Regulation of the Twist target gene tinman by modular cis-regulatory elements during early mesoderm development

Size: px
Start display at page:

Download "Regulation of the Twist target gene tinman by modular cis-regulatory elements during early mesoderm development"

Transcription

1 Development 124, (1997) Printed in Great Britain The Company of Biologists Limited 1997 DEV Regulation of the Twist target gene tinman by modular cis-regulatory elements during early mesoderm development Zhizhang Yin, Xiao-Lei Xu and Manfred Frasch* Brookdale Center for Developmental and Molecular Biology, Mount Sinai School of Medicine, Box 1126, One Gustave L. Levy Place, New York, NY 10029, USA *Author for correspondence ( SUMMARY The Drosophila tinman homeobox gene has a major role in early mesoderm patterning and determines the formation of visceral mesoderm, heart progenitors, specific somatic muscle precursors and glia-like mesodermal cells. These functions of tinman are reflected in its dynamic pattern of expression, which is characterized by initial widespread expression in the trunk mesoderm, then refinement to a broad dorsal mesodermal domain, and finally restricted expression in heart progenitors. Here we show that each of these phases of expression is driven by a discrete enhancer element, the first being active in the early mesoderm, the second in the dorsal mesoderm and the third in cardioblasts. We provide evidence that the early-active enhancer element is a direct target of twist, a gene encoding a basic helix-loop-helix (bhlh) protein, which is necessary for tinman activation. This 180 bp enhancer includes three E-box sequences which bind Twist protein in vitro and are essential for enhancer activity in vivo. Ectodermal misexpression of twist causes ectopic activation of this enhancer in ectodermal cells, indicating that twist is the only mesoderm-specific activator of early tinman expression. We further show that the 180 bp enhancer also includes negatively acting sequences. Binding of Evenskipped to these sequences appears to reduce twistdependent activation in a periodic fashion, thus producing a striped tinman pattern in the early mesoderm. In addition, these sequences prevent activation of tinman by twist in a defined portion of the head mesoderm that gives rise to hemocytes. We find that this repression requires the function of buttonhead, a head-patterning gene, and that buttonhead is necessary for normal activation of the hematopoietic differentiation gene serpentin the same area. Together, our results show that tinman is controlled by an array of discrete enhancer elements that are activated successively by differential genetic inputs, as well as by closely linked activator and repressor binding sites within an early-acting enhancer, which restrict twist activity to specific areas within the twist expression domain. Key words: Drosophila, tinman, mesoderm, patterning INTRODUCTION The molecular mechanisms that determine patterning and differentiation of the mesoderm are a major focus of current research. In Drosophila, genetic analysis has shown that the twistgene occupies a position at the top of a hierarchy of zygotically active genes that function in mesoderm development. twist encodes a basic helix-loop-helix (bhlh) protein that is expressed in the presumptive mesodermal cells on the ventral side of blastoderm embryos and, in the absence of twist function, no mesoderm is formed (Simpson, 1983; Thisse et al., 1988; Kosman et al., 1991; Leptin, 1991). In addition to its role in mesoderm formation, twist has a second function in stages after gastrulation, where it appears to be required for myogenesis of somatic muscles (Baylies and Bate, 1996). Because twist encodes a putative transcription factor, it is assumed to control mesoderm development through the activation of a large battery of target genes, either in the whole mesoderm or in specific portions of it. Candidates include the genes encoding the homeodomain proteins Tinman and Zfh-1 (Bodmer et al., 1990; Lai et al., 1991), the MADS-domain protein MEF-2 (Lilly et al., 1994; Nguyen et al., 1994; Taylor et al., 1995), the FGF-receptor Heartless (Shishido et al., 1993), the integrin PS2 (Leptin, 1991), the KH-domain protein Struthio (also named Who or How; Baehrecke, 1997; Zaffran et al., 1997; Lo and Frasch, 1997) and genes with as yet undefined functions (Casal and Leptin, 1996). The tinman gene has a key role in early mesoderm patterning and is essential for the formation of all dorsal mesodermal derivatives, including the heart, visceral musculature and dorsal somatic muscles (Azpiazu and Frasch, 1993; Bodmer, 1993). In addition, tinman is required for the formation of certain body wall muscles and glia-like cells that are derived from ventral areas of the mesoderm (Azpiazu and Frasch, 1993; Gorczyka et al., 1994). These functions of tin are reflected in its dynamic mesodermal expression pattern, which can be subdivided into three major phases. The early, twistdependent phase includes tin expression from late blastoderm until after gastrulation in all cells of the trunk mesoderm (Bodmer et al., 1990; Azpiazu and Frasch, 1993). During the

2 4972 Z. Yin, X.-L. Xu and M. Frasch second phase, upon internal spreading of the mesoderm, tin expression is restricted to a broad band of cells in the dorsal mesoderm. This restricted tin expression domain is controlled by inductive signals mediated through Dpp, which is secreted by dorsal ectodermal cells (Frasch, 1995). Most likely, it is during this phase when tin fulfills its major role in the determination of dorsal mesodermal derivatives. Finally, tinman expression is further restricted to heart precursors and may be involved in differentiation and diversification of cardioblasts and pericardial cells during this phase. In this study, we address the question of how mesodermautonomous regulation, involving activation by twist, and inductive inputs, which include those mediated by Dpp, are integrated molecularly at the promoter level of the tinman gene. Our functional dissection of regulatory regions from the tin locus reveals that tin is controlled by distinct enhancer modules, one of which is responsible for its early and broad mesodermal expression, another for its Dpp-mediated dorsal mesodermal activation, and a third for cardioblast-specific expression. We further show that the early-acting module is composed of positively and negatively acting subsequences that functionally antagonize each other. We propose that direct binding of Twist to its target sites within this element can activate tin expression in the whole mesoderm, but Twist activity is abrogated in defined mesodermal areas by the binding of negative regulators to adjacent sites. As a result, tin is activated in the trunk mesoderm which gives rise to visceral and somatic muscles, heart and glial-like cells, but not in areas of the head mesoderm that give rise to hemocytes. The exclusion of tinman from the prospective blood mesoderm is, at least in part, due to the activity of the head-patterning gene buttonhead. Moreover, negative inputs triggered by the binding of the pair-rule gene product Eve result in periodically modulated levels of tin along the anteroposterior embryo axis. MATERIALS AND METHODS Isolation of genomic clones and construction of P- transformation plasmids The tinman gene from Drosophila virilis was isolated from a genomic library obtained from W. Hanna-Rose through J.D. Licht (Hanna-Rose et al., 1997). The screen was performed with a tin cdna probe using hybridization conditions as in McGinnis et al. (1984). Washes were done three times 30 minutes at 50 C with 1 SSC, 0.5% SDS. Upstream fragments of tin (D.mel.) were cloned into P-transformation vectors with native orientations relative to the basal promoters. +1 bp of our map refers to the first base of the longest tin cdna available (Bodmer et al., 1990). Following upstream constructs were made and tested in vivo: tin-3436/tin1: A fragment from 1716 bp to +287 bp generated by PCR with BamHI-site containing primers and cloned into the BamHI site of pcasper AUG βgal (Thummel et al., 1988). tin-3668/tin1 contains a fragment from 984 bp to +287 bp that was generated by PCR and cloned as above. tin-pvu/tin1 contains a fragment from the PvuII site at 384 bp to +287 bp in the EcoRI/BamHI sites of the same vector. tin-3436/3437 was generated from a PCR fragment ( 1716 bp to 39 bp), with primers containing BamHI (5 ) and XhoI (3 ) sites, that was cloned into XhoI/BamHI of pcasper hs43 βgal (Thummel et al., 1988). tin-3436/3843 was generated similarly from a PCR fragment containing sequences from 1716 bp to 269 bp. tin-1.7 kb+int was generated from tin- 3436/3437 by inserting intron 1 into the NotI site. Intron fragments from D. melanogaster (Dm) or D. virilis (Dv) were cloned upstream of the basal hsp70 promoter of pcasper hs43 βgal. The final constructs were: tin-a/lac (Dm), a 500 bp HindIII/XhoI fragment cloned into NotI/XhoI; tin-b-374 bp/lac (Dm), a 374 bp fragment cloned into NotI/XhoI, previously generated by PCR from a pks+ clone containing the 2.5 kb HindIII fragment of tin, using the M13 primer (5 ) and ATGCGGCCGCCTGCGGGAAA (3 ) and cut with NotI and internally with XhoI; tin-b-180 bp/lac (Dm), a PCR fragment generated with the primer pair AGGAATTCGTCAACAT- GTGT (5 ) and AGCTCGAGTCTCGTATATGG (3 ) and cloned into EcoRI/XhoI; tin-b (Dv), a 390 bp PCR fragment generated with the primer pair AGGAATTCGACAAATCATCG (5 ) and AGGGATC- CTCGATTAGTTGGC (3 ) and cloned into EcoRI/BamHI. E1,2/lac and E3/lac were made from double stranded oligonucleotides. E1,2 was flanked by EcoRI (5 ) and BamHI(3 ) sites, E3 by BamHI (5 ) and XhoI (3 ) sites and cloned into the corresponding sites of pcasper hs43 β-gal. For E1,2-E3/lac, E3 was cloned into the BamHI/XhoI sites of E1,2/lac. For E1,2-R-E3/lac and E1,2-2R-E3/lac, one or two PCRderived fragments (using the primer BamHI site from the oligonucleotide GCGGATCCAGGGGCGTCCTT at 5 and the endogeneous BamHI site at 3 ) were added to E1,2-E3/lac, respectively. The final versions of constructs containing downstream fragments from the tin genes from D. mel. (Dm) and D.vir. (Dv) were made as follows: tin-c (Dm), a 300 bp PCR fragment generated with the primer pair AGGCGGCCGCCATGAACAGCTT (5 ) and AGGGATCCGAGGCAGGGAAA (3 ) and cloned into NotI/BamHI; tin-c (Dv), a 354 bp PCR fragment generated with the primer pair AGGCGGCCGCTCAGGCACGGATC (5 ) and AGCTCGAG- GCAAAACATTTTACAG (3 ) and cloned into NotI/XhoI; tin-d (Dm), a 346 bp PCR fragment generated with the primer pair GAGAATTCATGTCAAGTGGCACTA (5 ) and ACCTCGAG- GTGGGAGGCTCGCAGCT (3 ), cloned into EcoRI/XhoI; tin-d (Dv), a 2.3 kb SalI fragment cloned into XhoI. In vitro mutagenesis was performed with the Transformer TM mutagenesis kit from Clontech. P-element constructs were injected into embryos from yw flies together with 2-3 as the transposase source. Between two and five lines from each constructs were analyzed for expression. Embryo stainings In situ hybridizations were performed as described in Lo and Frasch (1997), and antibody stainings or combined antibody/in situ hybridization stainings as in Azpiazu et al. (1996). Tinman antibodies were raised in a rabbit against bacterially expressed His(6)-tagged proteins that were purified on a Ni-column (Quiagen). The expression construct contained a 1.1 kb BamHI (generated at the ATG of the first Met) to HindIII fragment in pqe11 (Qiagen). Peroxidase antibodies were obtained from L. Fessler (UCLA) and β-gal antibodies were from Sigma (mouse polyclonal) and Cappel (rabbit). Drosophila stocks Following mutant alleles were used: btd XG, Dfd 21, ems 7099, eve 1.27, otd D87. lacz-balancer chromosomes were used to identify homozygous mutant embryos, except for eve, where homozygous embryos were identified using Eve-antibodies. For ectopic expression of twist, we used a UAS-twist line (Baylies and Bate, 1996) and an en-gal4 line obtained from M. Baylies. DNaseI footprinting assays DNaseI protection assays were performed essentially as described by Heberlein et al. (1985) with following modifications: The reaction mixtures (50 µl) contained 110 mm KCl, 47.5 mm Hepes (ph 7.9), mm MgCl 2, 1 mm DTT, 17% glycerol, 0.05% NP-40, 1 µg poly[d(i-c)], and 5 ng of 3 end-labeled probe. Upon addition of purified GST/Twist fusion proteins (Ip et al., 1992a) or Eve proteins (gift from M. Biggin) and incubation for 60 minutes on ice, 50 µl of 10 mm MgCl 2/5 mm CaCl 2 were added, followed by 1 µl DNaseI (Boehringer) to a final concentration of 0.4 µg/ml. After 2.5 minutes

