Members of the Hedgehog (Hh) family of intercellular signaling molecules play crucial roles in animal development. Aberrant regulation of Hh signaling in humans causes developmental defects, and leads to various genetic disorders and cancers. We have characterized a novel regulator of Hh signaling through the analysis of the zebrafish midline mutant iguana (igu). Mutations in igu lead to reduced expression of Hh target genes in the ventral neural tube, similar to the phenotype seen in zebrafish mutants known to affect Hh signaling. Contradictory at first sight, igu mutations lead to expanded Hh target gene expression in somites. Genetic and pharmacological analyses revealed that the expression of Hh target genes in igu mutants requires Gli activator function but does not depend on Smoothened function. Our results show that the ability of Gli proteins to activate Hh target gene expression in response to Hh signals is generally reduced in igu mutants both in the neural tube and in somites. Although this reduced Hh signaling activity leads to a loss of Hh target gene expression in the neural tube, the same low levels of Hh signaling appear to be sufficient to activate Hh target genes throughout somites because of different threshold responses to Hh signals. We also show that Hh target gene expression in igu mutants is resistant to increased protein kinase A activity that normally represses Hh signaling. Together, our data indicate that igu mutations impair both the full activation of Gli proteins in response to Hh signals, and the negative regulation of Hh signaling in tissues more distant from the source of Hh. Positional cloning revealed that the igu locus encodes Dzip1, a novel intracellular protein that contains a single zinc-finger protein-protein interaction domain. Overexpression of Igu/Dzip1 proteins suggested that Igu/Dzip1 functions in a permissive way in the Hh signaling pathway. Taken together, our studies show that Igu/Dzip1 functions as a permissive factor that is required for the proper regulation of Hh target genes in response to Hh signals.

During animal development, cell fates are specified in response to gradients of signaling molecules within the embryo. Members of the Hedgehog(Hh) family of intercellular signaling molecules control a variety of developmental processes, ranging from segment patterning in Drosophila to organogenesis, left-right asymmetry and dorsoventral patterning of the spinal cord and forebrain in vertebrates (reviewed by Ingham and McMahon, 2001; McMahon et al., 2003). Aberrant regulation of Hh signaling in humans causes developmental defects such as holoprosencephaly (HPE) (reviewed by Wallis and Muenke, 2000) and postaxial polydactyly (Radhakrishna et al., 1997), and can also lead to various types of cancers,including basal cell carcinoma and medulloblastoma (reviewed by Goodrich and Scott, 1998; Ruiz i Altaba, 1999b; Villavicencio et al.,2000).

Because Hh signaling plays such a central role in development and disease,the Hh signaling pathway has been investigated in considerable detail. Genetic and in vitro studies in Drosophila have revealed that Hh signals are transduced by binding of Hh ligands to the Patched (Ptc) cell-surface receptor, resulting in the activation of the transmembrane protein Smoothened(Smo). In Drosophila, the intracellular regulation of Hh signaling is mediated by post-translational modifications of Cubitus interruptus (Ci), a zinc-finger-containing transcription factor of the Gli family that can be both an activator and a repressor of Hh target genes(Methot and Basler, 2001). In the absence of Hh signal, proteolytic cleavage converts Ci to a transcriptional repressor (Aza-Blanc et al., 1997; Wang and Holmgren,1999). In the presence of Hh signals, cleavage of Ci is inhibited and a full-length activator isoform predominates. In vertebrates, at least three Gli genes, Gli1, Gli2 and Gli3, mediate the transcriptional response to Hh signals(Hui et al., 1994; Ruiz i Altaba, 1998). The functions of these different Gli genes have been analyzed in mouse, Xenopus, zebrafish and cultured cells (reviewed by Ingham and McMahon, 2001; Koebernick and Pieler, 2002; Ruiz i Altaba et al., 2002). Although Gli1 is dispensable for normal mouse development(Park et al., 2000; Bai et al., 2002), analysis of Gli1–/–; Gli2–/+mutants (Park et al., 2000)and the rescue of Gli2 mutant by Gli1 (Bai and Joyner, 2001) suggest that Gli1 has an activator function. Consistently, gli1 mutations in zebrafish lead to a loss of Hh target gene expression (Karlstrom et al.,2003). As yet there is no evidence that Gli1 activity is regulated by protein processing, it appears to be solely an activator of the Hh response(Epstein et al., 1996; Marigo et al., 1996; Hynes et al., 1997; Lee et al., 1997; Dai et al., 1999). By contrast, Gli2 appears to be both an activator and repressor of Hh target genes, depending on the tissue being examined. Mouse Gli2 mutations are perinatal lethal and result in the downregulation of Hh target genes(Ding et al., 1998; Matise et al., 1998). The C-terminal region of Gli2 appears to be essential for the activation function,as the C-terminally truncated Gli2 proteins do not activate Hh target genes(Karlstrom et al., 1999; Ruiz i Altaba, 1999a; Sasaki et al., 1999; Karlstrom et al., 2003). Several studies have shown that Gli2 can also repress the expression of Hh target genes (von Mering and Basler,1999; Sasaki et al.,1999; Aza-Blanc et al.,2000; Karlstrom et al.,2003). Similarly, several lines of evidence suggest that Gli3 can act both as an activator and as a repressor of Hh signaling(Masuya et al., 1995; Dai et al., 1999; Ruiz i Altaba, 1999a; Sasaki et al., 1999; Shin et al., 1999; Aza-Blanc et al., 2000; Tole et al., 2000; Litingtung and Chiang, 2000; Wang et al., 2000; Persson et al., 2002). Despite suggestions that proteolytic processing may regulate Gli2 and Gli3 function,the in vivo cleavage of these proteins has not been directly demonstrated.

Thus, in both vertebrates and invertebrates, Hh signaling controls the expression of target genes by modulating the activity of the downstream Gli/Ci transcription factors. Studies in Drosophila have identified a large number of proteins that are involved in the regulation of this Gli/Ci activity(reviewed by McMahon, 2000; Ingham and McMahon, 2001; Nybakken and Perrimon, 2002). Within target cells, Ci forms a protein complex with Fused kinase (Fu)(Monnier et al., 1998; Stegman et al., 2000) and Costal2 (Cos2) (Robbins et al.,1997; Sisson et al.,1997) that is tethered to microtubules. Activated Smo signals to this cytoplasmic protein complex, resulting in the release of Ci and active transport into the nucleus where it can activate Hh target genes(Ohlmeyer and Kalderon, 1998; Ding et al., 1999; Kogerman et al., 1999; Methot and Basler, 2000; Murone et al., 2000; Wang et al., 2000). The transport of Ci to the nucleus is mediated by another protein, Suppressor of Fused [Su(Fu)]. In addition to such a positive regulation in response to Hh ligands, a negative pathway that includes cAMP-dependent protein kinase (PKA)also regulates Hh signaling (Li et al.,1995; Pan and Rubin,1995; Lepage et al.,1995; Hammerschmidt et al.,1996). In the absence of Hh ligands, PKA directly phosphorylates Ci to promote its proteolytic cleavage, generating the repressor isoform(Chen et al., 1998; Chen et al., 1999; Price and Kalderon, 1999). Studies in vertebrates have shown that these intracellular components of the Hh signaling pathway have been largely conserved through evolution(Hammerschmidt et al., 1996; Pearse et al., 1999; Kogerman et al., 1999; Ding et al., 1999; Murone et al., 2000).

Genetic studies of Hh signaling in zebrafish complement the analyses in fly and other vertebrate species, and provide an approach to look into the regulation of Hh signaling in vertebrates. In vertebrates, Sonic hedgehog(Shh) is expressed in the notochord and floor plate(Echelard et al., 1993; Krauss et al., 1993; Roelink et al., 1994; Ekker et al., 1995), and is essential for the induction of floor plate, motoneurons and a class of ventral interneurons in the neural tube (Chiang et al., 1996; Ericson et al.,1996). Shh signaling is also required for the induction of muscle and sclerotome cell types in somites (reviewed by Bumcrot and McMahon, 1995). A large number of zebrafish mutations, collectively called the midline mutants,have been identified that lead to ventral neural tube defects, absence of an optic chiasm and defects in slow muscle fiber formation(Brand et al., 1996; Chen et al., 1996; Karlstrom et al., 1996; van Eeden et al., 1996b). Many of these midline mutants have now been shown to encode components of the Hh signal cascade. shh is disrupted in sonic-you (syu)mutants (Schauerte et al.,1998), smo is disrupted in slow-muscle-omitted(smu) (Chen et al.,2001; Varga et al.,2001), gli2 is disrupted in you-too(yot) (Karlstrom et al.,1999), gli1 is disrupted in detour(dtr) (Karlstrom et al.,2003) and dispatched1 is disrupted in chameleon(con) (Nakano et al.,2004). As several molecularly uncharacterized midline mutants share many phenotypes with these known Hh pathway mutants, it is likely that they encode additional components of Hh signaling.

In this study, we show that igu mutations lead to reduced Hh target gene expression in the neural tube. Surprisingly, these same mutations cause the ectopic activation of Hh target genes in somites. Our analyses reveal that Igu function is required for the full activation of Hh signaling in response to Hh ligands. We also show that the igu mutations,directly or indirectly, affect the negative regulation of Hh signaling that is required for silencing the Hh target gene expression in the absence of Hh ligands. Positional cloning of the igu gene revealed that the gene encodes Dzip1, a novel component of the Hh signaling pathway. We show that Igu/Dzip1 acts as a necessary permissive factor for the proper regulation of Hh signaling.

Fish strains and genetics

The following zebrafish mutants were used, syu (tbx392), yot (ty119), dtr (ts269), con(tm15a) and igu (tms79a and ts294e). Two alleles of igu mutants were indistinguishable in all phenotypes,including marker gene expression. Therefore, we used iguts294e embryos for most of our analyses. For fine mapping, the DNA markers of simple sequence length polymorphisms (SSLP)(Knapik et al., 1998) and expressed sequence tag (EST) in the vicinity of igu locus on linkage group 6 (LG6)(http://zfin.org/)were selected for scoring meiotic recombination in iguts294e mutation(Postlethwait and Talbot,1997). Among 23 DNA markers that we used, the sox21 marker showed one recombination event and the EST marker unp172 showed no recombination in 422 meioses (see Fig. 6A). Searches of the GenBank sequence database using the Blast program(http://www.ncbi.nlm.nih.gov/blast/)identified overlapping PAC clones that contained unp172, z6624 and sox21. In the igu region, three genes, claudin10, a component of tight junctions, ATP-bindingcassette transporter 4(abcc4) and dzip1, were predicted by using the GenScan program(http://genes.mit.edu/GENSCAN.html).

