buttonhead


DEVELOPMENTAL BIOLOGY

Embryonic

The BTD transcript is first expressed in a stripe covering the head anlagen of the syncytial blastoderm embryo located between 65 and 77% egg-length (0% is the posterior pole). The BTD transcript overlaps the domain of Empty spiracles and Orthodenticle. The BTD strip persists until it decays in germ band extention. It is later expressed in a complex pattern (Wimmer, 1993).

During cellularization of the blastoderm a dorsal spot appears in the proneural region anterior to the head stripe. The head spot expression continues and splits up during germband retraction into several spots that become integrated into the developing brain, marking different brain areas. During the early phase of germ band extension btd starts to be expressed in a metameric pattern that decays at the fully extended germ band stage leaving single btd-expressing cells. The btd-expressing cells represent subgroups of neuroblasts, which finally end up in the ventral cord (Wimmer, 1996).

During germ band extension, a second metameric expression pattern of btd can be observed. It is restricted to the lateral region of the embryo, corresponding to the area of the proneural clusters from which the peripheral nervous system originates. During germ band extension btd is expressed in the leg anlagen located in the thoracic segments and in several restricted areas of the head. At this stage the pattern of btd expression resembles Distal-less, but btd expression is delayed compared to Dll expression. btd is expressed in the mandibular but not the labral segments, where Dll is expressed in the labral, but not in the mandibular segments (Wimmer, 1996).

Early tailless expression (blastoderm stage) covers the anlage of the entire brain. Beginning approximately with the onset of gastrulation, an anterior-dorsal region with a high expression level (called HL domain) can be distinguished from a posterior-ventral domain expressing tll at a somewhat lower level. The HL domain coincides with part of the central and anterior protocerebral neurectoderm. The low expression level LL domain covers the remaining part of the protocerebral neuroectoderm. orthodenticle is expressed in a circumferential domain of the cellular blastoderm but during gastrulation becomes restricted to a domain that encompasses most of the protocerebral neurectoderm and an adjacent part of the deuterocerebral neurectoderm. All neurobasts segregating from this domain transiently express otd during stages 10 and 11. buttonhead is initially expressed in a wide domain including the anlagen of the antennal, intercalary and mandibular segments, as well as the acron. With the beginning of gastrulation, expression disappears from most of the procephalon, except for small domains of the posterior part of the deuterocerebral and tritocerebral neurectoderm and a dorsoanterior patch that partially overlaps with the dorsoanterior protocerebrum. Both the late deutocerebral and tritocerebral btd domains contain few, if any neuroblasts. empty spiracles is in an asymmetric circumferential domain of the cellular blastoderm. During gastrulation, this pattern resolves into two stripes that occupy anterior portions of the deuterocerebral neuroectoderm and the mandibular metamere, respectively. In addition, a small circular domain corresponding to the tritocerebral neurectoderm appears ventral to the deuterocerebral stripe (Younossi-Hartenstein, 1997).

Loss of tll function results in the absence of all protocerebral neuroblasts and loss of all four coherent domains of Fas II expression in the protocerebrum. Also missing is the optic lobe. orthodenticle functions in a domain that includes a large part of the protocerebrum and a smaller part of the adjacent deuterocerebrum. Loss of otd results in loss of protocerebral P1, P2 and P4 coherent domains of Fas II expression. Also missing is a nerve that carries axons from the antennal organ. In buttonhead mutation the D/T cluster is missing; consequently a cervical connection is missing that normally sends nerves to the labral sensory organ, the hypopharyngeal sensory organ and the stomatogastric nervous system (Younossi-Hartenstein, 1997).

Association of tracheal placodes with leg primordia in Drosophila and implications for the origin of insect tracheal systems

Adaptation to diverse habitats has prompted the development of distinct organs in different animals to better exploit their living conditions. This is the case for the respiratory organs of arthropods, ranging from tracheae in terrestrial insects to gills in aquatic crustaceans. Although Drosophila tracheal development has been studied extensively, the origin of the tracheal system has been a long-standing mystery. Tracheal placodes and leg primordia arise from a common pool of cells in Drosophila, with differences in their fate controlled by the activation state of the wingless signalling pathway. Early events that trigger leg specification have been elucidated and it is shown that cryptic appendage primordia are associated with the tracheal placodes even in abdominal segments. The association between tracheal and appendage primordia in Drosophila is reminiscent of the association between gills and appendages in crustaceans. This similarity is strengthened by the finding that homologues of tracheal inducer genes are specifically expressed in the gills of crustaceans. It is concluded that crustacean gills and insect tracheae share a number of features that raise the possibility of an evolutionary relationship between these structures. An evolutionary scenario is proposed that accommodates the available data (Franch-Marro, 2006).

The Drosophila tracheal system has a clearly metameric origin, arising from clusters of cells, on either side of each thoracic and abdominal segment, that express the tracheal inducer genes trachealess (trh) and ventral veinless (vvl). Conversely, the leg precursors can be recognized as clusters of cells that express the Distal-less (Dll) gene, on either side of each thoracic segment; these will give rise both to the Keilin's Organs (KOs, the rudimentary legs of the larvae) and to the three pairs of imaginal discs that will give rise to the legs of the adult fly (Franch-Marro, 2006).

To investigate whether there is a direct physical association between the leg and tracheal primordia, Drosophila embryos co-stained for the expression of trh and early markers of leg primordia were examined. Although Dll is one of the most commonly used markers for the leg primordia, it is not the earliest gene required for their specification. Instead, a couple of related and apparently redundant genes, buttonhead (btd) and Sp1, act upstream of Dll in the specification of these primordia (Estella, 2003). Examining the specification of tracheal cells with respect to btd expression, tracheal cells were observed to appear in close apposition to btd-expressing cells, from the earliest stages of their appearance (by stage 9/early stage 10). Interestingly, unlike Dll, btd is initially expressed both in the thoracic and abdominal segments, and its expression is restricted to the thoracic segments later, under the influence of the BX-C. Thus, the cells of the respiratory system in Drosophila always arise in close proximity to the cells that are fated to give rise to the legs (Franch-Marro, 2006).

To fully endorse this conclusion it is necessary to show that the btd-expressing cells in the abdomen correspond to cryptic leg primordia. This may be a key point because, although many of the genes required for leg development are already known, it has not yet been possible to induce leg development in abdominal segments (except by transforming these segments into thoracic ones). In particular, although the Dll promoter contains BX-C binding sites that repress its expression in the abdominal segments, no ectopic appendage has been reported by misexpressing Dll in the abdomen. These observations have lead to some doubts as to whether a leg developmental program is at all compatible with abdominal segmental identity (Franch-Marro, 2006).

Since the initial expression of btd in the abdominal segments is downregulated by the BX-C genes, it was reasoned that sustained expression of btd might overcome the repressive effect of the BX-C genes and force the induction of leg structures in the abdomen. To test this, a btd-GAL4 driver was used to drive btd expression, expecting that the perdurance of the GAL4/UAS system would ensure a more persistent expression of btd in its endogenous expression domain. No sign was ever obtained of ectopic Dll expression or KOs in the abdominal segments, but the increased expression of btd had an effect on the KOs of the thoracic segments, which had more sensory hairs than the three normally found in wild-type KOs. Thus, on its own, btd seems unable to overcome BX-C repression of leg development (Franch-Marro, 2006).

One possibility would be that the BX-C genes could suppress appendage development in the abdomen by independently repressing both btd and Dll in this region. To assess this possibility, the same btd-GAL4 driver was used to simultaneously induce the expression of both btd and Dll. Under these circumstances, it was observed that KOs develop in otherwise normal abdominal segments; as in the previous experiment, the newly formed KOs have more than three sensory hairs. These results suggest that expression of btd and Dll in the btd-expressing abdominal primordia is sufficient to induce the development of leg structures in the abdomen, overcoming the repressive effect of the BX-C genes. Furthermore, these results demonstrate that these clusters of btd-expressing cells in the abdomen are indeed cryptic leg primordia. These results clearly show that tracheal cells are specified in close proximity to the leg primordia, in both thoracic and abdominal segments (Franch-Marro, 2006).

Previous results have shown that the leg primordia are specified straddling the segmental stripes of wingless (wg) expression in the early embryonic ectoderm, whereas tracheal cells are specified in between these stripes. To investigate whether wg might play a role in determining the fate of these primordia, what happens when the normal pattern of wg expression is disrupted was studied. In wg mutant embryos, trh and vvl from the earliest stages of their expression are no longer restricted to separate clusters of cells; instead larger patches of expression add up to a continuous band of cells running along the anteroposterior axis of the embryo, while btd expression is suppressed in this part of the embryonic ectoderm. Conversely, ubiquitous expression of wg suppresses trh expression, while causing an expansion of btd expression along the embryo. Restricted activation or inactivation of the wg pathway by the expression of a constitutive form of armadillo or a dominant-negative form of dTCF, respectively, are also able to specifically induce or repress trh and btd expression. trh/vvl and btd seem to respond independently to wg signalling and there is no sign of cross-regulation among them, since btd expression is normal in trh vvl double mutants, and trh and vvl expression is normal in mutants for a deficiency uncovering btd and Sp1 (Franch-Marro, 2006).

The role of wg as a repressor of the tracheal fate is further illustrated by looking at the behaviour of transformed cells: the clusters of cells that have lost btd expression and gained trh and vvl expression in wg mutant embryos begin a process of invagination that is characteristic of tracheal cells. Furthermore, these cells also express the dof (stumps) gene, a target gene of both trh and vvl in the tracheal cells. Although further development of these cells is hard to ascertain because of gross abnormalities in wg- embryos, these results indicate that they have been specified as tracheal cells. Thus, wg appears to act as a genetic switch that decides between two mutually exclusive fates in this part of the embryonic ectoderm: the tracheal fate, which is followed in the absence of wg signalling; and the leg fate, which is followed upon activation of the wg pathway. Given that there are no cell lineage restrictions setting apart the cells of the tracheal and leg primordia, these two cell populations could be considered as a single equivalence group, with the differences in their fate controlled by the activation state of the wg signalling pathway (Franch-Marro, 2006).

A link between respiratory organs and appendages is also found in many primitively aquatic arthropods, like crustaceans, where gills typically develop as distinct dorsal branches (or lobes) of appendages called epipods. Following the current observations, which suggest a link between respiratory organs and appendages in Drosophila, whether further similarities could be found between insect tracheal cells and crustacean gills was examined. Specifically, whether homologues of the tracheal inducing genes might have a role in the development of appendage-associated gills in crustaceans was considered (Franch-Marro, 2006).

