Molecular markers for identified neuroblasts and ganglion mother cells in the Drosophila central nervous system

Size: px
Start display at page:

Download "Molecular markers for identified neuroblasts and ganglion mother cells in the Drosophila central nervous system"

Transcription

1 Development 116, (1992) Printed in Great Britain The Company of Biologists Limited Molecular markers for identified neuroblasts and ganglion mother cells in the Drosophila central nervous system CHRIS Q. DOE Department of Cell and Structural Biology, University of Illinois, Urbana, Illinois 61801, USA Summary The first step in generating cellular diversity in the Drosophila central nervous system is the formation of a segmentally reiterated array of neural precursor cells, called neuroblasts. Subsequently, each neuroblast goes through an invariant cell lineage to generate neurons and/or glia. Using molecular lineage markers, I show that (1) each neuroblast forms at a stereotyped time and position; (2) the neuroblast pattern is indistinguishable between thoracic and abdominal segments; (3) the development of individual neuroblasts can be followed throughout early neurogenesis; (4) gene expression in a neuroblast can be reproducibly modulated during its cell lineage; (5) identified ganglion mother cells form at stereotyped times and positions; and (6) the cell lineage of four well-characterized neurons can be traced back to two identified neuroblasts. These results set the stage for investigating neuroblast specification and the mechanisms controlling neuroblast cell lineages. Key words: Drosophila, neuroblast, ganglion mother cell, cell lineage. Introduction Understanding the mechanisms that generate cellular diversity during development requires a combination of descriptive, experimental and molecular genetic analyses. The power of this approach is exemplified by studies on cell determination in C. elegans and neuronal determination in the Drosophila eye, where elegant descriptive studies (e.g. Sulston et al., 1983; Ready et al., 1976) paved the way for comprehensive molecular genetic analyses (e.g. Sternberg and Horvitz, 1991; Rubin, 1991). In this and the following paper (Cui and Doe, 1992), we combine a descriptive analysis of early neurogenesis with the molecular genetic characterization of a newly identified gene that controls cell specification in the Drosophila central nervous system (CNS). The Drosophila CNS develops from a monolayer of ventral ectodermal cells called the neurogenic region (Hartenstein and Campos-Ortega, 1984; Campos-Ortega and Hartenstein, 1985). The neurogenic region is bilaterally symmetrical, with the left and right halves separated by a narrow strip of specialized midline cells that generate both neurons and glia (Klambt et al., 1991). Individual cells within the neurogenic region delaminate into the embryo and enlarge, forming a subepidermal two-dimensional array of neural precursors, called neuroblasts (NBs; Wheeler, 1891, 1893). NBs divide asymmetrically to bud off smaller ganglion mother cells (GMCs) into the embryo (Bauer, 1904). Each GMC divides once to produce a pair of post-mitotic neurons. Most CNS precursors are NBs, but in every hemisegment of the neurogenic region there is at least one glial precursor (GP), which divides symmetrically to generate CNS longitudinal glia (Jacobs et al., 1989), and one midline precursor (MP2), which divides only once to produce the identified dmp2 and vmp2 neurons (Doe et al., 1988a; Thomas et al., 1984). In this paper, the term NB will be used as a general term for all CNS precursors, although several cells (e.g. MP2 and GP) clearly do not divide asymmetrically and some NBs may generate both neurons and glia (Ferdieu and Mahowald, 1989). NBs were first identified in grasshopper embryos (Wheeler, 1891). Much later it was shown that individual NBs can be uniquely identified, each forms at a characteristic time, and the final pattern of NBs in each segment is invariant (Bate, 1976; Doe and Goodman, 1985a). The grasshopper NB map has led to several important experimental results. Laser ablations of identified NBs show that positional cues within the neurogenic region control NB identity and that cell interactions determine which cells of the neurogenic region will form NBs (Doe and Goodman, 1985b). In addition, the lineage of several identified NBs have been traced, showing that each goes through an invariant cell lineage to generate a specific set of neurons or glia (Bate and Grunewald, 1981; Goodman et al., 1982; Raper et al., 1983; Goodman et al., 1984; Taghert and Goodman, 1984; Bastiani and Goodman, 1986). To identify the genes controlling each of these developmental events, attention has turned to the Drosophila embryo. Mutations have been identified that increase the number of NBs (Lehmann et al., 1983), decrease NB

2 856 C. Q. Doe number (Jimenez and Campos-Ortega, 1979, 1990) or change the fate of NB progeny (Doe et al., 1988a,b; Duffy et al., 1991; Doe et al., 1991; Vaessin et al., 1991; Cui and Doe, 1992). Previous studies show that Drosophila has segmentally reiterated patterns of NBs (Hartenstein et al., 1987; Doe et al., 1988a; Jimenez and Campos-Ortega, 1990); however, the paucity of NB-specific markers and the inability to follow individual NBs throughout development has limited the usefulness of these maps. In addition, the relationship between NBs at each stage has not been determined and only a few NBs have been uniquely identified (Doe et al., 1988a; Martin-Bermudo et al., 1991; Skeath and Carroll, 1992). To provide the necessary background for investigating cell specification in the Drosophila CNS, I have used antibody and enhancer trap markers to show that individual NBs and GMCs form at a stereotyped time and position, and that each NB can be uniquely identified throughout most of its embryonic cell lineage. In addition, even-skipped (eve) CNS expression is used to determine the complete cell lineage of the well characterized acc, pcc and RP2 neurons. Materials and methods Developmental stages and morphological landmarks are described in Campos-Ortega and Hartenstein (1985) and all developmental times are measured at 25 C. Expression of the following genes were used to identify NBs and/or GMCs: engrailed (en), even-skipped (eve), wingless (wg), achaete (ac), the promoter fusion fushi tarazu-lacz (ftz-lacz; insertions on either the second or third chromosome). In addition, the following enhancer trap lines were used: H162 (insertion at 87B4-6, detects the seven-up [svp] pattern), 1530 (insertion at 83C1-2, detects the ming pattern), 1912 (insertion at 45C) and 5953 (insertion at 82B3). Staged embryos were fixed and stained with antibodies to visualize subsets of NBs or their progeny. Standard methods were used to rear flies and collect embryos (Roberts, 1986); all procedures were done at room temperature except where noted. Embryos were dechorionated in 50% bleach (sodium hypochlorite; Chlorox) for 2-3 minutes and then rinsed under flowing tap water for 1 minute. Embryos were fixed in a 1:1 mix of heptane and fix (PEMFA; 100 mm Pipes, 2 mm EGTA, 1 mm MgSO 4, 3.7% formaldehyde, ph 6.9). Fixation was for minutes when staining for eve, for minutes when staining for en and minutes when staining for ac or the enhancer trap marker β-galactosidase (βgal). The fixative was removed and embryos were cracked out of their vitelline membranes in a 1:1 heptane:methanol mix with rapid shaking for 20 seconds. Embryos were washed twice in methanol, twice in phosphate-buffered saline (PBS ph 7.0) and then for 30 minutes in PBT (PBS, 2% BSA, 0.1% Triton X-100, ph 7.0). Primary antibodies (see below) were diluted in PBT and incubated overnight at 4 C with gentle rocking. Embryos were washed for 2 hours in PBT with the solution changed every minutes. PBT + 2% normal goat serum was used to block non-specific antibody binding for 30 minutes, and the secondary antibodies were added directly and incubated for 1.5 hours with gentle rocking. Embryos were washed for 1-2 hours in PBT with the solution changed every minutes. Fluorescently stained embryos were mounted in 80% glycerol containing 2% n-propyl gallate (Sigma) and the ventral surface visualized on a BioRad 600 confocal microscope. Detection of HRP-conjugated antibodies was in 0.5 mg/ml diaminobenzidine (Sigma), 0.003% H 2 O 2, in PBS (occasionally 0.03% CoCl 2 was included to intensify the reaction product). Embryos were dehydrated in a ethanol series, mounted in methyl salicylate, visualized on a Zeiss Axioplan microscope with differential interference contrast optics and photographed on Kodachrome ASA40 film. Primary antibodies were mouse anti-en used at 1:1 (N. Patel and C. Goodman); mouse anti-eve used at 1:1 (N. Patel and C. Goodman); mouse anti-ac used at 1:5 (J. Skeath and S. Carroll); mouse anti-βgal used at 1:1500-1:2000 (Promega); rabbit anti-wg used at 1:1500 (M. van den Heuvel and R. Nusse); rabbit anti-en used at 1:100 (S. DiNardo and P. O Farrell); and rabbit anti-βgal used at 1:1000 (Cappel/Organon-Teknika). The βgal antibodies were pre-absorbed at 1:10 in PBS against fixed embryos for 1 hour prior to use, which reduced background considerably. Secondary antibodies were HRP-conjugated goat anti-mouse used at 1:400 (BioRad); HRP-conjugated goat anti-rabbit used at 1:300 (Jackson Immunoresearch); lissamine rhodamine-conjugated goat anti-mouse used at 1:200 (Jackson Immunoresearch); and FITCconjugated goat anti-rabbit used at 1:500 (Jackson Immunoresearch). The secondary antibodies were often preabsorbed at 1:10 in PBS against fixed embryos for 1 hour prior to use to reduce background. Criteria for identifying NBs were (1) located in the subectodermal cell layer, (2) located in the neurogenic region (between the midline CNS and the tracheal pits), (3) relatively large size compared to superficial cells and (4) greater cytoplasm:nuclear ratio compared to GMCs or ectodermal cells. Criteria for identifying GMCs was simply eve, hb or βgal immunoreactivity in the cells adjacent to the dorsal surface of the NBs. Overlap between identified GMCs and NBs was based on camera-lucida tracings of embryos viewed from the ventral surface (en and wg expression were used as landmarks for identifying NBs). Results Morphological development of neuroblasts NBs form by the delamination and enlargement of individual ectodermal cells within the neurogenic region. To describe this process in detail, Nomarski microscopy was used to examine the development of a single NB (4-2). The site of NB 4-2 formation is always between the easily identified NB 3-2 and NB 5-3 (described in detail below). Immediately following formation of NB 3-2, ectodermal cells at the 4-2 position have a columnar morphology, with the nuclei located at ventral cell surface (Fig. 1A). After 3-2 formation, nuclei in one to four cells at the 4-2 position move dorsally (Fig. 1B). Soon after, one cell begins enlarging (Fig. 1C) and ultimately delaminates from the ventral ectoderm to form NB 4-2. Occasionally two cells will begin enlarging at the 4-2 position; one of these cells may die or revert back to an ectodermal fate, since embryos fixed at later stages always show a single NB 4-2. (While it is uncommon to observe dual enlarging cells at one NB position, it can occur at all stages, from S1 to S5 NBs; data not shown.) NB 4-2 divides asymmetrically, budding off a smaller GMC from its dorsal surface (Fig. 1D). Although the first GMC from NB 4-2 is displaced dorsally (inwards), other NB divisions show different, reproducible orientations (discussed in detail below). The dorsal processes of the ectodermal cells are withdrawn and re-establish a columnar morphology following GMC 4-2a formation.