3 Regulation of tinman during Drosophila mesoderm development 4973 digestion on ice, the reaction was stopped by adding 90 µl of 1% SDS, 20 mm EDTA, 200 mm NaCl, 250 µg/ml yeast trna, extracted twice with phenol/chloroform and ethanol precipitated. Electrophoretic separation was done in 8% polyacrylamide/7.5 M urea gels. RESULTS Distinct cis-elements drive each aspect of endogenous tinman expression As an initial step in the identification of enhancer sequences of the tinman gene, we isolated the tinman homolog and its flanking sequences from a distantly related species, Drosophila virilis, with the assumption that essential cis-regulatory sequences would be evolutionarily conserved. The tinman genes from D. melanogaster and D. virilis share a high degree of sequence similarity in their open reading frames, are organized in three exons with similar lengths and display almost identical patterns of expression in the two species (Fig. 1; Z. Y. and M. F., unpublished data). As illustrated in Fig. 2, the temporal and spatial expression of D. melanogaster tin protein expression can be subdivided into four major aspects. During invagination and early migration of the mesoderm, tin is expressed in all mesodermal cells, except for a small area in the head mesoderm that is negative (Fig. 2A,J). This early expression of tin depends on the function of the twist gene (Bodmer et al., 1990). tin expression then becomes restricted to the dorsal portion of the mesoderm, and both tin mrna and protein disappear from ventral cells during elongated germ band stages (Fig. 2D). It has been shown that restricted tin expression in the dorsal mesoderm is triggered by Dppmediated induction events (Frasch, 1995). In subsequent stages of embryogenesis, tin expression is limited to cells of the dorsal vessel. As shown in Fig. 2G, tin products are detected in generally four out of six pairs of cardioblasts per segment and in pericardial cells during dorsal closure (Jagla et al., 1997). The only non-mesodermal domain of tin expression is located at the anterior tip of the embryo, with highest expression levels in cells that are fated to become part of the pharynx and esophagus (Fig. 2A,D,J). The region containing essential tin enhancer elements was defined by a genomic construct, encompassing sequences from 6.2 kb to +4.6 kb with respect to the presumed transcription start site, that rescued the lethality of null mutations of tin (Azpiazu and Frasch, 1993). Since the closest gene upstream of tin, mod(mdg4), starts at 1.6 kb (Z. Y. and M. F., unpublished data), we tested fragments from the entire region between 1.7 kb and +4.8 kb for enhancer activity. Genomic restriction fragments, or PCR mod (mdg4) C fragments generated with primer pairs spanning stretches of sequences that were conserved between the two Drosophila species, were cloned upstream of a basal promoter and a β- galactosidase (β-gal) reporter gene and transformed into the germline (see Materials and Methods). Fragments showing enhancer activity in transgenic embryos were further dissected and tested similarly in additional rounds of analysis. From these studies, we identified four distinct enhancer elements from D. melanogaster, with lengths varying between 180 bp and ~500 bp, that activated reporter gene expression in patterns resembling specific aspects of the tinman pattern (Fig. 1). Surprisingly, all of these elements were located at positions downstream of the transcription start site. Element B (180 bp), located in the first intron, activated β-gal expression in the whole trunk mesoderm during gastrulation and germ-band elongation (Fig. 2B). An element from D. virilis located at a similar position had an identical temporal and spatial activity when tested in D. melanogaster embryos (Figs 1, 2C). The intensity of reporter gene expression with the B elements from both species was modulated in a pair-rule fashion along the anteroposterior axis (see below). A second enhancer element, D (~350 bp), that was located ~2 kb downstream of the 3 end of tin activated reporter gene expression specifically in cells in the dorsal portion of the mesoderm (Fig. 2E). Element D was active between stage 11 and early stage 12 of embryogenesis, corresponding to the period when maintenance of tinman expression in the dorsal mesoderm requires dpp. An analogous element was identified downstream of the D. virilis tinman gene and showed identical activities in transformed D. melanogaster embryos (Figs 1, 2F). A third element from D. melanogaster, C (300 bp; Fig. 1), was active in the dorsal vessel. This element activated β-gal expression from stage 12 on in four out of six cardioblasts per hemisegment, similar to the cardioblast expression of the endogenous tin gene (Fig. tinman 5' 3' A B C D R C H Xh B H H R R B Xb C 5' 3' B C D Pv H B R H Xh RV RV SB P S 1kb D. mel. D. vir. Fig. 1. Genomic organization and location of enhancer elements of the tinman genes from Drosophila melanogaster and Drosophila virilis. Exons are shown as black and introns as white boxes. The putative location of the transcription start site of the D. melanogaster tin gene was based on the position of the first nucleotide of the longest cdna (Bodmer et al., 1990). The approximate positions of the 5 end, exon boundaries and 3 end of the D. virilis genes were deduced from sequence comparisons with the D. melanogaster gene. Enhancer elements A to D, as described in the text, are shown as patterned boxes. Abbreviations: A, anterior head element; B; broad mesodermal element, C, cardioblast element; D, dorsal mesodermal (dpp-response) element; B, BamHI; C, ClaI; H, HindIII; P, PstI; Pv, PvuII; R, EcoRI; Xb, XbaI; Xh, XhoI; mod(mdg4), modifier of midget4 gene.

4 4974 Z. Yin, X.-L. Xu and M. Frasch 2H). A cardioblast element with similar activity was again found at a corresponding position downstream of the D. virilis tin gene (Figs 1, 2I). Elements C from both species did not exhibit any expression in the pericardial cells of the dorsal vessel. However, further dissection of the D element from D. mel. showed it to contain sequences that are indeed able to drive expression in pericardial cells, but this activity is obscured by the general dorsal expression with the intact D element (data not shown). Finally, we located an element (A, ~500 bp; Fig. 1) in the 5 portion of the first intron of the D. mel. tin gene that displayed activity in the anterior tip of the head (Fig. 2K). After invagination of the stomodeum, the bulk of the cells that are marked with β-gal from this element form the roof of the pharynx (Fig. 2L). In summary, the four enhancer elements from the D. melanogaster tin gene reflect the four major aspects of endogenous tin expression. At least three of these elements are conserved between D.mel and D. vir. with respect to sequences, relative positions, as well as spatial and temporal activities. Importantly, we were unable to find any enhancer activity in the 5 flanking and 5 untranslated regions of tin, in spite of testing a variety of constructs both with homologous tin and heterologous hsp70 promoters. Neither did we detect any enhancement of expression when 5 flanking sequences were combined with the first intron that contained elements A and B (see Materials and Methods). These findings are in agreement with the virtually normal expression of tin in embryos that are homozygous for deletions of 5 flanking regions (tin and tin , which delete sequences between 1546 bp and 162 bp or 23 bp, respectively; [Azpiazu and Frasch, 1993]; data not shown). Three conserved Twist-binding sites in the tin B enhancer are functional in vivo The early expression of tinman in the whole mesoderm has been shown to depend on twist (Bodmer et al., 1990). Since the tin B enhancer elements mimicked this pattern of expression, we focused on these elements and tested whether they are direct targets for the Twist protein. Sequence comparisons between the B enhancers from D. melanogaster and Fig. 2. Comparisons of tinman protein expression and the reporter gene expression driven by tinman enhancer elements from D. melanogaster and D. virilis. (A,D,G,J) tinman antibody stainings of D. melanogaster embryos; (B,E,H,K,L) β-gal antibody stainings of D. melanogaster embryos transformed with D. mel.-derived enhancer constructs and (C,F,I) similar stainings of D. melanogaster embryos transformed with D. vir.-derived enhancer constructs. Anterior is to the left and ventral is down. (A) In gastrulating embryos, tinman expression is observed in all cells of the trunk mesoderm and in a cap at the dorsal tip of the head. (B,C) In embryos of similar stages, the tin B enhancers from D. mel. and D. vir. drive β-gal expression in all cells of the trunk mesoderm. (D) At stage 11, tin expressing is restricted to an undulating band of dorsal mesodermal cells (d.ms.) and is absent in the ventral mesoderm (v.ms.). Head expression is enhanced. (E,F) The tin D enhancers from D. mel. and D. vir. are active only during stage 11 and early stage 12 in dorsal portions of the mesoderm. (G) Dorsal view of a stage 14 embryo, showing tin expression in the majority of cardioblasts (cbs) and in pericardial cells (pcs). (H,I) β-gal expression driven by the tin C enhancers from D. mel. and D. vir. in about four cardioblasts per hemisegment. (J) High magnification view of tin expression in the anterior portion of a stage 8 embryo, showing expression in the anterior head cap (arrowheads) and absence of expression in a region of the head mesoderm (bracket). (K) β-gal expression driven by the tin A enhancer from D. mel. in a head cap (arrowheads). (L) Embryo as in K at stage 12, showing perduring β-gal proteins in pharyngeal structures. The corresponding portion of the intron from D. vir. tin was not tested for enhancer activity.

