51
|
Adherens junctions in Drosophila retinal morphogenesis. Trends Cell Biol 2006; 17:26-35. [PMID: 17134901 DOI: 10.1016/j.tcb.2006.11.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2006] [Revised: 10/06/2006] [Accepted: 11/16/2006] [Indexed: 12/25/2022]
Abstract
Adherens junctions and their core molecular components, classic cadherins, make major contributions to animal morphogenesis. Although the significance of cadherins in development is generally accepted, the mechanisms regulating adherens junction function during morphogenesis remain a subject of intense research. Adherens junctions are involved in the organization of simple cellular patterns, and more complex cell shape changes and cell movements that depend on the dynamic modulation of adherens junctions.
Collapse
|
52
|
Gerke P, Benzing T, Höhne M, Kispert A, Frotscher M, Walz G, Kretz O. Neuronal expression and interaction with the synaptic protein CASK suggest a role for Neph1 and Neph2 in synaptogenesis. J Comp Neurol 2006; 498:466-75. [PMID: 16874800 DOI: 10.1002/cne.21064] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Formation, differentiation, and plasticity of synapses require interactions between pre- and postsynaptic partners. Recently, it was shown that the transmembrane immunoglobulin superfamily protein SYG-1 is required for providing synaptic specificity in C. elegans. However, it is unclear whether the mammalian orthologs of SYG-1 are also involved in local cell interactions to determine specificity during synapse formation. We used in situ hybridization, immunohistochemistry, and immunogold electron microscopy to study the temporal and spatial expression of Neph1 and Neph2 in the developing and adult mouse brain. Both proteins show similar patterns with neuronal expression starting around embryonic days 12 and 11, respectively. Expression is strongest in areas of high migratory activity. In the adult brain, Neph1 and Neph2 are predominantly seen in the olfactory nerve layer and the glomerular layer of the olfactory bulb, in the hippocampus, and in Purkinje cells of the cerebellum. At the ultrastructural level, Neph1 and Neph2 are detectable within the dendritic shafts of pyramidal neurons. To a lesser extent, there is also synaptic localization of Neph1 within the stratum pyramidale of the hippocampal CA1 and CA3 region on both pre- and postsynaptic sites. Here it colocalizes with the synaptic scaffolder calmodulin-associated serin/threonin kinase (CASK), and both Neph1 and Neph2 interact with the PDZ domain of CASK via their cytoplasmic tail. Our results show that Neph proteins are expressed in the developing nervous system of mammals and suggest that these proteins may have a conserved function in synapse formation or neurogenesis.
Collapse
Affiliation(s)
- Peter Gerke
- Renal Division, University of Freiburg, D-79104 Freiburg, Germany
| | | | | | | | | | | | | |
Collapse
|
53
|
Kreisköther N, Reichert N, Buttgereit D, Hertenstein A, Fischbach KF, Renkawitz-Pohl R. Drosophila Rolling pebbles colocalises and putatively interacts with alpha-Actinin and the Sls isoform Zormin in the Z-discs of the sarcomere and with Dumbfounded/Kirre, alpha-Actinin and Zormin in the terminal Z-discs. J Muscle Res Cell Motil 2006; 27:93-106. [PMID: 16699917 DOI: 10.1007/s10974-006-9060-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2005] [Accepted: 02/14/2006] [Indexed: 10/24/2022]
Abstract
The rolling pebbles gene of Drosophila encodes two proteins, one of which, Rols7, is essential for myoblast fusion. In addition, Rols 7 is expressed during myofibrillogenesis and in the mature muscles. Here it overlaps with alpha-Actinin (alpha-Actn) and the N-terminus of D-Titin/Kettin/Zormin in the Z-line of the sarcomeres. In the attachment sites of the somatic muscles, Rols7 and the immunoglobulin superfamily protein Dumbfounded/Kin of irreC (Duf/Kirre) colocalise. As Duf/Kirre is detectable only transiently, it may be involved in establishing the first contact of the outgrowing muscle fiber to the epidermal attachment site. We propose that Rols7 and Duf/Kirre link the terminal Z-disc to the cell membrane by direct interaction. This is supported by the fact that in yeast two hybrid assays the tetratricopeptide repeat E (TPR E) of Rols7 shows interaction with the intracellular domain of Duf/Kirre. The colocalisation of Rols7 with alpha-Actn and with D-Titin/Kettin/Zormin in the Z-dics is reflected in interactions with different domains of Rols7 in this assay. In summary, these data show that besides the role in myoblast fusion, Rols7 is a scaffold protein during myofibrillogenesis and in the Z-line of the sarcomere as well as in the terminal Z-disc linking the muscle to the epidermal attachment sites.
