sloppy paired 1


DEVELOPMENTAL BIOLOGY

Embryonic

Transcription of slp is first detected in the head, in a gap-like pattern during the syncytial blastoderm. Seven primary segmental stripes appear during the cellular blastoderm followed by seven additional secondary stripes between the primary stripes. By germ band expansion [Images], expression in the head is more complex, due to the development of the different head primordia. The metameric pattern fades at germ band retraction and transcript appears in the border of the dorsal epidermis, during dorsal closure. slp2 lags slp1 in many of these expression features (Grossnicklaus, 1992) .

The Drosophila brain develops from the procephalic neurogenic region of the ectoderm. About 100 neural precursor cells (neuroblasts) delaminate from this region on either side in a reproducible spatiotemporal pattern. Neuroblast maps have been prepared from different stages of the early embryo (stages 9, 10 and 11, when the entire population of neuroblasts has formed), in which about 40 molecular markers representing the expression patterns of 34 different genes are linked to individual neuroblasts. In particular, a detailed description is presented of the spatiotemporal patterns of expression in the procephalic neuroectoderm and in the neuroblast layer of the gap genes empty spiracles, hunchback, huckebein, sloppy paired 1 and tailless; the homeotic gene labial; the early eye genes dachshund, eyeless and twin of eyeless; and several other marker genes (including castor, pdm1, fasciclin 2, klumpfuss, ladybird, runt and unplugged). Based on the combination of genes expressed, each brain neuroblast acquires a unique identity, and it is possible to follow the fate of individual neuroblasts through early neurogenesis. Furthermore, despite the highly derived patterns of expression in the procephalic segments, the co-expression of specific molecular markers discloses the existence of serially homologous neuroblasts in neuromeres of the ventral nerve cord and the brain. Taking into consideration that all brain neuroblasts are now assigned to particular neuromeres and individually identified by their unique gene expression, and that the genes found to be expressed are likely candidates for controlling the development of the respective neuroblasts, these data provide a basic framework for studying the mechanisms leading to pattern and cell diversity in the Drosophila brain, and for addressing those mechanisms that make the brain different from the truncal CNS (Urbach, 2003).

The cephalic gap genes are expressed in large domains of the procephalon and play a crucial role not only in the patterning of the peripheral ectoderm, but also in regionalizing the brain primordium. The segmental organization of the Drosophila brain is based on the expression pattern of segment polarity and DV patterning genes. To see whether the cephalic gap genes respect the neuromeric boundaries segment polarity and DV patterning genes, and to provide a basis for studying their potential role in the formation or specification of brain precursor cells, the expression was studied of orthodenticle, empty spiracles, sloppy paired 1, tailless, huckebein, and hunchback in the developing head ectoderm, as well as in the entire population of identified NBs during stages 9-11 (Urbach, 2003).

The sloppy paired (slp) locus contains the two related genes slp1 and slp2. slp1, which acts as a head gap gene, plays a predominant role in head formation, while slp2 is largely dispensable. In the trunk neuroectoderm, where slp1 has a function as a pair-rule and segment polarity gene, it is segmentally expressed in neuroectodermal stripes as well as in NBs of row 4 and 5. This segmental appearance of slp1 expression is found to be conserved in parts of the procephalon. In the blastoderm, Slp1 protein is detected in a large domain of the procephalon anlage, which subsequently diminishes in its anterior/ventral part. As a result, only the posterior half of the original slp1 domain remains as a circumferential ring ('head stripe') and gets separated from the anterodorsal part ('head cap'). To follow the dynamics in the Slp1 expression pattern, Slp1/En double labelling was examined during stages 8-11. The 'head stripe' corresponds to the slp1 stripe of the prospective mandibular segment, and the posterior part of the 'head cap' to the Slp1-positive stripe in the prospective antennal segment (slp1 as). At the beginning of gastrulation, a new Slp1 ectodermal spot in the anterodorsal procephalon is observed; this spot later becomes part of the labral ectoderm. In addition, at stage 9, three new ectodermal domains become detectable: one stripe anterior to the en intercalary stripe belonging to the intercalary segment, and two small spots in the region of the ocular segment (anterior to the en head spot). Except for the labral domain, the slp1 domains contribute NBs to the brain. Thus, slp1 is segmentally expressed in the procephalic neuroectoderm and subsets of brain NBs, resembling the situation in the trunk. At stage 11 patchy expression of Slp1 becomes detectable within the ocular and labral ectoderm and in some underlying ocular and labral NBs. Some of these NBs initiate slp1 expression after delamination; e.g. Pcv6 and Pcd2 delaminate at stage 9 and do not express slp1 before stage 11. Slp1 expression is observed in the brain until the end of embryogenesis (Urbach, 2003).