RT-PCR was used to clone fragments of the vvl and trh homologues from Artemia franciscana and from Parhyale hawaiensis, representing two major divergent groups of crustaceans (members of the branchiopod and malacostracan crustaceans, respectively). In the case of Artemia vvl, a fragment was cloned that corresponds to the APH-1 gene and an antibody was generated for immunochemical staining in developing Artemia larvae. It was observed that Artemia Vvl is initially absent from early limb buds; it becomes weakly and uniformly expressed while the limb is developing its characteristic branching morphology, and becomes strongly upregulated in one of the epipods as its cells begin to differentiate. Uniform weak expression persists in mature limbs, but expression levels in the epipod are always significantly higher. Expression of the trh homologue from Artemia appears to be restricted to the same epipod as Vvl. Similarly, homologues of vvl and trh were cloned from Parhyale hawaiensis and their expression was studied by in situ hybridization. Both genes are specifically expressed in the epipods of developing thoracic appendages. Besides epipods, the Artemia trh and vvl homologues are also expressed in the larval salt gland, an organ with osmoregulatory functions during early larval stages of Artemia development (Franch-Marro, 2006).

What is the significance of the two Drosophila tracheal inducer genes being specifically expressed in crustacean epipods/gills? One possibility is that the expression of these two genes was acquired independently in insect tracheae and in crustacean gills. Alternatively, tracheal systems and gills may have inherited these expression patterns from a common evolutionary precursor, perhaps a respiratory/osmoregulatory structure that was already present in the common ancestors of crustaceans and insects (Franch-Marro, 2006).

The latter possibility is considered unlikely by conventional views, because of the structural differences between gills and tracheae (external versus internal organs, discrete segmental organs versus fused network of tubes), and the difficulty to conceive a smooth transition between these structures. Yet, analogous transformations have occurred during arthropod evolution: tracheae can be organized as large interconnected networks or as isolated entities in each segment (as in some apterygote insects), invagination of external respiratory structures is well documented among groups that have made the transition from aquatic to terrestrial environments (terrestrial crustaceans, spiders and scorpions), and conversely evagination of respiratory surfaces is common in animals that have returned to an aquatic environment (tracheal gills or blood gills in aquatic insect larvae). A very similar (but independent) evolutionary transition is, in fact, thought to have occurred in arachnids, where gills have been internalised to give rise to book lungs, and these in turn have been modified to give rise to tracheae in some groups of spiders. Thus, a relationship between insect tracheae and crustacean gills is plausible (Franch-Marro, 2006).

A particular type of epipod/gill has also been proposed as the origin of insect wings, a hypothesis that has received support from the specific expression in a crustacean epipod of the pdm/nubbin (nub) and apterous (ap) genes - that have wing-specific functions in Drosophila. In fact, the Artemia nub and ap homologues are expressed in the same epipod as trh and vvl, raising questions as to the specific relationship of this epipod with either tracheae or wings. A resolution to this conundrum becomes apparent when one considers the different types of epipods/gills found in aquatic arthropods, and their relative positions with respect to other parts of the appendage (Franch-Marro, 2006).

The primary branches of arthropod appendages, the endopod/leg and exopod, develop straddling the anteroposterior (AP) compartment boundary, which corresponds to a widely conserved patterning landmark in all arthropods. Different types of epipods/gills, however, differ in their position with respect to this boundary. For example, in the thoracic appendages of the crayfish, some epipods develop spanning the AP boundary [visualized by engrailed (en) expression running across the epipod], whereas others develop exclusively from anterior cells (with no en expression). Given that wing primordia comprise cells from both the anterior and posterior compartments, wings probably derived from structures that were straddling the AP boundary. Conversely, given that tracheal primordia arise exclusively from cells of the anterior compartment (anterior to en and even wg-expressing cells), it seems probable that tracheal cells evolved from a population of cells that was located in the anterior compartment. In this respect, it is interesting to note that the former type of epipods express nub, whereas the latter do not (Franch-Marro, 2006).

In summary, it is suggested that the ancestors of arthropods had specific areas on the surface of their body that were specialized for osmoregulation and gas exchange. Homologues of trh and vvl were probably expressed in all of these cells and played a role in their specification, differentiation or function. Some of these structures were probably associated with appendages, in the form of epipods/gills or other types of respiratory surfaces. A particular type of gill, straddling the AP compartment boundary, is likely to have given rise to wings, whereas respiratory surfaces arising from anterior cells only may have given rise to the tracheal system of insects. Confirmation of this hypothetical scenario may ultimately come from the discovery of new fossils, capturing intermediate states in the transition of insects from an aquatic to a terrestrial lifestyle (Franch-Marro, 2006).

The evolutionary conserved transcription factor Sp1 controls appendage growth through Notch signaling

The appendages of arthropods and vertebrates are not homologous structures, although the underlying genetic mechanisms that pattern them are highly conserved. Members of the Sp family of transcription factors are expressed in the developing limbs and their function is required for limb growth in both insects and chordates. Despite the fundamental and conserved role that these transcription factors play during appendage development, their target genes and the mechanisms in which they participate to control limb growth are mostly unknown. This study analyzed the individual contributions of two Drosophila Sp members, buttonhead (btd) and Sp1, during leg development. Sp1 plays a more prominent role controlling leg growth than btd. A regulatory function of Sp1 in Notch signaling was identified, and a genome wide transcriptome analysis was performed to identify other potential Sp1 target genes contributing to leg growth. The data suggest a mechanism by which the Sp factors control appendage growth through the Notch signaling (Cordoba, 2016).

Understanding the molecular mechanisms that control the specification and acquisition of the characteristic size and shape of organs is a fundamental question in biology. Of particular interest is the development of the appendages of vertebrates and arthropods, i.e., non-homologous structures that share a similar underlying genetic program to build them, a similarity that has been referred to as 'deep homology.' Some of the conserved genes include the Dll/Dlx genes, Hth/Meis and the family of Sp transcription factors. The Sp family is characterized by the presence of three highly conserved Cys2-His2-type zinc fingers and the presence of the Buttonhead (BTD) box just N-terminal of the zinc fingers (Cordoba, 2016).

Members of the Sp family have important functions during limb outgrowth in a range of species from beetles to mice. In vertebrates, Sp6, Sp8 and Sp9 are expressed in the limb bud and are necessary for Fgf8 expression and, therefore, for apical ectodermal ridge (AER) maintenance. Moreover, Sp6/Sp8 phenotypes have been related to the split-hand/foot malformation phenotype (SHFM) and, in the most severe cases, to amelia (the complete loss of the limb) (Cordoba, 2016).

In Drosophila, two members of this family, buttonhead (btd) and Sp1, are located next to each other on the chromosome and share similar expression patterns throughout development. Recently, another member of the family, Spps (Sp1-like factor for pairing sensitive-silencing) has been identified with no apparent specific function in appendage development. The phenotypic analysis of a btd loss-of-function allele and of a deletion that removes both btd and Sp1 led to the proposal that these genes have partially redundant roles during appendage development. However, the lack of a mutant for Sp1 has prevented the analysis of the specific contribution of this gene during development (Cordoba, 2016).

In Drosophila, leg development is initiated in the early embryo by the expression of the homeobox gene Distal-less (Dll) in a group of cells in each thoracic segment. Later on, Dll expression depends on the activity of the Decapentaplegic (Dpp) and Wingless (Wg) signaling pathways, which, together with btd and Sp1, restrict Dll expression to the presumptive leg territory. Therefore, the early elimination of btd and Sp1 completely abolishes leg formation and, in some cases, causes a leg-to-wing homeotic transformation (Estella, 2010). As the leg imaginal disc grows, a proximo-distal (PD) axis is formed by the differential expression of three leg gap genes, Dll, dachshund (dac) and homothorax (hth), which divides the leg into distal, medial and proximal domains, respectively. Once these genes have been activated, their expression is maintained, in part through an autoregulatory mechanism, and no longer relies on Wg and Dpp. Meanwhile, the distal domain of the leg is further subdivided along the PD axis by the activity of the epidermal growth factor receptor (EGFR) signaling pathway through the activation of secondary PD targets such as aristaless (al), BarH1 (B-H1) or bric-a-brac (bab). During these stages, btd and Sp1 control the growth of the leg but are no longer required for Dll expression (Estella, 2010). How btd and Sp1 contribute to the shape and size of the leg and the identity of their downstream effector targets is unknown (Cordoba, 2016).

One important consequence of the PD territorial specification is the generation of developmental borders that activate organizing molecules to control the growth and pattern of the appendage. In the leg, PD subdivision is necessary to localize the expression of the Notch ligands Delta (Dl) and Serrate (Ser), which in turn activate the Notch pathway in concentric rings at the borders between presumptive leg segments. However, it is still unknown how Notch controls leg growth and how the localization of its ligands is regulated. The present study generated a specific Sp1 null mutant, which, in combination with the btd mutant and a deletion that removes both btd and Sp1, allow analysis of the individual contributions of these genes to leg development. This study finds that Sp1 plays a fundamental role during patterning and growth of the leg disc, and that this function is not compensated by btd. The growth-promoting function of Sp1 depends in part on the regulation of the expression of Ser and, therefore, on Notch activity. In addition, other candidate targets of Sp1 affecting leg growth and morphogenesis were identified. Intriguingly, some of these Sp1 potential downstream targets are ecdysone-responding genes. These results highlight a mechanism by which btd and Sp1 control the size and shape of the leg, in part through regulation of the Notch pathway (Cordoba, 2016).

The two Sp family members in Drosophila, Sp1 and btd, display a similar spatial and temporal expression pattern during embryonic and imaginal development. Previous work suggested that btd and Sp1 have partially redundant functions during development. However, the lack of an Sp1 mutant has prevented the detailed analysis of the individual contributions of each gene. This study has generated an Sp1 null mutant that allowed elucidation unambiguously of the individual contributions of each of these genes to leg development (Cordoba, 2016).