Collapse
Affiliation(s)
- Nina Kreisköther
- Fachbereich Biologie, Entwicklungsbiologie, Philipps-Universität Marburg, Karl-von-Frisch-Str. 8, 35043 Marburg, Germany
| | | | | | | | | | | |
Collapse
|
54
|
Vishnu S, Hertenstein A, Betschinger J, Knoblich JA, Gert de Couet H, Fischbach KF. The adaptor protein X11Lα/Dmint1 interacts with the PDZ-binding domain of the cell recognition protein Rst in Drosophila. Dev Biol 2006; 289:296-307. [PMID: 16380111 DOI: 10.1016/j.ydbio.2005.09.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2004] [Revised: 07/04/2005] [Accepted: 09/09/2005] [Indexed: 12/22/2022]
Abstract
The Drosophila cell adhesion molecule Rst plays key roles during the development of the embryonic musculature, spacing of ommatidia in the compound eye and of sensory organs on the antenna, as well as in the neuronal wiring of the optic lobe. In rst(CT) mutants lacking the cytoplasmic domain of the Rst protein, cell sorting and apoptosis in the eye are affected, suggesting a requirement of this domain for Rst function. To identify potential interacting proteins, yeast two-hybrid screens were performed using the cytoplasmic domains of Rst and its paralogue Kirre as baits. Among several putative interactors, two paralogous Drosophila PDZ motif proteins related to X11/Mint were identified. X11/Mint family members in C. elegans (LIN-10) and vertebrates are believed to function as adaptor proteins and to regulate the assembly of multi-subunit complexes at the synapse, thereby linking the vesicle cycle to cell adhesion. Using genetic, cell biological, and biochemical approaches, we show that the interaction of Rst with X11Lalpha is of biological significance. The proteins interact, for example, in the context of cell sorting in the pupal retina.
Collapse
Affiliation(s)
- Smitha Vishnu
- Institut für Biologie III, Albert-Ludwigs Universität, D-79104 Freiburg im Breisgau, Germany
| | | | | | | | | | | |
Collapse
|
55
|
Apitz H, Strünkelnberg M, de Couet HG, Fischbach KF. Single-minded, Dmef2, Pointed, and Su(H) act on identified regulatory sequences of the roughest gene in Drosophila melanogaster. Dev Genes Evol 2005; 215:460-69. [PMID: 16096801 DOI: 10.1007/s00427-005-0005-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2005] [Accepted: 05/18/2005] [Indexed: 10/25/2022]
Abstract
Roughest (Rst) is a cell adhesion molecule of the immunoglobulin superfamily that has multiple and diverse functions during the development of Drosophila melanogaster. The pleiotropic action of Rst is reflected by its complex and dynamic expression during the development of Drosophila. By an enhancer detection screen, we previously identified several cis-regulatory modules that mediate specific expression of the roughest gene in Drosophila developmental processes. To identify trans-regulators of rst expression, we used the Gal4/UAS system to screen for factors that were sufficient to activate Rst expression when ectopically expressed. By this method we identified the transcription factors Single-minded, Pointed.P1, and Su(H)-VP16. Furthermore, we showed that these factors and, in addition, Dmef2 are able to ectopically activate rst expression via the previously described rst cis-regulatory modules. This fact and the use of mutant analysis allocates the action of the transcription factors to specific developmental contexts. In the case of Sim, we could show that it regulates rst expression in the embryonic midline, but not in the optic lobes. Mutagenesis of Sim consensus binding sites in the regulatory module required for rst expression in the embryonic midline, abolished rst expression; indicating that the regulation of rst by Sim is direct.
