariate Wilcoxon rank tests were run using R software and R Commander (R Foundation for Statistical Computing).

Wing discs SNAP labeling experiments

Third instar larva were dissected in S2 cell medium. They were then incubated for 30 min with the nonliposoluble Alexa 546 fluorescent substrate at a dilution of 1/600 in S2 medium to allow fixation of the substrate and then incubated for 10 min at 25°C. The larvae were fixed 20 min with PFA, followed by three 10 min washes (PBS with 0.3% Triton) before immunostaining or direct imaging.

We thank M. Crozatier, J. Jiang, D. Kalderon, T. Kornberg, S. Ogden, D. Robbins, P. Thérond, A. Zhu and their laboratories for flies and reagents. We are grateful to F. Schweisguth, L. Ruel, G. D'Angelo, P. Therond, A. Brigui and our colleagues from the Institut Jacques Monod for insightful discussions and to T. Herbillon for technical help. Antibodies from the DSHB were developed under the auspices of the NICHD and maintained by the University of Iowa. Drosophila embryo injections were carried out by BestGene Inc. We acknowledge the ImagoSeine core facility of Institut Jacques Monod, member of France-BioImaging (ANR-10-INBS-04) and certified IBiSA.

Author contributions

Conceptualization, M.S., I.B., L.H., R.A.H. and A.P.; methodology, M.S. and I.B.; validation, M.S. and I.B.; formal analysis, M.S., I.B., L.H. and A.P.; investigation, M.S., I.B., L.H., J.B., C.A., V.G., L.B. and R.A.H.; writing of original draft, M.S., I.B., L.H., R.A.H. and A.P.; writing, review and editing, M.S., I.B., R.A.H. and A.P.; visualization, M.S. and I.B.; supervision, A.P.; project administration, A.P.; and funding acquisition, A.P.

Funding

This work was supported by the Centre Nationnal de la Recherche Scientifique (University Paris 7) and the Fondation ARC pour la recherche sur le Cancer (155032). I.B. was supported by the ARC and by the Ligue Contre le Cancer; L.H. was supported by Université Paris Diderot and the Fondation ARC pour la recherche sur le Cancer; C.A. was supported by the Association Franco-argentine and by the Fondation ARC pour la recherche sur le Cancer; L.B. was supported by Paris Sorbonne Cité.