Fig. 6.

The zinc-finger gene dzip1 is disrupted in igu mutants.(A) Genetic map of igu mutation relative to known genes in the region. P1 artificial chromosome (PAC) clones (below) and predicted ORFs(arrows in the middle) are shown. (B) Sequence ferograms showing nonsense mutations in iguts294e and igutm79aalleles. (C) Schematic view of the structure of predicted Igu/Dzip1 protein. A conserved domain (yellow box) has 51% identity with the human DZIP1. Putative nuclear localization signal (NLS), zinc finger (red boxes) and PEST domains(blue boxes) are shown. (D) Amino acid sequence of zebrafish Igu/Dzip1 protein. The NLS (gray), zinc finger (yellow) and positions of nonsense mutations (green) are highlighted. The N-terminal conserved domain is boxed. PEST sequences are underlined.

Fig. 6.

The zinc-finger gene dzip1 is disrupted in igu mutants.(A) Genetic map of igu mutation relative to known genes in the region. P1 artificial chromosome (PAC) clones (below) and predicted ORFs(arrows in the middle) are shown. (B) Sequence ferograms showing nonsense mutations in iguts294e and igutm79aalleles. (C) Schematic view of the structure of predicted Igu/Dzip1 protein. A conserved domain (yellow box) has 51% identity with the human DZIP1. Putative nuclear localization signal (NLS), zinc finger (red boxes) and PEST domains(blue boxes) are shown. (D) Amino acid sequence of zebrafish Igu/Dzip1 protein. The NLS (gray), zinc finger (yellow) and positions of nonsense mutations (green) are highlighted. The N-terminal conserved domain is boxed. PEST sequences are underlined.

Sequence analysis and characterization of igu mutation

To identify the genetic lesions associated with the two iguallele, the dzip1-coding regions was amplified by PCR from reverse-transcribed RNAs made from wild-type and mutant embryos at 30 hours post fertilization (hpf), and these PCR products were directly sequenced. The point mutations were also confirmed by sequencing subcloned cDNAs and genomic DNAs. The sequences of dzip1 cDNAs were deposited in GenBank under Accession Numbers AB106357 and AB106358.

RNA injection

For RNA injections, PCR fragments of igu-coding sequences from wild-type and mutant alleles were cloned into pCS2+ vector. Capped mRNAs were synthesized in vitro using the mMessage Machine kit (Ambion) according to the manufacturer's instruction and injected into embryos (1- to 4-cell stage) that were obtained from igu+/– incrosses.

Cyclopamine and forskolin treatments

Cyclopamine and forskolin were dissolved in 100% dimethyl sulfoxide (DMSO)at 10 mM and 60.9 mM, respectively. Embryos were dechorinated with pronase at 50% epiboly and placed in 6 ml of embryo medium(Westerfield, 1993) containing 50 μM of cyclopamine or the desired concentrations of forskolin. Surviving embryos were fixed and processed for in situ hybridization.

In situ hybridization and genotyping

Whole-mount in situ hybridization was performed as described(Schier et al., 1997). Embryos were photographed in 80% glycerol, and genomic DNA was recovered from each embryo by proteinase K treatment (1 mg/ml) in 50 μl of lysis buffer (10 mM Tris-HCl pH 8, 50 mM KC1, 0.3% Tween 20, 0.3% NP40, 1 mM EDTA) for 5 hours to overnight at 55°C. For genotyping, 5 μl of genomic DNA was used for each PCR reaction. Mutant embryos were identified using tightly linked genetic markers. Primers used for genotyping were unp172F(5′-TCATGACGAAGCAGTTTGGA-3′) and unp172R(5′-CAGGTGTCGTTTTCAGGGTTA-3′) for iguts294e,z1660 for yotty119, and z14475 for dtrts269.

Transfection and antibody staining

PCR amplified wild-type and mutant (ts294e allele) igu/dzip1-coding sequences were subcloned into pcDNA4-HisMax TOPO(Invitrogen). Transfection into NIH3T3 or HEK293T cells was performed according to the manufacturer's instructions (Fugene, Roche). Cells were fixed at 2 days, permeabilized with methanol and stained with Omni-probe antibody(Santa Cruz Biotechnology) in combination with anti-rabbit FITC (Jackson ImmunoResearch). Lysosomes were visualized with anti-Lamp1 antibody(Hughes and August, 1981) in NIH3T3 cells. We also made FLAG-tagged constructs and obtained a similar result (data not shown).

Aberrant regulation of Hh signaling in igu mutants

Zebrafish midline mutants have a characteristic curved body shape and defects in cell fate specification in the ventral neural tube and somites. Careful analyses of the different midline mutants have revealed a range of phenotypes that help uncover the differential contributions of various Hh pathway components to overall Hh signal transduction(Brand et al., 1996; Chen et al., 1996; van Eeden et al., 1996a; van Eeden et al., 1996b; Karlstrom et al., 1996; Lewis et al., 1999; Odenthal et al., 2000; Sbrogna et al., 2003). To examine more carefully the regional regulation of Hh signaling, we examined the expression of Hh target genes in five midline mutants, syu/shh,dtr/gli1, yot/gli2, con and igu. In the neural tube, the expression of nk2.2, a Hh-induced marker for ventral neuroectoderm(Ericson et al., 1996), is variably reduced in all mutants (Fig. 1A1-F1,A2-F2). However, markers for adjacent dorsal regions, such as pax2 and lim5, are largely unaffected (data not shown). Based on defects in nk2.2 and pax6 expression, these mutants can be ranked in order of severity: con>yot/gli2>dtr/gli1> syu/shh>igu. Thus, nk2.2 expression defects are least severe in igu mutants, with punctate nk2.2 expression remaining in the trunk region (Fig. 1B1). nk2.2 expression defects are most severe in con and yot/gli2 (Fig. 1E1,F1). Consistently, the expression of pax6, a gene that is expressed in dorsally adjacent regions of neural tube and negatively regulated by Hh signaling (Ekker et al.,1995; Macdonald et al.,1995; Barth and Wilson,1995; Ericson et al.,1997), is expanded ventrally in yot/gli2 and conmutants, but not in igu or other mutants(Fig. 1A3-F3). We conclude that defects in Hh signaling are more severe in yot/gli2 and conmutants than in the other mutants examined, while igu mutations impair Hh signaling to a lesser degree. Intriguingly, in dtr/gli1,syu/shh and igu mutants, cells in the ventral spinal cord do not express both nk2.2 and pax6, suggesting that the induction of nk2.2 expression and inhibition of pax6 expression are independently regulated by different threshold responses to Hh signals.

Fig. 1.

Aberrant expression of Hh target genes in igu mutants and comparison with four other Hh signaling mutants. (A1-A7) Wild-type, (B1-B7) igu, (C1-C7) syu/shh, (D1-7) dtr/gli1, (E1-E7) yot/gli2 and (F1-F7) con embryos at 30 hpf. (A1-F1,A2-F2)Expression of nk2.2 in the ventral neural tube is reduced in all mutants. Only igu embryos retain a punctate nk2.2 expression(arrowheads in B1, inset is a higher magnification of boxed area). Lateral views are shown in A1-F1 and the respective cross-sections at the middle of the yolk extension are shown in A2-F2. (A3-F3) Expression of pax6 is ventrally expanded in yot/gli2 (E3) and con (F3), but not in other mutants. (A4-F4,A5-F5) Expression of en1 in somites is shown in lateral views and in cross-sections. en1 expression in MPs and MFFs is completely lost in con (F4, F5). A low level of expression persists in syu/shh (C4) and yot/gli2 (E4), and no defect of en1 expression is seen in dtr/gli1 mutants (D4,D5). In contrast to the other four mutants, en1 expression in somites is upregulated in igu embryos (B4,B5). (A6-F7,A7-F7) Expression of ptc1 is shown in whole mount and in cross-sections. Expression of ptc1 is reduced in syu/shh, dtr/gli1, yot/gli2 and con mutants (C7-F7). Expression is most severely reduced in con mutants, and is mildly reduced in the neural tube of syu/shh,dtr/gli and yot/gli2 mutants. Note that ptc1 expression is not maintained in somites of dtr/gli1 embryos, despite the presence of en1 expression, and that the ptc1 expression persists in the sclerotome regions of yot/gli2 (E7). In iguembryos, ptc1 expression is upregulated in the entire somites (B6,B7), whereas the expression in neural tube is unaffected or slightly reduced in the trunk, and slightly more reduced in the brain.

Fig. 1.

Aberrant expression of Hh target genes in igu mutants and comparison with four other Hh signaling mutants. (A1-A7) Wild-type, (B1-B7) igu, (C1-C7) syu/shh, (D1-7) dtr/gli1, (E1-E7) yot/gli2 and (F1-F7) con embryos at 30 hpf. (A1-F1,A2-F2)Expression of nk2.2 in the ventral neural tube is reduced in all mutants. Only igu embryos retain a punctate nk2.2 expression(arrowheads in B1, inset is a higher magnification of boxed area). Lateral views are shown in A1-F1 and the respective cross-sections at the middle of the yolk extension are shown in A2-F2. (A3-F3) Expression of pax6 is ventrally expanded in yot/gli2 (E3) and con (F3), but not in other mutants. (A4-F4,A5-F5) Expression of en1 in somites is shown in lateral views and in cross-sections. en1 expression in MPs and MFFs is completely lost in con (F4, F5). A low level of expression persists in syu/shh (C4) and yot/gli2 (E4), and no defect of en1 expression is seen in dtr/gli1 mutants (D4,D5). In contrast to the other four mutants, en1 expression in somites is upregulated in igu embryos (B4,B5). (A6-F7,A7-F7) Expression of ptc1 is shown in whole mount and in cross-sections. Expression of ptc1 is reduced in syu/shh, dtr/gli1, yot/gli2 and con mutants (C7-F7). Expression is most severely reduced in con mutants, and is mildly reduced in the neural tube of syu/shh,dtr/gli and yot/gli2 mutants. Note that ptc1 expression is not maintained in somites of dtr/gli1 embryos, despite the presence of en1 expression, and that the ptc1 expression persists in the sclerotome regions of yot/gli2 (E7). In iguembryos, ptc1 expression is upregulated in the entire somites (B6,B7), whereas the expression in neural tube is unaffected or slightly reduced in the trunk, and slightly more reduced in the brain.