3 Drosophila embryonic neuroblast maps 857 Fig. 1. Morphological differentiation of neuroblasts. The development of NB 4-2 is drawn based on camera-lucida tracings of embryos viewed with Nomarski optics. (A) Ectodermal cells at the 4-2 position have a uniform columnar morphology, with the nuclei located at the ventral surface of the cells. (B) The nuclei of one to four cells at the 4-2 position move towards the dorsal cell surface. (C) One cell begins to delaminate into the embryo; it shifts both cytoplasm and the nucleus dorsally relative to adjacent cells. (D) Delamination is complete and the new NB 4-2 divides asymmetrically, budding off a smaller ganglion mother cell (top cell) at its dorsal surface. Subsequently, the adjacent ectodermal cells withdraw their dorsal processes and regain a columnar morphology. Neuroblasts form in a stereotyped array Camera-lucida tracings were made of NBs in fixed, staged embryos (Fig. 2; for the criteria used to define a NB, see Materials and methods). Only the bilateral neurogenic region is addressed in this paper; development and lineage of the midline CNS has already been elegantly described (Klambt et al., 1991). NB names have been chosen to maximize homology to grasshopper NBs there are many similarities in timing of formation, pattern of gene expression and cell lineage of specific neural precursors (see Discussion). However, the nomenclature is not meant to imply that every Drosophila NB has a definitive grasshopper homolog. NB formation is divided into six stages: early S1 (es1), S1, S2, S3, S4 and S5. Phases S1-S3 are similar to previously described S1-S3 NB maps (Hartenstein and Campos- Ortega, 1984; Hartenstein et al., 1987). It is important to note that these stages are snapshots of the developing NB pattern and intermediate patterns can be observed (this is particularly true between the S3, S4 and S5 stages). The es1 NBs develop at late stage 8 (staging according to Campos-Ortega and Hartenstein, 1985). There are nine NBs per hemisegment, in an incomplete orthogonal array, with four NBs in the medial column, one NB (5-3) in the intermediate column and four NBs in the lateral column (Fig. 2A). Development of NB 3-2 at early stage 9 completes the S1 array (Fig. 2B); NBs 3-2 and 5-3 are usually closer together in the same segment than between segments, allowing all S1 NBs to be identified by position alone. The S2 NBs enlarge at stage 9, primarily in the intermediate column (Fig. 2C). Each S2 NB can be uniquely identified by position, with orientation provided by a slight ectodermal groove between NB rows 5 and 6 (the parasegmental groove), as well as the more medial position of NB 2-5 relative to the other three NBs in the lateral column (3-5, 5-6 and 7-4). The S3 NBs develop at stage 10, in columns at the medial and lateral edges of the NB array (Fig. 2D). One of the S3 NBs is actually a Glial Precursor (GP; Doe et al., 1988a; Jacobs et al., 1989). The mediolateral position, size and temporal profile of the GP is matched by a nearby neural precursor, called X. The fate of this cell is unknown; its similarity to GP suggests it may also generate glia. Although it is usually possible to identify each S3 NB based on its position relative to the GP and X cells, unambiguous NB identification at this stage requires the use of molecular markers (see below). The S4 and S5 NBs develop at stage 11 and late stage 11, respectively (Fig. 2E,F). Both S4 and S5 NBs form in columns between the existing intermediate and lateral columns of NBs; after these NBs form, it is difficult to discern clear columns or rows of NBs, due to variability in NB positions. Formation of the S5 NBs brings the total number of NBs to 31 in each hemisegment, in addition to the MNB. However, four of these precursors are not present in the S5 array due to reductive divisions to generate neurons or glia (MP2, GP) or unknown events (X, 2-2). The combination of variable NB positions and disappearance of some precursors makes it impossible to identify NBs by their position alone. Accurate identification of S4 and S5 NBs requires molecular markers (described below). Molecular markers allow individual neuroblasts to be uniquely identified Eight molecular markers have been used to assay a different, overlapping population of NBs at several stages of neurogenesis; expression of these markers is summarized in Fig. 3 and examples of each are shown in Fig. 4. Antibodies were used to detect the ac (Skeath and Carroll, 1992) (Fig. 4A), wg (van den Heuvel et al., 1989) (Fig. 4C) and en (DiNardo et al., 1985) (Fig. 4D) proteins. The product of the lacz gene was detected in the promoter fusion construct ftzlacz (Hiromi et al., 1985) (Fig. 4B). In addition, the lacz pattern was assayed in four enhancer trap lines: H162, which detects the svp expression pattern (Mlodzik et al., 1990) (Fig. 4F; Fig. 5); 1530, which detects the ming expression pattern (Cui and Doe, 1992) (Fig. 4G); 5953 (C. Q. D., unpublished results) (Fig. 4E); and 1912 (C. Q. D., unpublished results) (Fig. 4H). The expression of lacz in the enhancer trap or gene fusion lines will be referred to by the name of the line or adjacent gene (e.g and svp are expressed... indicates expression of lacz in embryos from the 1912 and H162 enhancer trap lines, respectively). The combination of position and gene expression is suf-