Appendage formation starts in early embryos by the activation of Dll (through its early enhancer, Dll-304), btd and Sp1 by Wg, and their expression is repressed posteriorly by the abdominal Hox genes. Some hours later, there is a molecular switch from the early Dll enhancer (Dll-304) to the late enhancer (Dll-LT) to keep Dll expression throughout the embryo-larvae transition restricted to the cells that will form the leg. At this developmental stage, Sp1 and btd play redundant roles in Dll activation, as only the elimination of both genes suppresses Dll expression and Dll-LT activity in the leg primordia. Once Dll expression is activated in the leg disc by the combined action of Wg, Dpp and Btd/Sp1, its expression is maintained in part through an autoregulatory mechanism. At this time point, during second instar, btd and Sp1 are co-opted to control the growth of the leg. The leg phenotype of Sp1 and btd single mutants demonstrates the divergent contributions of each gene to leg growth. Removing btd from the entire leg only slightly affects the growth of proximo-medial segments, whereas loss of Sp1 causes dramatic growth defects along the entire leg. The different phenotypes of Sp1 and btd mutant legs could be a consequence of their distinct expression pattern along the leg PD axis, with btd being expressed more proximally than Sp1 (Cordoba, 2016).

The growth defects observed in Sp1 mutant legs are not due to gross defects in the localization of the different transcription factors that subdivide the leg along the PD axis, nor to defects in the expression of the EGFR ligand vn. By contrast, the results suggest a role for Sp1 in the regulation of the Notch ligand Ser. Notch pathway activation is necessary for the formation of the joints and the growth of the leg, and defects in these two processes were observed in Sp1 mutant legs. Moreover, the results demonstrate that Sp1 is necessary and sufficient for Ser expression at least in the distal domain of the leg and is therefore required for the correct activation of the Notch pathway. These results are consistent with the proposed role of Sp8 in allometric growth of the limbs in the beetle where the number of Ser-expressing rings is reduced in Sp8 knockdown animals (Cordoba, 2016).

The regulation of Ser expression is controlled by multiple CREs that direct its transcription in different developmental territories. Interestingly, although the wing and leg are morphologically different appendages and express a diverse combination of master regulators (e.g. Sp1 selects for leg identity whereas Vg determines wing fate), the same set of enhancers are accessible in both appendages, with the exception of the ones that control the expression of the master regulators themselves. These results imply that appendage-specific master regulators differentially interact with the same enhancers to generate a specific expression pattern in each appendage. The current analysis of Ser CREs identified a specific sequence that is active in the wing and in the leg. In the leg, this CRE reproduced Ser expression in the fourth tarsal segment and require the combined inputs of Sp1 and Ap. It is proposed that Sp1, in coordination with the other leg PD transcription factors, interacts with different Ser CREs to activate Ser expression in concentric rings in the leg. Meanwhile, given the same set of Ser CREs in the wing, the presence of a different combination of transcription factors regulate Ser expression in the characteristic 'wing pattern' (Cordoba, 2016).

Transcriptome analysis identified additional candidate Sp1 target genes that contribute to control the size and shape of the leg. Appendage elongation depends on the steroid hormone ecdysone through several of its effectors, such as Sb. Sb, as well as other genes related to the ecdysone pathway, were misregulated in Sp1 mutant discs. The characteristic change in cell shape that normally occurs during leg eversion does not happen correctly in these mutants. Other genes identified in this study are the Notch pathway targets dys and Poxn, which are both required for the correct development of the tarsal joints. dys and Poxn downregulation is consistent with Sp1 regulation of the Notch ligand Ser. Interestingly, the upregulation of the antenna-specific gene danr in Sp1 mutants might explain the partial transformation of the distal leg to antennal-like structures observed when two copies of Sp1 and one of btd are mutated. Interestingly, btd and Sp1 are only expressed in the antenna disc in a single ring corresponding to the second antennal segment whereas in the leg both genes are more broadly expressed. Consistent with this, misexpression of Sp1 in the antenna transforms the distal domain to leg-like structures, suggesting that different levels or expression domains of Sp1 helps distinguish between these two homologous appendages (Cordoba, 2016).

A considerable group of Hsp-related genes were downregulated in Sp1 mutant legs. Although their contribution to Drosophila leg development is unknown, downregulation of DnaJ-1, the Drosophila ortholog of the human HSP40, affects joint development and leg size, suggesting a potential role of these genes during leg morphogenesis (Cordoba, 2016).

An ancient common mechanism for the formation of outgrowths from the body wall has been suggested. Members of the Sp family are expressed and required for appendage growth in a range of species from Tribolium to mice. Consistent with the current results, knockdown of Sp8/Sp9 in the milkweed bug or the beetle generated dwarfed legs with fused segments that maintain the correct PD positional values. As is the case for Drosophila Sp1 mutants, mouse Sp8-deficient embryos develop with truncated limbs. By contrast, loss of function of Sp6 results in milder phenotypes of limb syndactyly. A progressive reduction of the dose of Sp6 and Sp8 lead to increased severity of limb phenotypes from syndactyly to amelia, suggesting that these genes play partially redundant roles. This phenotypic analysis of Sp1 and btd are consistent with this model, in which Sp1 plays the predominant role in appendage growth and the complete elimination of btd and Sp1 together abolish leg formation. Therefore, Drosophila Sp1 mutants are phenotypically equivalent to vertebrate Sp8 mutants. In vertebrate Sp8 mutant limbs, Fgf8 expression is not maintained and a functional AER fails to form. In Drosophila, FGF signaling does not seem to be involved in appendage development. Nevertheless, another receptor tyrosine kinase, EGFR, is activated at the tip of the leg and act as an organizer to regulate the PD patterning of the tarsus. The current results suggest that Sp1 acts in parallel with the EGFR pathway, as the ligand vn and EGFR target genes maintain their PD positional information in Sp1 mutant legs. However, a potential relationship between Sp1 and the EGFR pathway in later stages of leg development cannot be ruled out (Cordoba, 2016).

The results suggest that the Notch ligand Ser is a target of Sp1, and mediates in part the growth-promoting function of Sp1. Interestingly, members of the Notch pathway in vertebrates, including the Ser ortholog jagged 2 and notch 1 are expressed in the AER and regulate the size of the limb. It would be interesting to investigate further the possible relationship between Sp transcription factors and the Notch pathway in vertebrates, and test whether the functional relationship described in this work is also maintained throughout evolution (Cordoba, 2016).

Effects of Mutation or Deletion

BTD is required for development of the antennal, intercalary and mandibular segments of the head. In btd mutants these segments are lacking and head involution [Images] is incomplete (Wimmer, 1993).

btd mutants show reduced numbers of thoracic and abdominal chordotonal organs. Adult flies with chordotonal defects are disabled, showing uncoordinated or sedentary behavior. Such behavior is also seen in transgene-rescued btd mutants, i.e. they never fly nor mate, they rarely move and show very uncoordinated footwork when moving. Part, but not all of these defects may be explained by leg malformations, which vary in penetrance and expressivity (Wimmer, 1996).

The role of buttonhead and Sp1 in the development of the ventral imaginal discs

The related genes buttonhead (btd) and Drosophila Sp1 (the Drosophila homolog of the human SP1 gene) encode zinc-finger transcription factors known to play a developmental role in the formation of the Drosophila head segments and the mechanosensory larval organs. A novel function of btd and Sp1 is reported: they induce the formation and are required for the growth of the ventral imaginal discs. They act as activators of the headcase (hdc) and Distal-less (Dll) genes, which allocate the cells of the disc primordia. The requirement for btd and Sp1 persists during the development of ventral discs: inactivation by RNA interference results in a strong reduction of the size of legs and antennae. Ectopic expression of btd in the dorsal imaginal discs (eyes, wings and halteres) results in the formation of the corresponding ventral structures (antennae and legs). However, these structures are not patterned by the morphogenetic signals present in the dorsal discs; the cells expressing btd generate their own signalling system, including the establishment of a sharp boundary of engrailed expression, and the local activation of the wingless and decapentaplegic genes. Thus, the Btd product has the capacity to induce the activity of the entire genetic network necessary for ventral imaginal discs development. It is proposed that this property is a reflection of the initial function of the btd/Sp1 genes that consists of establishing the fate of the ventral disc primordia and determining their pattern and growth (Estella, 2003).

In a search for genes with restricted expression in the adult cuticle, the MD808 Gal4 line was found to direct expression in the ventral derivatives of the adult body; proboscis, antennae, legs and genitalia. In the abdomen and analia no clear expression was discerned. It was also noticed that the insertion was located in the first chromosome and associated with a lethal mutation. The mutant larvae showed a head phenotype resembling that described for mutants at the btd gene: loss of antennal organ and the ventral arms of the cephalopharyngeal skeleton, and complementation analysis indicated that the chromosome carrying the insert contained a mutation at btd. The expression pattern found in MD808/UAS-lacZ embryos was also similar to that reported for btd, suggesting that the Gal4 insertion was located at this gene. In addition, the imaginal expression of MD808 and of btd was largely coincident (Estella, 2003).

Further to the genetic analysis and the expression data, DNA fragments at the insertion site were cloned to map the position of the P-element on the genome. It is located 753 bp 5' of the btd gene. The related gene Sp1 is immediately adjacent. It is likely that btd and Sp1 have originated by a tandem duplication of a primordial btd-like gene (Estella, 2003).

In early embryos btd is expressed in the head region, but by the extended germ band stage the expression domain has expanded to the ventral region of cephalic, thoracic and abdominal segments. During germ band retraction most of the abdominal and thoracic expression is lost, except in derivatives of the peripheral nervous system and the primordia of the imaginal discs. Sp1 is not expressed in early embryos, but from stage 11 onwards it shows the same pattern as btd (Estella, 2003).

Special attention was paid to the btd/Sp1 expression domain in the thoracic imaginal discs primordia, as it may suggest a novel function related to imaginal development. Double labelling with Dll and btd probes indicates that btd precedes Dll expression, but by stage 12 the two genes are co-expressed in a group of thoracic cells. However, the Dll domain is smaller and is included within the btd/Sp1 domain: there are cells expressing btd that do not show Dll activity, although all the cells expressing Dll express btd (Estella, 2003).

The ventral disc primordia include not only cells expressing Dll but also other cells containing expression of escargot (esg) and hdc, markers of the diploid cells that form the imaginal primordia. In late embryonic stages, esg is expressed in a ring domain surrounding the Dll-expressing cells and hdc is expressed in a similar pattern. Double label experiments were carried out with btd, hdc and esg probes; the expression of the two latter genes overlaps with that of btd (and with Sp1) in the thoracic disc primordia (Estella, 2003).

The overlap of the btd and of esg domains indicates that btd is also expressed in the hth domain, which is coincident with that of esg. As the hth/esg domain marks the precursor cells of the proximal region of the adult leg the embryonic expression data indicate that btd and Sp1 are active in the entire primordia of the ventral adult structures, including the distal and the proximal parts (Estella, 2003).