5 Regulation of tinman during Drosophila mesoderm development 4975 D. virilis showed strong similarities within multiple stretches of ~20 bp to ~50 bp length (Fig. 3A), which were candidates for conserved regulatory sequences. Importantly, the sequences corresponding to the minimal tin B enhancer (180 bp) from D. mel. include three conserved E-box sequences that could potentially bind the bhlh protein Twist. These E-boxes contain the sequences CATGTG or CATATG in both species, and while E-boxes 1 and E-boxes 2 are arranged in tandem, E- boxes 3 are located ~120 bp to ~150 bp further downstream (Fig. 3A). DNaseI footprinting assays with bacterially purified Twist fusion proteins showed clear protection in the vicinity of these sequences, demonstrating that Twist is able to bind to all three E-boxes in vitro (Fig. 4A,C). Binding specificity was confirmed by using fragments in which the E-boxes have been mutagenized in vitro. Upon altering the sequences of E-box 1 and 2 from D. mel. to TATGTA (E1,2mut I), a strong reduction of Twist-binding was observed, although there was still some protection of sequences containing the modified E-box 1 at the highest concentration of Twist that was tested (Fig. 4B). The modified sequence TATATA at the position of E-box 3 (E3mut I) lost its ability to bind Twist under these conditions (Fig. 4D). To test whether the in vitro Twist-binding sites are required for the activity of the tin B enhancer in vivo, we generated a series of reporter constructs with modified versions of B enhancer sequences and analyzed their activity in transgenic embryos. A B enhancer construct (374 bp; Fig. 3A) in which E-box 3 had been mutated to TATATA, showed reduced reporter gene expression (Fig. 5A; see Fig. 2B for comparison). Similar results were obtained with B enhancer constructs containing an intact E-box 3 but mutant E-box 1 and E-box 2 sequences (TATGTA; Fig. 5B). Most strikingly, the periodic modulation of β-gal expression, which was evident with the intact B element, became much more pronounced with the constructs containing one or two mutant E-box sequences. When all three E-boxes were mutated, enhancer activity was drastically reduced and only a few residual mesodermal cells expressed β-gal (Fig. 5C). The weak, residual activity may reflect the residual binding affinity of Twist seen in vitro with these modified sequences (Fig. 4B). Therefore, we tested a reporter construct in which the sequences of all three E-boxes were further modified (E1,2mut II and E3mut II; Fig. 3B). As predicted, this mutant version of B exhibited a complete lack of mesodermal enhancer activity (Fig. 5D). We next tested whether the conserved sequence blocks spanning Twist-binding sites are sufficient to activate reporter gene expression in the mesoderm. When the sequences E1,2 and E3 (Fig. 3B) were linked with each other, strong Fig. 3. Sequence comparison and molecular dissection of tin B enhancer elements from D. melanogaster and D. virilis. A. BESTFIT alignment 374 bp tin B enhancer from D. mel. with the 425 bp tin B enhancer from D. vir. The sequences corresponding to the 180 bp tin B enhancer from D. mel. are boxed. E-box sequences and consensus binding sites for homeodomain proteins (and more specifically, Evenskipped) are shaded in black. It may be significant that, in five of the six E-boxes, the 3 G is folllowed by a T (E-box 3 would be in reverse orientation). B. Shown are the normal sequences of the 29 bp E1,2 and E3 elements, as well as the 52 bp R element, that were used individually or in combination to test for enhancer activities. The modified sequences that were obtained in two steps of in vitro mutagenesis and tested in the context of the 374 bp tin B element are shown below the wild-type sequences.

6 4976 Z. Yin, X.-L. Xu and M. Frasch expression was observed in the whole mesoderm (Fig. 5E). Thus, a 58 bp element containing three putative Twist-binding sites was able to activate β-gal in all cells of the embryo that contain Twist. Even a 29 bp fragment, E1,2, containing the first two E-boxes was sufficient to drive reporter gene expression in this pattern, albeit at reduced levels (Fig. 3B; Fig. 5F). By contrast, the E3 sequence was inactive in the trunk mesoderm and showed expression only in small regions of the rostral and caudal mesoderm (Fig. 5G and unpublished data). In order to provide stronger support for the notion that Twist binds and transactivates gene expression through the three E-boxes in the B enhancer, we mis-expressed Twist with the binary GAL4 system (Brand and Perrimon, 1993) under the control of engrailed enhancers in embryos carrying the reporter construct with the combined E1,2 and E3 elements. As shown in Fig. 5H, ectopic expression of Twist in ectodermal stripes caused striped ectopic expression of β-gal in the ectoderm. Taken together, these experiments provide strong evidence that the three E-box sequences are in vivo targets of Twist and, in the context of surrounding sequences of ~50 bp, are responsible for the early activation of tinman in the trunk mesoderm. Periodic modulation of tin B enhancer activity depends on eve and an Eve-binding element During its earliest phase of expression, just prior to gastrulation, ventral tin expression is modulated in a pair-rule fashion along the anteroposterior embryo axis, with highest levels of expression seen in six mesodermal stripes (Fig. 6A). This feature may be reflected in the periodic expression that was observed with tin B enhancer elements, which was particularly obvious upon introducing mutations in individual E-box sequences that diminished Twist binding (Figs 2B,C, 5A,B). To determine the segmental register of reporter gene expression in embryos transformed with these constructs, we performed double stainings for β-gal and pair-rule gene products. As shown in Fig. 6B, the pattern of β-gal directed by the E1,2 mut I derivative of the B enhancer is complementary to that of the pair-rule gene even-skipped (Macdonald et al., 1986; Frasch et al., 1987). Peak levels of β-gal were observed in areas lacking Eve protein. Since this β-gal pattern could indicate repression of enhancer activity by Eve, we tested its activity in eve mutant embryos. Indeed, in the absence of eve activity, the same enhancer is able to drive strong expression throughout the trunk mesoderm without any periodic interruptions (Fig. 6C). A B C D E Fig. 4. DNA-binding of Twist and Even-skipped to tin B enhancer sequences. (A) DNase I footprinting with Twist using the 5 portion of the 374 bp tin B element (B1). Lane 1 shows a G + A sequence ladder, lanes 2 to 5 protection experiments with 200, 400, 800 and 1600 ng of GST- Twi fusion proteins, respectively, and lane 6 a control experiment with the GST fusion moiety only. The protected sequences, including E-box 1 and 2 (shadowed black), are shown on the left. (B) Similar footprinting experiment as in A, but with tin B sequences containing mutated E- boxes 1 and 2 (left column, shadowed sequences). Note that two times higher GST-Twi protein concentrations were used in each lane as compared to A. (C) Twist DNA-binding experiment as in A, using the 3 portion of the 374 bp tin B enhancer (B2) which includes E-box 3 (shadowed black). (D) Twist DNA-binding experiment as in B, using a tin B element with in vitro mutated E-box 3 (shadowed grey). (E) DNase I footprinting with Even-skipped (Eve) assaying sequences of the R portion of the 374 bp tin B element (BR). 50 ng, 100 ng, 200 ng, 400 ng, 800 ng and 1600 ng of Eve protein were used for lanes 2 to 7, respectively. The protected sequences, including a sequence matching a consensus binding site for Eve (shadowed black), are shown in the left column.

7 Regulation of tinman during Drosophila mesoderm development 4977 Fig. 5. Functional analysis of the in vitro Twistbinding sites from the tin B enhancer in vivo. (A) A 374 bp tin B enhancer element with E-box 3 mutated to TATATA shows reduced and periodically modulated levels of reporter gene expression along the anterioposterior axis. Weak anterior expression indicating the presence of a second head element in the 374 bp B enhancer is also evident. (B) A 374 bp tin B enhancer element with E-boxes 1 and 2 mutated to TATGTA, shows similar periodic reductions of expression as with the mutant element from A. (C) Mutation of all three E-box sequences (as in A and B) within the 374 bp element leads to a dramatic loss of enhancer activity. Arrows point to mesodermal cells with residual, weak expression. (D) Further modifications of E-box sequences within the 374 bp element (see Fig. 3B) completely abolish its enhancer activity. (E) A combination of the elements E1,2 and E3 with native orientations (see Fig. 3B) exhibits strong mesodermal enhancer activity. Ectopic expression in the head mesoderm is marked with arrowheads. (F) The 29 bp element E1,2 is sufficient to drive mesodermal gene expression. Note that the expression in E and F includes the head mesoderm and anterior and posterior endoderm primordia. (G) The 29 bp element E3 is not sufficient to drive expression in the trunk mesoderm, but expresses β-gal ectopically in a portion of the head mesoderm. (H) Ectopic Twist (expressed under the control of engrailed enhancers in ectodermal stripes) is sufficient to activate ectodermal β-gal expression in embryos with a reporter construct containing in vitro Twist-binding sequences only (E1,2-E3, as in E). Among the pair-rule mutant tested, this outcome was unique to eve. In mutants for fushi tarazu (ftz), hairy (h) and runt (run), we observed interruptions in the β-gal pattern along the anteroposterior axis, however the pattern of the interruptions was consistent with the previously reported alterations in the eve pattern in these mutants (data not shown; Frasch and Levine, 1987). Additional data indicated that Eve may repress B enhancer activity (and tin) through direct binding to sequences in this element. The conserved sequence block between E-box 2 and E-box 3 contained a sequence matching homeodomain binding sites, which was particularly close to a consensus binding site previously determined for Eve (Fig. 3A; Hoey et al., 1988). In vitro DNA-binding experiments confirmed that this sequence is able to bind Eve, both in the case of D. mel. (Fig. 4E) and D. vir. (data not shown). Enhancer constructs that lacked the R element containing this Eve-binding site showed uniform expression throughout the mesoderm (Fig. 5E, F). This pattern is similar to that of B enhancers with weakened Twist sites in eve mutant backgrounds (Fig. 6C). When the 52 bp R element was added to the E1,2 and E3 elements, with a configuration similar to that of the native B enhancer, periodic repression was restored (Fig. 6D). Moreover, addition of two R elements in tandem resulted in much stronger periodic repression and yielded a pattern of clean stripes of β-gal in the mesoderm. These results indicate that Eve represses B enhancer activity by direct binding to recognition sequences in the B enhancer and thereby reduces its activation by Twist in a periodic fashion. Regulation of tinman in trunk versus head mesoderm tin is expressed in the whole trunk mesoderm of late blastoderm and gastrulating embryos, but expression is excluded from portions of the mesoderm in the head (Figs 2A,J, 6A). This is in contrast to the expression of its upstream regulator twist, which is seen throughout the length of the mesoderm (Thisse et al., 1988), and indicates the presence of negative regulators of tin in the head mesoderm. Lack of head mesoderm expression was also observed with 180 bp tin B enhancer constructs and their derivatives, which suggested that, in addition to positively acting Twi-binding sites, there are negative elements that prevent activation by Twist in the anterior mesoderm. This possibility was confirmed with the 58 bp and 29 bp constructs, containing the Twist-binding sites plus immediately adjacent sequences, which lacked repression in the head mesoderm and showed a pattern identical to twist (Fig. 5E,F). Since the major difference between these latter constructs and the tin B enhancer constructs was the absence of the R element, it was likely that sequences within the R elements mediated the head repression. Indeed, when the R element was added to the E1,2 and E3 elements, repression in the head mesoderm was restored (Fig. 6D,E). These results show that the R elements mediate repression in the head