4 858 C. Q. Doe Fig. 2. Development of the embryonic neuroblast pattern. Camera-lucida tracings the NBs in one segment and the adjacent anterior en+ neuroblasts (rows 6, 7 and NB 1-1). Each panel is drawn from a single embryo of a different age. The newest NBs of each stage are shown in grey on the left hemisegment, highlighting the columnar organization of NB formation. Ventral midline is indicated by the vertical line. See text for discussion of individual NBs at each stage. (A) The early S1 pattern; late stage 8. (B) The S1 pattern; early stage 9. (C) The S2 pattern; stage 9. (D) The S3 pattern; stage 10. NB 6-4 is not visible in the left hemisegment. (E) The S4 pattern; stage 11. (F) The S5 pattern; late stage 11. NB 5-5 is not visible in this embryo. ficient to uniquely identify all NBs at each stage of neurogenesis. For example, among the medial column of S1 NBs, 2-2 expresses no marker genes, MP2 expresses ac, 5-2 expresses wg and svp, and 7-1 expresses ac and en (Fig. 3B). Among S2 lateral column NBs, 2-5 expresses svp, 3-5 expresses ac, 5-6 expresses wg and 7-4 expresses ac, svp and en (Fig. 3C). The molecular identification of NBs after S4 formation is possible, despite the complexity of the pattern. Markers for a scattered array of NBs (e.g and 5953) provide local landmarks that facilitate identification of NBs in most areas of the pattern. Based on previous NB maps in grasshopper (Bate, 1976; Doe and Goodman, 1985a) and Drosophila (Hartenstein and Campos-Ortega, 1984; Hartenstein et al., 1987), I expected differences in the NB array between thoracic and abdominal segments. In fact, each molecular marker for S5 NBs is expressed identically in thoracic and abdominal segments. However, many thoracic NBs form earlier than their abdominal homologs, such that abdominal segments can be up to one NB stage behind that observed in thoracic segments in a single embryo (data not shown). Segmental differences in the NB array can be found in the extreme termini of the embryo: the first subesophageal (S1) and ninth abdominal (A9) segments have a reduced number of NBs compared to the intervening segments (data not shown). Individual neuroblasts can be identified throughout neurogenesis In addition to identifying NBs at each stage of development, individual NBs can be followed from one stage to the next. With this information, it should be possible to determine the ultimate fate of a precursor, the changes in gene expression within a NB cell lineage and the fate of the GMCs produced from a single NB. Most NB markers are expressed in specific subsets of NBs over several hours of development and thus they allow identification of specific, individual NBs at most or all stages of neurogenesis. In addition, several markers have transient coexpression in specific NBs, allowing the NB to be initially identified by one marker and subsequently identified by the second marker. A good example of a marker that can be used to follow specific NBs throughout neurogenesis is svp (Fig. 5). svp is expressed in NB 5-2 from S1 to S5 stages and NB 7-4

5 Drosophila embryonic neuroblast maps 859 from S2 to S5 stages, allowing these NBs to be followed throughout neurogenesis, where they can be used as markers to identify surrounding NBs (Figs 4F, 5). Other genes are also expressed in the same NBs at multiple stages of development (e.g. en and wg; Fig. 3). To confirm the relative positions of identified NBs at each stage, embryos were double-labeled for different pairs of markers. Fig. 6A shows an embryo labeled for en and ming expression; clearly most en-expressing (en + ) NBs co-express ming, although several NBs express only ming. Similar double labels show that most en + NBs do not express 5953 (Fig. 6B). Double labels have also been done for the following pairs of genes: en/ac, en/wg, en/ftz, en/svp, en/1912, ac/svp and ac/ming (data summarized in Fig. 3). Molecular markers not only allow NBs to be followed from one stage to the next, they also can distinguish between the displacement of a NB to a new position and the disappearance of a NB. For example, ming expression can be used to follow the lateral displacement of NB 3-4 between stages S4 and S5 (Fig. 3). In contrast, some precursors disappear from the NB pattern altogether. ftz expression can be used to follow the disappearance of the MP2 and GP precursors from the NB pattern. MP2 delaminates and enlarges like a NB, but it moves dorsally and laterally before dividing symmetrically to produce the dmp2 and vmp2 neurons (Doe et al., 1988a), whereas the GP divides symmetrically and generates longitudinal glia (Doe et al., 1988a; Jacobs et al., 1989). Interestingly, NB 2-2 has a similar developmental profile to MP2: it delaminates, enlarges, moves dorsally and disappears from the NB pattern concurrently with MP2 (Fig. 3). It is not known whether 2-2 divides symmetrically, like MP2, or asymmetrically, like a NB. Neuroblasts reproducibly alter gene expression during their cell lineage Using the markers described above, it is possible to trace the pattern of gene expression in individual NBs throughout their early cell lineages. It is clear that gene expression can be reproducibly altered during the cell lineage of an identified NB. For example, ac is initially expressed in three NBs and MP2, but is turned off at about the time that the NBs first divide (Figs 3, 4A). In contrast, several genes are initially expressed midway through specific NB lineages. svp is detectable in NB 1-1 only after it has generated its first progeny (Fig. 3E). ming enhancer trap line and transcript are also expressed at different points within identified NB cell lineages (described in detail in Cui and Doe, 1992). These results clearly show that NBs reproducibly switch genes on or off at specific points in their cell lineage. The mechanisms regulating differential NB gene expression are unknown. Because NBs appear to change identity during the course of their lineage (as reflected by differential gene expression), it is necessary to develop a nomenclature that distinguishes between a NB at each point in its cell lineage. Here I propose a method for uniquely naming each cell in the developing CNS, using NB 1-1 as an example. At the time of formation, the NB is called 1-1; following its first division, the NB is termed 1-1A and the GMC is 1-1a (the same letter indicates sibling cells, with upper case denoting the NB and the lower case indicating the GMC). Subsequent NB divisions produce new NBs (1-1B, 1-1C, etc) and their sibling GMCs (1-1b, 1-1c, etc). The pair of neurons from each GMC are named according to their parental GMC (e.g. 1-1a1 and 1-1a2). This system allows the unique naming of every NB, GMC and neuron throughout neurogenesis. Several identified GMCs form in a stereotyped position due to the reproducible orientation of neuroblast divisions Individual GMCs were identified using a variety of molecular markers and camera-lucida tracings were used to establish the spatial relationship between NBs and GMCs (Fig. 7). A close association between GMC and NB is presumably because the NB has recently generated the GMC (i.e. it is the parental NB). Many NBs examined to date (1-1, 2-5, 3-5, 4-2, 5-2, 5-6 and 7-4) have a preferred orientation of NB cell division, ranging from perfectly dorsal (perpendicular to the embryo surface) to nearly lateral (parallel to the embryo surface); other identified GMCs have a more variable position (Fig. 7). The nuclear hunchback (hb) protein can be detected in all NBs and many of the earlyborn GMCs; the position of the first GMCs born from the lateral column of S2 NBs is summarized in Fig. 7A. Each of these NBs usually divides to produce a GMC positioned dorsolaterally from the NB (plane of division about 45 to the embryo surface), although GMC 2-5a tends to be located more dorsally than the other GMCs. The position of later-born GMCs can be assayed by svp and eve expression (Fig. 7B). Precise GMC positioning occurs following division of NB 5-2, where both svpexpressing GMCs 5-2a and 5-2b are generated from the lateral surface of the NB (only GMC 5-2b is shown in Fig. 7B). GMC 5-2b is distinguished from GMC 5-2a due to its closer position to NB 5-2 at the stage examined. svp can also be used to identify a GMC from NB 7-4, which is probably the second GMC (7-4b), based on its position next to the NB, its time of formation and the presence of an earlier GMC (7-4a) detectable by hb expression at the S2 NB stage (Fig. 7A). Both 7-4a and 7-4b have different, yet stereotyped, positions: 7-4a tends to be produced from the lateral surface of NB 7-4, whereas 7-4b is usually generated from the dorsal surface of the NB (Fig. 7). Expression of eve can also be used to identify uniquely three GMCs; two GMCs are located adjacent to NBs 1-1 and 4-2, whereas the third eve + GMCs has a more variable position (Fig. 7b). The lineage of the eve + GMCs is discussed below. Results from using hb, svp and eve expression to identify uniquely GMCs shows that some NB divisions have a characteristic or preferred orientation, which may change with each division. Development of identified neurons: cell lineage from neuroblast to neuron Camera-lucida tracings of eve + GMCs shows that two GMCs are reproducibly associated with a specific NB (Fig. 7B; Table 1). eve + GMCs are closely associated with NBs 1-1 and 4-2. The eve + GMC 1-1a is observed soon after formation of NB 1-1, suggesting that it is the first progeny of this NB, similar to the lineage observed in grasshopper embryos (Goodman et al., 1984). The eve + GMC near NB