Collapse
Affiliation(s)
- Holger Apitz
- Institut für Biologie III, Albert-Ludwigs-Universität Freiburg, Schänzlestr.1, 79104, Freiburg, Germany
| | | | | | | |
Collapse
|
56
|
Bao S, Cagan R. Preferential adhesion mediated by Hibris and Roughest regulates morphogenesis and patterning in the Drosophila eye. Dev Cell 2005; 8:925-35. [PMID: 15935781 DOI: 10.1016/j.devcel.2005.03.011] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2004] [Revised: 01/25/2005] [Accepted: 03/16/2005] [Indexed: 11/29/2022]
Abstract
Cell adhesion is essential for morphogenesis; however, the mechanisms by which cell adhesion coordinates precisely regulated morphogenesis are poorly understood. Here we analyze the morphogenetic processes that organize the interommatidial precursor cells (IPCs) of the Drosophila pupal eye. We demonstrate that the Drosophila immunoglobulin superfamily members Hibris and Roughest are essential for IPC morphogenesis in the eye. The two loci are expressed in complementary cell types, and Hibris and Roughest proteins bind directly in vivo. Primary pigment cells employ Hibris to function as organizers in this process; IPCs minimize contacts with neighboring IPCs and utilize Roughest to maximize contacts with primaries. In addition, we provide evidence that interactions between Hibris and Roughest promote junction formation and that levels of Roughest in individual cells determine their capacity for competition. Our results demonstrate that preferential adhesion mediated by heterophilic interacting cell-adhesion molecules can create a precise pattern by minimizing surface free energy.
Collapse
Affiliation(s)
- Sujin Bao
- Department of Molecular Biology and Pharmacology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, Missouri 63110, USA
| | | |
Collapse
|
57
|
Mehta SQ, Hiesinger PR, Beronja S, Zhai RG, Schulze KL, Verstreken P, Cao Y, Zhou Y, Tepass U, Crair MC, Bellen HJ. Mutations in Drosophila sec15 reveal a function in neuronal targeting for a subset of exocyst components. Neuron 2005; 46:219-32. [PMID: 15848801 DOI: 10.1016/j.neuron.2005.02.029] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2004] [Revised: 10/26/2004] [Accepted: 02/25/2005] [Indexed: 11/23/2022]
Abstract
The exocyst is a complex of proteins originally identified in yeast that has been implicated in polarized secretion. Components of the exocyst have been implicated in neurite outgrowth, cell polarity, and cell viability. We have isolated an exocyst component, sec15, in a screen for genes required for synaptic specificity. Loss of sec15 causes a targeting defect of photoreceptors that coincides with mislocalization of specific cell adhesion and signaling molecules. Additionally, sec15 mutant neurons fail to localize other exocyst members like Sec5 and Sec8, but not Sec6, to neuronal terminals. However, loss of sec15 does not cause cell lethality in contrast to loss of sec5 or sec6. Our data suggest a role of Sec15 in an exocyst-like subcomplex for the targeting and subcellular distribution of specific proteins. The data also show that functions of other exocyst components persist in the absence of sec15, suggesting that different exocyst components have separable functions.
Collapse
Affiliation(s)
- Sunil Q Mehta
- Program in Developmental Biology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Grzeschik NA, Knust E. IrreC/rst-mediated cell sorting duringDrosophilapupal eye development depends on proper localisation of DE-cadherin. Development 2005; 132:2035-45. [PMID: 15788453 DOI: 10.1242/dev.01800] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Remodelling of tissues depends on the coordinated regulation of multiple cellular processes, such as cell-cell communication, differential cell adhesion and programmed cell death. During pupal development, interommatidial cells (IOCs) of the Drosophila eye initially form two or three cell rows between individual ommatidia, but then rearrange into a single row of cells. The surplus cells are eliminated by programmed cell death, and the definitive hexagonal array of cells is formed, which is the basis for the regular pattern of ommatidia visible in the adult eye. Here, we show that this cell-sorting process depends on the presence of a continuous belt of the homophilic cell adhesion protein DE-cadherin at the apical end of the IOCs. Elimination of this adhesion belt by mutations in shotgun, which encodes DE-cadherin, or its disruption by overexpression of DE-cadherin, the intracellular domain of Crumbs, or by a dominant version of the monomeric GTPase Rho1 prevents localisation of the transmembrane protein IrreC-rst to the border between primary pigment cells and IOCs. As a consequence, the IOCs are not properly sorted and supernumerary cells survive. During the sorting process, Notch-mediated signalling in IOCs acts downstream of DE-cadherin to restrict IrreC-rst to this border. The data are discussed in relation to the roles of selective cell adhesion and cell signalling during tissue reorganisation.