In the mature antennal disc, btd expression is restricted mostly to the region corresponding to the second antennal segment, where it co-localizes with both Dll and hth. In the leg disc btd also overlaps in part with Dll and with hth. The latter result is significant, for the expression of Dll and hth define two major genetic domains, which are kept apart by antagonistic interactions. The fact that btd is expressed in the two domains suggests that its regulation and function is independent from the interactions between the two domains. This observation is consistent with the results obtained in embryos and suggests that the btd domain includes the precursors of the whole ventral thoracic region from the beginning of development (Estella, 2003).

This work demonstrates a novel and also redundant function of btd and Sp1: they are responsible for the formation of the ventral imaginal discs by activating the genetic network necessary for their development. Furthermore, Btd protein retains the capacity of inducing the entire ventral genetic network during the larval period. It is proposed that the activation of btd/Sp1 is the crucial event in the determination of the ventral structures of the adult fly (Estella, 2003).

This argument is based on the finding that btd and Sp1 appear to mediate all events connected with the formation of the ventral discs. The discussion deals with the leg disc, but there is evidence that antennal primordium also requires btd. Moreover, the genital primordium is lacking in Df(1)C52 embryos, suggesting that this disc is also under the same control. Most of the experiments concern the function of btd but given the expression and functional similarities between the two genes, it is assumed that Sp1 fulfils the same or a very similar role. Therefore, btd/Sp1 will be considered to carry out a single function (Estella, 2003).

One crucial feature is that btd is an upstream activator of Dll and hdc, which are considered developmental markers of disc primordia: (1) btd expression precedes that of Dll and of hdc; (2) the btd expression domain includes those of Dll and hdc; (3) in btd mutants, Dll and hdc activity is much reduced, and completely absent in Df(1)C52 embryos; (4) ectopic btd function induces ectopic activation of Dll and hdc (Estella, 2003).

The role of btd in embryogenesis can be illustrated in the light of the models of Dll regulation. Dll is activated by wg and its expression modulated by the EGF spitz and by dpp, whereas it is repressed in the abdominal segments by the BX-C genes. The current experiments suggest that Dll regulation is mediated by btd: in wg mutants there is no btd expression and hence neither Dll nor hdc activity. In dpp mutant embryos, btd expands to the dorsal region resembling the effect on Dll. In Ubx- embryos there is an additional group of cells in the first abdominal segment expressing btd; the same cells that also express Dll in those embryos. The interpretation of the role of btd mediating Dll regulation by Ubx is complicated by previous observations showing direct repression of Dll by the Ubx protein. It is possible that Ubx regulates Dll both directly and by controlling btd activity (Estella, 2003).

It is proposed that the localization of btd/Sp1 activity to a group of ventral cells is a major event in the specification of adult structures. btd and Sp1 are activated in response to spatial cues from Wg, Dpp, EGF and BX-C, and in turn their function induces the activity of the genes necessary for ventral imaginal development (Estella, 2003).

This hypothesis is strongly supported by the results obtained inducing ectopic btd activity in the dorsal discs; just the presence of the Btd product alone is sufficient to bring about ventral disc development. In the wing and the haltere discs, Btd induces a transformation into leg, whereas in the eye it induces antennal development. This indicates that it specifies ventral disc identity jointly with other factors, e.g., the Hox genes, possibly through the activation of subsidiary genes such as Dll, known to contribute to ventral appendage identity in combination with Hox genes (Estella, 2003).

The requirement for btd and Sp1 activity appears to be restricted only to the ventral discs, even during the early phases of the thoracic disc primordia. In this context it is worth considering the observation that in Df(1)C52 embryos there is esg expression in the wing and haltere disc primordia, even though it is absent in the leg discs. Thus, the wing and haltere discs are formed in the absence of btd and Sp1. Because in these embryos there is an almost complete lack of Dll expression, this observation raises the question of the origin of the dorsal thoracic discs, which are currently considered to derive from the original ventral primordium, formed by cells expressing Dll. Although some of the original group of ventral cells may contribute to the dorsal disc primordia, the data suggest that there may be cells recruited to form the dorsal discs that do not originate in the initial ventral primordium. Accordingly, it is worth considering that in the absence of Dll activity the leg and wing discs are formed, although the leg only differentiates proximal disc derivatives. Thus, the activity of Dll cannot be considered a reliable marker for imaginal discs (Estella, 2003).

RNA interference experiments also indicate that both btd and Sp1 are required for the growth of the antennal and leg discs. When the two gene functions are reduced simultaneously, leg segments fuse and there is an overall reduction in the size of antennae and legs. The reduction of growth affects the proximal and distal regions of the appendage, and assigns a role to the expression observed in the imaginal discs. The two genes are able to perform this function on their own, for the inactivation of only one is not sufficient to impair growth. This conclusion is also supported by the observation that mutant btd clones do not have any effect; they still possess Sp1 activity, which is sufficient for normal development. At this point the mechanism by which btd/Sp1 may affect growth is not known (Estella, 2003).

One particularly significant result about the mode of action of btd comes from the analysis of the ectopic leg patterns observed with ectopic btb expression in the wing and halteres. The clones of cells ectopically expressing btd tend to recapitulate the complete development of leg and antennal discs. For example, the whole genetic network necessary to make a leg appears to be activated. btd induces the activity of hth, dac and Dll, the domains of which account for the entire disc. Furthermore, hth, dac and Dll are activated in a spatially discriminated manner. The formation of the dac and Dll domains is dependent on signalling from Wg and Dpp, although they require different signal thresholds. In one clone, for example hth is expressed only in the peripheral region, resembling the normal expression in the leg disc; in another clone the discriminate expressions of dac and Dll define three distinct regions. The formation of the dac and Dll domains is dependent on signalling from Wg and Dpp, although they require different signal thresholds, but the hth domain is independent from Wg and Dpp (Estella, 2003).

The generation of distinct hth, dac and Dll domains within the clones suggested that btd-expressing cells in the wing and haltere generate their own signalling process. Indeed, within these clones there is local activation of en, the transcription factor that initiates Hh/Wg/Dpp signalling in imaginal discs. btd-expressing clones also acquire wg and dpp activity in subsets of cells. It is probably in the boundary of en-expressing with non expressing cells where the Wg and Dpp signals are generated de novo; subsequently, their diffusion initiates the same patterning mechanism which operates during normal leg development. The result of this process is that the hth, dac and Dll genes are expressed in different domains contributing to form leg patterns containing DV and PD axes. One question for which there is no clear answer is how the initial asymmetry is generated, so that a few cells within the group gain (or lose) en activity. The cells expressing en within the clones are those closer to the posterior compartment cells. It is conceivable that there might be an external signal, perhaps mediated by Hh, which triggers the initial asymmetry (Estella, 2003).

The ability of cells expressing btd to build their own patterning mechanism is also indicated by the observation that inducing btd activity in different parts of the wing disc results in the production of similar sets of leg pattern elements. For example, in MD743/UAS-btd and omb-Gal4/UAS-btd flies, btd is induced in different, non-overlapping wing regions, and yet all leg pattern elements are produced in both genotypes. Thus, the effect of btd is by and large independent of the position where it is induced, e.g., it does not depend on local positional signals (Estella, 2003).

A relevant issue is whether the ability of the Btd product to induce the formation of the full array of ventral structures has a functional significance in normal development. This property may be a faithful reflection of the original btd/Sp1 function: the activation of the developmental program to build the ventral adult patterns. btd/Sp1 function can be envisaged as follows. During the embryonic period, the conjunction of several regulatory factors (Wg, Dpp, EGF, Hox genes) allows activation of btd/Sp1 in a group of cells in each thoracic segment (it is assumed that a similar process takes place in the head). These cells become the precursors of the ventral imaginal discs and will eventually form the ventral thorax of the adult -- these include the trunk (the hth domain) and appendage (the Dll domain) regions. The activity of btd/Sp1 is instrumental in segregating these ventral discs precursors from those of the larval epidermis and determining their imaginal fate. It is involved in specifying their segment identity (in collaboration with the Hox genes) and in establishing their pattern and growth. To achieve the latter role btd/Sp1 induces the production of the growth signals Wg and Dpp, probably in response to localized activation of en and subsequent signalling by hedgehog (hh) (Estella, 2003).

A problem with this model is that in normal development all the genes involved, hth, en, hh, wg and dpp, are expressed in embryos prior to btd/Sp1. Why should a new round of activation be necessary? Although a totally satisfactory answer can not be provided, it is noted that clones of btd-expressing cells in wing or haltere lose their memory of en expression. Those that originated in the A compartment had no previous en expression, but gained it in some cells. Conversely, all cells in P compartment clones contained en activity but some lose it. The best explanation for this unexpected behavior is that btd provokes a 'naïve' cell state in which the previous commitment for en activity is lost. Later, en activity is re-established. This phenomenon may reflect the process that occurs in normal development. The initial btd/Sp1 domain may not inherit the previous developmental commitments and has to build a new developmental program. It is worth considering that the btd/Sp1function appears to determine ventral imaginal fate as different from larval fate. This is a major developmental decision, which may require de novo establishment of the genetic system responsible for pattern and growth. A key aspect of this would be the localized activation of en in part of the btd/Sp1 domain. It is speculated that there might be a short-range signal, perhaps Hh, emanating from nearby en-expressing embryonic cells, that acts as an inducer in the btd/Sp1 primordium. There is evidence that Hh can induce en activity (Estella, 2003).

The origins of the Drosophila leg revealed by the cis-regulatory architecture of the Distalless gene

Limb development requires the elaboration of a proximodistal (PD) axis, which forms orthogonally to previously defined dorsoventral (DV) and anteroposterior (AP) axes. In arthropods, the PD axis of the adult leg is subdivided into two broad domains, a proximal coxopodite and a distal telopodite. This study shows that the progressive subdivision of the PD axis into these two domains occurs during embryogenesis and is reflected in the cis-regulatory architecture of the Distalless (Dll) gene. Dll protein in the thorax was first detected during embryonic stage 11, and continues to be visualized in this region until the end of embryogenesis. Early Dll expression, governed by the Dll304 enhancer, is in cells that can give rise to both domains of the leg as well as to the entire dorsal (wing) appendage. A few hours after Dll304 is activated, the activity of this enhancer fades, and two later-acting enhancers assume control over Dll expression. The LT enhancer is expressed in cells that will give rise to the entire telopodite, and only the telopodite. By contrast, cells that activate the DKO ("Distalless Keilin Organ") enhancer will give rise to a leg-associated larval sensory structure known as the Keilin's organ (KO). Cells that activate neither LT nor DKO, but had activated Dll304, will give rise to the coxopodite. In addition, the trans-acting signals controlling the LT and DKO enhancers are described; surprisingly, the coxopodite progenitors begin to proliferate ~24 hours earlier than the telopodite progenitors. Together, these findings provide a complete and high-resolution fate map of the Drosophila appendage primordia, linking the primary domains to specific cis-regulatory elements in Dll (McKay, 2009).