Hh signaling is also required for cell fate determination in somitic mesoderm and is responsible for the induction of muscle pioneer cells (MPs)and a population of fast fibers around the MPs termed medial fast fibers(MFFs) (Currie and Ingham,1996; Hammerschmidt et al.,1996; Blagden et al.,1997; Roy et al.,2001; Wolff et al.,2003). These cell types are marked by strong and weak expression of engrailed1 (en1), respectively(Fig. 1A4,A5). In accordance with the defects of nk2.2 expression in the neural tube, en1expression in somites is completely lost in con mutants(Fig. 1F4,F5) and severely reduced in syu/shh and yot/gli2 mutants(Fig. 1C4,C5,E4,E5). As previously shown using an anti-En antibody(Wolff et al., 2003), dtr/gli1 mutations do not affect en1 expression in MPs and MFFs (Fig. 1D4,D5). In striking contrast to the defects seen in the other Hh pathway mutants, igumutations lead to an increase in en1 positive cells in somites. This suggests either that Hh signaling is increased in igu mutants, or that Hh signals propagate farther from the source of Hh.

To verify that defects in nk2.2 and en1 expression are correlated with the changes in Hh signaling, we examined the expression of patched1 (ptc1), a sensitive indicator of Hh signaling(Concordet et al., 1996; Goodrich et al., 1996). In agreement with the observed en1 phenotypes, ptc1 expression is reduced in somites of syu/shh, dtr/gli1, yot/gli2 and conmutants. More specifically, ptc1 expression is severely reduced in con mutants both in the neural tube and in somites(Fig. 1F6,F7), whereas it is less severely reduced in the neural tube in syu/shh, dtr/gli1 and yot/gli2 mutants (Fig. 1C6-E6,C7-E7). Intriguingly, in yot/gli2 mutant embryos,the medial-ventral regions of somites retain ptc1(Fig. 1E7). This region corresponds to developing sclerotomal tissues(Morin-Kensicki and Eisen,1997), which is also induced by Hh signals(Fan and Tessier-Lavigne,1994). In marked contrast to the other Hh pathway mutants, igu mutants show expanded ptc1 expression in somites(Fig. 1B4-B7), indicating that Hh signaling is ectopically activated in somites. The ectopic ptc1expression is evident as early as 12 hpf(Fig. 2I). By contrast, ptc1 expression in the ventral neural tube appears to be unchanged or slightly decreased compared with wild-type embryos(Fig. 1A7,B7). Thus, igu mutants show ectopic activation of Hh target genes in somites despite the reduction of the Hh target genes ptc1 and nk2.2in the neural tube. Such an aberrant regulation of Hh target gene expression suggests that the igu gene product may play an important role in both the positive and negative regulation of Hh signaling in different tissues.

Fig. 2.

Hh-independent target gene expression in igu embryos. (A-D) In control wild-type embryos treated with 5% DMSO, expression patterns of ptc1 at 14 hpf (A), myod at 14 hpf (B), en1 at 30hpf (C), ptc1 at 30 hpf (D) and nk2.2 at 30 hpf (E) are all normal. (F-J) Wild-type embryos treated with cyclopamine. Expression of ptc1 at 14 hpf (F), adaxial myod at 14 hpf (G), en1at 30hpf (H), ptc1 at 30 hpf (I) and nk2.2 (J) at 30 hpf, is efficiently suppressed. Note that a low level of ptc1 expression in the neural tube in I appears to be independent of Smo-mediated Hh signaling,since similar ptc1 activation is seen in smu/smo mutants at 30 hpf (data not shown). (K-O) igu embryos treated with cyclopamine. A broad activation of ptc1 expression in igu mutants at 14 hpf (K) is not affected by cyclopamine. Arrows indicate the adaxial cells. Expression of myod in adaxial cells is indistinguishable from normal wild-type embryos, and is not affected by cyclopamine (L). Likewise, en1 (M) and ptc1 (N) expression in 30 hpf embryos is not affected by cyclopamine. Despite the defect of nk2.2 expression in igu mutants, the residual expression of nk2.2 in the neural tube is not eliminated by cyclopamine (O). The genotypes of these embryos were confirmed using tightly linked PCR-based genetic markers after in situ staining.

Fig. 2.

Hh-independent target gene expression in igu embryos. (A-D) In control wild-type embryos treated with 5% DMSO, expression patterns of ptc1 at 14 hpf (A), myod at 14 hpf (B), en1 at 30hpf (C), ptc1 at 30 hpf (D) and nk2.2 at 30 hpf (E) are all normal. (F-J) Wild-type embryos treated with cyclopamine. Expression of ptc1 at 14 hpf (F), adaxial myod at 14 hpf (G), en1at 30hpf (H), ptc1 at 30 hpf (I) and nk2.2 (J) at 30 hpf, is efficiently suppressed. Note that a low level of ptc1 expression in the neural tube in I appears to be independent of Smo-mediated Hh signaling,since similar ptc1 activation is seen in smu/smo mutants at 30 hpf (data not shown). (K-O) igu embryos treated with cyclopamine. A broad activation of ptc1 expression in igu mutants at 14 hpf (K) is not affected by cyclopamine. Arrows indicate the adaxial cells. Expression of myod in adaxial cells is indistinguishable from normal wild-type embryos, and is not affected by cyclopamine (L). Likewise, en1 (M) and ptc1 (N) expression in 30 hpf embryos is not affected by cyclopamine. Despite the defect of nk2.2 expression in igu mutants, the residual expression of nk2.2 in the neural tube is not eliminated by cyclopamine (O). The genotypes of these embryos were confirmed using tightly linked PCR-based genetic markers after in situ staining.

Hh-independent expression of Hh target genes in igumutants

To further characterize the role of the igu gene in Hh signaling,we performed epistasis analyses using cyclopamine, an alkaloid that binds and inhibits the function of Smo (Incardona et al., 1998; Taipale et al.,2000). Cyclopamine has been shown to effectively and specifically block Hh signaling in zebrafish (Barresi et al., 2000; Chen et al.,2001; Sbrogna et al.,2003). Embryos treated with cyclopamine lose the expression of Hh target genes (Fig. 2F-J, Table 1) and resemble smu/smo loss-of-function mutants(Chen et al., 2001; Varga et al., 2001). Residual faint expression of ptc1 in the neural tube(Fig. 2I) appears to be independent of Smo, as similar weak ptc1 expression is seen in the neural tube of smu/smo mutants (data not shown). In contrast to wild-type embryos, cyclopamine treatment of igu mutants did not alter the ectopic expression of ptc1 and en1 in somites(Fig. 2K,M,N; Table 1). Surprisingly,seemingly normal expression of myod(Weinberg et al., 1996) in adaxial cells of igu mutants is also resistant to cyclopamine treatment (Fig. 2B,L). Importantly, cyclopamine treatment did not eliminate residual expression of nk2.2 in igu mutants(Fig. 2J). These results show that the activation of Hh target genes in igu mutants is independent of Hh signals in both somitic and neural tissue. Furthermore, the fact that blocking Smo function had no effect on the igu phenotypes suggests that Igu functions downstream of Smo or in a parallel pathway.

Table 1.

Summary of epistasis analyses

 myod (12-14hpf)
 
 ptc1 (12-14hpf)
 
 nk2.2
 
 ptc1
 
   
Injected mRNA Up igu Up igu Up igu Up igu   
shh (250 pg) 15 5 (25%) 16 4 (20%) 20 4 (17%) 20 10 (33%)   
dnPKA (250 pg) 26 9 (26%) 29 10 (26%) 60 21 (26%) 52 19 (27%)   
 myod (12-14hpf)
 
 ptc1 (12-14hpf)
 
 nk2.2
 
 ptc1
 
 en1
 
 
Inhibitors Down igu Down igu Down igu Down igu Down igu 
Cyclopamine (50 μM) 45 11 (20%) 33 10 (30%) 52 15 (22%) 62 22 (26%) 43 18 (30%) 
Forskolin (300 μM) 45 0 (0%) ND  64 0 (0%) 31 8 (20%) 66 1 (2%) 
(150 μM) 28 1 (3%) ND  45 8 (15%) 38 13 (25%) 44 5 (10%) 
(75 μM) 18 6 (25%) ND  19 8 (30%) 19 8 (30%) 22 7 (24%) 
 myod (12-14hpf)
 
 ptc1 (12-14hpf)
 
 nk2.2
 
 ptc1
 
   
Injected mRNA Up igu Up igu Up igu Up igu   
shh (250 pg) 15 5 (25%) 16 4 (20%) 20 4 (17%) 20 10 (33%)   
dnPKA (250 pg) 26 9 (26%) 29 10 (26%) 60 21 (26%) 52 19 (27%)   
 myod (12-14hpf)
 
 ptc1 (12-14hpf)
 
 nk2.2
 
 ptc1
 
 en1
 
 
Inhibitors Down igu Down igu Down igu Down igu Down igu 
Cyclopamine (50 μM) 45 11 (20%) 33 10 (30%) 52 15 (22%) 62 22 (26%) 43 18 (30%) 
Forskolin (300 μM) 45 0 (0%) ND  64 0 (0%) 31 8 (20%) 66 1 (2%) 
(150 μM) 28 1 (3%) ND  45 8 (15%) 38 13 (25%) 44 5 (10%) 
(75 μM) 18 6 (25%) ND  19 8 (30%) 19 8 (30%) 22 7 (24%) 

Embryos were obtained from igu/+ incrosses. Expected genotypes were 25% igu-/-, 50% igu-/+ and 25%homozygous wild type.

Up, respective Hh target genes are ectopically expressed. These embryos have +/+ or +/igu genotypes.

Down, Gene expression is downregulated.

igu, Hh target genes are expressed in the same pattern in igu mutants.

Percentages in parenthesis represent the ratios of embryos with igu-type gene expression to the total embryos tested for respective gene expressions. All of these embryos have igu/igu genotype. Note that igu embryos have a seemingly normal myod expression pattern.