8 4978 Z. Yin, X.-L. Xu and M. Frasch mesoderm, in addition to their function in Eve binding and periodic repression of Twist activation. Since head repression was maintained in eve mutant embryos (Fig. 6C), additional negative regulators must act on the R element. The absence of tin in the head mesoderm may be functionally important, since these cells have different developmental fates (Tepass et al., 1994) and express serpent (srp), a gene of the GATA family. serpent is required for the normal differentiation of these cells into hemocytes (Rehorn et al., 1996). srp is expressed in the cells of the head mesoderm that lack tin expression (Fig. 7A,C, arrowheads; note that srp has a second, more anterior domain that overlaps with endodermal tin expression). To identify upstream genes involved in the regulation of gene expression in the head mesoderm, we examined tin and srp expression in embryos mutant for early head-patterning genes, including buttonhead (btd; Wimmer et al., 1991), Deformed (Dfd; Merrill et al., 1987), empty spiracles (ems; Dalton et al., 1989), orthodenticle (otd; Cohen and Jürgens, 1990; Finkelstein and Perrimon, 1990) and ems,otd double mutants. Interestingly, in gastrulating embryos that are mutant for btd, tin expression was expanded into the head mesoderm, whereas the mesodermal domain of serpent was absent (Fig. 7B). During blastoderm stages, we observed severely reduced, transient expression of srp in the head mesoderm of btd mutants (Fig. 7D). None of the other mutations tested produced any obvious alterations of the tin and srp domains, indicating a major role of btd in the patterning of the head mesoderm. Similar to tin, expression of β-gal from B enhancer constructs expanded into the head mesoderm in the absence of btd function (Fig. 7E,F). Taken together, these results suggest that btd acts through the R element to prevent tin activation by twist in the head mesoderm. In addition, btd is required for normal activation of srp in the same area. Since both these activities of btd may be essential for normal blood cell development, we examined the distribution of hemocytes in btd mutant embryos. Normally, a large number of hemocytes, which stain for peroxidasin, are scattered throughout the body cavity of late stage embryos (Fig. 7G; Nelson et al., 1994; Tepass et al., 1994). By contrast, in btd mutant embryos, the number of hemocytes was reduced and virtually all of the residual cells remained near their place of origin in the embryonic head (Fig.7H). DISCUSSION Fig. 6. Periodic abrogation of B enhancer activity by eve and the R element. (A) tin mrna expression in a wild-type embryo at blastoderm, showing periodic reductions of tin levels along the anteroposterior axis. (B) Stage 8 embryo carrying the E1,2mutI/lac derivative of the tin B enhancer and stained for β-gal (brown) and Eve (black). β-gal staining is observed between the Eve stripes, which have split up into a strong and a weak component at this stage (indicated by solid parts of bracket). The absence of β-gal expression in areas between strong and weak eve stripes could be explained by the repressing activities of more broadly distributed eve products prior to this stage (indicated by entire bracket). (C) Stage 8 eve mutant embryo (eve 1.27 ) carrying the same transgene and stained as embryo in B. Absence of eve results in a uniform mesodermal activity of this enhancer. (D) Stage 9 embryo carrying an E1,2-R- E3/lac transgene. The presence of the R element leads to periodic reductions of mesodermal β-gal expression (compare Fig. 5E). (E) Stage 9 embryo carrying an E1,2-2R-E3/lac transgene. The presence of two R elements leads to wide periodic gaps of β-gal expression and, thus, a pattern of six mesodermal stripes. The mesoderm-patterning gene tinman has three major phases of mesodermal expression, the first occurring in the whole trunk mesoderm, the second in broad dorsal subdomains of the trunk mesoderm and the third in heart progenitors at the dorsal mesodermal crest. This refinement from an initially broad expression domain to progressively smaller areas could be explained by several different molecular mechanisms, including selective mrna stabilization or successive waves of transcriptional activation. Our analysis of regulatory regions from the tin gene demonstrates that each of these aspects of tin expression is indeed controlled by a separate enhancer module and, thus, reflects a distinct transcriptional activation event. The modular character of these enhancers was also evident in experiments with larger elements containing two different enhancers (e.g. A+B, C+D; data not shown), which produced additive patterns of reporter gene expression. Candidates for upstream components that trigger tin activation through some of these elements have been identified in previous genetic experiments (Bodmer et al., 1990; Frasch, 1995). Our present study shows that twist acts through the early (B) element (Fig. 8). dpp is likely to act

9 Regulation of tinman during Drosophila mesoderm development 4979 through the dorsally active (D) element, a notion reinforced by our observation that this element is inactive in dpp mutant embryos (data not shown). The upstream activators acting on the heart element (element C) are not yet known. Although tin expression in the heart progenitors, and the formation of heart progenitors proper, have been shown to require wingless (Wu et al., 1995), the critical period of wg requirement occurs earlier (at stage 10-11) than the activation of this enhancer, which does not begin until stage 12. Furthermore, element C does not contain any sequences closely matching those of binding sites for dtcf/pangolin, which is the Drosophila homolog of LEF-1 and appears to mediate responses to the wingless signal (Brunner et al., 1997; van de Wetering et al., 1997; Z. Y. and M. F., unpublished results). Therefore we favor the idea that wingless is required early, together with tinman during its expression in a broad, dorsal domain, to determine heart progenitor identities. In a subsequent step, these cells activate tin through as yet undefined mechanisms that involve element C in the case of the cardioblasts. In order to obtain further insights into the molecular mechanisms of mesoderm patterning, Dppmediated induction and heart differentiation, it will be of major importance to functionally dissect the C and D elements to a similar extent to that done in this study with the B element, and to identify the corresponding binding factors. Since tinmanrelated genes are expressed in the developing heart of vertebrate embryos and their expression depends on bone morphogenetic proteins (BMPs) that are homologous to Dpp (for a review, see Harvey, 1996; Schultheiss et al., 1997), it is conceivable that some of the molecular mechanisms involving the C and D elements are conserved. This possibility can now tested through sequence comparisons and reporter gene assays in heterologous systems. In addition to the regulatory elements that are active in the mesoderm, our analysis has also revealed the presence of distinct elements driving tin expression in cells at the anterior tip of the embryonic head. Tagging these cells with β-gal shows that they give mainly rise to pharynx and anterior endoderm, consistent with previous fate map studies for this area (Technau and Campos-Ortega, 1985). Although this aspect of tin expression has previously received less attention, it is interesting to note that the pharynx and anterior endoderm are also prominent sites of expression of tinmanrelated genes from vertebrates (Lints et al., 1993; Tonissen et al., 1994; Evans et al., 1995; Lee et al., 1996; Brand et al., 1997). Thus, some of the upstream regulators acting through element A are likely to be evolutionarily conserved as well. In Drosophila, candidates include Bicoid and D-gsc, the Zn-finger protein Tailless, and the Torsodependent Ras pathway (Berleth et al., 1988; Hahn and Jäckle, 1996; Goriely et al., 1996; Sprenger et al., 1989). The focus of the present study is the regulation of tin expression in the early trunk mesoderm. Early tin expression has several important biological functions. Notably, it appears to be required for the specification of cell fates in the ventral portion of the mesoderm, such as those of distinct muscle precursors and of mesodermally derived glia-like cells at the ventral midline (Azpiazu and Frasch, 1993; Gorczyka et al., Fig. 7. The role of btd in tin and srp regulation in the head mesoderm and in hemocyte development. (A) Stage 9 wild-type embryo stained for Tin protein (brown) and srp mrna (purple). Tin is excluded from a portion of the head mesoderm (between arrowheads) that expresses srp. A few cells at the border between the Tin and srp domains may express both genes. Cells in a second domain of srp, which constitute prospective endoderm of the anterior midgut (anterior to the left arrowhead), co-express Tin. (B) In a stage 9 btd mutant embryo, Tin expression is expanded into the head mesoderm (between arrowheads) and mesodermal srp expression is absent. The endodermal domains of expression are unaltered. (C) srp mrna expression in a wild-type embryo at blastoderm stage. The mesodermal srp domain giving rise to hemocytes is marked by arrowheads. (D) srp expression in a btd mutant embryo at blastoderm. Mesodermal srp expression (arrowheads) is severely reduced. (E) The 374 bp tin B enhancer is not active in the head mesoderm (arrowheads). (F) In a btd mutant embryo, the activity of the 374 bp tin B enhancer is derepressed in the head mesoderm (arrowheads). (G) Stage 16 wild-type embryo stained with antibodies against peroxidasin to detect hemocytes. Fat body and ring gland are also stained. (H) Stage 15 btd mutant embryo, stained as in G. There is a reduced number of hemocytes and all of them remain in the embryonic head region. Abbreviations: fb, fat body; hc, hemocytes; rg, ring gland.

