hairy mediates dominant repression in the Drosophila embryo (original) (raw)

Abstract

hairy encodes a bHLH repressor that regulates several developmental processes in Drosophila, including embryonic segmentation and neurogenesis. Segmentation repressors such as Krüppel and knirps have been shown to function over short distances, less than 50-100 bp, to inhibit or quench closely linked upstream activators. This mode of repression permits multiple enhancers to work independently of one another within a modular promoter. Here, we employ a transgenic embryo assay to present evidence that hairy acts as a dominant repressor, which can function over long distances to block multiple enhancers. hairy is shown to repress a heterologous enhancer, the rhomboid NEE, when bound 1 kb from the nearest upstream activator. Moreover, the binding of hairy to a modified NEE leads to the repression of both the NEE and a distantly linked mesoderm-specific enhancer within a synthetic modular promoter. Additional evidence that hairy is distinct from previously characterized embryonic repressors stems from the analysis of the gypsy insulator DNA. This insulator selectively blocks the hairy repressor, but not the linked activators, within a modified NEE. We compare hairy with previously characterized repressors and discuss the consequences of short-range and long-range repression in development.

Full Text

The Full Text of this article is available as a PDF (263.8 KB).

Selected References

These references are in PubMed. This may not be the complete list of references from this article.

  1. Akazawa C., Sasai Y., Nakanishi S., Kageyama R. Molecular characterization of a rat negative regulator with a basic helix-loop-helix structure predominantly expressed in the developing nervous system. J Biol Chem. 1992 Oct 25;267(30):21879–21885. [PubMed] [Google Scholar]
  2. Arnosti D. N., Barolo S., Levine M., Small S. The eve stripe 2 enhancer employs multiple modes of transcriptional synergy. Development. 1996 Jan;122(1):205–214. doi: 10.1242/dev.122.1.205. [DOI] [PubMed] [Google Scholar]
  3. Barbash D. A., Cline T. W. Genetic and molecular analysis of the autosomal component of the primary sex determination signal of Drosophila melanogaster. Genetics. 1995 Dec;141(4):1451–1471. doi: 10.1093/genetics/141.4.1451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Benezra R., Davis R. L., Lockshon D., Turner D. L., Weintraub H. The protein Id: a negative regulator of helix-loop-helix DNA binding proteins. Cell. 1990 Apr 6;61(1):49–59. doi: 10.1016/0092-8674(90)90214-y. [DOI] [PubMed] [Google Scholar]
  5. Bier E., Vaessin H., Younger-Shepherd S., Jan L. Y., Jan Y. N. deadpan, an essential pan-neural gene in Drosophila, encodes a helix-loop-helix protein similar to the hairy gene product. Genes Dev. 1992 Nov;6(11):2137–2151. doi: 10.1101/gad.6.11.2137. [DOI] [PubMed] [Google Scholar]
  6. Botas J., Moscoso del Prado J., García-Bellido A. Gene-dose titration analysis in the search of trans-regulatory genes in Drosophila. EMBO J. 1982;1(3):307–310. doi: 10.1002/j.1460-2075.1982.tb01165.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Brown N. L., Sattler C. A., Paddock S. W., Carroll S. B. Hairy and emc negatively regulate morphogenetic furrow progression in the Drosophila eye. Cell. 1995 Mar 24;80(6):879–887. doi: 10.1016/0092-8674(95)90291-0. [DOI] [PubMed] [Google Scholar]
  8. Cabrera C. V., Alonso M. C., Huikeshoven H. Regulation of scute function by extramacrochaete in vitro and in vivo. Development. 1994 Dec;120(12):3595–3603. doi: 10.1242/dev.120.12.3595. [DOI] [PubMed] [Google Scholar]
  9. Cai H. N., Arnosti D. N., Levine M. Long-range repression in the Drosophila embryo. Proc Natl Acad Sci U S A. 1996 Sep 3;93(18):9309–9314. doi: 10.1073/pnas.93.18.9309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Cai H., Levine M. Modulation of enhancer-promoter interactions by insulators in the Drosophila embryo. Nature. 1995 Aug 10;376(6540):533–536. doi: 10.1038/376533a0. [DOI] [PubMed] [Google Scholar]