Alves
,
G.
,
Limbourg-Bouchon
,
B.
,
Tricoire
,
H.
,
Brissard
,
Z. J.
,
Lamour
,
I. C.
and
Busson
,
D.
(
1998
).
Modulation of Hedgehog target gene expression by the Fused serine- threonine kinase in wing imaginal discs
.
Mech. Dev.
78
,
17
-
31
.
Ayers
,
K. L.
and
Thérond
,
P. P.
(
2010
).
Evaluating Smoothened as a G-protein-coupled receptor for Hedgehog signalling
.
Trends Cell Biol.
20
,
287
-
298
.
Aza
,
B. P.
and
Kornberg
,
T. B.
(
1999
).
Ci: a complex transducer of the hedgehog signal
.
Trends Genet.
15
,
458
-
462
.
Chen
,
Y.
and
Jiang
,
J.
(
2013
).
Decoding the phosphorylation code in Hedgehog signal transduction
.
Cell Res.
23
,
186
-
200
.
Chen
,
W.
,
Ren
,
X. R.
,
Nelson
,
C. D.
,
Barak
,
L. S.
,
Chen
,
J. K.
,
Beachy
,
P. A.
,
de Sauvage
,
F.
and
Lefkowitz
,
R. J.
(
2004
).
Activity-dependent internalization of smoothened mediated by beta-arrestin 2 and GRK2
.
Science
306
,
2257
-
2260
.
Claret
,
S.
,
Sanial
,
M.
and
Plessis
,
A.
(
2007
).
Evidence for a novel feedback loop in the Hedgehog pathway involving Smoothened and Fused
.
Curr. Biol.
17
,
1326
-
1333
.
Crozatier
,
M.
,
Glise
,
B.
,
Khemici
,
V.
and
Vincent
,
A.
(
2003
).
Vein-positioning in the Drosophila wing in response to Hh: new roles of Notch signaling
.
Mech. Dev.
120
,
529
-
535
. https://doi.org/10.1016/S0925-4773(03)00041-8
Fan
,
J.
,
Liu
,
Y.
and
Jia
,
J.
(
2012
).
Hh-induced Smoothened conformational switch is mediated by differential phosphorylation at its C-terminal tail in a dose- and position-dependent manner
.
Dev. Biol.
366
,
172
-
184
.
Fan
,
J.
,
Jiang
,
K.
,
Liu
,
Y.
and
Jia
,
J.
(
2013
).
Hrs promotes ubiquitination and mediates endosomal trafficking of smoothened in Drosophila hedgehog signaling
.
PLoS ONE
8
,
e79021
.
Ferrell
,
J. E.
, Jr
and
Ha
,
S. H.
(
2014a
).
Ultrasensitivity part II: multisite phosphorylation, stoichiometric inhibitors, and positive feedback
.
Trends Biochem. Sci.
39
,
556
-
569
.
Ferrell
,
J. E.
, Jr
and
Ha
,
S. H.
(
2014b
).
Ultrasensitivity part III: cascades, bistable switches, and oscillators
.
Trends Biochem. Sci.
39
,
612
-
618
.
Guerrero
,
I.
and
Kornberg
,
T. B.
(
2014
).
Hedgehog and its circuitous journey from producing to target cells
.
Semin. Cell Dev. Biol.
33
,
52
-
62
.
Ho
,
K. S.
,
Suyama
,
K.
,
Fish
,
M.
and
Scott
,
M. P.
(
2005
).
Differential regulation of Hedgehog target gene transcription by Costal2 and Suppressor of Fused
.
Development
132
,
1401
-
1412
.
Ingham
,
P. W.
,
Nakano
,
Y.
and
Seger
,
C.
(
2011
).
Mechanisms and functions of Hedgehog signalling across the metazoa
.
Nat. Rev. Genet.
12
,
393
-
406
.
Jia
,
J.
,
Tong
,
C.
and
Jiang
,
J.
(
2003
).
Smoothened transduces Hedgehog signal by physically interacting with Costal2/Fused complex through its C-terminal tail
.
Genes Dev.
17
,
2709
-
2720
.
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
,
1045
-
1050
.
Jia
,
H.
,
Liu
,
Y.
,
Xia
,
R.
,
Tong
,
C.
,
Yue
,
T.
,
Jiang
,
J.
and
Jia
,
J.
(
2010
).
Casein kinase 2 promotes Hedgehog signaling by regulating both smoothened and Cubitus interruptus
.
J. Biol. Chem.
285
,
37218
-
37226
.
Jiang
,
K.
,
Liu
,
Y.
,
Fan
,
J.
,
Epperly
,
G.
,
Gao
,
T.
,
Jiang
,
J.
and
Jia
,
J.
(
2014
).
Hedgehog-regulated atypical PKC promotes phosphorylation and activation of Smoothened and Cubitus interruptus in Drosophila
.
Proc. Natl. Acad. Sci. USA
111
,
E4842
-
E4850
.
Kupinski
,
A. P.
,
Raabe