Attenuated Gli activating function in igu mutants

To test whether the activation of Hh target genes in igu mutants depends on the activation of the Hh signaling pathway, as opposed to other signaling pathways, we made compound mutants between igu and mutants that affect the Gli transcription factors(Fig. 3). To reduce Gli activator function, we made compound mutants with dtr/gli1, which is a loss-of-function allele of the gli1 transcriptional activator(Karlstrom et al., 2003). In dtr/gli1–/+;igu–/– compound mutants, spinal cord expression of nk2.2 is completely absent,similar to the phenotype seen in dtr/gli1–/–mutants (Fig. 3A,D,G). In the somites, posterior expression of en1 is expanded in dtr/gli1–/+;igu–/–embryos, similar to igu–/– mutants, while the expression is reduced in anterior somites(Fig. 3B,E). In dtr/gli1–/–;igu–/–homozygous double mutants, en1 expression is further reduced(Fig. 3H). Similarly, the expansion of somitic ptc1 expression seen in igu mutants is reduced when one or two mutant copies of dtr/gli1 are added(Fig. 3C,F,I). Taken together,these data indicate that the activation of Hh target genes in igumutants partially depends on Gli1 activator function.

Fig. 3.

Decreased Gli activator function and/or increased Gli repressor function represses ectopic Hh target gene expression in igu embryos. (A-I)Introduction of dtr/gli1 mutant alleles into the igu genetic background suppressed ectopic Hh target gene expressions in somites. In dtr/gli1–/– embryos (A-C), the expression of nk2.2 and ptc1 is reduced, but en1 expression is normal. In dtr/gli1–/+;igu–/–embryos (D-F), nk2.2 expression is similar to dtr/gli1–/– embryos (D), with no nk2.2 expression in the posterior neural tube. By contrast, the expression of en1 (E) and ptc1 (F) are similar to that seen in igu–/– mutants; however, en1expression is reduced in anterior somites. In dtr/gli1–/–;igu–/–embryos (G-I), the decrease of en1 and ptc1 is more evident(H,I), and the overall phenotype including nk2.2 (G) is stronger than in dtr/gli1–/– embryos. Insets show higher magnification views of ptc1 expression in the trunk region. (J-O) A drastic reduction of Hh target gene expression is caused by the introduction of a yot/gli2 mutant allele into the igu genetic background. One copy of yot/gli2 completely suppressed ectopic target gene expression (M-O), and the resulting phenotype is similar to or even stronger than that seen in yot/gli2–/– embryos (J-L). An arrowhead marks a spot of nk2.2 expression, which is consistently seen only in yot/gli2–/– embryos (J) but not in yot/gli2–/+;igu–/– embryos(M). The genotypes of these embryos were confirmed by PCR after in situ staining.

Fig. 3.

Decreased Gli activator function and/or increased Gli repressor function represses ectopic Hh target gene expression in igu embryos. (A-I)Introduction of dtr/gli1 mutant alleles into the igu genetic background suppressed ectopic Hh target gene expressions in somites. In dtr/gli1–/– embryos (A-C), the expression of nk2.2 and ptc1 is reduced, but en1 expression is normal. In dtr/gli1–/+;igu–/–embryos (D-F), nk2.2 expression is similar to dtr/gli1–/– embryos (D), with no nk2.2 expression in the posterior neural tube. By contrast, the expression of en1 (E) and ptc1 (F) are similar to that seen in igu–/– mutants; however, en1expression is reduced in anterior somites. In dtr/gli1–/–;igu–/–embryos (G-I), the decrease of en1 and ptc1 is more evident(H,I), and the overall phenotype including nk2.2 (G) is stronger than in dtr/gli1–/– embryos. Insets show higher magnification views of ptc1 expression in the trunk region. (J-O) A drastic reduction of Hh target gene expression is caused by the introduction of a yot/gli2 mutant allele into the igu genetic background. One copy of yot/gli2 completely suppressed ectopic target gene expression (M-O), and the resulting phenotype is similar to or even stronger than that seen in yot/gli2–/– embryos (J-L). An arrowhead marks a spot of nk2.2 expression, which is consistently seen only in yot/gli2–/– embryos (J) but not in yot/gli2–/+;igu–/– embryos(M). The genotypes of these embryos were confirmed by PCR after in situ staining.

To examine how more severe gli mutations affect the iguphenotype, we generated compound mutants with yot/gli2 mutation. Existing yot/gli2 alleles encode truncated forms of Gli2 that have no activator function and that act as dominant repressors of Gli1-mediated Hh target gene activation (Karlstrom et al.,2003). Strikingly, introduction of one copy of yot/gli2into igu mutants suppressed the ectopic expression of Hh target genes in the somites and further reduced expression in the neural tube. The resultant phenotype was similar to or even stronger than that seen in yot/gli2–/– homozygous mutants(Fig. 3M-O). The phenotype of yot/gli2–/–;igu–/–homozygous double mutants is nearly identical to that seen in yot/gli2–/+;igu–/–embryos (data not shown). These results suggest that Igu functions at the same level as, or upstream of, the Gli proteins in the Hh signaling cascade. More importantly, our results show that Hh target gene expression in igumutants is sensitive to a decrease in Gli activator function. This suggests that the overall Gli mediated activating function of Hh target gene expression is reduced in igu mutants despite the ectopic activation of en1 and ptc1 genes in somites. We conclude that the normal Igu function is necessary for the full activation of Gli-mediated Hh signaling.

igu mutations impair both the proper activation and repression of Hh target genes

Previous studies have established that PKA negatively regulates Hh signaling by promoting the proteolytic processing of Ci/Gli proteins and their conversion to repressor isoforms (Ohlmeyer and Kalderon, 1998; Pan and Rubin, 1995; Lepage et al.,1995; Hammerschmidt et al.,1996). This processing is crucial for silencing target gene expression in the absence of Hh ligands. To explore the possible relationship between Igu and the negative regulation of Hh signaling, we manipulated the PKA activity in igu embryos. First, we inhibited PKA activity by overexpressing the dominant-negative regulatory subunit of PKA (dnPKA). This inhibition of PKA activity in wild-type embryos leads to the ectopic activation of Hh target genes in broad regions of the neural tube and somites(Fig. 4A-C)(Hammerschmidt et al., 1996). However, the expression of dnPKA in igu embryos did not induce expression of Hh-responsive genes (Fig. 4D-F; Table 1). As dnPKA is thought to increase full-length Gli activators by disturbing the processing of Gli proteins into repressor isoforms, our results suggest that igu mutations impair the positive regulation of Gli proteins into a fully active state. This is consistent with results from compound mutants(Fig. 3) and results from shh mRNA injections in which Shh overexpression did not affect the igu mutant phenotype either in somites or in the neural tube(Table 1; data not shown).

Fig. 4.

Inhibition of PKA does not activate Hh target genes. (A-C) Injection of 250 pg of dnPKA mRNA into wild-type embryos induced the ectopic expression of myod at 12 hpf (A), ptc1 at 30 hpf (B) and nk2.2at 30 hpf (C). (D-F) In igu embryos, overexpression of dnPKA did not induce the expression of myod (D), nk2.2 (E) and ptc1 (F), and Hh target gene expression is similar to that in igu mutants. Genotypes were confirmed by PCR after in situ analysis.

Fig. 4.

Inhibition of PKA does not activate Hh target genes. (A-C) Injection of 250 pg of dnPKA mRNA into wild-type embryos induced the ectopic expression of myod at 12 hpf (A), ptc1 at 30 hpf (B) and nk2.2at 30 hpf (C). (D-F) In igu embryos, overexpression of dnPKA did not induce the expression of myod (D), nk2.2 (E) and ptc1 (F), and Hh target gene expression is similar to that in igu mutants. Genotypes were confirmed by PCR after in situ analysis.

To further test the relationship between igu mutations and the negative regulation of Hh signaling, we inhibited Hh signaling using forskolin, which activates adenylate cyclase and increases the activity of PKA(Seamon and Daly, 1981). Treatment of wild-type embryos with forskolin in the range of 75-300 μM completely suppressed Hh target gene expression without producing gross morphological abnormalities (Fig. 5A-D; Table 1). If the igu mutation simply disrupted the positive regulation of Gli proteins, we would predict that forskolin treatment of igu mutants would suppress ectopic Hh target gene expression. Although high doses of forskolin (300 μM) did suppress the expression of Hh target genes in igu mutants (Table 1),a lower dose of forskolin that completely inhibits Hh target gene expression in wild-type embryos (75 μM) did not suppress Hh target gene expression in igu mutants (Fig. 5E-H). This result suggests two possibilities: (1) that the negative regulation of Gli proteins is directly impaired in igumutants; or (2) that Igu mutant proteins have acquired a new function that results in the constitutive production of a low level of Gli activator function and indirectly leads to the impaired negative regulation of Hh signaling. In either case, the ectopic activation of Hh target genes in igu mutants could be explained if igu mutations also affect Gli-mediated repression of Hh target genes in cells more distant from the source of Hh signals.

Fig. 5.

Activation of PKA does not efficiently suppress Hh target gene expression.(A-D) In wild-type embryos, the expression of myod at 14 hpf (A), nk2.2 at 30 hpf (B), en1 at 30 hpf (C) and ptc1 at 30 hpf (D) is repressed by 75 μM forskolin treatment. (E-H) In iguembryos, treatment with 75 μM forskolin did not block Hh target gene expression, with expression patterns similar to those seen in untreated igu mutants. Genotypes were confirmed by PCR after in situ analysis.

Fig. 5.

Activation of PKA does not efficiently suppress Hh target gene expression.(A-D) In wild-type embryos, the expression of myod at 14 hpf (A), nk2.2 at 30 hpf (B), en1 at 30 hpf (C) and ptc1 at 30 hpf (D) is repressed by 75 μM forskolin treatment. (E-H) In iguembryos, treatment with 75 μM forskolin did not block Hh target gene expression, with expression patterns similar to those seen in untreated igu mutants. Genotypes were confirmed by PCR after in situ analysis.