Larval

Very little information is available about gene expression during the larval period, a developmental interval critical to the formation of the adult. To what extent does gene expression during this period resemble that in the embryonic stages, and how does gene expression during the larval period contribute to segment polarity in the adult? In fact, all the genes expressed during embryonic segment polarity also play a similar role in the formation of the adult. Cells destined to form the cuticle of the adult abdomen are present as clusters of small, non-dividing diploid cells (the anterior dorsal, posterior dorsal and ventral histoblast nests) located at stereotyped postions in the larval epidermis. These cells, just as do their embryonic counterparts, express engrailed, hedgehog, wingless, patched, cubitus interruptus and sloppy paired in a stereotyped manner dependent on their positions within each segment. Each segment is subdivided into an anterior (A) and posterior (P) compartment, distinguished by activity of the selector gene engrailed (en) in P but not A compartment cells. The ventral epidermis of each abdominal segment forms a flexible cuticle, the pleura, with a small plate of sclerotised cuticle, the sternite, centered on the ventral midline. The pleura is covered with a uniform lawn of hairs, all pointed posteriorly, whereas the sternite contains a stereotyped pattern of bristles. Posterior compartments are to a large degree devoid of hairs and bristles, while the sternite cuticle of the A compartment consists of an anterior-to posterior progression of six types of cuticle distinguished by ornamentation and pigmentation. Just anterior to the posterior compartment, A6 is unpigmented, with hairs and none of the larger ornaments called bristles. A5 is darkly pigmented with hairs and bristles of large size. A4 and A3 are darkly and lightly pigmented respectively with moderately sized hairs and bristles. A2 is lightly pigmented with hairs, and A1, adjacent to the next more anteriorly located "posterior" compartment is unpigmented without hairs (Struhl, 1997a).

Hedgehog (Hh), a protein secreted by engrailed expressing P compartment cells, spreads into each A compartment across the anterior and the posterior boundaries to form opposing concentration gradients that organize cell pattern and polarity. Anteriorly and posteriorly situated cells within the A compartment respond in distinct ways to Hh: they express different combinations of genes and form different cell types. patched is expressed at both boundaries. patched is expressed in a graded fashion within each stripe, just anterior to each P compartment. ci peaks at high level in those cells abutting Hh- secreting cells of the P compartment and declines progressively in cells further away. wingless is also expressed in this domain and sloppy paired is expressed in the same region as wingless. decapentaplegic is expressed only in the ventral pleura in those A compartment cells neighboring P compartment cells within the same segment. dpp is not expressed immediately behind posterior compartments (Struhl, 1997).

A temporal mechanism that produces neuronal diversity in the Drosophila visual center

The brain consists of various types of neurons that are generated from neural stem cells; however, the mechanisms underlying neuronal diversity remain uncertain. A recent study demonstrated that the medulla, the largest component of the Drosophila optic lobe, is a suitable model system for brain development because it shares structural features with the mammalian brain and consists of a moderate number and various types of neurons. The concentric zones in the medulla primordium that are characterized by the expression of four transcription factors, including Homothorax (Hth), Brain-specific homeobox (Bsh), Runt (Run) and Drifter (Drf/Vvl), correspond to types of medulla neurons. This study examined the mechanisms that temporally determine the neuronal types in the medulla primordium. For this purpose, transcription factors were sought that are transiently expressed in a subset of medulla neuroblasts (NBs, neuronal stem cell-like neural precursor cells) and identified five candidates [Hth, Klumpfuss (Klu), Eyeless (Ey), Sloppy paired (Slp) and Dichaete (D)]. The results of genetic experiments at least explain the temporal transition of the transcription factor expression in NBs in the order of Ey, Slp and D. The results also suggest that expression of Hth, Klu and Ey in NBs trigger the production of Hth/Bsh-, Run- and Drf-positive neurons, respectively. These results suggest that medulla neuron types are specified in a birth order-dependent manner by the action of temporal transcription factors that are sequential ly expressed in NBs (Suzuki, 2013).