To determine how each of the cell fates in the limb primordia is specified, genetic experiments were carried out to identify the regulators of the LT and DKO enhancers. Consistent with LT's dependency on wg and dpp for leg disc expression, LT is activated in the embryo in cells that receive both inputs, as monitored by anti-Wg and anti-PMad staining. To determine whether wg is required for LT activity, a temperature-sensitive allele of wg was used to allow earlier Dll activation. Switching the embryos to the restrictive temperature at stage 11 resulted in the absence of LT activity, despite the presence of Dll protein (probably derived from Dll304 activity. In addition, ectopic activation of the wg pathway [using an activated form of armadillo (arm*)] resulted in more LT-lacZ-expressing cells (McKay, 2009).

Like wg, the dpp pathway is necessary for LT-lacZ expression in leg discs. Paradoxically, dpp signaling represses Dll in the embryo because dpp mutants show an expansion in Dll304-lacZ expression. By contrast, LT-lacZ is not expressed in dpp null embryos. LT-lacZ, but not Dll protein, was also repressed by two dpp pathway repressors, Dad and brk. Conversely, stimulation of the dpp pathway [using an activated form of the Dpp receptor (TkvQD)] resulted in ectopic activation of LT ventrally (McKay, 2009).

Taken together, these data demonstrate that LT is activated by Wg and Dpp in the embryonic limb primordia, just as it (and Dll) is in the leg disc. Similarly, DKO activity also requires Wg and Dpp input (McKay, 2009).

Although LT is activated by wg and dpp in the leg primordia, these signals are also present in each abdominal segment. Consequently, there must be additional factors that restrict LT activity to the thorax. One possibility is that LT is repressed by the abdominal Hox factors, such as Dll304. Alternatively, LT might be regulated by Dll, itself. In Dll null embryos LT-lacZ was initially expressed in a stripe of cells instead of a ring, but then expression decayed. Ectopic expression of Dll resulted in weak ectopic expression of LT-lacZ in the thorax and abdomen. These data suggest that LT activity is restricted to the thorax in part because of the earlier restriction of Dll304 activity to the thorax (McKay, 2009).

The related zinc-finger transcription factors encoded by buttonhead (btd) and Sp1 are also expressed in the limb primordia and are also required for ventral appendage specification. In strong btd hypomorphs, the activity of LT was still detected but the number of cells expressing LT-lacZ was decreased and its pattern was disrupted. LT-lacZ expression was completely eliminated in animals bearing a large deficiency that removes both btd and Sp1. By contrast, Dll304 was activated normally in these animals (data not shown). Importantly, LT-lacZ expression was rescued by expressing btd in these deficiency embryos. By contrast, expressing Dll, tkvQD, or arm* did not rescue LT expression in these deficiency embryos. Ectopic expression of btd resulted in weak ectopic activation of LT-lacZ in cells of the thorax and abdomen. Strikingly, the simultaneous expression of Dll and btd resulted in robust ectopic expression of LT-lacZ in abdominal segments in the equivalent ventrolateral position as the thoracic limb primordia. btd and Dll were not sufficient to activate LT in wg null embryos (data not shown). These data indicate that the thoracic-specific expression of the LT enhancer is controlled by the combined activities of btd and/or Sp1, Dll and the wg and dpp pathways (McKay, 2009).

Although the data suggest that LT is activated by a combination of Wg, Dpp, Btd and Dll, these activators are also present in the precursors of the KO, which activate DKO instead of LT. Because the KO is a sensory structure, the role of members of the achaete-scute complex (ASC) that are expressed in these cells was tested. In embryos hemizygous for a deficiency that removes the achaete-scute complex, LT-lacZ expression was expanded at the expense of the Ct-expressing cells. Consistently, ectopic expression of the ASC gene asense (ase) repressed LT and increased the number of Ct-expressing cells. These data suggest that there is a mutual antagonism between the progenitors of the telopodite and those of the KO. It was also found that DKO-lacZ expression in the leg primordia was lost in Dll or btd null embryos, consistent with the loss of KOs in these mutants. DKO activity was also lost from the limb primordia in embryos deficient for the ASC. These results indicate that DKO is activated by the same genes that promote LT expression but, in addition, requires proneural input from the ASC (McKay, 2009).

One of the most interesting findings from this work is that the temporal control of Dll expression in the limb primordia by three cis-regulatory elements is linked to cell-type specification. The earliest acting element, Dll304, is active throughout the appendage primordia. At the time Dll304 is active, the cells are multipotent and can give rise to any part of the dorsal or ventral appendages, or KO. A few hours later, Dll304 activity fades, and two alternative cis-regulatory elements become active. Together, these two elements allow for the uninterrupted and uniform expression of Dll within the appendage primordia. However, their activation correlates with a higher degree of refinement in cell fate potential: LT, active in only the outer ring of the appendage primordia, is only expressed in the progenitors of the telopodite. By contrast, DKO, active in the cells within the LT ring, is only expressed in the progenitors of the KO. Thus, although the pattern of Dll protein appears unchanged, the control over Dll expression has shifted from singular control by Dll304 to dual control by LT and DKO. Moreover, not only is there a molecular handoff from Dll304 to LT and DKO, the two later enhancers both require the earlier expression of Dll. Thus, the logic of ventral primordia refinement depends on a cascade of Dll regulatory elements in which the later ones depend on the activity of an earlier one (McKay, 2009).

The high-resolution view of the embryonic limb primordia provided in this study allows clarification of some contradictions that currently exist in the literature. Initial expression of Dll in the thorax overlaps entirely with Hth-nExd (referring to nuclear Extradenticle). Subsequently, hth expression is lost from most, but not all, of the Dll-expressing cells of the leg primordia. The first reports describing these changes failed to recognize the persistent overlap between Dll and Hth-nExd in some cells. As a result, and partly because of the analogy with the third instar leg disc, the predominant view of this fate map became that the Dll-positive, Hth-nExd-negative cells of the embryonic primordia gave rise to the telopodite, while the surrounding Hth-positive cells gave rise to the coxopodite. The expression pattern of esg, a gene required for the maintenance of diploidy, was also misinterpreted as being a marker exclusively of proximal leg fates. Counter to these earlier studies, the current experiments unambiguously show that the Dll-positive, Hth-nExd-negative cells in the center of the primordia give rise to the KO, the ring of Dll-positive, Esg-positive, Hth-nExd-positive cells gives rise to the telopodite, and the remaining Esg-positive, Dll-negative cells give rise to the coxopodite (McKay, 2009).

The spurious expression of DKO-lacZ in Dll-non-expressing cells outside the leg primorida complicates the interpretation of several experiments. Attempts to refine DKO activity by changing the size of the cloned fragment proved unsuccessful. Nevertheless, the evidence supports the idea that DKO-positive, Dll-positive cells of the leg primordia give rise to the Keilin's organ, and not the adult appendage (McKay, 2009).

The progenitors of the coxopodite begin to proliferate at approximately 48 hours of development, consistent with previous measurements of leg imaginal disc growth, whereas the progenitors of the telopodite do not resume proliferating for an additional 12 to 24 hours. According to estimates of the cell cycle time in leg discs, this difference in the onset of proliferation results in one to two additional cell divisions in the coxopodite, consistent with images of late second instar leg discs presented in this study. Why might the telopodite and coxopodite begin proliferation at different times? One possibility is that the cells of the coxopodite give rise to the peripodial epithelium that covers the leg imaginal disc, and therefore require additional cell divisions relative to the telopodite. It is also possible that the telopodite is delayed because the neurons of the Keilin's organ serve a pathfinding role for larval-born neurons that innervate the adult limb. Perhaps this pathfinding function requires that the KO and telopodite remain associated with each other through the second instar. Consistently, the leg is the only imaginal disc that has not invaginated as a sac-like structure in newly hatched first instar larvae (McKay, 2009).

A possible explanation for the delay in the onset of telopodite proliferation is the persistent co-expression of hth and Dll in these cells; hth (and tsh) expression is turned off in these cells at about the same time they begin to proliferate. Consistent with this idea, maintaining the expression of hth throughout the primordia blocks the proliferation of the telopodite. Also noteworthy is the finding that the genes no ocelli and elbow have been shown to mediate the ability of Wg and Dpp to repress coxopodite fates. Together with the current findings, it is possible that the activation of these two genes in the LT-expressing progenitors is the trigger that turns off hth and tsh in these cells (McKay, 2009).

The experiments suggest that once LT is activated, and under normal growth conditions, there is a lineage restriction between the telopodite and coxopodite. By contrast, previous lineage-tracing experiments using tsh-Gal4 concluded that the progeny of proximal cells could adopt more distal leg fates. However, tsh is still expressed in the telopodite progenitors far into the second instar, providing an explanation for these results. In contrast to this early restriction, there is no evidence for a later lineage restriction within the telopodite. For example, the progeny of a Dll-positive cell can lose Dll expression and contribute to the dac-only domain (McKay, 2009).

Interestingly, the lineage restriction between coxopodite and telopodite is not defined by the presence or absence of Hth-nExd or Tsh because both progenitor populations express hth and tsh after their fates have been specified. By contrast, when these two domains are specified, the telopodite expresses Dll, while the coxopodite does not, suggesting that Dll may be important for the lineage restriction. However, later in development, some cells in the telopodite lose Dll expression and express dac, but continue to respect the coxopodite-telopodite boundary. Thus, either Dll expression in the telopodite is somehow remembered or the telopodite-coxopodite boundary can be maintained by dac, which is expressed in place of Dll immediately adjacent to the telopodite-coxopodite boundary. Also noteworthy is the finding that clones originating in the coxopodite can contribute to the trochanter, the segment inbetween the proximal and distal components of the adult leg that expresses both Dll and hth in third instar imaginal discs. However, the progeny of such clones do not contribute to fates more distal than the trochanter. Likewise, a clone originating in the telopodite can also contribute to the trochanter, but will not grow more proximally into the coxa. Thus, the lineage restriction uncovered here seems to be determined by distinct combinations of transcription factors expressed in the coxopodite and telopodite progenitors at stage 14. The progeny of cells that express Dll, tsh and hth can populate the telopodite or trochanter, whereas the progeny of cells that express tsh and hth, but not Dll, can populate the coxopodite or trochanter. In light of Minute-positive results, however, the lineage restriction between coxopodite and telopodite does not satisfy the classical definition of a compartment boundary. A similar non-compartment lineage restriction has also been documented along the PD axis of the developing Drosophila wing (McKay, 2009).