Collapse
Affiliation(s)
- Nicola A Grzeschik
- Institut für Genetik, Heinrich-Heine Universität Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | | |
Collapse
|
59
|
Tamura S, Morikawa Y, Hisaoka T, Ueno H, Kitamura T, Senba E. Expression of mKirre, a mammalian homolog of Drosophila kirre, in the developing and adult mouse brain. Neuroscience 2005; 133:615-24. [PMID: 15908127 DOI: 10.1016/j.neuroscience.2005.03.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2005] [Revised: 03/15/2005] [Accepted: 03/25/2005] [Indexed: 12/01/2022]
Abstract
mKirre, a mammalian homolog of the Drosophila kirre, is expressed in bone marrow stromal cells and the brain. Although mKirre has been shown to support the hematopoietic stem cells, little is known about the function of mKirre in the brain. In the present study, to gain insights into the function of mKirre, we investigated the expression pattern of mKirre gene in the developing and adult mouse brain using in situ hybridization. In the adult brain, mKirre mRNA was highly expressed in the olfactory bulb, the piriform cortex, the cochlear nucleus, and the cerebellum. At embryonic day (E) 11.5, we could observe mKirre mRNA in the differentiating zones of various regions, such as the caudate-putamen, the geniculate body, the thalamus, the amygdala, and the brainstem. Its gene expression in these regions at E11.5 also persisted to the adult, in which its expression levels were much less prominent. After birth, we could first observe high expression of mKirre mRNA in the glomerular and mitral layers of the olfactory bulb, the cortical plate of the neocortex, the cochlear nucleus, and the molecular and granule cell layers of the cerebellum. In the hippocampus, its gene expression was first observed in the dentate gyrus at postnatal day 7. The spatiotemporal expression pattern of mKirre mRNA suggests important roles of mKirre in later developmental processes, especially the synapse formation.
Collapse
Affiliation(s)
- S Tamura
- Department of Anatomy and Neurobiology, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 641-8509, Japan
| | | | | | | | | | | |
Collapse
|
60
|
Galletta BJ, Chakravarti M, Banerjee R, Abmayr SM. SNS: adhesive properties, localization requirements and ectodomain dependence in S2 cells and embryonic myoblasts. Mech Dev 2004; 121:1455-68. [PMID: 15511638 DOI: 10.1016/j.mod.2004.08.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2004] [Revised: 07/20/2004] [Accepted: 08/04/2004] [Indexed: 11/30/2022]
Abstract
The body wall muscles in the Drosophila larva arise from interactions between Duf/Kirre and Irregular chiasm C-roughest (IrreC-rst)-expressing founder myoblasts and sticks-and-stones (SNS)-expressing fusion competent myoblasts in the embryo. Herein, we demonstrate that SNS mediates heterotypic adhesion of S2 cells with Duf/Kirre and IrreC-rst-expressing S2 cells, and colocalizes with these proteins at points of cell contact. These properties are independent of their transmembrane and cytoplasmic domains, and are observed quite readily with GPI-anchored forms of the ectodomains. Heterotypic interactions between Duf/Kirre and SNS-expressing S2 cells occur more rapidly and to a greater extent than homotypic interactions with other Duf/Kirre-expressing cells. In addition, Duf/Kirre and SNS are present in an immunoprecipitable complex from S2 cells. In the embryo, Duf/Kirre and SNS are present at points of contact between founder and fusion competent cells. Moreover, SNS clustering on the cell surface is dependent on Duf/Kirre and/or IrreC-rst. Finally, although the cytoplasmic and transmembrane domains of SNS are expendable for interactions in culture, they are essential for fusion of embryonic myoblasts.
