BarH1 and BarH2


DEVELOPMENTAL BIOLOGY

Embryonic and Larval

Early expression is found in anterior segments, the labrium, maxilla and procephalic lobes at 5.5 to 6.5 hours into development. Other staining is seen in the mandible and procephalic lobe [Image]. Later, BarH1 and BarH2 are coexpressed in cells of the embryonic central and peripheral nervous systems. Positive cells are found in the procephalic lobe, antenna-maxillary complex, labium, hypopharynx and clypeolabrum [Images]. Expression in the brain is apparent. In each case the Bar proteins are expressed in a subset of neurons. In external sensory organs, their expression is marked in thecogens (glial cells) and neurons late in development (Higashijima, 1992b).

BarH1 and BarH2 are not only specifically coexpressed in the developing eye, but are also functionally required in R1/R6 prephotoreceptors and primary pigment cells in developing ommatidia (see The Drosophila Adult Ommatidium: Illustration and explanation with Quicktime animation). They are also essential for normal lens and pigment cell formation, and for the elimination of excess cells from mature ommatidia (Higashijima, 1992a).

Transient overexpression of BarH1 or BarH2 in the morphogenetic furrow of the developing eye produces a characteristic Bar-like eye malformation. It is suggested that Bar overexpression results in suppression of the anterior progression of the morphogenetic furrow and inhibition of reinitiation of normal ommatidial differentiation (Kojima, 1993).

Mutation of roughex perturbs cell fate determination. Many rux mutant clusters contain multiple boss-expressing cells. In some of these clusters, R8 cells are missing. There is also a reduction in the number of cells expressing bar and Seven-up. This may be due to errors in cell fate determination. Alternatively, the reduced number of cells expressing these markers may reflect cell death. Extensive cell death is seen in rux mutants beginning with the MF and extending to the posterior edge of the disc. In rux mutant discs, neuronal differentiation is delayed by approximately 6 hours of development (Thomas, 1994).

The simplest external sensory organ (es) found in the thorax and abdomen consists of a neuron and a set of two support cells. The glial cell (or thecogen) forms a sheath around the tip of a dendrite, whereas the outer support cells, the trichogen and tormogen, secrete cuticule structures. It is the thecogen cell (a glial type) that specifically expresses BarH1. The es neurons express both BarH1 and BarH2. These cells also produce Prospero and Cut, but not under control of Bar (Higashijima, 1992b).

Effects of Mutation or Deletion

Simultaneous deletion of Bar genes leads to irregularly fused, bulging ommatidia, greatly differing in the number of rhabdomeres. Thus Bar genes are required for proper eye morphogenesis (Higashijima, 1992a and Kojima, 1993).

A new segment polarity gene of Drosophila melanogaster, oroshigane (oro) was identified as a dominant enhancer of Bar (B). The B1 allele of the Bar locus is associated with a tandem duplication of the division 16A of the X chromosome and causes the overexpression of the Bar (B) gene. decapentaplegic expression in the morphogenetic furrow is abolished in the B background, and therefore morphogenetic furrow progression prematurely ceases resulting in the characteristic bar-shapped compound eye. hedgehog expression also fades 8 hours after heat induction of the Bar protein, indicating that hh is another target for inhibition by B. Since the Bar protein is required for R1, R6 and primary pigment cell differentiion behind the furrow, the overexpressed B protein apparently interferes with furrow progression by inhibiting dpp expression in the furrow and hedgehog expression just behind the furrow. Overexpression of the Bar protein has little deleterious effect on differentiation of photoreceptor clusters that have already started to develop behind the morphogenetic furrow. Failure of morphogenetic furrow progression likely triggers programmed cell death in the undifferentiated cells ahead of the morphogenetic furrow (Epps, 1997).

oro is a recessive embryonic lethal, and homozygous oro embryos show variable substitution of denticles for naked cuticle. These patterns are distinctly similar to those of hedgehog and wingless mutant embryos, which indicates that oro functions in determining embryonic segment polarity. oro works downstream of hedgehog but upstream of dpp to enhance the Bar phenotypes. Although dpp expression is reduced in oro heterozygotes, hh expression remains the same as that found in wild-type discs. Evidence that oro function is involved in Hh signal transduction during embryogenesis is provided by its genetic interactions with the segment polarity genes patched and fused. ptcIN is a dominant suppressor of the oro embryonic lethal phenotype, suggesting a close and dose-dependent relationship between oro and ptc in Hh signal transduction. oro function is also required in imaginal development. The oro1 allele significantly reduces decapentaplegic (but not hh) expression in the eye imaginal disc. oro enhances the fused1 wing phenotype in a dominant manner. Based upon the interactions of oro with hh, ptc, and fu, it is proposed that the oro gene plays important roles in Hh signal transduction (Epps, 1997).