The zinc-finger protein Dzip1 is disrupted in igumutants

To further understand how the igu mutation affects Hh signaling,we sought to identify the igu gene. Fine genetic mapping placed igu in a small region on linkage group 6 that contained dzip1 as a possible candidate gene(Fig. 6A). Homologs of dzip1 are present in human, mouse and other vertebrates, but the function of these proteins in development has not yet been characterized. Sequence analysis of dzip1 cDNA amplified from wild-type and igu embryos revealed point mutations leading to changes in the protein sequence at amino acid 371 in iguts294e and amino acid 454 in igutm79a(Fig. 6B-D). Both of these mutations introduce premature stop codons and result in the C-terminally truncated Igu/Dzip1 proteins. Sequence analysis of the Igu/Dzip1 protein predicts a single zinc-finger domain, a nuclear localization signal (NLS) and two potential PEST sequences that may serve as signals for rapid protein turnover (Rechsteiner and Rogers,1996). The N-terminal domain of Igu/Dzip1, including the NLS and zinc-finger domain, has a significant higher homology (51% amino acid identity) to human DZIP1 than does the remainder of the protein. In situ expression analysis showed that igu/dzip1 is expressed throughout the embryo starting at 12 hours (data not shown), consistent with the igumutation affecting multiple tissue types.

This predicted protein structure suggested that Igu/Dzip1 may localize in the cytoplasm and/or nucleus. To characterize the subcellular localization of Igu/Dzip1, we expressed wild type and mutant igu/dzip1 in cultured cells. Wild type Igu/Dzip1 is present in the cytoplasm, and is found at especially high levels in large vesicles(Fig. 7A). These vesicles correspond to lysosomes and/or endosomes based on the co-localization with the lysosomal protein Lamp1 (Hughes and August, 1981) (Fig. 7B,C). By contrast, the mutant Igu/Dzip1 protein, which lacks the PEST sequences but retains the NLS, is strongly enriched in nuclei(Fig. 7D-F). Because a single zinc-finger domain does not bind to DNA, it is likely that Igu/Dzip1 is involved in protein-protein interactions. Indeed, human DZIP1 has been suggested to interact with DAZ, a protein required for spermatogenesis(Moore et al., 2003). Therefore, our results raise the possibility that Igu/Dzip1 might regulate the stability or the nuclear translocation of other proteins, including Gli proteins or other components of the Hh signaling pathway.

Fig. 7.

Subcellular localization of Igu/Dzip1 proteins in cultured cells. (A-C)Transfection of a construct encoding the His-tagged Igu/Dzip1 protein into NIH3T3 cells. Wild-type Igu/Dzip1 proteins (A) are located in the cytoplasm and enriched in punctate vesicles. These vesicles are also enriched with the lysosomal protein Lamp1 (B, C). (D-F) Transfection of a construct encoding the His-tagged Igu/Dzip1(ts294e) mutant protein. The truncated mutant Igu/Dzip1 proteins (D) are distributed in the cytoplasm and enriched in nuclei. (C,F) Merged images, in which nuclei are stained with 4′,6-diamidino-2-phenylindole (DAPI).

Fig. 7.

Subcellular localization of Igu/Dzip1 proteins in cultured cells. (A-C)Transfection of a construct encoding the His-tagged Igu/Dzip1 protein into NIH3T3 cells. Wild-type Igu/Dzip1 proteins (A) are located in the cytoplasm and enriched in punctate vesicles. These vesicles are also enriched with the lysosomal protein Lamp1 (B, C). (D-F) Transfection of a construct encoding the His-tagged Igu/Dzip1(ts294e) mutant protein. The truncated mutant Igu/Dzip1 proteins (D) are distributed in the cytoplasm and enriched in nuclei. (C,F) Merged images, in which nuclei are stained with 4′,6-diamidino-2-phenylindole (DAPI).

Igu is a permissive factor in Hh signaling

To further confirm that dzip1 is the gene mutated in igu,and to gain insights into the molecular nature of the igu mutations,we injected dzip1 mRNAs into wild-type and igu mutant embryos (Fig. 8). Injection of wild-type dzip1 mRNA (50 pg) into igu embryos dramatically rescued the igu phenotypes both in the neural tube and somites of almost all mutant embryos (Fig. 8A-E; Table 2),consistent with dzip1 being the gene disrupted in igumutants. Despite the dramatic rescue of the igu phenotype, no effect was observed upon injection of dzip1 mRNA into wild-type embryos(Fig. 8F-J), suggesting that Igu/Dzip1 is a permissive factor in Hh signaling. Importantly, the injection of mutant dzip1 mRNA into igu embryos[(Fig. 8K-O) or into wild-type embryos (Fig. 8P-T)] did not alter the Hh target gene expression (Table 2). Even a large amount of wild-type or mutant dzip1 mRNA(250 pg) did not produce a dominant effect on Hh target gene expression(Table 2). Thus, Igu/Dzip1,while being required for the proper regulation of Hh signaling, is not sufficient to activate or repress Hh target genes, and is therefore a permissive factor in the Hh signaling cascade

Fig. 8.

Rescue of igu mutant phenotype by igu/dzip1 mRNA and permissive nature of Igu/Dzip1 proteins. (A-J) Injection of wild-type igu/dzip1 mRNA. Nearly all igu embryos injected with wild-type igu/dzip1 mRNA completely recovered the normal expression of Hh target genes (A-E), including: ptc1 expression at 14 hpf (A), nk2.2 expression at 30 hpf (B), ptc1 expression at 30 hpf(C, D) and en1 at 30 hpf (E) (compare with Fig. 1). Wild-type embryos injected with wild type igu/dzip1 mRNA did not show any defects in Hh target gene expression (F-J). (K-T) Injection of mutant mRNA that encodes truncated igu/dzip1 proteins. Expression of mutant Igu/Dzip1 does not affect Hh target gene expression in igu mutants (K-O) or wild-type embryos (P-T). (A,D,F,I,K,N,P,S) Dorsal views and other panels are side views. Expression patterns in A-J are identical to those seen in wild-type embryos,while those in K-O are identical to igu mutant embryos (see Fig. 1 for comparison).

Fig. 8.

Rescue of igu mutant phenotype by igu/dzip1 mRNA and permissive nature of Igu/Dzip1 proteins. (A-J) Injection of wild-type igu/dzip1 mRNA. Nearly all igu embryos injected with wild-type igu/dzip1 mRNA completely recovered the normal expression of Hh target genes (A-E), including: ptc1 expression at 14 hpf (A), nk2.2 expression at 30 hpf (B), ptc1 expression at 30 hpf(C, D) and en1 at 30 hpf (E) (compare with Fig. 1). Wild-type embryos injected with wild type igu/dzip1 mRNA did not show any defects in Hh target gene expression (F-J). (K-T) Injection of mutant mRNA that encodes truncated igu/dzip1 proteins. Expression of mutant Igu/Dzip1 does not affect Hh target gene expression in igu mutants (K-O) or wild-type embryos (P-T). (A,D,F,I,K,N,P,S) Dorsal views and other panels are side views. Expression patterns in A-J are identical to those seen in wild-type embryos,while those in K-O are identical to igu mutant embryos (see Fig. 1 for comparison).

Table 2.

dzip1 mRNA rescues igu phenotypes

ptc1 (12-14 hpf)
ptc1 (30 hpf)
en1 (30 hpf)
nk2.2 (30 hpf)
RNA injectedNormaligu likeNormaligu likeNormaligu likeNormaligu like
dzip1 50 pg 19 0 (0%) 145 1 (1%) 101 0 (0%) 132 2 (2%) 
dzip1 (iguts294e) 50 pg 19 5 (21%) 53 19 (26%) 36 11 (23%) 58 21 (27%) 
dzip1 (iguts294e) 250 pg 19 5 (21%) 23 7 (23%) 16 7 (30%) 22 5 (19%) 
ptc1 (12-14 hpf)
ptc1 (30 hpf)
en1 (30 hpf)
nk2.2 (30 hpf)
RNA injectedNormaligu likeNormaligu likeNormaligu likeNormaligu like
dzip1 50 pg 19 0 (0%) 145 1 (1%) 101 0 (0%) 132 2 (2%) 
dzip1 (iguts294e) 50 pg 19 5 (21%) 53 19 (26%) 36 11 (23%) 58 21 (27%) 
dzip1 (iguts294e) 250 pg 19 5 (21%) 23 7 (23%) 16 7 (30%) 22 5 (19%) 

Embryos were obtained from igu/+ incrosses. Expected genotypes were 25% igu-/-, 50% igu-/+ and 25%homozygous wild type. Percentages in parenthesis represent the ratios of embryos with igu-type gene expression to the total embryo tested for respective gene expressions. All of these embryos were confirmed to have igu/igu genotype.

Igu is necessary for the Hh-dependent activation of Gli proteins

The zebrafish Hh pathway mutants dtr/gli1, yot/gli2, syu/shh and con all lead to a loss of Hh target gene expression in both the neural tube and in somites. However, igu is unique among midline mutants in that Hh target gene expression is reduced in the ventral neural tube and brain, whereas in somites the expression of Hh target genes is expanded. Cyclopamine treatments showed that the Hh-dependency of target gene activation is lost in igu mutants. This is probably not due to the activation of a different signaling pathway, as a decrease in Gli activator function (dtr/gli1;igu double mutants) and/or an increase in Gli repressor function (yot/gli2;igu double mutants) suppressed all Hh target gene expression in igu mutants. These results suggest that Gli proteins are responsible for the ectopic expression of Hh target genes in igu mutants, and that the Igu protein functions upstream of Gli1 and Gli2 in the Hh signaling cascade. As Gli protein activities are regulated in response to Hh signals, the Hh-independent activation of target genes in igu mutants suggests that Igu is not a transcriptional corepressor or co-activator of Gli proteins.

Our examination of compound mutant phenotypes also revealed that the activating ability of Gli proteins is generally reduced in igumutants, despite the apparent upregulation of en1 and ptc1expression in somites. This reduced Gli activator function explains the loss of Hh signaling in the neural tube of igu mutants and suggests that Igu/Dzip1 function is required for the full activation of Gli proteins in response to Hh signals.

Both positive and negative regulation of Hh signaling is impaired in igu mutants

Previous studies have shown that Hh signaling is tightly regulated in the embryo, with different intracellular mechanisms positively and negatively regulating Hh signal transduction. Hh signaling is positively regulated through the functions of Fu, Cos2 and Su(Fu) and the nuclear trafficking of Gli/Ci proteins (see Fig. 9)(Monnier et al., 1998; Ding et al., 1999; Kogerman et al., 1999; Methot and Basler, 2000; Murone et al., 2000; Stegman, et al., 2000; Wang et al., 2000). Negative regulation is thought to occur via PKA-mediated processing of Gli/Ci proteins into repressor isoforms in the absence of Hh ligands(Lepage et al., 1995; Ohlmeyer and Kalderon, 1998; Pan and Rubin, 1995; Hammerschmidt et al., 1996; Jiang and Struhl, 1998).