In the embryonic central nervous system, the heterochronic transcription factors suchas Hb, Kr, Pdm, Cas and Grh are expressed in NBs to regulate the temporal specification of neuronal identity. They regulate each other to achieve sequential changes in their expression in NBs without cell-extrinsic factors. However, expression of the embryonic heterochronic genes was not detected in the medulla NBs.Instead this study found that Hth, Klu, Ey, Slp and D are transiently and sequentially expressed in medulla NBs. The expression of Hth and Klu was observed in lateral NBs, while that of Ey/Slp and D was observed in intermediate and medial NBs, respectively. These observations suggest that the expression of heterochronic transcription factors changes sequentially as each NB ages, as observed in the development of the embryonic central nervous system (Suzuki, 2013).

This study demonstrates that at least three of the temporal factors Ey, Slp and D regulate each other to form a genetic cascade that ensures the transition from Ey expression to D expression in the medulla NBs. Ey expression in NBs activates Slp, while Slp inactivates Ey expression. Similarly, Slp expression in NBs activates D expression, while D inactivates Slp expression. In fact, the expression of Slp is not strong in newer NBs in which Ey is strongly expressed, but is up regulated in older NBs in which Ey is weakly expressed in the wildtype medulla. A similar relationship is found between Slp and D, supporting the idea that Ey, Slp and D regulate each other's expression to control the transition from Ey-expression to D-expression. In the embryonic central nervous system, similar interaction is mainly observed between adjacent genes of the cascade hb-Kr-pdm-cas-grh, and this concept may also be applied to the medulla primordium. The expression pattern and function of Ey, Slp and D suggest that they are adjacent to each other in the cascade of transcription factor expression in medulla NBs (Suzuki, 2013).

However, no such relationship was found between Hth, Klu and the other temporal factors.The sequential expression of Hth and Klu could be regulated by an unidentified mechanism that is totally different from the genetic cascade that controls the transition through Ey-Slp-D. Or, there might be unidentified temporal factors that are expressed in lateral NBs which act upstream of Hth and Klu to regulate their expression. It is necessary to identify additional transcription factors that are transiently expressed in medulla NBs (Suzuki, 2013).

The expression of concentric transcription factors in the medulla neurons correlates with the temporal sequence of neuron production from the medulla NBs (Hasegawa, 2011). In the larval medulla primordium, the neurons are located in the order of Hth/Bsh-, Run- and Drf-positive cells from inside to outside, and these domains are adjacent to each other (Hasegawa, 2011). Given that NBs generate neurons toward the center of the developing medulla, Hth/Bsh-positive neurons are produced at first, and then Run-positive and Drf-positive neurons. Thus Hth/Bsh, Run and Drf were used as markers to examine roles of Hth, Klu, Ey, Slp and D expressed in NBs in specifying types of medulla neurons. The continuous expression of Hth and Ey from NBs to neurons and the results of clonal analyses that visualize the progeny of NBs expressing each one of the temporal transcription factors suggest that the temporal windows of NBs expressing Hth, Klu and Ey approximately correspond to the production of Hth/Bsh-, Run- and Drf- positive neurons, respectively. Indeed, the results of the genetic study suggest that Hth and Ey are necessary and sufficient to induce the production of Hth/Bsh- and Drf-positive neurons,respectively (Hasegawa, 2011, 2013). Ectopic Klu expression at least induces the produc tion of Run-positive neurons (Suzuki, 2013).

Slp and D expression in NBs may correspond to the temporal windows that produce medulla neurons in the outer domains of the concentric zones, which are most likely produced after the production of Drf-positive neurons. The results at least suggest that Slp is necessary and sufficient and D is sufficient to repress the production of Drf-positive neurons. Identification of additional markers that are expressed in the outer concentric zones compared to the Drf-positive domain would be needed to elucidate the roles of Slp and D in specification of medulla neuron types (Suzuki, 2013).

D mutant clones did not produce any significant phenotype except for derepression of Slp expression in NBs. Drf expression in neurons was not affected either. Since D is a Sox family transcription factor, SoxN, another Sox family transcription factor, is a potential candidate molecule that acts together with D in the medulla NBs. However, its expression was found in neuroepithelia cells and lateral NBs that overlap with Hth-positive cells but not with D-positive cells. All the potential heterochronic transcription factors examined in this study are expressed in three to five cell rows of NBs. Nevertheless, one NB has been observed to produce one Bsh- positive and one Run-positive neuron (Hasegawa, 2011). Therefore, the expression pattern of the heterochronic transcription factors is not sufficient to explain the stable production of one Bsh-positive and one Run-positive neuron from a single NB.The combinatorial action of multiple temporal factors expressed in NBs may play important roles in the specification of Bsh- and Run- positive neurons (Suzuki, 2013).