In the developing Drosophila eye, BarH1 and BarH2, paired homeobox genes expressed in R1/R6 outer photoreceptors and primary pigment cells, are essential for normal eye morphogenesis. BarH1 was ectopically expressed under the control of the sevenless enhancer (sev-BarH1). The sev enhancer drives gene expression strongly, not only in R7 precursors but also in R3/R5 and cone cell precursors. Evidence is presented that sev-BarH1 causes two types of cone cell transformation: transformation of anterior/posterior cone cells into outer photoreceptors and transformation of equatorial/polar cone cells into primary pigment cells. The ectopic primary pigment cells are partially similar in morphology to cone cells. sev-BarH1 represses the endogenous expression of the rough homeobox gene in R3/R4 photoreceptors, while the BarH2 homeobox gene is activated by sev-BarH1 in an appreciable fraction of extra outer photoreceptors. In primary pigment cells generated by cone cell transformation, the expression of cut, a homeobox gene specific to cone cells, is completely replaced with that of Bar homeobox genes. Extra outer photoreceptor formation is either suppressed or enhanced, respectively, by reducing the activity of Ras/MAPK signaling or by dosage reduction of yan, a negative regulator of the pathway, suggesting interactions between Bar homeobox genes (cell fate determinants) and Ras/MAPK signaling in eye development. It is concluded that cone cell precursors may adopt four different cell fates: an outer photoreceptor fate, a primary pigment cell fate, a cone cell fate, or the fate of disappearance f from ommatidia (X-cell fate). Cone cell precursors appear to be divided into two subgroups with respect to sensitivity to sev-BarH1: either anterior/posterior or equatorial/polar cone cell precursors. sev-BarH1 causes transformation of a fraction of anterior/posterior cone cells into outer photoreceptors partially expressing R1/R6-specific genes and also causes the transformation of a fraction of equatorial/polar cone cells into primary pigment cells; this suggests that BarH1 serves as a determinant of R1/R6 and primary pigment cell fates in normal eye development (Hayashi, 1998).

The progression of the morphogenetic furrow in the developing Drosophila eye is an early metamorphic, ecdysteroid-dependent event. Although Ecdysone receptor-encoded nuclear receptor isoforms are the only known ecdysteroid receptors, it has been shown that the Ecdysone receptor gene is not required for furrow function. DHR78, which encodes another candidate ecdysteroid receptor, is also not required. In contrast, zinc finger-containing isoforms encoded by the early ecdysone response gene Broad-complex regulate furrow progression and photoreceptor specification. br-encoded Broad-complex subfunctions are required for furrow progression and proper R8 specification, and are antagonized by other subfunctions of Broad-complex. There is a switch from Broad complex Z2 to Z1 zinc-finger isoform expression at the furrow that requires Z2 expression and responds to Hedgehog signals. These results suggest that a novel hormone transduction hierarchy involving an uncharacterized receptor operates in the eye disc (Brennan, 2001).

Bar is a dominant mutation causing premature arrest of the furrow, which results in the deep anterior nick in the adult eye. Since Bar has the dominant effect of stopping the furrow early, one might expect loss-of-function mutations at other loci that normally act to promote furrow progression to be genetic enhancers of Bar and loss-of-function mutations in genes that normally antagonize the furrow to act as genetic suppressors of Bar. Thus, genetic interactions between BR-C sub loci and Bar were examined. Mutants defective for different BR-C subfunctions display unexpected heterogeneity in their genetic interactions with Bar, suggesting that the role of the BR-C in the regulation of furrow function might be complex. BR-C has several recessive lethal complementation groups that correspond to mutations that remove the function of all or individual zinc finger-containing isoforms subgroups. npr1 mutations lack all function, whereas rbp, br and 2Bc mutant groups correspond to the loss of Z1-, Z2-, and Z3-containing isoforms, respectively. Both npr1/Bar and br/Bar eyes are significantly smaller than +/Bar, indicating a dominant enhancement of the Bar furrow-stop phenotype, consistent with the earlier reports that the BR-C is required for furrow progression. However, br/Bar eyes are smaller than npr1/Bar, suggesting that the BR-C might encode isoforms that act antagonistically during furrow progression, so that the effect of losing isoforms that positively regulate furrow progression is more severe than losing all isoforms. This idea is supported by the observation that rbp/Bar and 2Bc/Bar eyes are larger than +/Bar, suppressing the phenotype, and possibly representing furrow-antagonistic functions of rbp- or br-encoded BR-C isoforms (Brennan, 2001).