10 4980 Z. Yin, X.-L. Xu and M. Frasch 5' 3' HEAD (pharynx, ant.endoderm)? twi wg btd eve A B C D TRUNK MESODERM CARDIOBLASTS dpp DORSAL MESODERM Fig. 8. Summary of tinman enhancer elements and their regulatory inputs. 1994). It is further required in an autoregulatory fashion to allow tin expression at high levels in the second, dppdependent phase in the dorsal mesoderm (X.-L. X., Z. Y. and M. F., unpublished data). Our experiments demonstrate that early mesodermal tin expression is driven by a distinct regulatory module, element B, that is composed of closely linked positively and negatively acting sequences. The basic helixloop-helix protein Twist appears to be the principal activator of this element, which has three E-box-containing binding sites. Two of these are arranged in tandem, with the E-boxes being only seven base pairs apart. Strikingly, a minimal element, E1,2, containing these two E-boxes and only 17 base pairs of additional sequences is able to activate transcription in the mesoderm, suggesting that activation by Twist does not require many additional DNA-binding factors. Moreover, ectodermal expression of the reporter gene upon mis-expression of Twist suggests that Twist is the only mesodermally restricted factor that is required to activate tin through this module, although generally expressed proteins could act as co-factors. For example, given that bhlh proteins tend to form heterodimers (Murre et al., 1989b), it is possible that, in vivo, Twist binds as a heterodimer with ubiquitously expressed bhlh proteins to each of these E-box sequences. A candidate for a Twist partner could be the the ubiquitously expressed bhlh protein Daughterless (Da; Cronmiller et al., 1988; Murre et al., 1989a). However, we found that the early expression of tin in embryos lacking both the maternal and zygotic functions of da is normal and, thus, Da does not appear to be an essential Twist partner (M. F., unpublished data). Although Twist could bind as a homodimer, it probably requires one or more co-factors that bind to immediately flanking sequences for transcriptional activation. This is suggested by the inability of other fragments from the tin locus to activate early mesodermal gene expression, even though they contain E-box clusters that bind Twist in vitro with similar affinities (Lee et al., 1997; Z. Y. and M. F., unpublished data). Since vertebrate homologs of tinman are not broadly expressed in the early mesoderm, the Twist-dependent activation of tin-related genes may not be conserved between insects and vertebrates. However, Twist homologs are expressed in the early mesoderm of vertebrate embryos, which suggests that at least some molecular aspects of Twist function have been conserved. Previous studies have focused on inhibitory functions of mouse Twist during myogenesis, which appear to involve competition for binding of myogenic factors, titration of E proteins, and the formation of inhibitory complexes with promoter sequences (Spicer et al., 1996; Hebrok et al., 1997). It will be important to determine whether mouse Twist has the ability to also activate target genes, and our identification of an activating Twist-response element in Drosophila may provide a means to address this question. The 52 bp R element, which is interspersed between the E-boxes, counteracts the activating functions of the E1,2 and E3 sequences by preventing tin activation in specific areas of the twist expression domain. This arrangement is reminiscent of an enhancer module from the rhomboid (rho) promoter that contains closely linked positively and negatively acting binding sites (Ip et al., 1992b). In this case, the Dorsal protein serves as an activating factor, with a minor contribution of Twist, while in the mesoderm, Snail repressor molecules interfere with this activation, leading to defined on/off states of rho expression along the dorsoventral axis. In the case of tin, Twist is the key activator, and negative regulators determine off domains at specific positions along the anteroposterior axis. One repressor molecule, Even-skipped, reduces activation by Twist in odd-numbered parasegments. Eve has been previously described as a transcriptional repressor (Han and Manley, 1993). In the tin B element, it could interfere with the formation of an active complex, which includes Twist bound to the three E-boxes, or it could reduce transcriptional activation through interactions with basal transcription factors (Austin and Biggin, 1995; Um et al., 1995). We do not know whether the pair-rule modulation of tin is functionally relevant, or whether Eve is merely pirating a homeodomain binding site without any major consequences for mesoderm patterning. The major role of the R element may be to prevent activation of tin in the portion of the head mesoderm that is fated to form hemocytes. We have identified the head-patterning gene btd as a negative regulator of tin and as a positive regulator of the hematopoietic differentiation gene srp in this region. This and additional genetic experiments with tin and srp mutants (Z. Y. and M. F., unpublished data) suggest that the complementary patterns of srp and tin are not achieved by mutual inhibition, but rather by an overlapping set of upstream regulators that affect tin and srp expression in an opposite manner. It should be noted that other regulators in addition to btd must be involved, because we observe some residual activation of srp in a small mesodermal domain in btd mutants that disappears prematurely during gastrulation. Similarly, tin mrna expression is initially absent in the head mesoderm of btd mutants, but expands into this area after blastoderm. The small amount of srp products in btd appears to be sufficient to allow the formation of some hemocytes. However, the premature disappearance of srp expression and perhaps the ectopic expression of tinman seem to interfere with normal hemocyte differentiation, as indicated by the failure of the remnant cells to migrate into the embryonic body cavity. There is genetic evidence for a distinct enhancer driving srp expression in the blood mesoderm (Rehorn et al., 1996). Once available, it will be interesting to compare this enhancer element to the tin B enhancer and to determine how reciprocal patterns of tin and srp are achieved on the molecular level. Since we did not detect any specific binding of Btd protein to the R element (although it bound well to control sequences containing a SP1-binding site; data not shown), it is possible that tin repression by btd is

Axis determination in flies. Sem 9.3.B.5 Animal Science

Axis determination in flies. Sem 9.3.B.5 Animal Science Axis determination in flies Sem 9.3.B.5 Animal Science All embryos are in lateral view (anterior to the left). Endoderm, midgut; mesoderm; central nervous system; foregut, hindgut and pole cells in yellow.

More information

Midterm 1. Average score: 74.4 Median score: 77

Midterm 1. Average score: 74.4 Median score: 77 Midterm 1 Average score: 74.4 Median score: 77 NAME: TA (circle one) Jody Westbrook or Jessica Piel Section (circle one) Tue Wed Thur MCB 141 First Midterm Feb. 21, 2008 Only answer 4 of these 5 problems.

More information

Gonadal mesoderm and fat body initially follow a common developmental path in Drosophila

Gonadal mesoderm and fat body initially follow a common developmental path in Drosophila Development 125, 837-844 (1998) Printed in Great Britain The Company of Biologists Limited 1998 DEV7627 837 Gonadal and fat body initially follow a common developmental path in Drosophila Lisa A. Moore

More information

Supplementary Materials for

Supplementary Materials for www.sciencesignaling.org/cgi/content/full/6/301/ra98/dc1 Supplementary Materials for Regulation of Epithelial Morphogenesis by the G Protein Coupled Receptor Mist and Its Ligand Fog Alyssa J. Manning,

More information

Segment boundary formation in Drosophila embryos

Segment boundary formation in Drosophila embryos Segment boundary formation in Drosophila embryos Development 130, August 2003 Camilla W. Larsen, Elizabeth Hirst, Cyrille Alexandre and Jean Paul Vincent 1. Introduction: - Segment boundary formation:

More information

Why Flies? stages of embryogenesis. The Fly in History

Why Flies? stages of embryogenesis. The Fly in History The Fly in History 1859 Darwin 1866 Mendel c. 1890 Driesch, Roux (experimental embryology) 1900 rediscovery of Mendel (birth of genetics) 1910 first mutant (white) (Morgan) 1913 first genetic map (Sturtevant

More information

Axis Specification in Drosophila

Axis Specification in Drosophila Developmental Biology Biology 4361 Axis Specification in Drosophila November 2, 2006 Axis Specification in Drosophila Fertilization Superficial cleavage Gastrulation Drosophila body plan Oocyte formation

More information

Unicellular: Cells change function in response to a temporal plan, such as the cell cycle.

Unicellular: Cells change function in response to a temporal plan, such as the cell cycle. Spatial organization is a key difference between unicellular organisms and metazoans Unicellular: Cells change function in response to a temporal plan, such as the cell cycle. Cells differentiate as a

More information

Drosophila Life Cycle

Drosophila Life Cycle Drosophila Life Cycle 1 Early Drosophila Cleavage Nuclei migrate to periphery after 10 nuclear divisions. Cellularization occurs when plasma membrane folds in to divide nuclei into cells. Drosophila Superficial

More information

RNA Synthesis and Processing

RNA Synthesis and Processing RNA Synthesis and Processing Introduction Regulation of gene expression allows cells to adapt to environmental changes and is responsible for the distinct activities of the differentiated cell types that

More information

Axis Specification in Drosophila

Axis Specification in Drosophila Developmental Biology Biology 4361 Axis Specification in Drosophila November 6, 2007 Axis Specification in Drosophila Fertilization Superficial cleavage Gastrulation Drosophila body plan Oocyte formation

More information

Axis Specification in Drosophila

Axis Specification in Drosophila Developmental Biology Biology 4361 Axis Specification in Drosophila July 9, 2008 Drosophila Development Overview Fertilization Cleavage Gastrulation Drosophila body plan Oocyte formation Genetic control

More information

Segmental patterning of heart precursors in Drosophila

Segmental patterning of heart precursors in Drosophila Development 121, 4303-4308 (1995) Printed in Great Britain The Company of Biologists Limited 1995 DEV5025 4303 Segmental patterning of heart precursors in Drosophila Peter A. Lawrence 1, *, Rolf Bodmer

More information

MOLECULAR CONTROL OF EMBRYONIC PATTERN FORMATION

MOLECULAR CONTROL OF EMBRYONIC PATTERN FORMATION MOLECULAR CONTROL OF EMBRYONIC PATTERN FORMATION Drosophila is the best understood of all developmental systems, especially at the genetic level, and although it is an invertebrate it has had an enormous

More information

Homeotic genes in flies. Sem 9.3.B.6 Animal Science

Homeotic genes in flies. Sem 9.3.B.6 Animal Science Homeotic genes in flies Sem 9.3.B.6 Animal Science So far We have seen that identities of each segment is determined by various regulators of segment polarity genes In arthopods, and in flies, each segment

More information

Developmental Biology 3230 Midterm Exam 1 March 2006

Developmental Biology 3230 Midterm Exam 1 March 2006 Name Developmental Biology 3230 Midterm Exam 1 March 2006 1. (20pts) Regeneration occurs to some degree to most metazoans. When you remove the head of a hydra a new one regenerates. Graph the inhibitor

More information

Autonomous concentration-dependent activation and repression of Krüppel by hunchback in the Drosophila embryo

Autonomous concentration-dependent activation and repression of Krüppel by hunchback in the Drosophila embryo Development 120, 3043-3049 (1994) Printed in Great Britain The Company of Biologists Limited 1994 3043 Autonomous concentration-dependent activation and repression of Krüppel by hunchback in the Drosophila

More information

Morphogens in biological development: Drosophila example

Morphogens in biological development: Drosophila example LSM5194 Morphogens in biological development: Drosophila example Lecture 29 The concept of morphogen gradients The concept of morphogens was proposed by L. Wolpert as a part of the positional information

More information

Two distinct mechanisms for differential positioning of gene expression borders involving the Drosophila gap protein giant

Two distinct mechanisms for differential positioning of gene expression borders involving the Drosophila gap protein giant Development 125, 3765-3774 (1998) Printed in Great Britain The Company of Biologists Limited 1998 DEV5218 3765 Two distinct mechanisms for differential positioning of gene expression borders involving

More information

Chapter 4 Evaluating a potential interaction between deltex and git in Drosophila: genetic interaction, gene overexpression and cell biology assays.