Non-redundant selector and growth-promoting functions of two sister genes, buttonhead and Sp1, in Drosophila leg development

The radically distinct morphologies of arthropod and tetrapod legs argue that these appendages do not share a common evolutionary origin. Yet, despite dramatic differences in morphology, it has been known for some time that transcription factors encoded by the Distalless (Dll)/Dlx gene family play a critical role in the development of both structures. This study shows that a second transcription factor family encoded by the Sp8 gene family, previously implicated in vertebrate limb development, also plays an early and fundamental role in arthropod leg development. By simultaneously removing the function of two Sp8 orthologs, buttonhead (btd) and Sp1, during Drosophila embryogenesis, adult leg development was found to be completely abolished. Remarkably, in the absence of these factors, transformations from ventral to dorsal appendage identities are observed, suggesting that adult dorsal fates become derepressed when ventral fates are eliminated. Further, Sp1 was shown to play a much more important role in ventral appendage specification than btd, and Sp1 lies genetically upstream of Dll. In addition to these selector-like gene functions, Sp1 and btd are also required during larval stages for the growth of the leg. Vertebrate Sp8 can rescue many of the functions of the Drosophila genes, arguing that these activities have been conserved, despite more than 500 million years of independent evolution. These observations suggest that an ancient Sp8/Dlx gene cassette was used in an early metazoan for primitive limb-like outgrowths and that this cassette was co-opted multiple times for appendage formation in multiple animal phyla (Estella, 2010).

Prior to this study, understanding of the roles that btd and Sp1 play in ventral appendage development in Drosophila was largely derived from ectopic expression experiments showing that btd could induce ectopic leg development when expressed in dorsal imaginal discs. In addition, based on a large deficiency that removes >50 genes, it was suggested that these genes may function upstream of Dll in ventral appendage specification. What was lacking in this previous study was the ability to specifically analyze the functions of these genes, both in embryogenesis and during adult development, using loss-of-function null alleles. Using a newly derived deficiency, together with rescue experiments, this study showed that these Zn-finger transcription factors play non-redundant roles in ventral appendage development. Moreover, for all of the readouts examined in this study (leg allocation, leg growth, proliferation, and PD axis formation) btd plays a much more minor or no role compared to Sp1. Early, Sp1, but not btd, is required to define the group of cells that will give rise to the legs and perhaps additional ventral body structures as well. Thus, Sp1 is a selector-like gene for the entire ventral appendage. Later in development, both genes are required for the proper growth of the leg, although to very different degrees. It was also shown that vertebrate Sp8 retains both the selector and growth-promoting functions, suggesting that there has been a remarkable amount of functional conservation between the vertebrate and fly genes (Estella, 2010).

During larval development, it was found that Sp1 is required for the proper growth of the entire leg, from the coxa through the tarsus. In contrast, btd plays a much more limited role in the tibia and femur. At this stage, neither gene is required for leg identity, nor are they required for the development of ventral body structures that arise from the most proximal cells in the leg imaginal disc. These 'late' phenotypes are consistent with the expression patterns of these genes in the third instar leg imaginal discs, where they appear to mark the entire presumptive leg, but not more proximal cells. This is interesting, because prior to these observations there were no markers that distinguished between the hth-expressing cells that give rise to the coxa from the hth-expressing cells that give rise to the ventral body wall. Dll, for example, is expressed in the cells that give rise to the distal tibia and tarsus, and lineage tracing with the Dll-LT element marks the entire telopodite (trochanter, femur, tibia, and tarsus). The addition of the btd and Sp1 expression patterns and mutant phenotypes to previously characterized PD genes therefore adds an important demarcation that distinguishes leg from body fates (Estella, 2010).

This analysis also reveals dramatic differences in the post-embryonic functions of btd and Sp1. Specifically, most of the growth phenotypes observed when both genes are removed can be phenocopied by knocking down only Sp1. In contrast, btdXG81 clones (or btdXA clones) have no phenotypes in the antenna, and, in the leg, result in only partial fusions between the femur and tibia. Thus, Sp1, not btd, plays an important and non-redundant function in ventral appendage development at this stage (Estella, 2010).

Selector and selector-like genes have the property that they specify an entire organ or body part. The classic example is engrailed (en) which 'selects' posterior compartment identities in Drosophila. Another example is eyeless (ey), which is both necessary and sufficient for eye development in Drosophila. In the leg, previous work highlighted the role of Dll in ventral appendage specification. In the absence of Dll, the distal portion of the leg fails to develop, while dorsal appendages remain wild type. Moreover, ectopic expression of Dll can induce distal legs to develop in dorsal positions. Taken together, these observations suggested that Dll is a selector-like gene for the distal leg (Estella, 2010).

Despite the requirement for Dll in leg development, it has been known for sometime that the ventral appendage primordia form in the absence of Dll. Moreover, homeotic transformations are not observed in the absence of Dll. Thus, Dll cannot be considered a selector-like gene for the entire ventral appendage. These observations raise the question of what factor or factors initially specify the cells that will give rise to the ventral appendage. It is proposed that Sp1 fulfills this selector-like role (Estella, 2010).

The suggestion that Sp1 is a selector-like gene for the entire ventral appendage stems in part from the observation that when the function of this gene is removed early in development, ∼10% of the animals have dramatic transformations of ventral structures to dorsal structures. In many of these cases, both wing and notum tissue were observed developing in ventral positions. Molecularly, Dll and dac expression is lost in transformed leg discs, and ectopic expression of vg and eyg, two markers for the dorsal appendages, are observed instead. The expression of Dll-304, which is traditionally been considered a marker for the ventral appendage, in Df(btd,Sp1) embryos may seem at odds with the idea that Sp1 is required for the initial specification of leg fates. However, fate-mapping studies show that Dll-304-expressing cells give rise to both the ventral (leg) and dorsal (wing and haltere) appendages. Thus, Dll-304 cannot be considered a ventral marker, and its activity in Df(btd,Sp1) embryos only confirms the establishment of appendage primordia without ventral or dorsal identity (Estella, 2010).

In sum, the striking transformations of fate seen in Df(btd,Sp1) animals suggest that Sp1 promotes ventral fates, both the entire leg and ventral body wall, and that in the absence of this gene, dorsal fates are de-repressed. This change in developmental fate is analogous to other classical homeotic transformations, for example, when the leg is transformed to antenna in the absence of Antennapedia (Antp). Note that btd null clones made at the same early time in development only result in mild growth defects, but legs are still generated. Thus, btd is not required for this function. However, because an Sp1 null allele (btd+) is not currently available, we cannot at this time be completely certain that btd plays no role in this process (Estella, 2010).

Because wing development is normally limited to T2, it was unexpected to observe leg to wing transformations in the T1 and, to a lesser extent, T3 segments. One potential explanation for this violation of antero-posterior identity is due to the timing of clone induction. Although the Hox genes are responsible for determining the segmental identities of the dorsal appendages, it may be that they are deployed at different times in the ventral and dorsal primordia in the different thoracic segments. If this is the case, then the resulting transformations may be very sensitive to the time they were generated and to their segmental origins. It is also worth noting that the wing primordia and T2 identity can be generated in the absence of Hox input. Thus, wing fates, as opposed to haltere or humeral (dorsal T1) fates, represent a Hox-free default state, which may predominate in these aberrant developmental situations (Estella, 2010).

Together with previous studies, these findings allow a presentation of a more complete view of ventral appendage specification, which is brocken down into three main phases. In the first phase, Sp1, btd, and Dll (via it's early Dll-304 enhancer) are initially activated in parallel in a ventral domain in each thoracic hemisegment of stage 11 embryos. The activation of all three genes is dependent on Wg signaling. This early, Dll-304-driven expression of Dll does not require either btd or Sp1. This initial group of cells is fated to give rise to both the entire ventral and dorsal thoracic imaginal discs, in other words, the entire adult thorax. In the second phase, which begins at stage 14, Dll-304 is no longer active and Dll is controlled by late-acting enhancers such as Dll-LT, which is activated by Wg and Dpp signaling. Interestingly, as shown in this study, these late-acting Dll enhancers also require Sp1, but not btd, thus placing Sp1 genetically upstream of Dll. At this stage, the Dll+ cells will only give rise to the leg telopodite. Sp1 is also required for telopodite formation but is carrying out at least two additional functions. One is that, unlike Dll, Sp1 is required to specify more proximal leg segments (the coxapodite). Second, the ventral to dorsal homeotic transformations described above suggest that Sp1 is also required to repress dorsal fates. Finally, in the third phase, Dll begins to autoactivate it's expression and no longer depends on Wg and Dpp inputs. At this stage, Dll also no longer requires Sp1 to be expressed. Instead of working through Dll, btd and Sp1 continue to play a critical role in leg development but now work in parallel to Dll to promote the growth of the entire leg. Thus, the specification of the ventral primordia depends on a feed-forward logic in which Sp1 activates late embryonic Dll expression followed by a phase in which both btd and Sp1 contribute to appendage growth in parallel to Dll (Estella, 2010).