Hemizygous males of all BR-C mutant groups survived through the third instar: the eye discs of these males displayed defects consistent with the genetic interactions with Bar. npr1/Y discs show ommatidial disorganization, and signs of furrow failure, including mature ommatidial clusters at the furrow. br/Y discs show a much more dramatic failure of furrow progression, as well as ommatidial disorganization. rbp/Y and 2Bc/Y discs do not show any defects. While rbp does interact strongly with Bar it shows no eye disc defect alone -- it may be that rbp is a redundant function (Brennan, 2001).


REFERENCES

Brennan, C. A., et al. (2001). Broad-complex, but not Ecdysone receptor, is required for progression of the morphogenetic furrow in the Drosophila eye. Development 128: 1-11. PubMed ID: 11092806

Bulfone, A., Menguzzato, E., Broccoli, V., Marchitiello, A., Gattuso, C., Mariani, M., Consalez, G. G. Martinez, S., Ballabio, A. and Banfi, S. (2000). Barhl1, a gene belonging to a new subfamily of mammalian homeobox genes, is expressed in migrating neurons of the CNS. Hum. Mol. Genet. 9: 1443-1452. PubMed ID: 10814725

Campbell, G. (2002). Distalization of the Drosophila leg by graded EGF-receptor activity. Nature 418: 781-785. PubMed ID: 12181568

Campbell, G. (2005). Regulation of gene expression in the distal region of the Drosophila leg by the Hox11 homolog, C15. Dev. Biol. 278(2): 607-18. PubMed ID: 15680373

Crew, J. R., Batterham, P. and Pollock, J. A. (1997). Developing compound eye in lozenge mutants of Drosophila: lozenge expression in the R7 equivalence group. Dev. Genes and Evol. 206(8): 481-493

Daga, A., et al. (1996). Patterning of cells in the Drosophila eye by Lozenge, which shares homologous domains with AML1. Genes Dev 10: 1194-1205. PubMed ID: 8675007

Del Signore, S. J., Hayashi, T. and Hatini, V. (2012). odd-skipped genes and lines organize the notum anterior-posterior axis using autonomous and non-autonomous mechanisms. Mech Dev 129: 147-161. PubMed ID: 22613630

Edelman, D. B., Meech, R. and Jones, F. S. (2000). The homeodomain protein Barx2 contains activator and repressor domains and interacts with members of the CREB family. J. Biol. Chem. 275: 21737-21745. PubMed ID: 10781615

Epps, J. L., Jones, J. B. and Tanda, S. (1997). oroshigane, a new segment polarity gene of Drosophila melanogaster, functions in Hedgehog signal transduction. Genetics 145 (4): 1041-1052. PubMed ID: 9093856

Fu, W. and Noll, M. (1997). The Pax2 homolog sparkling is required for development of cone and pigment cells in the Drosophila eye. Genes Dev. 11(16): 2066-2078. PubMed ID: 9284046

Garces, A., Bogdanik, L., Thor, S. and Carroll, P. (2006). Expression of Drosophila BarH1-H2 homeoproteins in developing dopaminergic cells and segmental nerve a (SNa) motoneurons. Eur. J. Neurosci. 24(1): 37-44. PubMed ID: 16882006

Gauchat, D., et al. (2000). Evolution of Antp-class genes and differential expression of Hydra Hox/paraHox genes in anterior patterning. Proc. Natl. Acad. Sci. 97: 4493-4498. PubMed ID: 10781050