Chapter 4 Evaluating a potential interaction between deltex and git in Drosophila: genetic interaction, gene overexpression and cell biology assays. Evaluating a potential interaction between deltex and git in Drosophila: genetic interaction, gene overexpression and cell biology assays. The data described in chapter 3 presented evidence that endogenous

More information

Chapter 18 Lecture. Concepts of Genetics. Tenth Edition. Developmental Genetics

Chapter 18 Lecture. Concepts of Genetics. Tenth Edition. Developmental Genetics Chapter 18 Lecture Concepts of Genetics Tenth Edition Developmental Genetics Chapter Contents 18.1 Differentiated States Develop from Coordinated Programs of Gene Expression 18.2 Evolutionary Conservation

More information

purpose of this Chapter is to highlight some problems that will likely provide new

purpose of this Chapter is to highlight some problems that will likely provide new 119 Chapter 6 Future Directions Besides our contributions discussed in previous chapters to the problem of developmental pattern formation, this work has also brought new questions that remain unanswered.

More information

Developmental Biology

Developmental Biology Developmental Biology 317 (2008) 417 429 Contents lists available at ScienceDirect Developmental Biology journal homepage: www.elsevier.com/developmentalbiology Daughterless dictates Twist activity in

More information

Development of Drosophila

Development of Drosophila Development of Drosophila Hand-out CBT Chapter 2 Wolpert, 5 th edition March 2018 Introduction 6. Introduction Drosophila melanogaster, the fruit fly, is found in all warm countries. In cooler regions,

More information

Three types of RNA polymerase in eukaryotic nuclei

Three types of RNA polymerase in eukaryotic nuclei Three types of RNA polymerase in eukaryotic nuclei Type Location RNA synthesized Effect of α-amanitin I Nucleolus Pre-rRNA for 18,.8 and 8S rrnas Insensitive II Nucleoplasm Pre-mRNA, some snrnas Sensitive

More information

Control of cell fates and segmentation in the Drosophila mesoderm

Control of cell fates and segmentation in the Drosophila mesoderm Development 124, 2915-2922 (1997) Printed in Great Britain The Company of Biologists Limited 1997 DEV7576 2915 Control of cell fates and segmentation in the Drosophila mesoderm Veit Riechmann, Uwe Irion,

More information

Development Team. Developmental Biology Axis Specification in Drosophila. Head, Department of Zoology, University of Delhi

Development Team. Developmental Biology Axis Specification in Drosophila. Head, Department of Zoology, University of Delhi Paper No. : 11 Module : 6 Development Team Principal Investigator: Prof. Neeta Sehgal Head, Department of Zoology, University of Delhi Paper Coordinator: Prof. Namita Agrawal Department of Zoology, University

More information

BIS &003 Answers to Assigned Problems May 23, Week /18.6 How would you distinguish between an enhancer and a promoter?

BIS &003 Answers to Assigned Problems May 23, Week /18.6 How would you distinguish between an enhancer and a promoter? Week 9 Study Questions from the textbook: 6 th Edition: Chapter 19-19.6, 19.7, 19.15, 19.17 OR 7 th Edition: Chapter 18-18.6 18.7, 18.15, 18.17 19.6/18.6 How would you distinguish between an enhancer and

More information

Sonic hedgehog (Shh) signalling in the rabbit embryo

Sonic hedgehog (Shh) signalling in the rabbit embryo Sonic hedgehog (Shh) signalling in the rabbit embryo In the first part of this thesis work the physical properties of cilia-driven leftward flow were characterised in the rabbit embryo. Since its discovery

More information

178 Part 3.2 SUMMARY INTRODUCTION

178 Part 3.2 SUMMARY INTRODUCTION 178 Part 3.2 Chapter # DYNAMIC FILTRATION OF VARIABILITY WITHIN EXPRESSION PATTERNS OF ZYGOTIC SEGMENTATION GENES IN DROSOPHILA Surkova S.Yu. *, Samsonova M.G. St. Petersburg State Polytechnical University,

More information

A Direct Contact between the Dorsal rel Homology Domain and Twist May Mediate Transcriptional Synergy

A Direct Contact between the Dorsal rel Homology Domain and Twist May Mediate Transcriptional Synergy MOLECULAR AND CELLULAR BIOLOGY, June 1997, p. 3345 3355 Vol. 17, No. 6 0270-7306/97/$04.00 0 Copyright 1997, American Society for Microbiology A Direct Contact between the Dorsal rel Homology Domain and

More information

Lecture 7. Development of the Fruit Fly Drosophila

Lecture 7. Development of the Fruit Fly Drosophila BIOLOGY 205/SECTION 7 DEVELOPMENT- LILJEGREN Lecture 7 Development of the Fruit Fly Drosophila 1. The fruit fly- a highly successful, specialized organism a. Quick life cycle includes three larval stages

More information

Drosophila Somatic Anterior-Posterior Axis (A-P Axis) Formation

Drosophila Somatic Anterior-Posterior Axis (A-P Axis) Formation Home Biol 4241 Luria-Delbruck 1943 Hershey-Chase 1952 Meselson-Stahl 1958 Garapin et al. 1978 McClintock 1953 King-Wilson 1975 Sanger et al. 1977 Rothberg et al. 2011 Jeffreys et al. 1985 Bacterial Genetics

More information

Transcription Regulation And Gene Expression in Eukaryotes UPSTREAM TRANSCRIPTION FACTORS

Transcription Regulation And Gene Expression in Eukaryotes UPSTREAM TRANSCRIPTION FACTORS Transcription Regulation And Gene Expression in Eukaryotes UPSTREAM TRANSCRIPTION FACTORS RG. Clerc March 26. 2008 UPSTREAM TRANSCRIPTION FACTORS Experimental approaches DNA binding domains (DBD) Transcription

More information

MCB 141 Midterm I Feb. 14, 2012

MCB 141 Midterm I Feb. 14, 2012 Write your name and student ID# on EVERY PAGE of your exam MCB 141 Midterm I Feb. 14, 2012 Question #1 Question #2 Question #3 Question #4 BONUS / 28 pts / 27 pts / 25 pts / 20 pts / 1 pt TOTAL / 100 pts

More information

Question Set # 4 Answer Key 7.22 Nov. 2002

Question Set # 4 Answer Key 7.22 Nov. 2002 Question Set # 4 Answer Key 7.22 Nov. 2002 1) A variety of reagents and approaches are frequently used by developmental biologists to understand the tissue interactions and molecular signaling pathways

More information

Illegitimate translation causes unexpected gene expression from on-target out-of-frame alleles

Illegitimate translation causes unexpected gene expression from on-target out-of-frame alleles Illegitimate translation causes unexpected gene expression from on-target out-of-frame alleles created by CRISPR-Cas9 Shigeru Makino, Ryutaro Fukumura, Yoichi Gondo* Mutagenesis and Genomics Team, RIKEN

More information

Introduction. Gene expression is the combined process of :

Introduction. Gene expression is the combined process of : 1 To know and explain: Regulation of Bacterial Gene Expression Constitutive ( house keeping) vs. Controllable genes OPERON structure and its role in gene regulation Regulation of Eukaryotic Gene Expression

More information

Chapter 18 Regulation of Gene Expression

Chapter 18 Regulation of Gene Expression Chapter 18 Regulation of Gene Expression Differential gene expression Every somatic cell in an individual organism contains the same genetic information and replicated from the same original fertilized

More information

factor receptor (DFR1/DFGF-R2) involved in the directional migration of early mesodermal cells in the

factor receptor (DFR1/DFGF-R2) involved in the directional migration of early mesodermal cells in the heartless encodes a fibroblast growth factor receptor (DFR1/DFGF-R2) involved in the directional migration of early mesodermal cells in the Drosophila embryo Stephen Gisselbrecht, 1 James B. Skeath, 2

More information

Developmental genetics: finding the genes that regulate development

Developmental genetics: finding the genes that regulate development Developmental Biology BY1101 P. Murphy Lecture 9 Developmental genetics: finding the genes that regulate development Introduction The application of genetic analysis and DNA technology to the study of

More information

Thoracic Patterning by the Drosophila Gap Gene hunchback

Thoracic Patterning by the Drosophila Gap Gene hunchback Developmental Biology 237, 79 92 (2001) doi:10.1006/dbio.2001.0355, available online at http://www.idealibrary.com on Thoracic Patterning by the Drosophila Gap Gene hunchback Xuelin Wu,* Vikram Vasisht,*

More information

PRACTICE EXAM. 20 pts: 1. With the aid of a diagram, indicate how initial dorsal-ventral polarity is created in fruit fly and frog embryos.