  11. Carroll S. B., Scott M. P. Zygotically active genes that affect the spatial expression of the fushi tarazu segmentation gene during early Drosophila embryogenesis. Cell. 1986 Apr 11;45(1):113–126. doi: 10.1016/0092-8674(86)90543-x. [DOI] [PubMed] [Google Scholar]
  12. Cooper J. P., Roth S. Y., Simpson R. T. The global transcriptional regulators, SSN6 and TUP1, play distinct roles in the establishment of a repressive chromatin structure. Genes Dev. 1994 Jun 15;8(12):1400–1410. doi: 10.1101/gad.8.12.1400. [DOI] [PubMed] [Google Scholar]
  13. Dawson S. R., Turner D. L., Weintraub H., Parkhurst S. M. Specificity for the hairy/enhancer of split basic helix-loop-helix (bHLH) proteins maps outside the bHLH domain and suggests two separable modes of transcriptional repression. Mol Cell Biol. 1995 Dec;15(12):6923–6931. doi: 10.1128/mcb.15.12.6923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Edmondson D. G., Smith M. M., Roth S. Y. Repression domain of the yeast global repressor Tup1 interacts directly with histones H3 and H4. Genes Dev. 1996 May 15;10(10):1247–1259. doi: 10.1101/gad.10.10.1247. [DOI] [PubMed] [Google Scholar]
  15. Feder J. N., Jan L. Y., Jan Y. N. A rat gene with sequence homology to the Drosophila gene hairy is rapidly induced by growth factors known to influence neuronal differentiation. Mol Cell Biol. 1993 Jan;13(1):105–113. doi: 10.1128/mcb.13.1.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Fisher A. L., Ohsako S., Caudy M. The WRPW motif of the hairy-related basic helix-loop-helix repressor proteins acts as a 4-amino-acid transcription repression and protein-protein interaction domain. Mol Cell Biol. 1996 Jun;16(6):2670–2677. doi: 10.1128/mcb.16.6.2670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Gray S., Levine M. Short-range transcriptional repressors mediate both quenching and direct repression within complex loci in Drosophila. Genes Dev. 1996 Mar 15;10(6):700–710. doi: 10.1101/gad.10.6.700. [DOI] [PubMed] [Google Scholar]
  18. Gray S., Levine M. Transcriptional repression in development. Curr Opin Cell Biol. 1996 Jun;8(3):358–364. doi: 10.1016/s0955-0674(96)80010-x. [DOI] [PubMed] [Google Scholar]
  19. Gray S., Szymanski P., Levine M. Short-range repression permits multiple enhancers to function autonomously within a complex promoter. Genes Dev. 1994 Aug 1;8(15):1829–1838. doi: 10.1101/gad.8.15.1829. [DOI] [PubMed] [Google Scholar]
  20. Grbavec D., Stifani S. Molecular interaction between TLE1 and the carboxyl-terminal domain of HES-1 containing the WRPW motif. Biochem Biophys Res Commun. 1996 Jun 25;223(3):701–705. doi: 10.1006/bbrc.1996.0959. [DOI] [PubMed] [Google Scholar]
  21. Han K., Levine M. S., Manley J. L. Synergistic activation and repression of transcription by Drosophila homeobox proteins. Cell. 1989 Feb 24;56(4):573–583. doi: 10.1016/0092-8674(89)90580-1. [DOI] [PubMed] [Google Scholar]
  22. Herschbach B. M., Johnson A. D. Transcriptional repression in eukaryotes. Annu Rev Cell Biol. 1993;9:479–509. doi: 10.1146/annurev.cb.09.110193.002403. [DOI] [PubMed] [Google Scholar]
  23. Huang J. D., Dubnicoff T., Liaw G. J., Bai Y., Valentine S. A., Shirokawa J. M., Lengyel J. A., Courey A. J. Binding sites for transcription factor NTF-1/Elf-1 contribute to the ventral repression of decapentaplegic. Genes Dev. 1995 Dec 15;9(24):3177–3189. doi: 10.1101/gad.9.24.3177. [DOI] [PubMed] [Google Scholar]
  24. Ip Y. T., Park R. E., Kosman D., Bier E., Levine M. The dorsal gradient morphogen regulates stripes of rhomboid expression in the presumptive neuroectoderm of the Drosophila embryo. Genes Dev. 1992 Sep;6(9):1728–1739. doi: 10.1101/gad.6.9.1728. [DOI] [PubMed] [Google Scholar]
  25. Ishibashi M., Sasai Y., Nakanishi S., Kageyama R. Molecular characterization of HES-2, a mammalian helix-loop-helix factor structurally related to Drosophila hairy and Enhancer of split. Eur J Biochem. 1993 Aug 1;215(3):645–652. doi: 10.1111/j.1432-1033.1993.tb18075.x. [DOI] [PubMed] [Google Scholar]