6

7

8

9

10 Drosophila embryonic neuroblast maps 861 rons from adjacent NBs intermingle in the CNS, as was first observed by Hartenstein et al. (1987). This does not mean that all GMC positions are random. Using markers for identified GMCs, it is clear that some (but not all) GMCs are born in a stereotyped position, reflecting a reproducible orientation of the NB division. It is not clear whether a NB divides with the same orientation every time. NB 5-2 generates its first two progeny (GMCs 5-2a and 5-2b) from its lateral surface, whereas NB 7-4 produces two GMCs at slightly different dorsal and dorsolateral positions. The mechanisms regulating oriented NB divisions are unknown. Development of the neuroblast pattern The pattern of NBs in Drosophila has been partially characterized (Hartenstein and Campos-Ortega, 1984; Campos- Ortega and Hartenstein, 1985; Hartenstein et al., 1987; Doe et al., 1988a; Jimenez and Campos-Ortega, 1990; Skeath and Carroll, 1992). Each study described the pattern of NBs at one or more stages of development, but usually did not determine the relationship between NBs at each of the stages. In contrast, the pattern of NBs has been well characterized in the grasshopper embryo (Bate, 1976; Doe and Goodman, 1985a) and there are many similarities to the pattern of Drosophila NBs described in this paper. Both organisms have about 30 NBs per hemisegment, including a glial precursor (GP) that generates longitudinal glia, and the MP2 precursor that produces the dmp2 and vmp2 neurons. All of the cell types described in Drosophila are present in grasshopper. (The grasshopper has one cell type not present in Drosophila, the cap cell, which is attached to the ventral surface of every NB and is required for their asymmetric divisions [Kawamura and Carlson, 1962].) As first described by Hartenstein and Campos-Ortega (1984), Drosophila NBs form in waves, with each new population of NBs arranged roughly in columns. This is true for grasshopper embryos as well (Doe and Goodman, 1985a), and is confirmed for the later-forming S4 and S5 NBs described in this paper. Drosophila NBs are named after their approximate homolog in grasshopper, based on primarily on relative position (although time of formation, gene expression and cell lineage data are considered as well). For example, in both grasshopper and Drosophila the en gene is expressed in NBs of rows 6 and 7, and the eve gene is expressed in the first progeny of NBs 1-1, 4-2 and 6-2 (this paper; N. H. Patel, personal communication). Similarly, in both grasshopper and Drosophila, the MP2 and GP appear in the same position, divide similarly and generate the identical neuronal or glial progeny. Thus at least 1-1, 4-2, 6-2, GP, MP2 and the NBs in rows 6 and 7 share some degree of homology between organisms. Nevertheless, identically named NBs in each organism are not necessarily completely homologous. For example, NB 1-1 produces the acc and pcc neurons in both organisms, yet the en gene is expressed in NB 1-1 in Drosophila but not in grasshopper (this paper; Patel et al., 1989b). The extent of similarity between positionally homologous precursors in grasshopper and Drosophila will require cell lineage analysis and gene expression studies in both organisms. Grasshopper embryos have clear, but subtle, differences between thoracic and abdominal NB arrays (Bate, 1976; Doe and Goodman, 1985a), yet in Drosophila these differences are not apparent. Segment-specific neurons or glia in Drosophila may be generated by segmental differences in NB lineages or by postembryonic NB formation. Alternatively, segmental differences in embryonic NBs may exist in Drosophila,but go undetected due to lack of markers, the smaller size of the precursors and/or NB formation after the S5 stage. Neuroblast gene expression Eight molecular markers have been used to map the array of NBs throughout early neurogenesis. Many NBs at various stages can be uniquely identified based on expression of one or more genes (and all NBs can be identified by their position relative to nearby, marked NBs). Greater precision in NB identification should be possible after the mapping of at least 9 other genes expressed in subsets of NBs (runt [Duffy et al., 1991], gooseberry-early and gooseberry-late [N. H. Patel, personal communication], pox-neuro [Dambly-Chaudiere et al., 1992], dpou-19 and dpou-28 [Dick et al., 1991], cut [C. Q. D., unpublished results], scute [Cabrera, 1990] and lethal-of-scute [Martin-Bermudo et al., 1991]). In addition to providing markers for specific NBs, gene expression patterns may reveal co-expression that reflects regulatory interactions occurring in NBs and/or GMCs. For example, the ming and en genes show substantial co-expression in NBs and GMCs, which led to the observation that ming function is required for normal en expression in the CNS (Cui and Doe, 1992). Many genes are expressed in NBs only during portions of their cell lineage, either only early in the lineage (e.g. ac) or only late in the lineage (e.g. ming, svp, 5953). The change in gene expression is extremely reproducible, but the function of differential gene expression within a NB lineage is not clear. It is likely that differential gene expression within a NB lineage contributes to unique GMC specification. While this remains to be tested in detail, it is known that the ming gene, which is expressed in portions of the NB 7-4 lineage, appears to be required for the correct expression of en within this lineage (Cui and Doe, 1992). The mechanisms controlling differential gene activity in a NB throughout its lineage are unknown. Neuroblast cell lineages Cell lineage analysis in the grasshopper embryo shows that specific NBs go through invariant lineages to generate stereotyped family of neurons (Goodman et al., 1984; Raper et al., 1983; Doe and Goodman, 1985b; Bate and Grunewald, 1981; Goodman et al., 1982; Raper et al., 1983; Taghert and Goodman, 1984). The lineage studies described here suggests that this is also true in Drosophila. The 1-1 and 4-2 NBs are closely associated with an eve + GMC. This correlation suggests that the 1-1 and 4-2 NBs generate eve + GMCs, although direct lineage studies will be required to absolutely prove this relationship. These GMCs generate the acc, pcc, RP2 and RP2sib neurons. The axon morphology, muscle target and certain gene expression profiles are known for the acc and RP2 motoneurons. The lineage studies described here provide a starting point for linking many previously disparate events of neurogenesis: from formation and specification of an individual NB (e.g. NB 4-