Besides having a PD axis, arthropod and vertebrate appendage morphologies have little in common. Moreover, the developmental logic of limb formation in Drosophila is very different from that of vertebrate limb development. In flies, Hedgehog signaling induces two antagonistic secondary signals, Dpp and Wg, which in turn establish the PD axis by activating genes such as Dll and dac. In vertebrate limb development, Sonic hedgehog induces the activity of fibroblast growth factor-like molecules such as FGF8 in the ectoderm, which drives the proliferation of the underlying mesenchyme and the nested expression of Hox genes to create a PD axis. Despite these differences, it is striking that multiple vertebrate orthologs of both Sp1 and Dll are expressed during vertebrate limb development. In addition, orthologs of both hth and exd (Meis and pbx, respectively) are expressed in the proximal domain of the developing mouse limb. Although the existence of multiple Dll and Sp1 orthologs (Dlx1/Dlx2/Dlx5/Dlx6 and Sp8/Sp9, respectively) makes it much more challenging to assess their functions in detail, the available data demonstrate that, as in flies, both sets of genes are critical for vertebrate limb development. The current results, illustrating that vertebrate Sp8 can rescue many of the Sp1 and btd loss of function phenotypes in Drosophila, support the idea that appendage development in these two phyla represents a case of 'deep homology'. Interestingly, that orthologs of both Sp1 and Dll gene families are used in both phyla argue that, for appendage development, the functions of these transcription factors have been much more conserved than those of the signaling pathways used in limb development. The same conclusion holds for eye development where the transcription factors, more than the deployment of specific signaling pathways, have been conserved over vast evolutionary distances. These observations imply that, once established, transcription factor networks may be very stable, while the organization of signaling pathway networks may be much more plastic and easily modified to accommodate radically distinct morphologies (Estella, 2010).


REFERENCES

Adnane, J., Shao, Z. and Robbins, P. D. (1999). Cyclin D1 associates with the TBP-associated factor TAF(II)250 to regulate Sp1-mediated transcription. Oncogene 18(1): 239-47. PubMed ID: 9926939

Ahlgren, R., et al. (1999). Role of Sp1 in cAMP-dependent transcriptional regulation of the bovine CYP11A gene. J. Biol. Chem. 274: 19422-19428. PubMed ID: 10383457

Armstrong, S. A., et al. (1997). Casein kinase II-mediated phosphorylation of the C terminus of Sp1 decreases its DNA binding activity. J. Biol. Chem. 272 (21): 13489-13495. PubMed ID: 9153193

Athanikar, J. N., Sanchez, H. B. and Osborne, T. F. (1997). Promoter selective transcriptional synergy mediated by sterol regulatory element binding protein and Sp1: a critical role for the Btd domain of Sp1. Mol. Cell. Biol. 17(9): 5193-5200. PubMed ID: 9271397

Beermann, A., Aranda, M. and Schroder, R. (2004). The Sp8 zinc-finger transcription factor is involved in allometric growth of the limbs in the beetle Tribolium castaneum. Development 131(4): 733-42. PubMed ID: 14724124

Bigger, C. B., Melnikova, I. N. and Gardner, P. D. (1997). Sp1 and Sp3 regulate expression of the neuronal nicotinic acetylcholine receptor beta4 subunit gene. J. Biol. Chem. 272(41): 25976-25982. PubMed ID: 9325332

Brown, J. L., Grau, D. J., DeVido, S. K. and Kassis, J. A. (2005). An Sp1/KLF binding site is important for the activity of a Polycomb group response element from the Drosophila engrailed gene. Nucleic Acids Res. 33(16): 5181-9. PubMed ID: 16155187

Borja, P. F. de, et al. (2001). Cyclin A-CDK phosphorylates Sp1 and enhances Sp1-mediated transcription. EMBO J. 20: 5737-5747. PubMed ID: 11598016

Bouwman, P. and Philipsen, S. (2002). Regulation of the activity of Sp1-related transcription factors. Mol. Cell Endocrinol. 195: 27-38. PubMed ID: 12354670

Cao, F., Chen, Y., Cierpicki, T., Liu, Y., Basrur, V., Lei, M. and Dou, Y. (2010). An Ash2L/RbBP5 heterodimer stimulates the MLL1 methyltransferase activity through coordinated substrate interactions with the MLL1 SET domain. PLoS One 5: e14102. PubMed ID: 21124902

Chen, J. L., et al. (1994). Assembly of recombinant TFIID reveals differential coactivator requirements for distinct transcriptional activators. Cell 79(1): 93-105. PubMed ID: 7923382

Chen-Plotkin, A. S., et al. (2006). Decreased association of the transcription factor Sp1 with genes downregulated in Huntington's disease. Neurobiol. Dis. 22(2): 233-41. PubMed ID: 16442295

Cordoba, S., Requena, D., Jory, A., Saiz, A. and Estella, C. (2016). The evolutionary conserved transcription factor Sp1 controls appendage growth through Notch signaling. Development 143(19):3623-3631. PubMed ID: 27578786

Crozatier, M., et al. (1996). Collier, a novel regulator of Drosophila head development, is expressed in a single mitotic domain. Curr. Biol. 6(6): 707-718. PubMed ID: 8793297

Crozatier, M., et al. (1999). Head versus trunk patterning in the Drosophila embryo; collier requirement for formation of the intercalary segment. Development 126: 4385-4394. PubMed ID: 10477305

Dunah, A. W., et al. (2002). Sp1 and TAFII130 transcriptional activity disrupted in early Huntington's disease, Science 296: 2238-2243. PubMed ID: 11988536

Estella, C., et al. (2003). The role of buttonhead and Sp1 in the development of the ventral imaginal discs of Drosophila. Development 130: 5929-5941. PubMed ID: 14561634

Estella, C. and Mann, R. S. (2010). Non-redundant selector and growth-promoting functions of two sister genes, buttonhead and Sp1, in Drosophila leg development. PLoS Genet 6: e1001001. PubMed ID: 20585625

Feng, X.-H., Lin, X. and Derynck, R. (2000). Smad2, Smad3 and Smad4 cooperate with Sp1 to induce p15Ink4B transcription in response to TGF-beta. EMBO J. 19: 5178-5193. PubMed ID: 11013220

Flora, A., et al. (2001). SP proteins and PHOX2B regulate the expression of the human PHOX2a gene. J. Neurosci. 21(18): 7037-7045. PubMed ID: 11549713

Franch-Marro, X., Martin, N., Averof, M. and Casanova, J. (2006). Association of tracheal placodes with leg primordia in Drosophila and implications for the origin of insect tracheal systems. Development 133(5): 785-90. PubMed ID: 16469971

Gill, G., et al. (1994). A glutamine-rich hydrophobic patch in transcription factor Sp1 contacts the dTAFII110 component of the Drosophila TFIID complex and mediates transcriptional activation. Proc. Natl. Acad. Sci. 91(1): 192-6. PubMed ID: 8278363

Gollner, H., et al. (2001). Impaired ossification in mice lacking the transcription factor Sp3. Mech. Dev. 106: 77-83. PubMed ID: 11472836

Greenwel, P., et al. (1997). Sp1 is required for the early response of alpha2(I) collagen to transforming growth factor-beta1. J. Biol. Chem. 272(32): 19738-19745. PubMed ID: 9242631

Harrison, S. M. (2000). Sp5, a new member of the Sp1 family, is dynamically expressed during development and genetically interacts with Brachyury. Dev. Bio. 227: 358-372. PubMed ID: 11071760

Hilton, T. L. and Wang, E. H. (2003). Transcription factor IID recruitment and Sp1 activation. Dual function of TAF1 in cyclin D1 transcription. J. Biol. Chem. 278(15): 12992-3002. PubMed ID: 12569092

Hilton, T. L., Li, Y., Dunphy, E. L. and Wang, E. H. (2005). TAF1 histone acetyltransferase activity in Sp1 activation of the cyclin D1 promoter. Mol. Cell. Biol. 25(10): 4321-32. PubMed ID: 15870300

Hirano, F., et al. (1998). Functional interference of Sp1 and NF-kappaB through the same DNA binding site. Mol. Cell. Biol. 18(3): 1266-1274. PubMed ID: 9488441

Hoey, T., et al. (1993). Molecular cloning and functional analysis of Drosophila TAF110 reveal properties expected of coactivators. Cell 72(2): 247-60. PubMed ID: 7678780

Jackson, B., et al. (2011). TALE homeodomain proteins regulate site-specific terminal differentiation, LCE genes and epidermal barrier. J. Cell Sci. 124: 1681-1690. PubMed ID: 21511732

Ji, C., et al. (1997). Multiple and essential Sp1 binding sites in the promoter for transforming growth factor-beta type I receptor. J. Biol. Chem. 272(34): 21260-21267. PubMed ID: 9261136

Kadonaga, J. T., et al. (1988). Distinct regions of Sp1 modulate DNA binding and transcriptional activation. Science 242(4885): 1566-70. PubMed ID: 3059495

Kardassis, D., et al. (1999). c-Jun transactivates the promoter of the human p21WAF1/Cip1 gene by acting as a superactivator of the ubiquitous transcription factor Sp1. J. Biol. Chem. 274: 29572-29581. PubMed ID: 10506225

Kawakami, J., et al. (2004). Sp8 and Sp9, two closely related buttonhead-like transcription factors, regulate Fgf8 expression and limb outgrowth in vertebrate embryos. Development 131: 4763-4774. PubMed ID: 15358670

Komori, H., Xiao, Q., Janssens, D. H., Dou, Y. and Lee, C. Y. (2014). Trithorax maintains the functional heterogeneity of neural stem cells through the transcription factor Buttonhead. Elife 3 [Epub ahead of print]. PubMed ID: 25285447

Krainc, D., et al. (1998). Synergistic activation of the N-methyl-D-aspartate receptor subunit 1 promoter by myocyte enhancer factor 2C and Sp1. J. Biol. Chem. 273(40): 26218-24. PubMed ID: 9748305

Kwon, H. S., et al. (1999). Sp3 and Sp4 can repress transcription by competing with Sp1 for the core cis-elements on the human ADH5/FDH minimal promoter. J. Biol. Chem. 274(1): 20-8. PubMed ID: 9867805

Lee, J.-A., et al. (2005). Transcriptional activity of Sp1 is regulated by molecular interactions between the zinc finger DNA binding domain and the inhibitory domain with corepressors, and this interaction is modulated by MEK. J. Biol. Chem. 280: 28061-28071. PubMed ID: 15878880

Lee, Y.-H., et al. (1997). The ability of C/EBPbeta but not C/EBPalpha to synergize with an SP1 protein is specified by the leucine zipper and activation domain. Mol. Cell. Biol. 17: 2038-47. PubMed ID: 9121452

Lécuyer, E., et al. (2002). The SCL complex regulates c-kit expression in hematopoietic cells through functional interaction with Sp1. Blood 100: 2430-2440. PubMed ID: 12239153

Li, S. H., et al. (2002). Interaction of Huntington disease protein with transcriptional activator Sp1. Mol. Cell. Biol. 22(5): 1277-87. PubMed ID: 11839795