Fig. 9.

Model of possible Igu function in Hh signaling. (A) In wild-type embryos,Igu function is required for the positive and negative regulation of Gli protein function in response to Hh signals. Components in green have activating functions, and those in red have repressor functions. (B) Without the normal Igu/Dzip1 protein function (light blue), Hh-dependent regulation is lost and Gli proteins cannot fully activate the expression of Hh target genes. In addition, the negative regulation of Hh signaling is also reduced. igu mutations could act directly on PKA-mediated negative regulation of Hh signals (broken arrows and lines in B) and/or could act indirectly,possibly by causing the constitutive nuclear import of Gli proteins (thin arrows). The resulting constitutive, but weak, activating function of Gli proteins is sufficient to induce en1 and ptc1 in broad regions in somites. This same low level of activator function is not sufficient to activate genes such as nk2.2 in the ventral neural tube the transcription of which requires a higher level of Hh signals.

Fig. 9.

Model of possible Igu function in Hh signaling. (A) In wild-type embryos,Igu function is required for the positive and negative regulation of Gli protein function in response to Hh signals. Components in green have activating functions, and those in red have repressor functions. (B) Without the normal Igu/Dzip1 protein function (light blue), Hh-dependent regulation is lost and Gli proteins cannot fully activate the expression of Hh target genes. In addition, the negative regulation of Hh signaling is also reduced. igu mutations could act directly on PKA-mediated negative regulation of Hh signals (broken arrows and lines in B) and/or could act indirectly,possibly by causing the constitutive nuclear import of Gli proteins (thin arrows). The resulting constitutive, but weak, activating function of Gli proteins is sufficient to induce en1 and ptc1 in broad regions in somites. This same low level of activator function is not sufficient to activate genes such as nk2.2 in the ventral neural tube the transcription of which requires a higher level of Hh signals.

Although our analysis of compound mutants suggested that the overall Gli activating function is reduced in igu mutants, it is probably not due to the direct decrease of full-length Gli proteins. If igu mutations simply lead to a reduction of full-length Gli activators, then we would predict that injection of dnPKA would still induce Hh responsive gene expression in igu mutants as it does in wild-type embryos. However,we found that dnPKA injections did not induce Hh responsive genes in igu mutants. This suggests that the positive regulation of Gli proteins, which may be either a modification of Gli proteins or nuclear trafficking, is disrupted in igu mutants.

We also showed that igu mutations reduce the ability of PKA to repress Hh target gene expression. igu mutants were resistant to forskolin, which blocks Hh signaling by activating PKA. This reduced sensitivity to forskolin treatment cannot be simply explained by defects in the positive regulation of Gli proteins. In Drosophila, it has been shown that mutations in PKA lead to the accumulation of Ci and the upregulation of Hh signaling (Ohlmeyer and Kalderon, 1998). Likewise, the upregulation of target genes in igu mutants may partly be due to a defect in the negative regulation of Gli proteins. Taken together, our analyses suggest that both the positive and negative regulation of Hh signaling is impaired in igu mutants(Fig. 9).

Igu protein structure and function

We showed that the igu gene encodes a single zinc-finger protein,Dzip1. Although igu/dzip1 is conserved among vertebrate species such as human, mouse, rat, chicken and frog, we could not identify a homologous gene in Drosophila. Considering the high degree of conservation in the Hh signaling pathway during evolution, a functionally equivalent protein may present in invertebrates, though the sequence might be highly diverged. Intriguingly, human postaxial polydactyly type A2 (PAPA2), a congenital defect that is caused by the ectopic activation of Hh signaling(Villavicencio et al., 2000),maps to a region containing human DZIP1 at 13q32. DZIP1 might be the gene disrupted in PAPA2.

Single zinc-finger domain proteins are not thought to bind DNA, instead they are implicated in mediating protein-protein interactions. The presence of PEST domains in the Dzip1 protein sequence suggests that the protein may be a target for rapid degradation and is consistent with our observation that Igu/Dzip1 accumulates in lysosomes in cultured cells. These facts suggest the possibility that Igu/Dzip1 is involved in mediating the rapid turnover of interacting proteins, perhaps components of the Hh signaling cascade. However,the Igu/Dzip1 protein may not simply be a cytoplasmic protein, as it has a nuclear localization signal (NLS) within the N-terminal conserved region. Indeed, the truncated Igu/Dzip1 proteins encoded by igu are enriched in nuclei in cultured cells, suggesting that the NLS is also functional. This finding raises the possibility that wild type Igu/Dzip1 proteins shuttle between the cytoplasm and the nucleus, and they could thus affect the nuclear import of Hh pathway proteins. Although Gli proteins are possible candidates for Igu/Dzip1 interacting partners, our preliminary data suggest that Igu proteins do not physically interact with Gli proteins in vitro, and they do not alter the transcriptional activation of Gli proteins in a luciferase reporter assay (H.S. and A.K., unpublished). Igu proteins might interact with other components of the Hh signaling pathway such as Fu, Cos2 or Su(Fu). Intriguingly, Su(Fu) is involved in the nuclear trafficking of Gli proteins and also contains a PEST sequence (Pham et al., 1995; Pearse et al.,1999). Moreover, it was recently shown that reducing Su(Fu)function in zebrafish using morpholino oligonucleotides resulted in the upregulation of En expression in somites(Wolff et al., 2003). From these data, we speculate that Igu/Dzip1 might be involved in the regulation of the Su(Fu), Fu and Cos2 cytoplasmic protein complex that is known to regulate Gli protein activities.

Our results also revealed that both wild-type and mutant versions of Igu/Dzip1 proteins do not have a dominant function when overexpressed in wild-type embryos. This, combined with our mutant analysis, indicates that Igu/Dzip1 function is a permissive factor required for the regulation of Hh signaling, and that mutant Igu/Dzip1 proteins do not function dominantly to interfere with the regulation of Hh signaling. These results support the idea that Igu/Dzip1 function is directly required for the negative regulation of Gli proteins. However, we cannot exclude the possibility that Igu mutant proteins have a new function that is masked in injected embryos by the presence of the wild-type protein. In fact, nuclear localization of mutant Igu/Dzip1 proteins in vitro could suggest a gained function for the mutant Igu/Dzip1 proteins. Although we attempted to generate a loss-of-function phenotype by blocking Igu/Dzip1 translation with antisense morpholino oligonucleotides, these morpholinos produced no phenotype in wild-type embryos, and did not alter Hh target gene expression in igu mutants. Considering the weak and ubiquitous expression of the igu gene and the permissive nature of Igu/Dzip1, it is possible that small amounts of Igu/Dzip1 proteins are sufficient for normal development, and that morpholinos are therefore unable to block expression sufficiently to produce a phenotype. Further biochemical analysis of Igu/Dzip1 will be needed to fully elucidate its function.

We thank R. Geisler for providing unpublished primer sequences for ESTs,the zebrafish community for providing probes and H. Yakushi for fish care. We also thank A. F. Schier and W. S. Talbot for critical reading of the manuscript. Lamp1 antibody was obtained from the Developmental Studies Hybridoma Bank. This work was supported by grants from the Ministry of Education, Culture, Sports, Science and Technology of Japan (MEXT) and the Kato Memorial Bioscience Foundation to A.K., and by NIH RO1NS39994(R.O.K.).