Another possible mechanism that guarantees the production of a limited number of the same neuronal type from multiple rows of NBs expressing a temporal transcription factor could be a mutual repression between concentric transcription factors expressed in medulla neurons. For example, Hth/Bsh, Run and Drf may repress each other to restrict the number of neurons that express either of these transcription factors. However, expression of Run and Drf was not essentially affected in hth mutant clones and in clones expressing Hth (Hasegawa, 2011). Similarly, expression of Hth and Drf was not essentially affected in clones expressing run RNAi under the control of AyGal4, in which Run expression is eliminated. Hth and Run expression was not affected in drf mutant clones (Hasegawa, 2011). These results suggest that Hth/Bsh, Run and Drf do not essentially regulate each other during the formation of concentric zones in the medulla (Suzuki, 2013).

During embryonic development, the heterochronic genes that are expressed in NBs (hb-Kr-pdm-cas-grh) are maintained and act in GMCs to specify neuronal type. Similarly, Hth and Ey are continuously expressed from NBs to neurons, suggesting that their expression may also be inherited through GMCs (Hasegawa, 2011). However, this type of regulatory mechanism may be somewhat modified in the case of Klu, Slp and D (Suzuki, 2013).

Klu is expressed in NBs and GMCs, but not in neurons. Slp and D are predominantly detected in NBs and neurons visualized by Dpn and Elav, respectively. Occasionally, however, expression of D was found in putative GMCs, which are situated between NBs and neurons. Additionally, both D-positive and D-negative cells were found among Miranda-positive GMCs. Slp expression was not found in Miranda-positive GMCs. Finally, D is expressed in medulla neurons forming a concentric zone in addition to its expression in medial NBs. However, D expression was abolished in slp mutant NBs but remained in the mutant neurons, suggesting that D expression in medulla neurons is not inherited from the NBs. These results suggest that Slp and D expression are not maintained from NBs to neurons and that not all the temporal transcription factors expressed in NBs are inherited through GMCs. However, it is possible to speculate that Klu, Slp and D regulate expression of unidentified transcription factors in NBs that are inherited from NBs to neurons through GMCs (Suzuki, 2013).

Effects of Mutation or Deletion

A severe segmentation phenotype is obtained only when both slp genes are deleted. The phenotypes of embryos containing various combinations of functional slp genes suggest that for early slp function, at least until gastrulation, only slp1 is required. At later times, there is still a greater requirement for slp1, but in many respects the two slp genes are completely redundant. Both slp genes produce similar phenotypes when ubiquitously expressed via a heat shock promoter. It has been suggested that the SLP proteins are biochemically equivalent and that the greater requirement for slp1 in some functions can be explained in large part by its earlier expression (Cadigan, 1994b).

Structures derived from the mandibular and labial segments are missing in slp mutants. These deficiencies are due to partial and defective expression of wingless and engrailed in slp mutants (Grossnicklaus, 1994).


REFERENCES

Ahlgren, S., Vogt, P., Bronner-Fraser, M. (2003). Excess FoxG1 causes overgrowth of the neural tube. J. Neurobiol. 57(3): 337-49. 14608667

Alexandre, C. and Vincent, J. P. (2003). Requirements for transcriptional repression and activation by Engrailed in Drosophila embryos. Development 130: 729-739. 12506003

Andrioli, L. P. M., et al. (2002). Anterior repression of a Drosophila stripe enhancer requires three position-specific mechanisms. Development 129: 4931-4940. 12397102

Andrioli, L. P., Digiampietri, L. A., de Barros, L. P. and Machado-Lima, A. (2012). Huckebein is part of a combinatorial repression code in the anterior blastoderm. Dev. Biol. 361(1): 177-85. PubMed Citation: 22027434

Azpiazu, N., et al. (1996). Segmentation and specification of the Drosophila mesoderm. Genes Dev. 10: 3183-94. PubMed Citation: 8985186

Baumgartner, S., Martin, D., Hagios, C. and Chiquet-Ehrismann, R. (1994). Tenm, a Drosophila gene related to tenascin, is a new pair-rule gene. EMBO J 13: 3728-3740. PubMed Citation: 8070401