11 862 C. Q. Doe 2), to lineal determination of NB progeny (e.g. GMC 4-2a), to the morphological differentiation of an identified neuron (e.g. RP2). Future directions The markers described here should be useful for investigating several areas of neurogenesis. (1) How is NB formation regulated? The neurogenic and proneural genes control NB formation (Campos-Ortega and Knust, 1990), yet it is not known if these genes affect all NBs identically. It may be that each mutation affects the formation of different subsets of NBs. (2) How are NBs uniquely specified? Due to the lack of markers for identified NBs, genes positionally expressed along the anterior-posterior or dorsal-ventral axes of the neurogenic region have yet to be tested for a role in NB specification. (3) What mechanisms control NB cell lineages? Genes expressed in subsets of NBs and their progeny, could be examined to determine relative expression patterns; overlaps would highlight potential regulatory interactions. For example, the ming gene is co-expressed with en in several NBs and GMCs, and indeed we find that ming is required for normal en CNS expression (Cui and Doe, 1992). (4) What is the complete cell lineage of identified NBs? Individual NBs could be labeled using a variety of techniques (e.g. transplantation, injection, photoactivation) and their neuronal or glial progeny identified. Lineage studies may reveal functional relationships between lineally related cells, such as neurotransmitter choice (Goodman et al., 1979; Taghert and Goodman, 1984). In addition, lineage studies will provide a foundation for interpreting experimental and genetic perturbations. Many thanks to L. Rost, K. Schuske and M. P. Scott for assistance in generating the 1530 and 1912 lines, and for providing the 5953 line. Antibodies were generously supplied by N. H. Patel and C. S. Goodman, M. van den Heuvel and R. Nusse, J. Skeath and S. Carroll, and S. DiNardo and P. O Farrell. A slide of embryos beautifully stained for hb was loaned by S. Macedonio (Zeiss technical representative) who received it from P. Mac- Donald and G. Struhl several years ago. Thanks to K. Doe for drawing Fig. 1; to Q. Chu-LaGraff for embryos stained for wg; to Steve Paddock and Sean Carroll for assistance with confocal microscopy; and to Volker Hartenstein and Xuan Cui for comments on the manuscript. This work was supported by the Searle Scholars Foundation, an NSF Presidential Young Investigator Award and the NIH (R ). References Bastiani, M. J. and Goodman, C. S. (1986). Guidance of neuronal growth cones in the grasshopper embryo. III. Recognition of specific glial pathways. J. Neurosci. 6, Bate, C. M. (1976). Embryogenesis of an insect nervous system I. A map of the thoracic and abdominal neuroblasts in Locusta migratoria. J. Embryol. Exp. Morph.35, Bate, C. M. and Grunewald, E. B. (1981). Embryogenesis of an insect nervous system. II. A second class of neuron precursor cells and the origin of the intersegmental connectives. J. Embryol. Exp. Morph. 61, Bauer, V. (1904). Zur inneren Metamorphose des Centralnervensystems der Insecten. Zool. Jahrb. Abt. Anat. Ontog. Tiere 20, Cabrera, C. V. (1990). Lateral inhibition and cell fate during neurogenesis in Drosophila: the interactions between scute, Notch and Delta. Development 109, Campos-Ortega, J. A. and Hartenstein, V. (1985). The Embryonic Development of Drosophila melanogaster. Berlin: Springer-Verlag. Campos-Ortega, J. A. and Knust, E. (1990). Genetic mechanisms in early neurogenesis of Drosophilamelanogaster. Ann. Rev. Genet.24, Carroll, S. B. and Scott, M. P. (1985). Localization of the fushi tarazu protein during Drosophila embryogenesis. Cell 43, Cui, X. and Doe, C. Q. (1992). ming is expressed in neuroblast sublineages and regulates gene expression in the Drosophila central nervous system. Development 116, Dambly-Chaudiere, C., Jamet, E., Burri, M., Bopp, D., Basler, K., Hafen, E., Dumont, N., Spielmann, P., Ghysen, A. and Noll, M. (1992). The paired box gene pox neuro: a determinant of poly-innervated sense organs in Drosophila. Cell 69, Dick, T., Yang, X., Yeo, S. and Chia, W. (1991). Two closely linked Drosophila POU domain genes are expressed in neuroblasts and sensory elements. Proc. Natl. Acad. Sci. USA 88, DiNardo, S., Kuner, J. M., Theis, J. and O Farrell, P. H. (1985). Development of embryonic pattern in Drosophila melanogaster as revealed by accumulation of the nuclear engrailed protein. Cell 43, Doe, C. Q. and Goodman, C. S. (1985a). Early events in insect neurogenesis. I. Development and segmental differences in the pattern of neuronal precursor cells. Dev. Biol. 111, Doe, C. Q. and Goodman, C. S. (1985b). Early events in insect neurogenesis. II. The role of cell interactions and cell lineage in the determination of neuronal precursor cells. Dev. Biol. 111, Doe, C. Q., Hiromi, Y., Gehring, W. J. and Goodman, C. S. (1988a). Expression and function of the segmentation gene fushi tarazu during Drosophila neurogenesis. Science 239, Doe, C. Q., Smouse, D. and Goodman, C. S. (1988b). Control of neuronal fate by the Drosophila segmentation gene even-skipped. Nature 333, Doe, C. Q., Chu-LaGraff, Q., Wright, D. M. and Scott, M. P. (1991). The prospero gene specifies cell fates in the Drosophila central nervous system. Cell 65, Duffy, J. B., Kania, M. A. and Gergen, J. P. (1991). Expression and function of the Drosophila gene runt in early stages of neural development. Development 113, Ferdieu, J. R. and Mahowald, A. P. (1989). Glial interactions with neurons during Drosophila embryogenesis. Development 106, Goodman, C. S., O Shea, M., McCaman, R. E. and Spitzer, N. C. (1979). Embryonic development of identified neurons: Temporal pattern of morphological and biochemical differentiation. Science 204, Goodman, C. S., Raper, J. A., Ho, R. K. and Chang, S. (1982). Pathfinding of neuronal growth cones in grasshopper embryos. In Developmental Order: Its Origin and Regulation. (eds. S. Subtelny and P. B. Green). pp New York: Alan R. Liss. Goodman, C. S., Bastiani, M. J., Doe, C. Q., DuLac, S., Helfand, S. L., Kuwada, J. Y. and Thomas, J. B. (1984). Cell recognition during neuronal development. Science 225, Halpern, M. E., Chiba, A., Johansen, J. and Keshishian, H. (1991). Growth cone behavior underlying the development of stereotypic synaptic connections in Drosophila embryos. J. Neurosci. 11, Hartenstein, V. and Campos-Ortega, J. A. (1984). Early neurogenesis in wild-type Drosophila melanogaster. Roux s Arch. Dev. Biol. 193, Hartenstein, V., Rudloff, E. and Campos-Ortega, J. A. (1987). The pattern of proliferation of the neuroblasts in the wild-type embryo of Drosophilamelanogaster. Roux s Arch. Dev. Biol. 196, Hiromi, Y., Kuroiwa, A. and Gehring, W. J. (1985). Control elements of the Drosophila segmentation gene fushi tarazu. Cell 43, Jacobs, J. R., Hiromi, Y., Patel, N. H. and Goodman, C. S. (1989). Lineage, migration and morphogenesis of longitudinal glia in the Drosophila CNS as revealed by a molecular lineage marker. Neuron 2, Jimenez, F. and Campos-Ortega, J. A. (1979). A region of the Drosophila genome necessary for CNS development. Nature 282, Jimenez, F. and Campos-Ortega, J. A. (1990). Defective neuroblast commitment in mutants of the achaete-scute complex and adjacent genes of D. melanogaster. Neuron 5, Kawamura, K. and Carlson, J. G. (1962). Studies on cytokinesis in neuroblasts of the grasshopper, Chortophaga viridifasciata (de geer). III. Factors determining the location of the cleavage furrow. Exp. Cell Res. 26., Klambt, C., Jacobs, J. R. and Goodman, C. S. (1991). The midline of the

12 Drosophila embryonic neuroblast maps 863 Drosophila central nervous system. A model for the genetic analysis of cell fate, cell migration, and growth cone guidance. Cell 64, Lehmann, R., Jimenez, R., Dietrich, V. and Campos-Ortega, J. A. (1983). On the phenotype and development of mutants of early neurogenesis in Drosophila melanogaster. Roux s Arch. Dev. Biol. 192, Martin-Bermudo, M. D., Martinez, C., Rodriguez, A. and Jimenez, F. (1991). Distribution and function of the lethal of scute gene product during early neurogenesis in Drosophila. Development 113, Mlodzik, M., Hiromi, Y., Weber, U., Goodman, C. S. and Rubin, G. M. (1990). The Drosophilaseven-up gene, a member of the steroid receptor gene superfamily, controls photoreceptor cell fates. Cell 60, Patel, N. H., Schafer, B., Goodman, C. S. and Holmgren, R. (1989a). The role of segment polarity genes during Drosophila neurogenesis. Genes and Dev. 3, Patel, N. H., Martin-Blanco, E., Coleman, K. G., Poole, S. J., Ellis, M. C., Kornberg, T. B. and Goodman, C. S. (1989b). Expression of engrailed proteins in arthropods, annelids and chordates. Cell 58, Patel, N. H., Ball, E. E. and Goodman, C. S. (1992). Changing role of even-skipped during the evolution of insect pattern formation. Nature 357, Raper, J. A., Bastiani, M. J. and Goodman, C. S. (1983). Pathfinding by neuronal growth cones in grasshopper embryos I. Divergent choices made by growth cones of sibling neurons. J. Neurosci. 3, Ready, D. F., Hanson, T. E. and Benzer, S. (1976). Development of the Drosophila retina, a neurocrystalline lattice. Dev. Biol. 53, Roberts, D. B. (1986). Drosophila, a Practical Approach. Eynsham, England: IRL Press Limited. Rubin, G. M. (1991). Signal transduction and the fate of the R7 photoreceptor in Drosophila. Trends Genet. 7, Sink, H. and Whitington, P. M. (1990). Location and connectivity of abdominal motoneurons in the embryo and larva of Drosophila melanogaster. J. Neurobiol. 12, Sink, H. and Whitington, P. M. (1991). Pathfinding in the central nervous system and periphery by identified embryonic Drosophila motor axons. Development 112, Skeath, J. B. and Carroll S. B. (1992). Regulation of proneural gene expression and cell fate during neuroblast segregation in the Drosophila embryo. Development 114, Sternberg, P. W. and Horvitz, H. R. (1991). Signal transduction during C. elegans vulval induction. Trends Genet. 7, Sulston, J. E., Schierenberg, E., White, J. G. and Thompson, J. N. (1983). The embryonic cell lineage of the nematode C. elegans. Dev. Biol. 100, Taghert, P. H. and Goodman, C. S. (1984). Cell determination and differentiation of identified serotonin-containing neurons in the grasshopper embryo. J. Neurosci. 4, Thomas, J. B., Bastiani, M. J., Bate, C. M. and Goodman, C. S. (1984). From grasshopper to Drosophila: a common plan for neuronal development. Nature 310, van den Heuvel, M., Nusse, R., Johnston, P. and Lawrence, P. A. (1989). Distribution of the wingless gene product in Drosophila embryos: a protein involved in cell-cell communication. Cell 59, Vaessin, H., Grell, E., Wolff, E., Bier, E., Jan, L. Y. and Jan, Y. N. (1991). prospero is expressed in neuronal precursors and encodes a nuclear protein that is involved in the control of axonal outgrowth in Drosophila. Cell 67, Wheeler, W. M. (1891). Neuroblasts in the arthropod embryo. J. Morph. 4, Wheeler, W. M. (1893). A contribution to insect embryology. J. Morph. 8, (Accepted 11 September 1992)