Collapse
Affiliation(s)
- Brian J Galletta
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | | | | | | |
Collapse
|
61
|
Apitz H, Kambacheld M, Höhne M, Ramos RGP, Straube A, Fischbach KF. Identification of regulatory modules mediating specific expression of the roughest gene in Drosophila melanogaster. Dev Genes Evol 2004; 214:453-9. [PMID: 15278452 DOI: 10.1007/s00427-004-0423-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2004] [Accepted: 06/15/2004] [Indexed: 10/26/2022]
Abstract
Roughest (Rst) is a cell adhesion molecule of the immunoglobulin superfamily with pleiotropic functions during the development of Drosophila melanogaster. It has been shown to be involved in cell sorting before apoptosis in the developing compound eye, in fusion processes of embryonic muscle development and in axonal pathfinding. In accordance with its multiple functions, the rst gene shows a dynamic expression pattern throughout the development of Drosophila. In order to understand the transcriptional regulation of rst expression we have identified rst cis regulatory sequences in an enhancer detection screen. By dissection of the identified rst cis regulatory sequences we identified several distinct rst regulatory modules. Among others these include elements for expression in interommatidial cells of the pupal eye disc at a time when apoptotic decisions are made in these cells and elements for expression in the embryonic mesoderm. The expression of rst in the embryonic mesoderm is regulated by at least two separate modules.
Collapse
Affiliation(s)
- Holger Apitz
- Institut für Biologie III, Albert-Ludwigs-Universität Freiburg, Schänzlestr.1, 79104 Freiburg, Germany
| | | | | | | | | | | |
Collapse
|
62
|
Shen K, Fetter RD, Bargmann CI. Synaptic specificity is generated by the synaptic guidepost protein SYG-2 and its receptor, SYG-1. Cell 2004; 116:869-81. [PMID: 15035988 DOI: 10.1016/s0092-8674(04)00251-x] [Citation(s) in RCA: 221] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2003] [Revised: 01/08/2004] [Accepted: 01/15/2004] [Indexed: 11/24/2022]
Abstract
Synaptic connections in the nervous system are directed onto specific cellular and subcellular targets. Synaptic guidepost cells in the C. elegans vulval epithelium drive synapses from the HSNL motor neuron onto adjacent target neurons and muscles. Here, we show that the transmembrane immunoglobulin superfamily protein SYG-2 is a central component of the synaptic guidepost signal. SYG-2 is expressed transiently by primary vulval epithelial cells during synapse formation. SYG-2 binds SYG-1, the receptor on HSNL, and directs SYG-1 accumulation and synapse formation to adjacent regions of HSNL. syg-1 and syg-2 mutants have defects in synaptic specificity; the HSNL neuron forms fewer synapses onto its normal targets and forms ectopic synapses onto inappropriate targets. Misexpression of SYG-2 in secondary epithelial cells causes aberrant accumulation of SYG-1 and synaptic markers in HSNL adjacent to the SYG-2-expressing cells. Our results indicate that local interactions between immunoglobulin superfamily proteins can determine specificity during synapse formation.
Collapse
Affiliation(s)
- Kang Shen
- Howard Hughes Medical Institute, Department of Anatomy, The University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | |
Collapse
|
63
|
Abstract
Neurons are connected with a high degree of specificity in neuronal circuits. Axon guidance mechanisms are responsible for directing axons to their approximate target region. It is not well understood how precise synaptic connections form between specific pre- and postsynaptic neurons within the target area. Recent analysis of a group of cell surface proteins in different systems has shed light on the diverse cellular and molecular mechanisms that generate the precise patterns of connectivity.