Bertet, C., Li, X., Erclik, T., Cavey, M., Wells, B., Desplan, C. (2014) Temporal patterning of neuroblasts controls Notch-mediated cell survival through regulation of Hid or Reaper. Cell 158: 1173-1186. PubMed ID: 25171415

Bhat, K. M., van Beers, E. H. and Bhat, P. (2000). Sloppy paired acts as the downstream target of Wingless in the Drosophila CNS and interaction between sloppy paired and gooseberry inhibits sloppy paired during neurogenesis. Development 127(3): 655-665. PubMed Citation: 10631185

Bourguignon, C., Li, J. and Papalopulu, N. (1998). XBF-1, a winged helix transcription factor with dual activity, has a role in positioning neurogenesis in Xenopus competent ectoderm. Development 125(24): 4889-900. PubMed Citation: 9811573

Buescher, M., Svendsen, P. C., Tio, M., Miskolczi-McCallum, C., Tear, G., Brook, W. J. and Chia, W. (2004). Drosophila T box proteins break the symmetry of hedgehog-dependent activation of wingless. Curr. Biol. 14(19): 1694-702. 15458640

Cadigan, K. M., Grossniklaus, U. and Gehring, W, J., (1994a). Localized expression of sloppy paired protein maintains the polarity of Drosophila parasegments. Genes Dev 8: 899-913. PubMed Citation: 7926775

Cadigan, K. M., Grossniklaus, U. and Gehring, J. W. (1994b). Functional redundancy: the respective roles of the two sloppy paired genes in Drosophila segmentation. Proc. Natl. Acad. Sci. 91: 6324-6328. PubMed Citation: 8022780

Choe, C. P. and Brown, S. J. (2007). Evolutionary flexibility of pair-rule patterning revealed by functional analysis of secondary pair-rule genes, paired and sloppy-paired in the short-germ insect, Tribolium castaneum. Dev. Biol. 302(1): 281-94. PubMed citation: 17054935

Danesin, C., et al. (2009). Integration of telencephalic Wnt and Hedgehog signaling center activities by Foxg1. Dev. Cell 16: 576-587. PubMed Citation: 19386266

Fujioka, M., Jaynes, J. B. and Goto, T. (1995). Early even-skipped stripes act as morphogenetic gradients at the single cell level to establish engrailed expression. Development 121: 4371-4382. PubMed Citation: 8575337

Fujioka, M. and Jaynes, J. B. (2012a). Regulation of a duplicated locus: Drosophila sloppy paired is replete with functionally overlapping enhancers. Dev. Biol. 362: 309-319. PubMed Citation: 22178246

Fujioka, M., Gebelein, B., Cofer, Z. C., Mann, R. S. and Jaynes, J. B. (2012b). Engrailed cooperates directly with Extradenticle and Homothorax on a distinct class of homeodomain binding sites to repress sloppy paired. Dev. Biol. 366(2): 382-92. PubMed Citation: 22537495

Gebelein. B., McKay, D. J. and Mann, R. S. (2004). Direct integration of Hox and segmentation gene inputs during Drosophila development. Nature 431: 653-659. 16556799

Goldstein, R. E., et al. (2005). An eh1-like motif in Odd-skipped mediates recruitment of Groucho and repression in vivo. Mol. Cell. Biol. 25(24): 10711-20. 16314497

Goriely, A., et al. (1996). A functional homologue of goosecoid in Drosophila. Development 122: 1641-1650

Grossniklaus, U., Pearson, R. K. and Gehring, W. J. (1992). The Drosophila sloppy paired locus encodes two proteins involved in segmentation that show homology to mammalian transcription factors. Genes Dev 6: 1030-51

Grossniklaus, U., Cadigan, K. M. and Gehring, W. J. (1994). Three maternal coordinate systems cooperate in the patterning of the Drosophila head. Development 120: 3155-3171

Hacker, U., et al. (1995). The Drosophila fork head domain protein crocodile is required for the establishment of head structures. EMBO J 14: 5306-5317

Hanashima, C., Li, S. C., Shen, L., Lai, E. and Fishell, G. (2004). Foxg1 suppresses early cortical cell fate. Science 303(5654): 56-9. 14704420

Hardcastle, Z. and Papalopulu, N. (2000). Distinct effects of XBF-1in regulating the cell cycle inhibitor p27 XIC1 and imparting a neural fate. Development 127: 1303-1314