Neuronal cell death in grasshopper embryos: variable patterns in different species, clutches, and clones

Neuronal cell death in grasshopper embryos: variable patterns in different species, clutches, and clones J. Embryol. exp. Morph. 78, 169-182 (1983) Printed in Great Britain The Company of Biologists Limited 1983 Neuronal cell death in grasshopper embryos: variable patterns in different species, clutches,

More information

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

The Origin, Location, and Projections of the Embryonic Abdominal Motorneurons of Drosophila

The Origin, Location, and Projections of the Embryonic Abdominal Motorneurons of Drosophila The Journal of Neuroscience, December 15, 1997, 17(24):9642 9655 The Origin, Location, and Projections of the Embryonic Abdominal Motorneurons of Drosophila Matthias Landgraf, 1 Torsten Bossing, 2 Gerd

More information

Nirupama Deshpande, Rainer Dittrich, Gerhard M. Technau and Joachim Urban* SUMMARY

Nirupama Deshpande, Rainer Dittrich, Gerhard M. Technau and Joachim Urban* SUMMARY Development 128, 3253-3261 (2001) Printed in Great Britain The Company of Biologists Limited 2001 DEV8801 3253 Successive specification of Drosophila neuroblasts NB 6-4 and NB 7-3 depends on interaction

More information

Cell division genes promote asymmetric interaction between Numb and Notch in the Drosophila CNS

Cell division genes promote asymmetric interaction between Numb and Notch in the Drosophila CNS Development 126, 2759-2770 (1999) Printed in Great Britain The Company of Biologists Limited 1999 DEV5300 2759 Cell division genes promote asymmetric interaction between Numb and Notch in the Drosophila

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

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

The prospero transcription factor is asymmetrically localized to the cell cortex during neuroblast mitosis in Drosophila

The prospero transcription factor is asymmetrically localized to the cell cortex during neuroblast mitosis in Drosophila Development 121, 3187-3195 (1995) Printed in Great Britain The Company of Biologists Limited 1995 3187 The prospero transcription factor is asymmetrically localized to the cell cortex during neuroblast

More information

Cellular Neurobiology BIPN 140 Fall 2016 Problem Set #8

Cellular Neurobiology BIPN 140 Fall 2016 Problem Set #8 Cellular Neurobiology BIPN 140 Fall 2016 Problem Set #8 1. Inductive signaling is a hallmark of vertebrate and mammalian development. In early neural development, there are multiple signaling pathways

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

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

Embryogenesis of an insect nervous system II: A second class of neuron precursor cells and the origin of the intersegmental connectives

Embryogenesis of an insect nervous system II: A second class of neuron precursor cells and the origin of the intersegmental connectives J. Embryol. exp. Morph. Vol. 61,pp. 317-330, 1981 3J7 Printed in Great Britain @ Company of Biologists Limited 1981 Embryogenesis of an insect nervous system II: A second class of neuron precursor cells

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

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

Proneural clusters: equivalence groups in the epithelium of Drosophila

Proneural clusters: equivalence groups in the epithelium of Drosophila Development. 927-932 (199) Printed in Great Britain The Company of Biologists Limited 199 927 Proneural clusters: equivalence groups in the epithelium of Drosophila PAT SIMPSON and CATHIE CARTERET Laboratoire

More information

Pioneering and pathfinding by an identified neuron in the embryonic leech

Pioneering and pathfinding by an identified neuron in the embryonic leech J. Embryol. exp. Morph. 86, 155-167, (1985) 155 Printed in Great Britain The Company of Biologists Limited 1985 Pioneering and pathfinding by an identified neuron in the embryonic leech JOHN Y. KUWADA

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

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

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

The Midline Protein Regulates Axon Guidance by Blocking the Reiteration of Neuroblast Rows within the Drosophila Ventral Nerve Cord

The Midline Protein Regulates Axon Guidance by Blocking the Reiteration of Neuroblast Rows within the Drosophila Ventral Nerve Cord The Midline Protein Regulates Axon Guidance by Blocking the Reiteration of Neuroblast Rows within the Drosophila Ventral Nerve Cord Mary Ann Manavalan, Ivana Gaziova, Krishna Moorthi Bhat* Department of

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

Asymmetric Prospero localization is required to generate mixed neuronal/glial lineages in the Drosophila CNS

Asymmetric Prospero localization is required to generate mixed neuronal/glial lineages in the Drosophila CNS Development 128, 4103-4112 (2001) Printed in Great Britain The Company of Biologists Limited 2001 DEV3509 4103 Asymmetric Prospero localization is required to generate mixed neuronal/glial lineages in

More information

Patterns of peanut agglutinin binding within the developing grasshopper central nervous system

Patterns of peanut agglutinin binding within the developing grasshopper central nervous system /. Embryol. exp. Morph. 90, 49-56 (1985) Printed in Great Britain The Company of Biologists Limited 1985 49 Patterns of peanut agglutinin binding within the developing grasshopper central nervous system

More information

Trans-regulatory functions in the Abdominal-B gene of the bithorax complex

Trans-regulatory functions in the Abdominal-B gene of the bithorax complex Development 101, 117-122 (1987) Printed in Great Britain The Company of Biologists Limited 1987 117 Trans-regulatory functions in the Abdominal-B gene of the bithorax complex JORDI CASANOVA 1 and ROBERT

More information

Expression of engrailed during segmentation in grasshopper and crayfish

Expression of engrailed during segmentation in grasshopper and crayfish Development 107, 201-212 (1989) Printed in Great Britain The Company of Biologists Limited 201 Expression of engrailed during segmentation in grasshopper and crayfish NIPAM H. PATEL 1, THOMAS B. KORNBERG

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

Reading. Lecture VI. Making Connections 9/17/12. Bio 3411 Lecture VI. Making Connections. Bio 3411 Monday September 17, 2012

Reading. Lecture VI. Making Connections 9/17/12. Bio 3411 Lecture VI. Making Connections. Bio 3411 Monday September 17, 2012 Lecture VI. Making Connections Bio 3411 Monday September 17, 2012!! 1! Reading NEUROSCIENCE: 5 th ed, pp!507?536! 4 th ed, pp 577-609 Bentley, D., & Caudy, M. (1983). Nature, 304(5921), 62-65. Dickson,

More information

Glia in the fly wing are clonally related to epithelial cells and use the nerve as a pathway for migration

Glia in the fly wing are clonally related to epithelial cells and use the nerve as a pathway for migration Development 120, 523-534 (1994) Printed in Great Britain The Company of Biologists Limited 1994 523 Glia in the fly wing are clonally related to epithelial cells and use the nerve as a pathway for migration

More information

Asymmetry and cell fate in the Drosophila embryonic CNS

Asymmetry and cell fate in the Drosophila embryonic CNS Int. J. Dev. Biol. 42: 379-383 (1998) Asymmetry and cell fate in the Drosophila embryonic CNS SAL FUERSTENBERG, JULIE BROADUS and CHRIS Q. DOE Department of Cell and Structural Biology, University of Illinois,

More information

Lineage, cell polarity and inscuteable function in the peripheral nervous system of the Drosophila embryo

Lineage, cell polarity and inscuteable function in the peripheral nervous system of the Drosophila embryo Development 128, 631-643 (2001) Printed in Great Britain The Company of Biologists Limited 2001 DEV5434 631 Lineage, cell polarity and inscuteable function in the peripheral nervous system of the Drosophila

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

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

The pattern of cell death in fushi tarazu, a segmentation gene of Drosophila

The pattern of cell death in fushi tarazu, a segmentation gene of Drosophila Development 104, 447-451 (1988) Printed in Great Britain The Company of Biologists Limited 1988 447 The pattern of cell death in fushi tarazu, a segmentation gene of Drosophila LORENZO MAGRASSI and PETER

More information

Reading: Chapter 5, pp ; Reference chapter D, pp Problem set F

Reading: Chapter 5, pp ; Reference chapter D, pp Problem set F Mosaic Analysis Reading: Chapter 5, pp140-141; Reference chapter D, pp820-823 Problem set F Twin spots in Drosophila Although segregation and recombination in mitosis do not occur at the same frequency

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

The maternal sex determination gene daughterless has zygotic activity necessary for the formation of peripheral neurons in Drosophila