Aza-Blanc, P., Ramirez-Weber, F.-A., Laget, M.-P., Schwartz, C. andKornberg, T. B. (
1997
). Proteolysis that is inhibited by hedgehog targets Cubitus interruptus protein to the nucleus and converts it to a repressor.
Cell
89
,
1043
-1053.
Aza-Blanc, P., Lin, H. Y., Ruiz i Altaba, A. and Kornberg, T. B. (
2000
). Expression of the vertebrate Gli proteins in Drosophila reveals a distribution of activator and repressor activities.
Development
127
,
4293
-4301.
Bai, C. B. and Joyner, A. L. (
2001
). Gli1 can rescue the in vivo function of Gli2.
Development
128
,
5161
-5172.
Bai, C. B., Auerbach, W., Lee, J. S., Stephen, D. and Joyner, A. L. (
2002
). Gli2, but not Gli1, is required for initial Shh signaling and ectopic activation of the Shh pathway.
Development
129
,
4753
-4761.
Barth, K. A. and Wilson, S. W. (
1995
). Expression of zebrafish nk2.2 is influenced by sonic hedgehog/vertebrate hedgehog-1 and demarcates a zone of neuronal differentiation in the embryonic forebrain.
Development
121
,
1755
-1768.
Barresi, M. J., Stickney, H. L. and Devoto, S. H.(
2000
). The zebrafish slow-muscle-omitted gene product is required for Hedgehog signal transduction and the development of slow muscle identity.
Development
127
,
2189
-2199.
Blagden, C. S., Currie, P. D., Ingham, P. W. and Hughes, S. M. (
1997
). Notochord induction of zebrafish slow muscle mediated by Sonic hedgehog.
Genes Dev.
11
,
2163
-2175.
Brand, M., Heisenberg, C. P., Warga, R. M., Pelegri, F.,Karlstrom, R. O.,Beuchle, D., Picker, A., Jiang, Y. J.,Furutani-Seiki, M., van Eeden, F. J. et al. (
1996
). Mutations affecting development of the midline and general body shape during zebrafish embryogenesis.
Development
123
,
129
-142.
Bumcrot, D. A. and McMahon, A. P. (
1995
). Somite differentiation. Sonic signals somites.
Curr. Biol.
5
,
612
-614.
Chen, J. N., Haffter, P., Odenthal, J., Vogelsang, E., Brand,M., van Eeden,F. J., Furutani-Seiki, M., Granato, M., Hammerschmidt,M., Heisenberg, C. P. et al. (
1996
). Mutations affecting the cardiovascular system and other internal organs in zebrafish.
Development
123
,
293
-302.
Chen, Y., Gallaher, N., Goodman, R. and Smolik, S.(
1998
). Protein kinase A directly regulates the activity and proteolysis of Cubitus interruptus.
Proc. Natl. Acad. Sci. USA
95
,
2349
-2354.
Chen, Y., Cardinaux, J., Goodman, R. and Smolik, S.(
1999
). Mutants of cubitus interruptus that are independent of PKA regulation are independent of hedgehog signaling.
Development
126
,
3607
-3616.
Chen, W., Burgess, S. and Hopkins, N. (
2001
). Analysis of the zebrafish smoothened mutant reveals conserved and divergent functions of hedgehog activity.
Development
128
,
2385
-2396.
Chiang, C., Litingtung, Y., Lee, E., Young, K. E., Corden, J. L., Westphal,H. and Beachy, P. A. (
1996
). Cyclopia and defective axial patterning in mice lacking sonic hedgehog gene function.
Nature
383
,
407
-413.
Concordet, J. P., Lewis, K. E., Moore, J. W., Goodrich, L. V.,Johnson, R.L., Scott, M. P. and Ingham, P. W. (
1996
). Spatial regulation of a zebrafish patched homologue reflects the roles of sonic hedgehog and protein kinase A in neural tube and somite patterning.
Development
122
,
2835
-2846.
Currie, P. D. and Ingham, P. W. (
1996
). Induction of a specific muscle cell type by a hedgehog-like protein in zebrafish.
Nature
382
,
452
-455.
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.
19
,
8143
-8152.
Ding, Q., Motoyama, J., Gasca, S., Rong, M., Sasaki, H.,Rossant, J. andHui, C.-C. (
1998
). Diminished Sonic hedgehog signaling and lack of floor plate differentiation in Gli2 mutant mice.
Development
125
,
2533
-2543.
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
,
1119
-1122.
Echelard, Y., Epstein, D. J., St-Jacques, B., Shen, L., Mohler,J.,McMahon, J. A. and McMahon, A. P. (
1993
). Sonic hedgehog, a member of a family of putative signaling molecules, is implicated in the regulation of CNS polarity.
Cell
75
,
1417
-1430.
Ekker, S. C., Ungar, A. R., Greenstein, P., von Kessler, D. P.,Porter, J. A.,Moon, R. T. and Beachy, P. A. (
1995
). Patterning activities of vertebrate Hedgehog proteins in the developing eye and brain.
Curr. Biol.
5
,
944
-955.
Epstein, D. J., Marti, E., Scott, M. P. and McMahon, A. P.(
1996
). Antagonizing cAMP-dependent protein kinase A in the dorsal CNS activates a conserved sonic hedgehog signaling pathway.
Development
122
,
2885
-2894.
Ericson, J., Morton, S., Kawakami, A., Roelink, H. and Jessell,T. M. (
1996
). Two critical periods of sonic hedgehog signaling required for the specification of motor neuron identity.
Cell
87
,
661
-673.
Ericson, J., Rashbass, P., Schedl, A., Brenner-Morton, S.,Kawakami, A., van Heyningen, V., Jessell, T. M. and Briscoe, J.(
1997
). Pax6 controls progenitor cell identity and neuronal fate in response to graded Shh signaling.
Cell
90
,
169
-180.
Fan, C. M. and Tessier-Lavigne, M. (
1994
). Patterning of mammalian somites by surface ectoderm and notochord: evidence for sclerotome induction by a hedgehog homolog.
Cell
79
,
1175
-1186.
Goodrich, L. V., Johnson, R. L., Milenkovic, L., McMahon, J. A. and Scott,M. P. (
1996
). Conservation of the hedgehog/patched signaling pathway from flies to mice: induction of a mouse patched gene by hedgehog.
Genes Dev.
10
,
301
-312.
Goodrich, L. V. and Scott, M. P. (
1998
). Hedgehog and patched in neural development and disease.
Neuron
21
,
1243
-1257.
Hammerschmidt, M., Bitgood, M. J. and McMahon, A. P.(
1996
). Protein kinase A is a common negative regulator of hedgehog signaling in the vertebrate embryo.
Genes Dev.
10
,
647
-658.
Hughes, E. N. and August, J. T. (
1981
). Characterization of plasma membrane proteins identified by monoclonal antibodies.
J. Biol. Chem.
256
,
664
-671.
Hui, C. C., Slusarski, D., Platt, K. A., Holmgren, R. and Joyner, A. L. (
1994
). Expression of three mouse homologs of the Drosophila segment polarity gene cubitus interruptus, Gli, Gli-2, and Gli-3, in ectoderm and mesoderm-derived tissues suggests multiple roles during postimplantation development.
Dev. Biol.
62
,
402
-413.
Hynes, M., Stone, D. M., Dowd, M., Pitts-Meek, S., Goddard, A.,Gurney,A. and Rosenthal, A. (
1997
). Control of cell pattern in the neural tube by the zinc finger transcription factor and oncogene gli-1.
Neuron
19
,
15
-26.
Incardona, J. P., Gaffield, W., Kapur, R. P. and Roelink, H.(
1998
). The teratogenic Veratrum alkaloid cyclopamine inhibits sonic hedgehog signal transduction.
Development
125
,
3553
-3562.
Ingham, P. W. and McMahon, A. P. (
2001
). Hedgehog signaling in animal development: paradigms and principles.
Genes Dev.
15
,
3059
-3087.
Jiang, J. and Struhl, G. (
1998
). Regulation of the Hedgehog and Wingless signaling pathways by the F-box/WD40-repeat protein Slimb.
Nature
391
,
493
-496.
Karlstrom, R. O., Trowe, T., Klostermann, S., Baier, H., Brand,M.,Crawford, A. D., Grunewald, B., Haffter, P., Hoffmann, H., Meyer,S. U. et al. (
1996
). Zebrafish mutations affecting retinotectal axon pathfinding.
Development
123
,
427
-438.
Karlstrom, R. O., Talbot, W. S. and Schier, A. F.(
1999
). Comparative synteny cloning of zebrafish you-too:mutations in the Hedgehog target gli2 affect ventral forebrain patterning.
Genes Dev.
13
,
388
-393.
Karlstrom, R. O., Tyurina, O. V., Kawakami, A., Nishioka, N.,Talbot, W.S., Sasaki, H., Schier, A. F. (
2003
). Genetic analysis of zebrafish gli1 and gli2 reveals divergent requirements for gli genes in vertebrate development.
Development
130
,
1549
-1564.
Knapik, E. W., Goodman, A., Ekker, M., Chevrette, M., Delgado,J.,Neuhauss, S., Shimoda, N., Driever, W., Fishman, M. C. and Jacob,H. J. (
1998
). A microsatellite genetic linkage map for zebrafish (Danio rerio).
Nat. Genet.
18
,
338
-343.
Koebernick, K. and Pieler, T. (
2002
). Gli-type zinc finger proteins as bipotential transducers of Hedgehog signaling.
Differentiation
70
,
69
-76.
Kogerman, P., Grimm, T., Kogerman, L., Krause, D., Unden, A. B.,Sandstedt, B., Toftgard, R., and Zaphiropoulos, P. G.(
1999
). Mammalian suppressor-of-fused modulates nuclear cytoplasmic shuttling of Gli-1.
Nat. Cell Biol.
1
,
312
-319.
Krauss, S., Concordet, J. P. and Ingham, P. W.(
1993
). A functionally conserved homolog of the Drosophila segment polarity gene hh is expressed in tissues with polarizing activity in zebrafish embryos.
Cell
75
,
1431
-1444.
Lee, J., Platt, K. A., Censullo, P. and Ruiz i Altaba, A.(
1997
). Gli1 is a target of sonic hedgehog that induces ventral neural tube development.
Development
124
,
2537
-2552.
Lepage, T., Cohen, S. M., Diaz-Benjumea, F. J. and Parkhurst, S. M. (
1995
). Signal transduction by cAMP-dependent protein kinase A in Drosophila limb patterning.
Nature
373
,
711
-715.
Lewis, K. E., Currie, P. D., Roy, S., Schauerte, H., Haffter, P. and Ingham,P. W. (
1999
). Control of muscle cell-type specification in the zebrafish embryo by Hedgehog signaling.
Dev. Biol.
216
,
469
-480.
Li, W., Ohlmeyer, J. T., Lane, M. E. and Kalderon, D.(
1995
). Function of protein kinase A in hedgehog signal transduction and Drosophila imaginal disc development.
Cell
80
,
553
-562.
Litingtung, Y. and Chiang, C. (
2000
). Specification of ventral neuron types is mediated by an antagonistic interaction between Shh and Gli3.
Nat. Neurosci.
3
,
979
-985.
Macdonald, R., Barth, K. A., Xu, Q., Holder, N., Mikkola, I. and Wilson,S. W. (
1995
). Midline signaling is required for Pax gene regulation and patterning of the eyes.
Development
121
,
3267
-3278.
Marigo, V., Johnson, R. L., Vortkamp, A. and Tabin, C. J.(
1996
). Sonic hedgehog differentially regulates expression of gli and gli3 during limb development.
Dev. Biol.
180
,
273
-283.