  26. Jiang J., Rushlow C. A., Zhou Q., Small S., Levine M. Individual dorsal morphogen binding sites mediate activation and repression in the Drosophila embryo. EMBO J. 1992 Aug;11(8):3147–3154. doi: 10.1002/j.1460-2075.1992.tb05387.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Keleher C. A., Redd M. J., Schultz J., Carlson M., Johnson A. D. Ssn6-Tup1 is a general repressor of transcription in yeast. Cell. 1992 Feb 21;68(4):709–719. doi: 10.1016/0092-8674(92)90146-4. [DOI] [PubMed] [Google Scholar]
  28. Knust E., Schrons H., Grawe F., Campos-Ortega J. A. Seven genes of the Enhancer of split complex of Drosophila melanogaster encode helix-loop-helix proteins. Genetics. 1992 Oct;132(2):505–518. doi: 10.1093/genetics/132.2.505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Levine M., Manley J. L. Transcriptional repression of eukaryotic promoters. Cell. 1989 Nov 3;59(3):405–408. doi: 10.1016/0092-8674(89)90024-x. [DOI] [PubMed] [Google Scholar]
  30. Nüsslein-Volhard C., Wieschaus E. Mutations affecting segment number and polarity in Drosophila. Nature. 1980 Oct 30;287(5785):795–801. doi: 10.1038/287795a0. [DOI] [PubMed] [Google Scholar]
  31. Ohsako S., Hyer J., Panganiban G., Oliver I., Caudy M. Hairy function as a DNA-binding helix-loop-helix repressor of Drosophila sensory organ formation. Genes Dev. 1994 Nov 15;8(22):2743–2755. doi: 10.1101/gad.8.22.2743. [DOI] [PubMed] [Google Scholar]
  32. Orenic T. V., Held L. I., Jr, Paddock S. W., Carroll S. B. The spatial organization of epidermal structures: hairy establishes the geometrical pattern of Drosophila leg bristles by delimiting the domains of achaete expression. Development. 1993 May;118(1):9–20. doi: 10.1242/dev.118.1.9. [DOI] [PubMed] [Google Scholar]
  33. Pan D. J., Huang J. D., Courey A. J. Functional analysis of the Drosophila twist promoter reveals a dorsal-binding ventral activator region. Genes Dev. 1991 Oct;5(10):1892–1901. doi: 10.1101/gad.5.10.1892. [DOI] [PubMed] [Google Scholar]
  34. Paroush Z., Finley R. L., Jr, Kidd T., Wainwright S. M., Ingham P. W., Brent R., Ish-Horowicz D. Groucho is required for Drosophila neurogenesis, segmentation, and sex determination and interacts directly with hairy-related bHLH proteins. Cell. 1994 Dec 2;79(5):805–815. doi: 10.1016/0092-8674(94)90070-1. [DOI] [PubMed] [Google Scholar]
  35. Rushlow C. A., Hogan A., Pinchin S. M., Howe K. M., Lardelli M., Ish-Horowicz D. The Drosophila hairy protein acts in both segmentation and bristle patterning and shows homology to N-myc. EMBO J. 1989 Oct;8(10):3095–3103. doi: 10.1002/j.1460-2075.1989.tb08461.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Sasai Y., Kageyama R., Tagawa Y., Shigemoto R., Nakanishi S. Two mammalian helix-loop-helix factors structurally related to Drosophila hairy and Enhancer of split. Genes Dev. 1992 Dec;6(12B):2620–2634. doi: 10.1101/gad.6.12b.2620. [DOI] [PubMed] [Google Scholar]
  37. Small S., Blair A., Levine M. Regulation of even-skipped stripe 2 in the Drosophila embryo. EMBO J. 1992 Nov;11(11):4047–4057. doi: 10.1002/j.1460-2075.1992.tb05498.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Studer M., Pöpperl H., Marshall H., Kuroiwa A., Krumlauf R. Role of a conserved retinoic acid response element in rhombomere restriction of Hoxb-1. Science. 1994 Sep 16;265(5179):1728–1732. doi: 10.1126/science.7916164. [DOI] [PubMed] [Google Scholar]
  39. Wainwright S. M., Ish-Horowicz D. Point mutations in the Drosophila hairy gene demonstrate in vivo requirements for basic, helix-loop-helix, and WRPW domains. Mol Cell Biol. 1992 Jun;12(6):2475–2483. doi: 10.1128/mcb.12.6.2475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Weintraub S. J., Chow K. N., Luo R. X., Zhang S. H., He S., Dean D. C. Mechanism of active transcriptional repression by the retinoblastoma protein. Nature. 1995 Jun 29;375(6534):812–815. doi: 10.1038/375812a0. [DOI] [PubMed] [Google Scholar]