PRACTICE EXAM. 20 pts: 1. With the aid of a diagram, indicate how initial dorsal-ventral polarity is created in fruit fly and frog embryos. PRACTICE EXAM 20 pts: 1. With the aid of a diagram, indicate how initial dorsal-ventral polarity is created in fruit fly and frog embryos. No Low [] Fly Embryo Embryo Non-neural Genes Neuroectoderm Genes

More information

10/03/2014. Eukaryotic Development. + Differentiation vs. Development. Differentiation. Development

10/03/2014. Eukaryotic Development. + Differentiation vs. Development. Differentiation. Development Differentiation vs. Development What comes to mind when you think of differentiation? Eukaryotic Development What about development? Presented by: Sean, Daria, Emily, and Maggie Example: Human Development

More information

Organization of Genes Differs in Prokaryotic and Eukaryotic DNA Chapter 10 p

Organization of Genes Differs in Prokaryotic and Eukaryotic DNA Chapter 10 p Organization of Genes Differs in Prokaryotic and Eukaryotic DNA Chapter 10 p.110-114 Arrangement of information in DNA----- requirements for RNA Common arrangement of protein-coding genes in prokaryotes=

More information

F. Frémion, M. Astier, S. Zaffran, A. Guillèn, V. Homburger, and M. Sémériva

F. Frémion, M. Astier, S. Zaffran, A. Guillèn, V. Homburger, and M. Sémériva Published Online: 31 May, 1999 Supp Info: http://doi.org/10.1083/jcb.145.5.1063 Downloaded from jcb.rupress.org on April 28, 2018 The Heterotrimeric Protein G o Is Required for the Formation of Heart Epithelium

More information

Big Idea 3: Living systems store, retrieve, transmit and respond to information essential to life processes. Tuesday, December 27, 16

Big Idea 3: Living systems store, retrieve, transmit and respond to information essential to life processes. Tuesday, December 27, 16 Big Idea 3: Living systems store, retrieve, transmit and respond to information essential to life processes. Enduring understanding 3.B: Expression of genetic information involves cellular and molecular

More information

18.4 Embryonic development involves cell division, cell differentiation, and morphogenesis

18.4 Embryonic development involves cell division, cell differentiation, and morphogenesis 18.4 Embryonic development involves cell division, cell differentiation, and morphogenesis An organism arises from a fertilized egg cell as the result of three interrelated processes: cell division, cell

More information

A regulatory code for neurogenic gene expression in the Drosophila embryo

A regulatory code for neurogenic gene expression in the Drosophila embryo Research article 2387 A regulatory code for neurogenic gene expression in the Drosophila embryo Michele Markstein 1, *,, Robert Zinzen 1, *, Peter Markstein 2, Ka-Ping Yee 3, Albert Erives 1, Angela Stathopoulos

More information

Mesoderm Induction CBT, 2018 Hand-out CBT March 2018

Mesoderm Induction CBT, 2018 Hand-out CBT March 2018 Mesoderm Induction CBT, 2018 Hand-out CBT March 2018 Introduction 3. Books This module is based on the following books: - 'Principles of Developement', Lewis Wolpert, et al., fifth edition, 2015 - 'Developmental

More information

CHAPTER 13 PROKARYOTE GENES: E. COLI LAC OPERON

CHAPTER 13 PROKARYOTE GENES: E. COLI LAC OPERON PROKARYOTE GENES: E. COLI LAC OPERON CHAPTER 13 CHAPTER 13 PROKARYOTE GENES: E. COLI LAC OPERON Figure 1. Electron micrograph of growing E. coli. Some show the constriction at the location where daughter

More information

Complex interactions between cis-regulatory modules in native conformation are critical for Drosophila snail expression

Complex interactions between cis-regulatory modules in native conformation are critical for Drosophila snail expression 4566 CORRIGENDUM Development 138, 4566 (2011) doi:10.1242/dev.074377 2011. Published by The Company of Biologists Ltd Complex interactions between cis-regulatory modules in native conformation are critical

More information

Exam 1 ID#: October 4, 2007

Exam 1 ID#: October 4, 2007 Biology 4361 Name: KEY Exam 1 ID#: October 4, 2007 Multiple choice (one point each) (1-25) 1. The process of cells forming tissues and organs is called a. morphogenesis. b. differentiation. c. allometry.

More information

Developmental Biology

Developmental Biology Developmental Biology 376 (2013) 99 112 Contents lists available at SciVerse ScienceDirect Developmental Biology journal homepage: www.elsevier.com/locate/developmentalbiology Genomes and Developmental

More information

BILD7: Problem Set. 2. What did Chargaff discover and why was this important?

BILD7: Problem Set. 2. What did Chargaff discover and why was this important? BILD7: Problem Set 1. What is the general structure of DNA? 2. What did Chargaff discover and why was this important? 3. What was the major contribution of Rosalind Franklin? 4. How did solving the structure

More information

AP Biology Gene Regulation and Development Review

AP Biology Gene Regulation and Development Review AP Biology Gene Regulation and Development Review 1. What does the regulatory gene code for? 2. Is the repressor by default active/inactive? 3. What changes the repressor activity? 4. What does repressor

More information

Multiple signaling pathways and a selector protein sequentially regulate Drosophila wing development

Multiple signaling pathways and a selector protein sequentially regulate Drosophila wing development Research article 285 Multiple signaling pathways and a selector protein sequentially regulate Drosophila wing development Shian-Jang Yan 1,2, Yi Gu 1, *, Willis X. Li 2, and Robert J. Fleming 1,, 1 Department

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION med!1,2 Wild-type (N2) end!3 elt!2 5 1 15 Time (minutes) 5 1 15 Time (minutes) med!1,2 end!3 5 1 15 Time (minutes) elt!2 5 1 15 Time (minutes) Supplementary Figure 1: Number of med-1,2, end-3, end-1 and

More information

Karen Beckett, Mary K. Baylies

Karen Beckett, Mary K. Baylies Developmental Biology 299 (2006) 176 192 www.elsevier.com/locate/ydbio Parcas, a regulator of non-receptor tyrosine kinase signaling, acts during anterior posterior patterning and somatic muscle development

More information

Bypass and interaction suppressors; pathway analysis

Bypass and interaction suppressors; pathway analysis Bypass and interaction suppressors; pathway analysis The isolation of extragenic suppressors is a powerful tool for identifying genes that encode proteins that function in the same process as a gene of

More information

Genetics 275 Notes Week 7

Genetics 275 Notes Week 7 Cytoplasmic Inheritance Genetics 275 Notes Week 7 Criteriafor recognition of cytoplasmic inheritance: 1. Reciprocal crosses give different results -mainly due to the fact that the female parent contributes

More information

Hindgut visceral mesoderm requires an ectodermal template for normal development in Drosophila

Hindgut visceral mesoderm requires an ectodermal template for normal development in Drosophila Development 128, 233-242 (2001) Printed in Great Britain The Company of Biologists Limited 2001 DEV7838 233 Hindgut visceral mesoderm requires an ectodermal template for normal development in Drosophila

More information

!!!!!!!! DB3230 Midterm 2 12/13/2013 Name:

!!!!!!!! DB3230 Midterm 2 12/13/2013 Name: 1. (10 pts) Draw or describe the fate map of a late blastula stage sea urchin embryo. Draw or describe the corresponding fate map of the pluteus stage larva. Describe the sequence of gastrulation events

More information

Tissue- and stage-specific control of homeotic and segmentation gene expression in Drosophila embryos by the polyhomeotic gene

Tissue- and stage-specific control of homeotic and segmentation gene expression in Drosophila embryos by the polyhomeotic gene Development 103, 733-741 (1988) Printed in Great Britain The Company of Biologists Limited 1988 733 Tissue- and stage-specific control of homeotic and segmentation gene expression in Drosophila embryos

More information

Eukaryotic Gene Expression

Eukaryotic Gene Expression Eukaryotic Gene Expression Lectures 22-23 Several Features Distinguish Eukaryotic Processes From Mechanisms in Bacteria 123 Eukaryotic Gene Expression Several Features Distinguish Eukaryotic Processes

More information

Evolution of Transcription factor function: Homeotic (Hox) proteins

Evolution of Transcription factor function: Homeotic (Hox) proteins Evolution of Transcription factor function: Homeotic (Hox) proteins Hox proteins regulate morphology in cellular zones on the anterior-posterior axis of embryos via the activation/repression of unknown

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION doi:10.1038/nature11589 Supplementary Figure 1 Ciona intestinalis and Petromyzon marinus neural crest expression domain comparison. Cartoon shows dorsal views of Ciona mid gastrula (left) and Petromyzon

More information

Chapter 10 Development and Differentiation

Chapter 10 Development and Differentiation Part III Organization of Cell Populations Chapter Since ancient times, people have wondered how organisms are formed during the developmental process, and many researchers have worked tirelessly in search

More information

Chapter 11. Development: Differentiation and Determination

Chapter 11. Development: Differentiation and Determination KAP Biology Dept Kenyon College Differential gene expression and development Mechanisms of cellular determination Induction Pattern formation Chapter 11. Development: Differentiation and Determination

More information

Caenorhabditis elegans

Caenorhabditis elegans Caenorhabditis elegans Why C. elegans? Sea urchins have told us much about embryogenesis. They are suited well for study in the lab; however, they do not tell us much about the genetics involved in embryogenesis.

More information

Developmental Biology Lecture Outlines

Developmental Biology Lecture Outlines Developmental Biology Lecture Outlines Lecture 01: Introduction Course content Developmental Biology Obsolete hypotheses Current theory Lecture 02: Gametogenesis Spermatozoa Spermatozoon function Spermatozoon

More information

REVIEW SESSION. Wednesday, September 15 5:30 PM SHANTZ 242 E

REVIEW SESSION. Wednesday, September 15 5:30 PM SHANTZ 242 E REVIEW SESSION Wednesday, September 15 5:30 PM SHANTZ 242 E Gene Regulation Gene Regulation Gene expression can be turned on, turned off, turned up or turned down! For example, as test time approaches,

More information

Role of Organizer Chages in Late Frog Embryos

Role of Organizer Chages in Late Frog Embryos Ectoderm Germ Layer Frog Fate Map Frog Fate Map Role of Organizer Chages in Late Frog Embryos Organizer forms three distinct regions Notochord formation in chick Beta-catenin localization How does beta-catenin

More information

Attractive and repulsive functions of Slit are mediated by different receptors in the Drosophila trachea

Attractive and repulsive functions of Slit are mediated by different receptors in the Drosophila trachea Development 129, 4941-4951 (2002) Printed in Great Britain The Company of Biologists Limited 2002 DEV7966 4941 Attractive and repulsive functions of Slit are mediated by different receptors in the Drosophila

More information

10/2/2015. Chapter 4. Determination and Differentiation. Neuroanatomical Diversity

10/2/2015. Chapter 4. Determination and Differentiation. Neuroanatomical Diversity Chapter 4 Determination and Differentiation Neuroanatomical Diversity 1 Neurochemical diversity: another important aspect of neuronal fate Neurotransmitters and their receptors Excitatory Glutamate Acetylcholine

More information

UNIVERSITY OF YORK BIOLOGY. Developmental Biology

UNIVERSITY OF YORK BIOLOGY. Developmental Biology Examination Candidate Number: UNIVERSITY OF YORK BSc Stage 2 Degree Examinations 2017-18 Department: BIOLOGY Title of Exam: Developmental Biology Desk Number: Time allowed: 1 hour and 30 minutes Total

More information

Honors Biology Reading Guide Chapter 11

Honors Biology Reading Guide Chapter 11 Honors Biology Reading Guide Chapter 11 v Promoter a specific nucleotide sequence in DNA located near the start of a gene that is the binding site for RNA polymerase and the place where transcription begins

More information

9/4/2015 INDUCTION CHAPTER 1. Neurons are similar across phyla Thus, many different model systems are used in developmental neurobiology. Fig 1.