Masuya, H., Sagai, T., Wakana, S., Moriwaki, K. and Shiroishi,T. (
1995
). A duplicated zone of polarizing activity in polydactylous mouse mutants.
Genes Dev.
9
,
1645
-1653.
Matise, M., Epstein, D., Park, H., Platt, K. and Joyner, A.(
1998
). Gli2 is required for the induction of floor plate and adjacent cells, but not most ventral neurons in the mouse central nervous system.
Development
125
,
2759
-2770.
McMahon, A. P. (
2000
). More surprises in the hedgehog signaling pathway.
Cell
100
,
185
-188.
McMahon, A., Ingham, P. and Tabin, C. (
2003
). The developmental roles and clinical significance of Hedeghog signaling.
Curr. Top. Dev. Biol.
53
,
1
-114.
Methot, N. and Basler, K. (
2000
). Suppressor of fused opposes hedgehog signal transduction by impeding nuclear accumulation of the activator form of Cubitus interruptus.
Development
127
,
4001
-4010.
Methot, N. and Basler, K. (
2001
). An absolute requirement for Cubitus interruptus in Hedgehog signaling.
Development
128
,
733
-742.
Morin-Kensicki, E. M. and Eisen, J. S. (
1997
). Sclerotome development and peripheral nervous system segmentation in embryonic zebrafish.
Development
124
,
159
-167.
Monnier, V., Dussillol, F., Alves, G., Lamour-Isnard, C. and Plessis, A. (
1998
). Molecular interactions between three members of the Drosophila Hedgehog signaling pathway: Suppressor of fused links the ser-thr kinase Fused and the transcription factor Cubitus interruptus.
Curr. Biol.
8
,
583
-586.
Moore, F. L., Jaruzelska, J., Fox, M. S., Urano, J., Firpo, M. T., Turek, P. J., Dorfman, D. M. and Pera, R. A. (
2003
). Human Pumilio-2 is expressed in embryonic stem cells and germ cells and interacts with DAZ (Deleted in AZoospermia) and DAZ-like proteins.
Proc. Natl. Acad. Sci. USA
100
,
538
-543.
Murone, M., Luoh, S. M., Stone, D., Li, W., Gurney, A.,Armanini, M.,Grey, C., Rosenthal, A. and de Sauvage, F. J.(
2000
). Gli regulation by the opposing activities of fused and suppressor of fused.
Nat. Cell Biol.
2
,
310
-312.
Nakano, Y., Kim, H. R., Kawakami, A., Roy, S., Schier, A. and Ingham,P. W. (
2004
). Inactivation of dispatched 1 by the chameleon mutation disrupts Hedgehog signaling in the zebrafish embryo.
Dev. Biol.
(in press).
Nybakken, K. and Perrimon, N. (
2002
). Hedgehog signal transduction: recent findings
Curr. Opin. Genet. Dev.
12
,
503
-511.
Odenthal, J., van Eeden, F. J., Haffter, P., Ingham, P. W. and Nusslein-Volhard, C. (
2000
). Two distinct cell populations in the floor plate of the zebrafish are induced by different pathways.
Dev. Biol.
219
,
350
-363.
Ohlmeyer, J. and Kalderon, D. (
1998
). Hedgehog stimulates maturation of Cubitus interruptus into a labile transcriptional activator.
Nature
396
,
749
-753.
Pan, D. and Rubin, G. M. (
1995
). cAMP-dependent protein kinase and hedgehog act antagonistically in regulating decapentaplegic transcription in Drosophila imaginal discs.
Cell
80
,
543
-552.
Park, H. L., Bai, C., Platt, K. A., Matise, M. P., Beeghly, A.,Hui, C. C.,Nakashima, M. and Joyner, A. L. (
2000
). Mouse Gli1 mutants are viable but have defects in SHH signaling in combination with a Gli2 mutation.
Development
127
,
1593
-1605.
Pearse, R. V., II, Collier, L. S., Scott, M. P. and Tabin, C. J. (
1999
). Vertebrate homologs of Drosophila Suppressor of fused interact with the Gli family of transcriptional regulators.
Dev. Biol.
212
,
323
-336.
Persson, M., Stamataki, D., te Welscher, P., Andersson, E.,Bose, J.,Ruther, U., Ericson, J. and Briscoe, J.(
2002
). Dorsal-ventral patterning of the spinal cord requires Gli3 transcriptional repressor activity.
Genes Dev.
16
,
2865
-2878.
Pham, A., Therond, P., Alves, G., Tournier, F. B., Busson, D.,Lamour-Isnard, C., Bouchon, B. L., Preat, T. and Tricoire, H.(
1995
). The Suppressor of fused gene encodes a novel PEST protein involved in Drosophila segment polarity establishment.
Genetics
140
,
587
-598.
Postlethwait, J. H. and Talbot, W. S. (
1997
). Zebrafish genomics: from mutants to genes.
Trends Genet.
13
,
183
-190.
Price, M. and Kalderon, D. (
1999
). Proteolysis of cubitus interruptus in Drosophila requires phosphorylation by protein kinase A.
Development
126
,
4331
-4339.
Radhakrishna, U., Wild, A., Grzeschik, K.-H. and Antonarakis, S. E. (
1997
). Mutation in Gli3 in postaxial polydactyly type A.
Nat. Genet.
17
,
269
-271.
Rechsteiner, M. and Rogers, S. W. (
1996
). PEST sequences and regulation by proteolysis.
Trends Biochem. Sci.
21
,
267
-271.
Robbins, D., Nybakken, K., Kobayashi, R., Sisson, J., Bishop, J. andTherond, P. (
1997
). Hedgehog elicits signal transduction by means of a large complex containing the kinesin-related protein Costal2.
Cell
90
,
225
-234.
Roelink, H., Augsburger, A., Heemskerk, J., Korzh, V., Norlin,S., Ruiz iAltaba, A., Tanabe, Y., Placzek, M., Edlund, T., Jessell, T. M. et al. (
1994
). Floor plate and motor neuron induction by vhh-1, a vertebrate homolog of hedgehog expressed by the notochord.
Cell
76
,
761
-775.
Roy, S., Wolff, C. and Ingham, P. W. (
2001
). The u-boot mutation identifies a Hedgehog-regulated myogenic switch for fiber-type diversification in the zebrafish embryo.
Genes Dev.
15
,
1563
-1576.
Ruiz i Altaba, A. (
1998
). Combinatorial Gli function in floor plate and neuronal inductions by Sonic hedgehog.
Development
125
,
2203
-2212.
Ruiz i Altaba, A. (
1999a
). Gli proteins encode context-dependent positive and negative functions: implications for development and disease.
Development
126
,
3205
-3216.
Ruiz i Altaba, A. (
1999b
). Gli proteins and hedgehog signaling–development and cancer.
Trends Genet.
15
,
418
-425.
Ruiz i Altaba, A., Sanchez, P. and Dahmane, N.(
2002
). Gli and hedgehog in cancer: tumours, embryos and stem cells.
Nat. Rev. Cancer
2
,
361
-372.
Sasaki, H., Nishizaki, Y., Hui, C., Nakafuku, M. and Kondoh,H. (
1999
). Regulation of Gli2 and Gli3 activities by an amino-terminal repression domain: implication of Gli2 and Gli3 as primary mediators of Shh signaling.
Development
126
,
3915
-3924.
Sbrogna, J. L., Barresi, M. J. F. and Karlstrom, R. O.(
2003
). Multiple roles for Hedgehog signaling in zebrafish pituitary development.
Dev. Biol.
254
,
19
-35.
Schauerte, H. E., van Eeden, F. J., Fricke, C., Odenthal, J.,Strahle, U. andHaffter, P. (
1998
). Sonic hedgehog is not required for the induction of medial floor plate cells in the zebrafish.
Development
125
,
2983
-2993.
Schier, A. F., Neuhauss, S. C. F., Helde, K. A., Talbot, W. S. and Driever,W. (
1997
). The one-eyed pinhead gene functions in mesoderm and endoderm formation in zebrafish and interacts with no tail.
Development
124
,
327
-342.
Seamon, K. B. and Daly, J. W. (
1981
). Forskolin: a unique diterpene activator of cyclic AMP-generating systems.
J. Cyclic Nucl. Res.
7
,
201
-224.
Shin, S. H., Kogerman, P., Lindstrom, E., Toftgard, R. and Biesecker, L.G. (
1999
). GLI3 mutations in human disorders mimic Drosophila cubitus interruptus protein functions and localization.
Proc. Natl. Acad. Sci. USA
96
,
2880
-2884.
Sisson, J. C., Ho, K., S., Suyama, K. and Scott, M. P.(
1997
). Costal2, a novel kinesin-related protein in the hedgehog signaling pathway.
Cell
90
,
235
-245.
Stegman, M., Vallance, J., Elangovan, G., Sosinski, J., Cheng,Y. andRobbins, D. (
2000
). Identification of a tetrameric hedgehog signaling complex.
J. Biol. Chem.
275
,
21809
-21812.
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
,
1005
-1009.
Tole, S., Ragsdale, C. W. and Grove, E. A.(
2000
). Dorsoventral patterning of the telencephalon is disrupted in the mouse mutant extra-toes(J).
Dev. Biol.
217
,
254
-265.
van Eeden, F. J., Granato, M., Schach, U., Brand, M.,Furutani-Seiki, M.,Haffter, P., Hammerschmidt, M., Heisenberg, C. P.,Jiang, Y. J., Kane, D. A. et al. (
1996a
). Genetic analysis of fin formation in the zebrafish, Danio rerio.
Development
123
,
255
-262.
van Eeden, F. J., Granato, M., Schach, U., Brand, M.,Furutani-Seiki, M.,Haffter, P., Hammerschmidt, M., Heisenberg, C. P.,Jiang, Y. J., Kane, D. A. et al. (
1996b
). Mutations affecting somite formation and patterning in the zebrafish, Danio rerio.
Development
123
,
153
-164.
Varga, Z. M., Amores, A., Lewis, K. E., Yan, Y. L.,Postlethwait, J. H.,Eisen, J. S. and Westerfield, M.(
2001
). Zebrafish smoothened functions in ventral neural tube specification and axon tract formation.
Development
128
,
3497
-3509.
Villavicencio, E. H, Walterhouse, D. O. and Iannaccone, P. M. (
2000
). The sonic hedgehog-patched-gli pathway in human development and disease.
Am. J. Hum. Genet.
67
,
1047
-1054.
von Mering, C. and Basler, K. (
1999
). Distinct and regulated activities of human Gli proteins in Drosophila.
Curr. Biol.
9
,
1319
-1322.
Wallis, D. and Muenke, M. (
2000
). Mutations in holoprosencephaly.
Hum. Mutat.
16
,
99
-108.
Wang, B., Fallon, J. F. and Beachy, P. A.(
2000
). Hedgehog-regulated processing of Gli3 produces an anterior/posterior repressor gradient in the developing vertebrate limb.
Cell
100
,
423
-434.
Wang, Q. T. and Holmgren, R. A. (
1999
). The subcellular localization and activity of Drosophila cubitus interruptus are regulated at multiple levels.
Development
126
,
5097
-5106.
Weinberg, E. S., Allende, M. L., Kelly, C. S., Abdelhamid, A.,Murakami,T., Andermann, P., Doerre, O. G., Grunwald, D. J. and Riggleman, B. (
1996
). Developmental regulation of zebrafish MyoD in wild-type, no tail and spadetail embryos.
Development
122
,
271
-280.
Westerfield, M. (
1993
).
The Zebrafish Book
. Eugene: University of Oregon Press.
Wolff, C., Roy, S. and Ingham, P. W. (
2003
). Multiple muscle cell identities induced by distinct levels and timing of hedgehog activity in the zebrafish embryo.
Curr. Biol.
13
,
1169
-1181.