Collapse
Affiliation(s)
- Kang Shen
- Department of Biological Sciences, 371 Serra Mall, Herrin Labs, Room 150, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
64
|
Babcock MC, Stowers RS, Leither J, Goodman CS, Pallanck LJ. A Genetic Screen for Synaptic Transmission Mutants Mapping to the Right Arm of Chromosome 3 in Drosophila. Genetics 2003; 165:171-83. [PMID: 14504225 PMCID: PMC1462763 DOI: 10.1093/genetics/165.1.171] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Abstract
Neuronal function depends upon the proper formation of synaptic connections and rapid communication at these sites, primarily through the regulated exocytosis of chemical neurotransmitters. Recent biochemical and genomic studies have identified a large number of candidate molecules that may function in these processes. To complement these studies, we are pursuing a genetic approach to identify genes affecting synaptic transmission in the Drosophila visual system. Our screening approach involves a recently described genetic method allowing efficient production of mosaic flies whose eyes are entirely homozygous for a mutagenized chromosome arm. From a screen of 42,500 mutagenized flies, 32 mutations on chromosome 3R that confer synaptic transmission defects in the visual system were recovered. These mutations represent 14 complementation groups, of which at least 9 also appear to perform functional roles outside of the eye. Three of these complementation groups disrupt photoreceptor axonal projection, whereas the remaining complementation groups confer presynaptic defects in synaptic transmission without detectably altering photoreceptor structure. Mapping and complementation testing with candidate mutations revealed new alleles of the neuronal fate determinant svp and the synaptic vesicle trafficking component lap among the collection of mutants recovered in this screen. Given the tools available for investigation of synaptic function in Drosophila, these mutants represent a valuable resource for future analysis of synapse development and function.
Collapse
Affiliation(s)
- Michael C Babcock
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195-7730, USA
| | | | | | | | | |
Collapse
|
65
|
Sun C, Kilburn D, Lukashin A, Crowell T, Gardner H, Brundiers R, Diefenbach B, Carulli JP. Kirrel2, a novel immunoglobulin superfamily gene expressed primarily in beta cells of the pancreatic islets. Genomics 2003; 82:130-42. [PMID: 12837264 DOI: 10.1016/s0888-7543(03)00110-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
A novel immunoglobulin superfamily (Igsf) protein gene was discovered by computational analysis of human draft genomic DNA, and multiple cDNA clones were obtained. The protein encoded by this gene contains five Ig domains, one transmembrane domain, and an intracellular domain. It has significant similarity with several known Igsf proteins, including Drosophila RST (irregular chiasm C-roughest) protein and mammalian KIRREL (kin of irregular chiasm C-roughest), NEPH1, and NPHS1 (nephrin) proteins. All these proteins have multiple Ig domains, possess properties of cell adhesion molecules, and play important roles in organ development. RT-PCR and Northern blot results indicate this gene is predominantly expressed in pancreas, and public sequence databases indicate there is also expression in the nervous system. We have named this gene Kirrel2 (kin of irregular chiasm-like 2), to reflect its similarity to irregular chiasm C-roughest and Kirrel. Four splice forms of Kirrel2 were observed, including two that we cloned from pancreas mRNA as well as two GenBank entries, one from the brain and one from a retinoblastoma cell line. A partial cDNA clone of the mouse orthologue was obtained by RT-PCR from mouse brain, and the inferred protein sequence has 85% sequence identity to the human protein. Immunohistochemical staining results indicate that the KIRREL2 protein is conserved from rodents to primates, and it is highly expressed in pancreatic islets. RT-PCR results on mouse pancreatic cell lines indicate that expression in the pancreas is restricted to beta cells. Thus, KIRREL2 protein is a beta-cell-expressed Ig domain protein and may be involved in pancreas development or beta cell function.