Lin, J. X. and Leonard, W. J. (1997). The immediate-early gene product Egr-1 regulates the human interleukin-2 receptor beta-chain promoter through noncanonical Egr and Sp1 binding sites. Mol. Cell. Biol. 17(7): 3714-3722. PubMed ID: 9199305

Liu, W. L., et al. (2009). Structures of three distinct activator-TFIID complexes. Genes Dev. 23(13): 1510-21. PubMed ID: 19571180

Marin, M., et al. (1997). Transcription factor Sp1 is essential for early embryonic development but dispensable for cell growth and differentiation. Cell 89: 619-628. PubMed ID: 9160753

McDonough, P. M., et al. (1997). Collaborative Roles for c-Jun N-terminal Kinase, c-Jun, Serum Response Factor, and Sp1 in Calcium-regulated Myocardial Gene Expression. J. Biol. Chem. 272(38): 24046-24053. PubMed ID: 9295358

McKay, D. J., Estella, C. and Mann, R. S. (2009). The origins of the Drosophila leg revealed by the cis-regulatory architecture of the Distalless gene. Development 136(1): 61-71. PubMed ID: 19036798

Merika, M. and Orkin, S. H. (1995). Functional synergy and physical interactions of the erythroid transcription factor GATA-1 with the Kruppel family proteins Sp1 and EKLF. Mol. Cell. Biol. 15(5): 2437-2447. PubMed ID: 7739528

Mortensen, E. R., et al. (1997). Epidermal growth factor and okadaic acid stimulate Sp1 proteolysis. J. Biol. Chem. 272(26): 16540-16547. PubMed ID: 9195964

Mohler, J. (1993). Genetic regulation of CNC expression in the pharyngeal primordia of Drosophila blastoderm embryos. Roux's Arch. Dev. Biol. 202: 214-23.

Mohler, J. (1995). Spatial regulation of segment polarity gene expression in the anterior terminal region of the Drosophila blastoderm embryo. Mech. Dev. 50: 151-161. PubMed ID: 7619727

Moustakas, A. and Kardassis, D. (1998). Regulation of the human p21/WAF1/Cip1 promoter in hepatic cells by functional interactions between Sp1 and Smad family members. Proc. Natl. Acad. Sci. 95(12): 6733-6738. PubMed ID: 9618481

Muller, C., et al. (1999). Cloning of the cyclin A1 genomic structure and characterization of the promoter region. GC boxes are essential for cell cycle-regulated transcription of the cyclin A1 gene. J. Biol. Chem. 274(16): 11220-8. PubMed ID: 10196209

Ochoa-Espinosa, A., Yu, D., Tsirigos, A., Struffi, P. and Small, S. (2009). Anterior-posterior positional information in the absence of a strong Bicoid gradient. Proc. Natl. Acad. Sci. 106(10): 3823-8. PubMed ID: 19237583

Pagliuca, A., Cannada-Bartoli, P. and Lania, L. (1998). A role for Sp and helix-loop-helix transcription factors in the regulation of the human Id4 gene promoter activity. J. Biol. Chem. 273(13): 7668-7674. PubMed ID: 9516472

Piedrafita, F. J. and Pfahl, M. (1997). Retinoid-induced apoptosis and Sp1 cleavage occur independently of transcription and require caspase activation. Mol. Cell. Biol. 17(11): 6348-6358. PubMed ID: 9343396

Pieri, I., et al. (1999). Regulation of the murine NMDA-receptor-subunit NR2C promoter by Sp1 and fushi tarazu factor1 (FTZ-F1) homologues. Eur. J. Neurosci. 11(6): 2083-92. PubMed ID: 10336677

Qin, H., Sun, Y. and Benveniste, E. N. (1999). The transcription factors Sp1, Sp3, and AP-2 are Required for constitutive matrix metalloproteinase-2 gene expression in astroglioma cells. J. Biol. Chem. 274: 29130-29137. PubMed ID: 10506168

Rafty, L. A., Santiago, F. S. and Khachigian, L. M. (2002). NF1/X represses PDGF A-chain transcription by interacting with Sp1 and antagonizing Sp1 occupancy of the promoter. EMBO J. 21: 334-343. PubMed ID: 11823426

Rosmarin, A. G., et al. (1998). Sp1 Cooperates with the ets transcription factor, GABP, to activate the CD18 (beta2 Leukocyte Integrin) promoter. J. Biol. Chem. 273(21): 13097-13103. PubMed ID: 9582348

Rusch, D. B. and Kaufman, T. C. (2000). Regulation of proboscipedia in Drosophila by homeotic selector genes. Genetics 156: 183-194. PubMed ID: 10978284

Ryu S., et al. (1999). The transcriptional cofactor complex CRSP is required for activity of the enhancer-binding protein Sp1. Nature 397(6718): 446-50. PubMed ID: 9989412

Santini, M. P., et al. (2001). Cross talk among calcineurin, Sp1/Sp3, and NFAT in control of p21WAF1/CIP1 expression in keratinocyte differentiation Proc. Natl. Acad. Sci. 98: 9575-9580. PubMed ID: 11493684

Schock, F., et al. (1999a). Drosophila head segmentation factor buttonhead interacts with the same TATA box-binding protein-associated factors and in vivo DNA targets as human Sp1 but executes a different biological program. Proc. Natl. Acad. Sci. 96(9): 5061-5. PubMed ID: 10220418

Schock, F., et al. (1999b). Common and diverged functions of the Drosophila gene pair D-Sp1 and buttonhead. Mech. Dev. 89: 125-132. PubMed ID: 10559487

Schock, F., et al. (2000). Phenotypic suppression of empty spiracles is prevented by buttonhead. Nature 405: 351-354. PubMed ID: 10830964

Shandala, T., et al. (1999). The Drosophila dead ringer gene is required for early embryonic patterning through regulation of argos and buttonhead expression. Development 126: 4341-4349. PubMed ID: 10477301

Smale, S. T., et al. (1990). Transcriptional activation by Sp1 as directed through TATA or initiator: specific requirement for mammalian transcription factor IID. Proc. Natl. Acad. Sci. (12): 4509-13. PubMed ID: 2141169

Spahn, P., Huelsmann, S., Rehorn, K. P., Mischke, S., Mayer, M., Casali, A. and Reuter, R. (2013). Multiple regulatory safeguards confine the expression of the GATA factor serpent to the hemocyte primordium within the Drosophila mesoderm. Dev Biol 386(1): 272-9. PubMed ID: 24360907

Supp, D. M., et al. (1996). Sp4, a member of the Sp1-family of zinc finger transcriptions factors, is required for normal murine growth, viability and male fertility. Dev. Biol 176: 284-299 8660867

Tallafuss, A., et al. (2001). The zebrafish buttonhead-like factor Bts1 is an early regulator of pax2.1 expression during mid-hindbrain development. Development 128: 4021-4034. PubMed ID: 11641225

Thorpe, C. J., Weidinger, G. and Moon, R. T. (2005). Wnt/ß-catenin regulation of the Sp1-related transcription factor sp5l promotes tail development in zebrafish. Development 132: 1763-1772. PubMed ID: 15772132

Thottassery, J. V., et al. (1999). Sp1 and egr-1 have opposing effects on the regulation of the rat Pgp2/mdr1b gene. J. Biol. Chem. 274(5): 3199-206. PubMed ID: 9915860

Treichel, D., Schöck, F., Jäckle, H., Gruss, P. and Mansouri, A. (2003). mBtd is required to maintain signaling during murine limb development. Genes and Development 17: 2630-2635. PubMed ID: 14597661

Utley, R. T., et al. (1997). SWI/SNF stimulates the formation of disparate activator-nucleosome complexes but is partially redundant with cooperative binding. J. Biol. Chem. 272(19): 12642-12649. PubMed ID: 9139720

Vinals, F., et al. (1997). Myogenesis and MyoD down-regulate Sp1. A mechanism for the repression of GLUT1 during muscle cell differentiation. J. Biol. Chem. 272 (20): 12913-12921. PubMed ID: 9148896

Vincent, A., Blankenship, J. T. and Wieschaus, E. (1997). Integration of the head and trunk segmentation systems controls cephalic furrow formation in Drosophila. Development 124(19): 3747-3754. PubMed ID: 9367430

Watanabe, G., et al. (1998). Inhibition of cyclin D1 kinase activity is associated with E2F-mediated inhibition of cyclin D1 promoter activity through E2F and Sp1. Mol. Cell. Biol. 18(6): 3212-3222. PubMed ID: 9584162

Wimmer, E. A., et al. (1993). A Drosophila homologue of human Sp1 is a head-specific segmentation gene. Nature 366: 690-4 . PubMed ID: 8259212

Wimmer, E. A., et al., (1995). Trans- and cis-acting requirements for blastodermal expression of the head gap gene buttonhead. Mech. Dev. 53: 235-245 8562425

Wimmer, E. A., et al. (1996). buttonhead and D-Sp1: a novel Drosophila gene pair. Mech. Dev. 59: 53-62. PubMed ID: 8892232

Wimmer, E. A., et al. (1997).buttonhead does not contribute to a combinatorial code proposed for Drosophila head development. Development 124 (8): 1509-1517. PubMed ID: 9108367

Xie, Y., Li, X., Zhang, X., Mei, S., Li, H., Urso, A. and Zhu, S. (2014). The Sp8 transcription factor Buttonhead prevents premature differentiation of intermediate neural progenitors. Elife 3 [Epub ahead of print]. PubMed ID: 25285448

Yin, Z., Xu, X. L. and Frasch, M. (1997). Regulation of the Twist target gene tinman by modular cis-regulatory elements during early mesoderm development. Development 124(24): 4971-4982. PubMed ID: 9362473

Younossi-Hartenstein, A., et al. (1997). Control of early neurogenesis in the Drosophila brain by the head gap genes tll, otd, ems, and btd. Dev. Biol 182: 270-283. PubMed ID: 9070327

Yu, J. H., et al. (1999). Role of the Sp family of transcription factors in the ontogeny of growth hormone receptor gene expression. J. Biol. Chem. 274: 34327-34336. PubMed ID: 10567409

Zembrzycki, A., et al. (2007). Genetic interplay between the transcription factors Sp8 and Emx2 in the patterning of the forebrain. Neural Devel. 2: 8. PubMed ID: 17470284


buttonhead: Biological Overview | Evolutionary Homologs | Regulation | Developmental Biology | Effects of Mutation

date revised: 25 November 2014

Home page: The Interactive Fly © 1997 Thomas B. Brody, Ph.D.

The Interactive Fly resides on the
Society for Developmental Biology's Web server.