Hasegawa, E., Kitada, Y., Kaido, M., Takayama, R., Awasaki, T., Tabata, T. and Sato, M. (2011). Concentric zones, cell migration and neuronal circuits in the Drosophila visual center. Development 138: 983-993. PubMed ID: 21303851

Hasegawa, E., Kaido, M., Takayama, R. and Sato, M. (2013). Brain-specific-homeobox is required for the specification of neuronal types in the Drosophila optic lobe. Dev Biol 377: 90-99. PubMed ID: 23454478

He, A., Kong, S. W., Ma, Q., and Pu, W. T. (2011). Co-occupancy by multiple cardiac transcription factors identifies transcriptional enhancers active in heart. Proc. Natl. Acad. Sci. 108: 5632-5637. PubMed Citation: 21415370

Junion, G., et al. (2012). A transcription factor collective defines cardiac cell fate and reflects lineage history. Cell 148(3): 473-86. PubMed Citation: 22304916

Kobayashi, M., et al. (2001). Groucho augments the repression of multiple Even skipped target genes in establishing parasegment boundaries. Development 128(10): 1805-1815. 11311161

Kobayashi, M., et al. (2003). Engrailed cooperates with extradenticle and homothorax to repress target genes in Drosophila. Development 130: 741-751. 12506004

Konstantinides, N., Holguera, I., ...., Walldorf, U., Roussos, P. and Desplan, C. (2022). A complete temporal transcription factor series in the fly visual system. Nature 604(7905): 316-322. PubMed ID: 35388222

Lee, H.-H. and Frasch, M. (2000). Wingless effects mesoderm patterning and ectoderm segmentation events via induction of its downstream target sloppy paired. Development 127: 5497-5508

Lee, H. H. and Frasch, M. (2005). Nuclear integration of positive Dpp signals, antagonistic Wg inputs and mesodermal competence factors during Drosophila visceral mesoderm induction. Development 132: 1429-1442. 15750188

Li, X., Erclik, T., Bertet, C., Chen, Z., Voutev, R., Venkatesh, S., Morante, J., Celik, A. and Desplan, C. (2013). Temporal patterning of Drosophila medulla neuroblasts controls neural fates. Nature 498: 456-462. PubMed ID: 23783517

Lujan, E., et al. (2012). Direct conversion of mouse fibroblasts to self-renewing, tripotent neural precursor cells. Proc. Natl. Acad. Sci. 109(7): 2527-32. PubMed Citation: 22308465

Manuel, M., et al. (2010). The transcription factor Foxg1 regulates the competence of telencephalic cells to adopt subpallial fates in mice. Development 137(3): 487-97. PubMed Citation: 20081193

Mariani, F. V. and Harland, R. M. (1998). XBF-2 is a transcriptional repressor that converts ectoderm into neural tissue. Development 125(24): 5019-31. PubMed Citation: 9811586

Molin, L., et al. (2000). Evolutionary conservation of redundancy between a diverged pair of forkhead transcription factor homologues. Development 127: 4825-4835. PubMed Citation: 11044397

Nasiadka, A. and Krause, H. M. (1999). Kinetic analysis of segmentation gene interactions in Drosophila embryos. Development 126: 1515-1526. PubMed Citation: 10068644

Ochoa-Espinosa, A., Yucel, G., Kaplan, L., Pare, A., Pura, N., Oberstein, A., Papatsenko, D. and Small S. (2005). The role of binding site cluster strength in Bicoid-dependent patterning in Drosophila. Proc. Natl. Acad. Sci. 102(14): 4960-5. 15793007

Plyte, S. E., et al. (1999). Glycogen synthase kinase-3 (GSK-3) is regulated during Dictyostelium development via the serpentine receptor cAR3. Development 126(2): 325-333. PubMed Citation: 9847246

Pratt, T., et al. (2004). The winged helix transcription factor Foxg1 facilitates retinal ganglion cell axon crossing of the ventral midline in the mouse. Development 131: 3773-3784. 15240555

Prazak, L., Fujioka. M. and Gergen, J. P. (2010). Non-additive interactions involving two distinct elements mediate sloppy-paired regulation by pair-rule transcription factors. Dev. Biol. 344: 1048-1059. PubMed Citation: 20435028

Ray, A. and Li, X. (2022). A Notch-dependent transcriptional mechanism controls expression of temporal patterning factors in Drosophila medulla. Elife 11. PubMed ID: 36040415