The maternal sex determination gene daughterless has zygotic activity necessary for the formation of peripheral neurons in Drosophila The maternal sex determination gene daughterless has zygotic activity necessary for the formation of peripheral neurons in Drosophila Michael Caudy/ Ellsworth H. Grell/ Christine Dambly-Chaudiere,^ Alain

More information

The big brain gene of Drosophila functions to control the number of neuronal precursors in the peripheral nervous system

The big brain gene of Drosophila functions to control the number of neuronal precursors in the peripheral nervous system Development 116, 31-40 (1992) Printed in Great Britain The Company of Biologists Limited 1992 31 The big brain gene of Drosophila functions to control the number of neuronal precursors in the peripheral

More information

Pathfinding in the central nervous system and periphery by identified embryonic Drosophila motor axons

Pathfinding in the central nervous system and periphery by identified embryonic Drosophila motor axons Development 112, 307-316 (1991) Pnnted in Great Britain The Company of Biologists Limited 1991 307 Pathfinding in the central nervous system and periphery by identified embryonic Drosophila motor axons

More information

Kriippel, a Drosophila segmentation gene, participates in the specification of neurons and glial cells

Kriippel, a Drosophila segmentation gene, participates in the specification of neurons and glial cells ELSEVIER Mechanisms of Development 60 (1996) 95-107 Kriippel, a Drosophila segmentation gene, participates in the specification of neurons and glial cells Susana Romania**, Fernando Jimenezb, Michael Hocha,

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

A glial cell arises from an additional division within the mechanosensory lineage during development of the microchaete on the Drosophila notum

A glial cell arises from an additional division within the mechanosensory lineage during development of the microchaete on the Drosophila notum Development 126, 4617-4622 (1999) Printed in Great Britain The Company of Biologists Limited 1999 DEV8615 4617 A glial cell arises from an additional division within the mechanosensory lineage during development

More information

Pathfinding by zebrafish motoneurons in the absence of normal pioneer axons

Pathfinding by zebrafish motoneurons in the absence of normal pioneer axons Development 114, 825-831 (1992) Printed in Great Britain The Company of Biologists Limited 1992 825 Pathfinding by zebrafish motoneurons in the absence of normal pioneer axons SUSAN H. PIKE, ELOINE F.

More information

Specifying the path of the intersegmental nerve of the Drosophila embryo: a role for Delta and Notch

Specifying the path of the intersegmental nerve of the Drosophila embryo: a role for Delta and Notch Development 117, 431-440 (1993) Printed in Great Britain The Company of Biologists Limited 1993 431 Specifying the path of the intersegmental nerve of the Drosophila embryo: a role for Delta and Notch

More information

Development and organization of glial cells in the peripheral nervous system of Drosophila melanogaster

Development and organization of glial cells in the peripheral nervous system of Drosophila melanogaster Development 117, 895-904 (1993) Printed in Great Britain The Company of Biologists Limited 1993 895 Development and organization of glial cells in the peripheral nervous system of Drosophila melanogaster

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

Nature Biotechnology: doi: /nbt Supplementary Figure 1. Overexpression of YFP::GPR-1 in the germline.

Nature Biotechnology: doi: /nbt Supplementary Figure 1. Overexpression of YFP::GPR-1 in the germline. Supplementary Figure 1 Overexpression of YFP::GPR-1 in the germline. The pie-1 promoter and 3 utr were used to express yfp::gpr-1 in the germline. Expression levels from the yfp::gpr-1(cai 1.0)-expressing

More information

Questions in developmental biology. Differentiation Morphogenesis Growth/apoptosis Reproduction Evolution Environmental integration

Questions in developmental biology. Differentiation Morphogenesis Growth/apoptosis Reproduction Evolution Environmental integration Questions in developmental biology Differentiation Morphogenesis Growth/apoptosis Reproduction Evolution Environmental integration Representative cell types of a vertebrate zygote => embryo => adult differentiation

More information

Axon guidance I. Paul Garrity March 15, /9.013

Axon guidance I. Paul Garrity March 15, /9.013 Axon guidance I Paul Garrity March 15, 2004 7.68/9.013 Neuronal Wiring: Functional Framework of the Nervous System Stretch reflex circuit Early theories of axonogenesis Schwann: many neurons link to form

More information

Peripheral Glia Direct Axon Guidance across the CNS/PNS Transition Zone

Peripheral Glia Direct Axon Guidance across the CNS/PNS Transition Zone Developmental Biology 238, 47 63 (2001) doi:10.1006/dbio.2001.0411, available online at http://www.idealibrary.com on Peripheral Glia Direct Axon Guidance across the CNS/PNS Transition Zone Katharine J.

More information

Building the brain (1): Evolutionary insights

Building the brain (1): Evolutionary insights Building the brain (1): Evolutionary insights Historical considerations! Initial insight into the general role of the brain in human behaviour was already attained in antiquity and formulated by Hippocrates

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

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

SPATIAL PROGRAMMING OF GENE EXPRESSION IN EARLY DROSOPHILA EMBRYOGENESIS

SPATIAL PROGRAMMING OF GENE EXPRESSION IN EARLY DROSOPHILA EMBRYOGENESIS Ann. Rev. Cell Biol. 1986.2 : 49-80 Copyright 0 1986 by Annual Reviews Inc. All rights reserved SPATIAL PROGRAMMING OF GENE EXPRESSION IN EARLY DROSOPHILA EMBRYOGENESIS Matthew P. Scott Department of Molecular,

More information

glide directs glial fate commitment and cell fate switch between neurones and

glide directs glial fate commitment and cell fate switch between neurones and Development 122, 131-139 (1996) Printed in Great Britain The Company of Biologists Limited 1996 DEV7433 131 glide directs glial fate commitment and cell fate switch between neurones and glia Stéphane Vincent,

More information

Developmental Dynamics of Peripheral Glia in Drosophila melanogaster

Developmental Dynamics of Peripheral Glia in Drosophila melanogaster GLIA 30:122 133 (2000) Developmental Dynamics of Peripheral Glia in Drosophila melanogaster KATHARINE J. SEPP, JOOST SCHULTE, AND VANESSA J. AULD* Department of Zoology, University of British Columbia,

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

Evidence for differential and redundant function of the Sox genes Dichaete and SoxN during CNS development in Drosophila

Evidence for differential and redundant function of the Sox genes Dichaete and SoxN during CNS development in Drosophila Development 129, 4219-4228 (2002) Printed in Great Britain The Company of Biologists Limited 2002 DEV7977 4219 Evidence for differential and redundant function of the Sox genes Dichaete and SoxN during

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

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

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

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

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

Follow this and additional works at: Part of the Medical Sciences Commons

Follow this and additional works at:   Part of the Medical Sciences Commons Bucknell University Bucknell Digital Commons Master s Theses Student Theses 2010 The overexpression of homeotic complex gene Ultrabithorax in the post-embryonic neuronal lineages of the ventral nervous

More information

A role for wingless in the segmental gradient of Drosophila?

A role for wingless in the segmental gradient of Drosophila? Development 117, 677-687 (1993) Printed in Great Britain The Company of Biologists Limited 1993 677 A role for wingless in the segmental gradient of Drosophila? Javier Sampedro, Paul Johnston and Peter

More information

Baz, Par-6 and apkc are not required for axon or dendrite specification in Drosophila

Baz, Par-6 and apkc are not required for axon or dendrite specification in Drosophila Baz, Par-6 and apkc are not required for axon or dendrite specification in Drosophila Melissa M. Rolls and Chris Q. Doe, Inst. Neurosci and Inst. Mol. Biol., HHMI, Univ. Oregon, Eugene, Oregon 97403 Correspondence

More information

Single-cell mapping of neural and glial gene expression in the developing Drosophila CNS midline cells

Single-cell mapping of neural and glial gene expression in the developing Drosophila CNS midline cells Developmental Biology 294 (2006) 509 524 www.elsevier.com/locate/ydbio Genomes & Developmental Control Single-cell mapping of neural and glial gene expression in the developing Drosophila CNS midline cells

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

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

Formation of the Cortex

Formation of the Cortex Formation of the Cortex Neuronal Birthdating with 3 H-thymidine 3H-thymidine is incorporated into the DNA during the S-phase (replication of DNA). It marks all mitotic cells Quantitative technique. (you

More information

Roles of wingless in patterning the larval epidermis of Drosophila

Roles of wingless in patterning the larval epidermis of Drosophila Development 113, 471-485 (1991) Printed in Great Britain (E) The Company of Biologists Limited 1991 471 Roles of wingless in patterning the larval epidermis of Drosophila AMY BEJSOVEC* and ALFONSO MARTINEZ