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. PubMed ID: 16314497

Hayashi, T., Kojima, T. and Saigo, K. (1998). Specification of primary pigment cell and outer photoreceptor fates by BarH1 homeobox gene in the developing Drosophila eye. Dev. Biol. 200(2): 131-145. PubMed ID: 9705222

Higashijima, S., Kojima, T., Michiue, T., Ishimaru, S., Emori, Y. and Saigo, K. (1992a). Dual Bar homeo box genes of Drosophila required in two photoreceptor cells, R1 and R6, and primary pigment cells for normal eye development. Genes & Dev. 6(1): 50-60. PubMed ID: 1346120

Higashijima, S., Michiue, T., Emori, Y. and Saigo, K. (1992b). Subtype determination of Drosophila embryonic external sensory organs by redundant homeo box genes BarH1 and BarH2. Genes and Development 6(6): 1005-18 . PubMed ID: 1350558

Hou, K., Jiang, H., Karim, M. R., Zhong, C., Xu, Z., Liu, L., Guan, M., Shao, J. and Huang, X. (2019). A critical E-box in Barhl1 3' enhancer is essential for auditory hair cell differentiation. Cells 8(5). PubMed ID: 31096644

Hjalt, T. A. and Murray, J. C. (1999). The human BARX2 gene: genomic structure, chromosomal localization, and single nucleotide polymorphisms. Genomics 62: 456-459. PubMed ID: 10644443

Jones, F. S., et al (1997). Barx2, a new homeobox gene of the Bar class, is expressed in neural and craniofacial structures during development. Proc. Natl. Acad. Sci. 94 (6): 2632-2637. PubMed ID: 9122247

Kim, B. M., Buchner, G., Miletich, I., Sharpe, P. T., Shivdasani, R. A. (2005). The stomach mesenchymal transcription factor Barx1 specifies gastric epithelial identity through inhibition of transient Wnt signaling. Dev. Cell 8(4): 611-22. PubMed ID: 15809042

Kojima, T., Ishimaru, S., Higashijima, S., Takayama, E., Akimaru, H., Sone, M., Emori, Y. and Saigo, K. (1991). Identification of a different-type homeobox gene, BarH1, possible causing Bar (B) and Om(1D) mutations in Drosophila. Proc. Natl. Acad. Sci. 88(10): 4343-7. PubMed ID: 1674606

Kojima, T., Sone, M., Michiue, T and Saigo, K. (1993). Mechanism of induction of Bar-like eye malformation by transient overexpression of Bar homeobox genes in Drosophila melanogaster. Genetics 88: 85-91. PubMed ID: 7901124

Kojima, T., Sato, M. and Saigo, K. (2000). Formation and specification of distal leg segments in Drosophila by dual Bar homeobox genes, BarH1 and BarH2. Development 127: 769-778. PubMed ID: 10648235

Kojima, T., Tsuji, T. and Saigo, K. (2005). A concerted action of a paired-type homeobox gene, aristaless, and a homolog of Hox11/tlx homeobox gene, clawless, is essential for the distal tip development of the Drosophila leg. Dev. Biol. 279(2): 434-45. PubMed ID: 15733670

Li, S., et al. (2002). Hearing loss caused by progressive degeneration of cochlear hair cells in mice deficient for the Barhl1 homeobox gene. Development 129: 3523-3532. PubMed ID: 12091321

Lim, J. and Choi, K. W., et al. (2003). Bar homeodomain proteins are anti-proneural in the Drosophila eye: transcriptional repression of atonal by Bar prevents ectopic retinal neurogenesis. Development 130: 5965-5974. PubMed ID: 14573515

Lim, J. and Choi, K.-W. (2004). Induction and autoregulation of the anti-proneural gene Bar during retinal neurogenesis in Drosophila. Development 131: 5573-5580. PubMed ID: 15496446

Lopes, C., et al. (2006). BARHL1 homeogene, the human ortholog of the mouse Barhl1 involved in cerebellum development, shows regional and cellular specificities in restricted domains of developing human central nervous system Biochem. Biophy. Res. Comm. 339: 296-304. PubMed ID: 16307728

Meech, R., et al. (2005). The homeobox transcription factor Barx2 regulates chondrogenesis during limb development. Development 132: 2135-2146. PubMed ID: 1580000