,
I.
,
Michel
,
M.
,
Ail
,
D.
,
Brusch
,
L.
,
Weidemann
,
T.
and
Bokel
,
C.
(
2013
).
Phosphorylation of the Smo tail is controlled by membrane localisation and is dispensable for clustering
.
J. Cell Sci.
126
,
4684
-
4697
.
Lander
,
A. D.
(
2007
).
Morpheus unbound: reimagining the morphogen gradient
.
Cell
128
,
245
-
256
.
Lee
,
R. T.
,
Zhao
,
Z.
and
Ingham
,
P. W.
(
2016
).
Hedgehog signalling
.
Development
143
,
367
-
372
.
Li
,
S.
,
Chen
,
Y.
,
Shi
,
Q.
,
Yue
,
T.
,
Wang
,
B.
and
Jiang
,
J.
(
2012
).
Hedgehog-regulated ubiquitination controls smoothened trafficking and cell surface expression in Drosophila
.
PLoS Biol.
10
,
e1001239
.
Li
,
S.
,
Li
,
S.
,
Han
,
Y.
,
Tong
,
C.
,
Wang
,
B.
,
Chen
,
Y.
and
Jiang
,
J.
(
2016
).
Regulation of smoothened phosphorylation and high-level hedgehog signaling activity by a plasma membrane associated kinase
.
PLoS Biol.
14
,
e1002481
.
Liu
,
Y.
,
Cao
,
X.
,
Jiang
,
J.
and
Jia
,
J.
(
2007
).
Fused-Costal2 protein complex regulates Hedgehog-induced Smo phosphorylation and cell-surface accumulation
.
Genes Dev.
21
,
1949
-
1963
.
Lum
,
L.
,
Zhang
,
C.
,
Oh
,
S.
,
Mann
,
R. K.
,
von Kessler
,
D. P.
,
Taipale
,
J.
,
Weis-Garcia
,
F.
,
Gong
,
R.
,
Wang
,
B.
and
Beachy
,
P. A.
(
2003
).
Hedgehog signal transduction via Smoothened association with a cytoplasmic complex scaffolded by the atypical kinesin, Costal-2
.
Mol. Cell
12
,
1261
-
1274
. https://doi.org/10.1016/S1097-2765(03)00426-X
Maier
,
D.
,
Cheng
,
S.
and
Hipfner
,
D. R.
(
2012
).
The complexities of G-protein-coupled receptor kinase function in Hedgehog signaling
.
Fly (Austin)
6
,
135
-
141
.
Maier
,
D.
,
Cheng
,
S.
,
Faubert
,
D.
and
Hipfner
,
D. R.
(
2014
).
A broadly conserved g-protein-coupled receptor kinase phosphorylation mechanism controls Drosophila smoothened activity
.
PLoS Genet.
10
,
e1004399
.
Malpel
,
S.
,
Claret
,
S.
,
Sanial
,
M.
,
Brigui
,
A.
,
Piolot
,
T.
,
Daviet
,
L.
,
Martin-Lannerée
,
S.
and
Plessis
,
A.
(
2007
).
The last 59 amino acids of Smoothened cytoplasmic tail directly bind the protein kinase Fused and negatively regulate the Hedgehog pathway
.
Dev. Biol.
303
,
121
-
133
.
Martin
,
V.
,
Carrillo
,
G.
,
Torroja
,
C.
and
Guerrero
,
I.
(
2001
).
The sterol-sensing domain of Patched protein seems to control Smoothened activity through Patched vesicular trafficking
.
Curr. Biol.
11
,
601
-
607
. https://doi.org/10.1016/S0960-9822(01)00178-6
Monnier
,
V.
,
Dussillol
,
F.
,
Alves
,
G.
,
Lamour-Isnard
,
C.
and
Plessis
,
A.
(
1998
).
Suppressor of fused links fused and Cubitus interruptus on the hedgehog signalling pathway
.
Curr. Biol.
8
,
583
-
586
.
Monnier
,
V.
,
Ho
,
K. S.
,
Sanial
,
M.
,
Scott
,
M. P.
and
Plessis
,
A.
(
2002
).
Hedgehog signal transduction proteins: contacts of the Fused kinase and Ci transcription factor with the kinesin-related protein Costal2
.
BMC Dev. Biol.
2
,
4
.
Motzny
,
C. K.
and
Holmgren
,
R.
(
1995
).
The Drosophila cubitus interruptus protein and its role in the wingless and hedgehog signal transduction pathways
.
Mech. Dev.
52
,
137
-
150
. https://doi.org/10.1016/0925-4773(95)00397-J
Nakano
,
Y.
,
Guerrero
,
I.
,
Hidalgo
,
A.
,
Taylor
,
A.
,
Whittle
,
J. R. S.
and
Ingham
,
P. W.
(
1989
).
A protein with several possible membrane-spanning domains encoded by the Drosophila segment polarity gene patched
.
Nature
341
,
508
-
513
.
Ogden
,
S. K.
,
Ascano
,
M.
, Jr
,