9/4/2015 INDUCTION CHAPTER 1. Neurons are similar across phyla Thus, many different model systems are used in developmental neurobiology. Fig 1. INDUCTION CHAPTER 1 Neurons are similar across phyla Thus, many different model systems are used in developmental neurobiology Fig 1.1 1 EVOLUTION OF METAZOAN BRAINS GASTRULATION MAKING THE 3 RD GERM LAYER

More information

orthodenticle homeobox gene

orthodenticle homeobox gene Development 121, 3561-3572 (1995) Printed in Great Britain The Company of Biologists Limited 1995 DEV8275 3561 Pattern formation in Drosophila head development: the role of the orthodenticle homeobox gene

More information

Biology 218, practise Exam 2, 2011

Biology 218, practise Exam 2, 2011 Figure 3 The long-range effect of Sqt does not depend on the induction of the endogenous cyc or sqt genes. a, Design and predictions for the experiments shown in b-e. b-e, Single-cell injection of 4 pg

More information

tinman, a Drosophila homeobox gene required for heart and visceral

tinman, a Drosophila homeobox gene required for heart and visceral Development 121, 3889-3899 (1995) Printed in Great Britain The Company of Biologists Limited 1995 DEV4511 3889 tinman, a Drosophila homeobox gene required for heart and visceral mesoderm specification,

More information

Lesson Overview. Gene Regulation and Expression. Lesson Overview Gene Regulation and Expression

Lesson Overview. Gene Regulation and Expression. Lesson Overview Gene Regulation and Expression 13.4 Gene Regulation and Expression THINK ABOUT IT Think of a library filled with how-to books. Would you ever need to use all of those books at the same time? Of course not. Now picture a tiny bacterium

More information

The Complex Spatio-Temporal Regulation of the Drosophila Myoblast Attractant Gene duf/kirre

The Complex Spatio-Temporal Regulation of the Drosophila Myoblast Attractant Gene duf/kirre The Complex Spatio-Temporal Regulation of the Drosophila Myoblast Attractant Gene duf/kirre K. G. Guruharsha 1,2, Mar Ruiz-Gomez 3, H. A. Ranganath 2, Rahul Siddharthan 4, K. VijayRaghavan 1 * 1 National

More information

Activation and repression by the C-terminal domain of Dorsal

Activation and repression by the C-terminal domain of Dorsal Development 128, 1869-1879 (2001) Printed in Great Britain The Company of Biologists Limited 2001 DEV5435 1869 Activation and repression by the C-terminal domain of Dorsal Rubén Darío Flores-Saaib*, Songtao

More information

Analysis of the gooseberry locus in Drosophila embryos: gooseberry determines the cuticular pattern and activates gooseberry neuro

Analysis of the gooseberry locus in Drosophila embryos: gooseberry determines the cuticular pattern and activates gooseberry neuro Development 118, 21-31 (1993) Printed in Great Britain The Company of Biologists Limited 1993 21 Analysis of the gooseberry locus in Drosophila embryos: gooseberry determines the cuticular pattern and

More information

1. What are the three general areas of the developing vertebrate limb? 2. What embryonic regions contribute to the developing limb bud?

1. What are the three general areas of the developing vertebrate limb? 2. What embryonic regions contribute to the developing limb bud? Study Questions - Lecture 17 & 18 1. What are the three general areas of the developing vertebrate limb? The three general areas of the developing vertebrate limb are the proximal stylopod, zeugopod, and

More information

MCB 141 Midterm I Feb. 19, 2009

MCB 141 Midterm I Feb. 19, 2009 Write your name and student ID# on EVERY PAGE of your exam MCB 141 Midterm I Feb. 19, 2009 Circle the name of your TA Jessica Lyons Alberto Stolfi Question #1 Question #2 Question #3 Question #4 TOTAL

More information

MCDB 4777/5777 Molecular Neurobiology Lecture 29 Neural Development- In the beginning

MCDB 4777/5777 Molecular Neurobiology Lecture 29 Neural Development- In the beginning MCDB 4777/5777 Molecular Neurobiology Lecture 29 Neural Development- In the beginning Learning Goals for Lecture 29 4.1 Describe the contributions of early developmental events in the embryo to the formation

More information

Conclusions. The experimental studies presented in this thesis provide the first molecular insights

Conclusions. The experimental studies presented in this thesis provide the first molecular insights C h a p t e r 5 Conclusions 5.1 Summary The experimental studies presented in this thesis provide the first molecular insights into the cellular processes of assembly, and aggregation of neural crest and

More information

Initiation of translation in eukaryotic cells:connecting the head and tail

Initiation of translation in eukaryotic cells:connecting the head and tail Initiation of translation in eukaryotic cells:connecting the head and tail GCCRCCAUGG 1: Multiple initiation factors with distinct biochemical roles (linking, tethering, recruiting, and scanning) 2: 5

More information

Newly made RNA is called primary transcript and is modified in three ways before leaving the nucleus:

Newly made RNA is called primary transcript and is modified in three ways before leaving the nucleus: m Eukaryotic mrna processing Newly made RNA is called primary transcript and is modified in three ways before leaving the nucleus: Cap structure a modified guanine base is added to the 5 end. Poly-A tail

More information

Development of Developmental System System(Mathematical Topics in Biolo. Citation 数理解析研究所講究録 (1993), 827:

Development of Developmental System System(Mathematical Topics in Biolo. Citation 数理解析研究所講究録 (1993), 827: Title Development of Developmental System System(Mathematical Topics in Biolo Author(s) Takeda, Yasuhiko Citation 数理解析研究所講究録 (1993), 827: 57-74 Issue Date 1993-03 URL http://hdl.handle.net/2433/83294 Right

More information

Spatio-temporal Registration of the Expression Patterns of Drosophila Segmentation Genes

Spatio-temporal Registration of the Expression Patterns of Drosophila Segmentation Genes From: ISMB-99 Proceedings. Copyright 1999, AAAI (www.aaai.org). All rights reserved. Spatio-temporal Registration of the Expression Patterns of Drosophila Segmentation Genes Ekaterina M. Myasnikova David

More information

EST1 Homology Domain. 100 aa. hest1a / SMG6 PIN TPR TPR. Est1-like DBD? hest1b / SMG5. TPR-like TPR. a helical. hest1c / SMG7.

EST1 Homology Domain. 100 aa. hest1a / SMG6 PIN TPR TPR. Est1-like DBD? hest1b / SMG5. TPR-like TPR. a helical. hest1c / SMG7. hest1a / SMG6 EST1 Homology Domain 100 aa 853 695 761 780 1206 hest1 / SMG5 -like? -like 109 145 214 237 497 165 239 1016 114 207 212 381 583 hest1c / SMG7 a helical 1091 Sc 57 185 267 284 699 Figure S1:

More information

Patterns of Gene Expression During Drosophila Mesoderm Development

Patterns of Gene Expression During Drosophila Mesoderm Development Patterns of Gene Expression During Drosophila Mesoderm Development Eileen E. M. Furlong, 1 Erik C. Andersen, 1 * Brian Null, 1 Kevin P. White, 2 Matthew P. Scott 1 The transcription factor Twist initiates

More information

10/15/09. Tetrapod Limb Development & Pattern Formation. Developing limb region is an example of a morphogenetic field

10/15/09. Tetrapod Limb Development & Pattern Formation. Developing limb region is an example of a morphogenetic field Tetrapod Limb Development & Pattern Formation Figure 16.5(1) Limb Bud Formation derived from lateral plate (somatic) & paraxial (myotome) Fig. 16.2 Prospective Forelimb Field of Salamander Ambystoma maculatum

More information

Wan-Ju Liu 1,2, John S Reece-Hoyes 3, Albertha JM Walhout 3 and David M Eisenmann 1*

Wan-Ju Liu 1,2, John S Reece-Hoyes 3, Albertha JM Walhout 3 and David M Eisenmann 1* Liu et al. BMC Developmental Biology 2014, 14:17 RESEARCH ARTICLE Open Access Multiple transcription factors directly regulate Hox gene lin-39 expression in ventral hypodermal cells of the C. elegans embryo

More information

Biology 4361 Developmental Biology The Genetics of Axis Specification in Drosophila November 2, 2006

Biology 4361 Developmental Biology The Genetics of Axis Specification in Drosophila November 2, 2006 Biology 4361 Developmental Biology The Genetics of Axis Specification in Drosophila November 2, 2006 EARLY DROSOPHILA DEVELOPMENT Fertilization 1) Drosophila egg activation occurs at ovulation - eggs are

More information

Chapter 4. DROSOPHILA bhlh-pas DEVELOPMENTAL REGULATORY PROTEINS 1. INTRODUCTION

Chapter 4. DROSOPHILA bhlh-pas DEVELOPMENTAL REGULATORY PROTEINS 1. INTRODUCTION Chapter 4 DROSOPHILA bhlh-pas DEVELOPMENTAL REGULATORY PROTEINS Stephen T. Crews The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 1. INTRODUCTION Drosophila bhlh-pas proteins play

More information

Functions for Drosophila brachyenteron and forkhead in mesoderm specification and cell signalling

Functions for Drosophila brachyenteron and forkhead in mesoderm specification and cell signalling Development 126, 3991-4003 (1999) Printed in Great Britain The Company of Biologists Limited 1999 DEV7738 3991 Functions for Drosophila brachyenteron and forkhead in mesoderm specification and cell signalling

More information