Riechmann, V., et al. (1997). Control of cell fates and segmentation in the Drosophila mesoderm. Development 124(15): 2915-2922. PubMed Citation: 9247334

Roth, M., et al. (2010). FoxG1 and TLE2 act cooperatively to regulate ventral telencephalon formation. Development 137(9): 1553-62. PubMed Citation: 20356955

Sasaki, H. and Hogan, B. L. M. (1993). Differentail expression of multiple fork head related genes during gastrulation and axial pattern formation in the mouse embryo. Development 118: 47-59

Schroeder, M. D., Greer, C. and Gaul, U. (2011). How to make stripes: deciphering the transition from non-periodic to periodic patterns in Drosophila segmentation. Development 138(14): 3067-78. PubMed Citation: 21693522

Shimamura, K. and Rubenstein, J. L. (1997). Inductive interactions direct early regionalization of the mouse forebrain. Development 124(14): 2709-2718.

Skeath, J. B., et al. (1992). Gene regulation in two dimensions: the proneural achaete-scute genes are controlled by combinations of axis-patterning genes through a common intergenic control region. Genes and Dev. 6: 2606-2619.

Schlesinger, J., et al. (2011). The cardiac transcription network modulated by Gata4, Mef2a, Nkx2.5, Srf, histone modifications, and microRNAs. PLoS Genet. 7: e1001313. PubMed Citation: 21379568

Struhl, G., Barbash, D. A. and Lawrence, P. A. (1997). Hedgehog organizes the pattern and polarity of epidermal cells in the Drosophila abdomen. Development 124 (11): 2143-2154. PubMed Citation: 9187141

Suzuki, T., Kaido, M., Takayama, R. and Sato, M. (2013). A temporal mechanism that produces neuronal diversity in the Drosophila visual center. Dev Biol 380: 12-24. PubMed ID: 23665475

Swantek, D. and Gergen, J. P. (2004). Ftz modulates Runt-dependent activation and repression of segment-polarity gene transcription. Development 131: 2281-2290. 15102703

Tan, K., et al. (2003). Human PLU-1 Has transcriptional repression properties and interacts with the developmental transcription factors BF-1 and PAX9. J. Biol. Chem. 278(23): 20507-13. 12657635

Tao, W. and Lai, E. (1992). Telencephalon-restricted expression of BF-1, a new member of the HNF-3/fork head gene family, in the developing rat brain. Neuron 8(5): 957-966. PubMed Citation: 1350202

Thomas, G. M., et al. (1999). A GSK3-binding peptide from FRAT1 selectively inhibits the GSK3-catalysed phosphorylation of Axin and beta-catenin. FEBS Lett. 458(2): 247-251. PubMed Citation: 10481074

Toresson H., et al. (1998). Conservation of BF-1 expression in amphioxus and zebrafish suggests evolutionary ancestry of anterior cell types that contribute to the vertebrate telencephalon. Dev. Genes Evol. 208(8): 431-439. PubMed Citation: 9799423

Urbach, R. and Technau, G. M. (2003). Molecular markers for identified neuroblasts in the developing brain of Drosophila. Development 130: 3621-3637. 12835380

Wang, X., Lee, C., Gilmour, D. S. and Gergen, J. P. (2007). Transcription elongation controls cell fate specification in the Drosophila embryo. Genes Dev. 21(9): 1031-6. Medline abstract: 17473169

Wu, C. H., Lee, C., Fan, R., Smith, M. J., Yamaguchi, Y., Handa, H., and Gilmour, D. S. (2005). Molecular characterization of Drosophila NELF. Nucleic Acids Res. 33: 1269-1279. Medline abstract: 15741180

Xiang, J., Reding, K., Heffer, A. and Pick, L. (2017) Conservation and variation in pair-rule gene expression and function in the intermediate-germ beetle, Dermestes maculatus. Development 144(24):4625-4636. PubMed ID: 29084804

Yao, J., Lai, E. and Stifani, S. (2001). The winged-helix protein Brain Factor 1 interacts with Groucho and Hes proteins to repress transcription. Mol. Cell. Biol. 21: 1962-1972. 11238932

Yu, Y., et al. (1999). A double interaction screen identifies positive and negative ftz gene regulators and Ftz-interacting proteins. Mech. Dev. 83(1-2): 95-105. PubMed Citation: 10381570


sloppy paired 1: Biological Overview | Evolutionary Homologs | Regulation | Developmental Biology | Effects of Mutation

date revised: 12 April 2018

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

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