Collapse
Affiliation(s)
- Chao Sun
- Gene Discovery Department, Biogen, Inc., 12 Cambridge Center, Cambridge, MA 02142, USA.
| | | | | | | | | | | | | | | |
Collapse
|
66
|
Araujo H, Machado LCH, Octacílio-Silva S, Mizutani CM, Silva MJF, Ramos RGP. Requirement of the roughest gene for differentiation and time of death of interommatidial cells during pupal stages of Drosophila compound eye development. Mech Dev 2003; 120:537-47. [PMID: 12782271 DOI: 10.1016/s0925-4773(03)00040-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The roughest locus of Drosophila melanogaster encodes a transmembrane protein of the immunoglobulin superfamily required for several developmental processes, including axonal pathfinding in the developing optic lobe, mechanosensory bristle differentiation and myogenesis. In the compound eye, rst was previously shown to be required for establishing the correct number and spacing of secondary and tertiary pigment cells during the final steps of ommatidial assembly. We have further investigated its function in the developing pupal retina by performing a developmental and molecular analysis of a novel dominant rst allele, rst(D). In addition to showing evidence that rst(D) is a regulatory mutant, the results strongly suggest a previously unnoticed role of the rst gene in the differentiation of secondary/tertiary pigment cell fate as well as establishing the correct timing of surplus cell removal by programmed cell death in the compound eye.
Collapse
Affiliation(s)
- Helena Araujo
- Departamento de Embriologia e Histologia, Universidade Federal do Rio de Janeiro, 21949-900, Rio de Janeiro, Brazil
| | | | | | | | | | | |
Collapse
|
67
|
Shen K, Bargmann CI. The immunoglobulin superfamily protein SYG-1 determines the location of specific synapses in C. elegans. Cell 2003; 112:619-30. [PMID: 12628183 DOI: 10.1016/s0092-8674(03)00113-2] [Citation(s) in RCA: 233] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
During nervous system development, neurons form reproducible synapses onto specific targets. Here, we analyze the development of stereotyped synapses of the C. elegans HSNL neuron in vivo. Postsynaptic neurons and muscles were not required for accurate synaptic vesicle clustering in HSNL. Instead, vulval epithelial cells that contact HSNL act as synaptic guidepost cells that direct HSNL presynaptic vesicles to adjacent regions. The mutant syg-1(ky652) has defects in synapse formation that resemble those in animals that lack vulval epithelial cells: HSNL synaptic vesicles fail to accumulate at normal synaptic locations and form ectopic anterior clusters. syg-1 encodes an immunoglobulin superfamily protein that acts in the presynaptic HSNL axon. SYG-1 protein is localized to the site of future synapses, where it initiates synapse formation and localizes synaptic connections in response to the epithelial signal. SYG-1 is related to Drosophila IrreC and vertebrate NEPH1 proteins, which mediate cell-cell recognition in diverse developmental contexts.
Collapse
Affiliation(s)
- Kang Shen
- Department of Anatomy, Howard Hughes Medical Institute, Box 0452, University of California, San Francisco, San Francisco, CA 94143, USA
| | | |
Collapse
|
68
|
Abstract
Early development in many tissues is characterized by a rapid expansion in cell number. Excess cells are removed through activation of their intrinsic apoptotic machinery. This over-expansion followed by selective removal is important for the sculpting of these tissues, and how specific cells are selected to die is one of the central questions in development. The Drosophila eye is a unique example of such patterning through cell death. Because of its remarkable reiterative design, the fly eye lends itself to studies of mutants with increased or decreased apoptosis. We know that the process of elimination of lattice cells is highly regulated. And we have learned that each ommatidial unit is involved in the life-death decision of lattice cells through cell-cell signaling. But, we have yet to understand how this signaling is regulated spatially to result in such precision. In this article, we describe and speculate on the role of selective cell death during maturation of the fly eye.
Collapse
Affiliation(s)
- Carrie Baker Brachmann
- Developmental and Cell Biology, University of California, Irvine, 5205 McGaugh Hall, Irvine, CA 92697-2300, USA
| | | |
Collapse
|
69
|
Abstract
Somatic muscle formation is an unusual process as it requires the cells involved, the myoblasts, to relinquish their individual state and fuse with one another to form a syncitial muscle fiber. The potential use of myoblast fusion therapies to rebuild damaged muscles has generated continuing interest in elucidating the molecular basis of the fusion process. Yet, until recently, few of the molecular players involved in this process had been identified. Now, however, it has been possible to couple a detailed understanding of the cellular basis of the fusion process with powerful classical and molecular genetic strategies in the Drosophila embryo. We review the cellular studies, and the recent genetic and biochemical analyses that uncovered interacting extracellular molecules present on fusing myoblasts and the intracellular effectors that facilitate fusion. With the conservation of proteins and protein functions across species, it is likely that these findings in Drosophila will benefit understanding of the myoblast fusion process in higher organisms.