Stegman
,
M. A.
,
Suber
,
L. M.
,
Hooper
,
J. E.
and
Robbins
,
D. J.
(
2003
).
Identification of a functional interaction between the transmembrane protein Smoothened and the kinesin-related protein Costal2
.
Curr. Biol.
13
,
1998
-
2003
.
Ohlmeyer
,
J. T.
and
Kalderon
,
D.
(
1998
).
Hedgehog stimulates maturation of Cubitus interruptus into a labile transcriptional activator
.
Nature
396
,
749
-
753
.
Pak
,
E.
and
Segal
,
R. A.
(
2016
).
Hedgehog signal transduction: key players, oncogenic drivers, and cancer therapy
.
Dev. Cell
38
,
333
-
344
.
Ranieri
,
N.
,
Therond
,
P. P.
and
Ruel
,
L.
(
2014
).
Switch of PKA substrates from Cubitus interruptus to Smoothened in the Hedgehog signalosome complex
.
Nat. Commun.
5
,
5034
.
Ruel
,
L.
,
Gallet
,
A.
,
Raisin
,
S.
,
Truchi
,
A.
,
Staccini-Lavenant
,
L.
,
Cervantes
,
A.
and
Therond
,
P. P.
(
2007
).
Phosphorylation of the atypical kinesin Costal2 by the kinase Fused induces the partial disassembly of the Smoothened-Fused-Costal2-Cubitus interruptus complex in Hedgehog signalling
.
Development
134
,
3677
-
3689
.
Shi
,
Q.
,
Li
,
S.
,
Jia
,
J.
and
Jiang
,
J.
(
2011
).
The Hedgehog-induced Smoothened conformational switch assembles a signaling complex that activates Fused by promoting its dimerization and phosphorylation
.
Development
138
,
4219
-
4231
.
Su
,
Y.
,
Ospina
,
J. K.
,
Zhang
,
J.
,
Michelson
,
A. P.
,
Schoen
,
A. M.
and
Zhu
,
A. J.
(
2011
).
Sequential phosphorylation of smoothened transduces graded hedgehog signaling
.
Sci. Signal.
4
,
ra43
.
Tirat
,
A.
,
Freuler
,
F.
,
Stettler
,
T.
,
Mayr
,
L. M.
and
Leder
,
L.
(
2006
).
Evaluation of two novel tag-based labelling technologies for site-specific modification of proteins
.
Int. J. Biol. Macromol.
39
,
66
-
76
.
Vervoort
,
M.
(
2000
).
hedgehog and wing development in Drosophila: a morphogen at work?
BioEssays
22
,
460
-
468
.
Wang
,
Q. T.
and
Holmgren
,
R. A.
(
2000
).
Nuclear import of cubitus interruptus is regulated by hedgehog via a mechanism distinct from Ci stabilization and Ci activation
.
Development
127
,
3131
-
3139
.
Xia
,
R.
,
Jia
,
H.
,
Fan
,
J.
,
Liu
,
Y.
and
Jia
,
J.
(
2012
).
USP8 promotes smoothened signaling by preventing its ubiquitination and changing its subcellular localization
.
PLoS Biol.
10
,
e1001238
.
Zadorozny
,
E. V.
,
Little
,
J. C.
and
Kalderon
,
D.
(
2015
).
Contributions of Costal 2-Fused interactions to Hedgehog signaling in Drosophila
.
Development
142
,
931
-
942
.
Zhao
,
Y.
,
Tong
,
C.
and
Jiang
,
J.
(
2007
).
Hedgehog regulates smoothened activity by inducing a conformational switch
.
Nature
450
,
252
-
258
.
Zhao
,
Z.
,
Lee
,
R. T. H.
,
Pusapati
,
G. V.
,
Iyu
,
A.
,
Rohatgi
,
R.
and
Ingham
,
P. W.
(
2016
).
An essential role for Grk2 in Hedgehog signalling downstream of Smoothened
.
EMBO Rep.
17
,
739
-
752
.
Zhou
,
Q.
and
Kalderon
,
D.
(
2010
).
Costal 2 interactions with Cubitus interruptus (Ci) underlying Hedgehog-regulated Ci processing
.
Dev. Biol.
348
,
47
-
57
.
Zhou
,
Q.
and
Kalderon
,
D.
(
2011
).
Hedgehog activates fused through phosphorylation to elicit a full spectrum of pathway responses
.
Dev. Cell
20
,
802
-
814
.
Zhu
,
A. J.
,
Zheng
,
L.
,
Suyama
,
K.
and
Scott
,
M. P.
(
2003
).
Altered localization of Drosophila Smoothened protein activates Hedgehog signal transduction
.
Genes Dev.
17
,
1240
-
1252
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information