Meech, R., Edelman, D. B., Jones, F. S. Makarenkova, H. P. (2005). The homeobox transcription factor Barx2 regulates chondrogenesis during limb development. Development 132(9): 2135-46. PubMed ID: 15800003

Mo, A., Li, S., Yang, X. and Xiang, M. (2004). Role of the Barhl2 homeobox gene in the specification of glycinergic amacrine cells. Development 131: 1607-1618. PubMed ID: 14998930

Nusinow, D., Greenberg, L. and Hatini, V. (2008). Reciprocal roles for bowl and lines in specifying the peripodial epithelium and the disc proper of the Drosophila wing primordium. Development 135: 3031-3041. PubMed ID: 18701548

Offner, N., et al. (2005). The pro-apoptotic activity of a vertebrate Bar-like homeobox gene plays a key role in patterning the Xenopus neural plate by limiting the number of chordin- and shh-expressing cells. Development 132: 1807-1818. PubMed ID: 15772136

Patterson, K. D., et al. (2000). Distinct expression patterns for two Xenopus Bar homeobox genes. Dev. Genes Evol. 210: 140-144. PubMed ID: 11180814

Peden, E., et al. (2007). Control of sex-specific apoptosis in C. elegans by the BarH homeodomain protein CEH-30 and the transcriptional repressor UNC-37/Groucho. Genes Dev 21: 3195-3207. PubMed ID: 18056429

Poggi, L., et al. (2004). The homeobox gene Xbh1 cooperates with proneural genes to specify ganglion cell fate within the Xenopus neural retina. Development 131: 2305-2315. PubMed ID: 15102701

Pueyo, J. I., et al. (2000). Proximal-distal leg development in Drosophila requires the apterous gene and the Lim1 homologue dlim1 Development 127: 5391-5402. PubMed ID: 11076760

Pueyo, J. I. and Couso, J. P. (2004). Chip-mediated partnerships of the homeodomain proteins Bar and Aristaless with the LIM-HOM proteins Apterous and Lim1 regulate distal leg development. Development 131: 3107-3120. PubMed ID: 15175252

Saba, R., Johnson, J. E. and Saito, T. (2005). Commissural neuron identity is specified by a homeodomain protein, Mbh1, that is directly downstream of Math1. Development 132: 2147-2155. PubMed ID: 15788459

Saito, T., Sawamoto, K., Okano, H., Anderson, D. J. and Mikoshiba, K. (1998). Mammalian BarH homologue is potential regulator of neural bHLH genes. Dev. Biol. 199: 216-225. PubMed ID: 9698441

Sato, A., et al. (1999). Dfrizzled-3, a new Drosophila Wnt receptor, acting as an attenuator of Wingless signaling in wingless hypomorphic mutants. Development 126: 4421-4430. PubMed ID: 10498678

Sato, M, et al. (1999). Bar homeobox genes are latitudinal prepattern genes in the developing Drosophila notum whose expression is regulated by the concerted functions of decapentaplegic and wingless. Development 126: 1457-1466. PubMed ID: 10068639

Stevens, T. A., Iacovoni, J. S., Edelman, D. B. and Meech, R. (2004). Identification of novel binding elements and gene targets for the homeodomain protein BARX2. J. Biol. Chem. 279(15): 14520-30. PubMed ID: 14744868

Thomas, B. J., et al. (1994). Cell cycle progression in the developing Drosophila eye: roughex encodes a novel protein required for the establishment of G1. Cell 77: 1003-1014. PubMed ID: 8020091

Tissier-Seta, J.-P., et al. (1995). Barx1, a new mouse homeodomain transcription factor expressed in cranio-facial ectomesenchyme and the stomach. Mech. Dev. 51: 3-15. PubMed ID: 7669690

Tsuji, T., et al. (2000). Requirements of Lim1, a Drosophila LIM-homeobox gene, for normal leg and antennal development. Development 127: 4315-4323. PubMed ID: 11003832

Tucker, A. S, Matthews, K.L. and Sharpe, P. T. (1998). Transformation of tooth type induced by inhibition of BMP signaling. Science 282(5391): 1136-8. PubMed ID: 9804553


BarH1 and BarH2 : Biological Overview | Evolutionary Homologs | Regulation | Developmental Biology | Effects of Mutation

date revised: 10 July 2019

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

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