More information

Early Development in Invertebrates

Early Development in Invertebrates Developmental Biology Biology 4361 Early Development in Invertebrates October 25, 2006 Early Development Overview Cleavage rapid cell divisions divisions of fertilized egg into many cells Gastrulation

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

16 The Cell Cycle. Chapter Outline The Eukaryotic Cell Cycle Regulators of Cell Cycle Progression The Events of M Phase Meiosis and Fertilization

16 The Cell Cycle. Chapter Outline The Eukaryotic Cell Cycle Regulators of Cell Cycle Progression The Events of M Phase Meiosis and Fertilization The Cell Cycle 16 The Cell Cycle Chapter Outline The Eukaryotic Cell Cycle Regulators of Cell Cycle Progression The Events of M Phase Meiosis and Fertilization Introduction Self-reproduction is perhaps

More information

Conservation of arthropod midline netrin accumulation revealed with a cross-reactive antibody provides evidence for midline cell homology

Conservation of arthropod midline netrin accumulation revealed with a cross-reactive antibody provides evidence for midline cell homology EVOLUTION & DEVELOPMENT 11:3, 260 268 (2009) DOI: 10.1111/j.1525-142X.2009.00328.x Conservation of arthropod midline netrin accumulation revealed with a cross-reactive antibody provides evidence for midline

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

Drosophila Neuroblasts Sequentially Express Transcription Factors which Specify the Temporal Identity of Their Neuronal Progeny

Drosophila Neuroblasts Sequentially Express Transcription Factors which Specify the Temporal Identity of Their Neuronal Progeny Cell, Vol. 106, 511 521, August 24, 2001, Copyright 2001 by Cell Press Drosophila Neuroblasts Sequentially Express Transcription Factors which Specify the Temporal Identity of Their Neuronal Progeny Takako

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

Formation and specification of a Drosophila dopaminergic precursor cell

Formation and specification of a Drosophila dopaminergic precursor cell 3316 RESEARCH ARTICLE AND STEM CELLS Development 139, 3316-3325 (2012) doi:10.1242/dev.079525 2012. Published by The Company of Biologists Ltd Formation and specification of a Drosophila dopaminergic precursor

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

Lateral inhibition and the development of the sensory bristles of the adult peripheral nervous system of Drosophila

Lateral inhibition and the development of the sensory bristles of the adult peripheral nervous system of Drosophila Development 109, 509-519 (1990) Printed in Great Britain The Company of Biologists Limited 1990 Review Article 509 Lateral inhibition and the development of the sensory bristles of the adult peripheral

More information

melanogaster The development of indirect flight muscle innervation in Drosophila Joyce Fernandes 1 and K. VijayRaghavan 1,2, * SUMMARY

melanogaster The development of indirect flight muscle innervation in Drosophila Joyce Fernandes 1 and K. VijayRaghavan 1,2, * SUMMARY Development 118, 215-227 (1993) Printed in Great Britain The Company of Biologists Limited 1993 215 The development of indirect flight muscle innervation in Drosophila melanogaster Joyce Fernandes 1 and

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 processes Differential gene expression Introduction to determination The model organisms used to study developmental processes

Developmental processes Differential gene expression Introduction to determination The model organisms used to study developmental processes Date Title Topic(s) Learning Outcomes: Sept 28 Oct 3 1. What is developmental biology and why should we care? 2. What is so special about stem cells and gametes? Developmental processes Differential gene

More information

Nature Neuroscience: doi: /nn.2662

Nature Neuroscience: doi: /nn.2662 Supplementary Figure 1 Atlastin phylogeny and homology. (a) Maximum likelihood phylogenetic tree based on 18 Atlastin-1 sequences using the program Quicktree. Numbers at internal nodes correspond to bootstrap

More information

Cell Type-Specific Expression of Fasciclin II Isoforms Reveals Neuronal Glial Interactions during Peripheral Nerve Growth

Cell Type-Specific Expression of Fasciclin II Isoforms Reveals Neuronal Glial Interactions during Peripheral Nerve Growth Developmental Biology 234, 24 41 (2001) doi:10.1006/dbio.2001.0247, available online at http://www.idealibrary.com on Cell Type-Specific Expression of Fasciclin II Isoforms Reveals Neuronal Glial Interactions

More information

split ends encodes large nuclear proteins that regulate neuronal cell fate and

split ends encodes large nuclear proteins that regulate neuronal cell fate and Development 127, 1517-1529 (2000) Printed in Great Britain The Company of Biologists Limited 2000 DEV8693 1517 split ends encodes large nuclear proteins that regulate neuronal cell fate and axon extension

More information

The bhlh genes in neural development

The bhlh genes in neural development Int. J. Dev. Biol. 42: 269-273 (1998) The bhlh genes in neural development CHRISTINE DAMBLY-CHAUDIÈRE* 1 and MICHEL VERVOORT 2 1 Laboratoire de Neurogénétique, Unité 432 INSERM, Université Montpellier

More information

Biology 340 Comparative Embryology Lecture 4 Dr. Stuart Sumida. Overview of Pre-Metazoan. and Protostome Development (Insects)

Biology 340 Comparative Embryology Lecture 4 Dr. Stuart Sumida. Overview of Pre-Metazoan. and Protostome Development (Insects) Biology 340 Comparative Embryology Lecture 4 Dr. Stuart Sumida Overview of Pre-Metazoan and Protostome Development (Insects) Plants Fungi Animals In1998 fossilized animal embryos were reported from the

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

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

The Microscopic Observation of Mitosis in Plant and Animal Cells

The Microscopic Observation of Mitosis in Plant and Animal Cells The Microscopic Observation of Mitosis in Plant and Animal Cells Prelab Assignment Before coming to lab, read carefully the introduction and the procedures for each part of the experiment, and then answer

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

MCN. Complex Genetic Interactions among Four Receptor Tyrosine Phosphatases Regulate Axon Guidance in Drosophila

MCN. Complex Genetic Interactions among Four Receptor Tyrosine Phosphatases Regulate Axon Guidance in Drosophila MCN Molecular and Cellular Neuroscience 17, 274 291 (2001) doi:10.1006/mcne.2000.0939, available online at http://www.idealibrary.com on Complex Genetic Interactions among Four Receptor Tyrosine Phosphatases

More information

MBios 401/501: Lecture 14.2 Cell Differentiation I. Slide #1. Cell Differentiation

MBios 401/501: Lecture 14.2 Cell Differentiation I. Slide #1. Cell Differentiation MBios 401/501: Lecture 14.2 Cell Differentiation I Slide #1 Cell Differentiation Cell Differentiation I -Basic principles of differentiation (p1305-1320) -C-elegans (p1321-1327) Cell Differentiation II

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

Analysis of the effects of encystment and excystment on incomplete doublets of Oxytricha fallax

Analysis of the effects of encystment and excystment on incomplete doublets of Oxytricha fallax /. Embryo/, exp. Morph. Vol. 59, pp. 19-26, 1980 19 Printed in Great Britain Company of Biologists Limited 1980 Analysis of the effects of encystment and excystment on incomplete doublets of Oxytricha

More information

1(1)pole hole is required maternally for pattern formation in the terminal regions of the embryo

1(1)pole hole is required maternally for pattern formation in the terminal regions of the embryo Development 106, 145-158 (1989) Printed in Great Britain The Company of Biologists Limited 1989 145 1(1)pole hole is required maternally for pattern formation in the terminal regions of the embryo LINDA

More information

Introduction Principles of Signaling and Organization p. 3 Signaling in Simple Neuronal Circuits p. 4 Organization of the Retina p.

Introduction Principles of Signaling and Organization p. 3 Signaling in Simple Neuronal Circuits p. 4 Organization of the Retina p. Introduction Principles of Signaling and Organization p. 3 Signaling in Simple Neuronal Circuits p. 4 Organization of the Retina p. 5 Signaling in Nerve Cells p. 9 Cellular and Molecular Biology of Neurons

More information

Asymmetric cell division: from A to Z

Asymmetric cell division: from A to Z REVIEW Asymmetric cell division: from A to Z Nancy Hawkins 1 and Gian Garriga Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3204 USA Cell divisions producing

More information

Supplementary Figures

Supplementary Figures Supplementary Figures Supplementary Fig. S1: Normal development and organization of the embryonic ventral nerve cord in Platynereis. (A) Life cycle of Platynereis dumerilii. (B-F) Axonal scaffolds and

More information