Collapse
Affiliation(s)
- Heather A Dworak
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | | |
Collapse
|
70
|
Machado RC, Pereira RNR, Costa MSA, Ramos RGP. "In vivo" toxicity of a truncated version of the Drosophila Rst-IrreC protein is dependent on the presence of a glutamine-rich region in its intracellular domain. AN ACAD BRAS CIENC 2002; 74:285-95. [PMID: 12098754 DOI: 10.1590/s0001-37652002000200007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The roughest-irregular chiasm C ( rst-irreC) gene of Drosophila melanogaster encodes a transmembrane glycoprotein containing five immunoglobulin-like domains in its extracellular portion and an intracytoplasmic tail rich in serine and threonine as well some conserved motifs suggesting signal transduction activity. In the compound eye, loss-of-function rst-irreC mutants lack the characteristic wave of programmed cell death happening in early pupa and which is essential for the elimination of the surplus interommatidial cells. Here we report an investigation on the role played by the Rst-irreC molecule in triggering programmed cell death. "In vivo" transient expression assays showed that deletion of the last 80 amino acids of the carboxyl terminus produces a form of the protein that is highly toxic to larvae. This toxicity is suppressed if an additional 47 amino acid long, glutamine-rich region ("opa-like domain"), is also removed from the protein. The results suggest the possibility that the opa-like domain and the carboxyl terminus act in concert to modulate rst-irreC function in apoptosis, and we discuss this implication in the context of the general mechanisms causing glutamine-rich neurodegenerative diseases in humans.
Collapse
Affiliation(s)
- Ricardo C Machado
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-900
| | | | | | | |
Collapse
|
71
|
Gorski S, Marra M. Programmed cell death takes flight: genetic and genomic approaches to gene discovery in Drosophila. Physiol Genomics 2002; 9:59-69. [PMID: 12006672 DOI: 10.1152/physiolgenomics.00114.2001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Programmed cell death (PCD) is an essential and wide-spread physiological process that results in the elimination of cells. Genes required to carry out this process have been identified, and many of these remain the subjects of intense investigation. Here, we describe PCD, its functions, and some of the consequences when it goes awry. We review PCD in the model system, the fruit fly, Drosophila melanogaster, with a particular emphasis on cell death gene discovery resulting from both genetics and genomics-based approaches.
Collapse
Affiliation(s)
- S Gorski
- Genome Sequence Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada V5Z 4E6.
| | | |
Collapse
|
72
|
Iwai Y, Hirota Y, Ozaki K, Okano H, Takeichi M, Uemura T. DN-cadherin is required for spatial arrangement of nerve terminals and ultrastructural organization of synapses. Mol Cell Neurosci 2002; 19:375-88. [PMID: 11906210 DOI: 10.1006/mcne.2001.1081] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We studied roles of DN-cadherin, the Drosophila major neuronal cadherin, in neuronal connections in the visual system. In DN-cadherin mutants, axon terminals of a large subset of photoreceptor cells reached and associated with their target interneurons, but their characteristic spatial arrangement was disrupted as synaptogenesis proceeded. Although synapses were formed at contact sites between the axon terminals and target neurons, underlying cytoplasmic structures were not fully specialized at both pre- and postsynaptic terminals and synaptic vesicles appeared to accumulate at the presynapses. These results suggest that the cadherin adhesion system is required for interaction between pre- and postsynaptic terminals and for generation of the mature synaptic structures.
Collapse
Affiliation(s)
- Youichi Iwai
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa, Sakyo-ku, Kyoto 606-8502, Japan
| | | | | | | | | | | |
Collapse
|
73
|
Strünkelnberg M, Bonengel B, Moda LM, Hertenstein A, de Couet HG, Ramos RG, Fischbach KF. rstand its paraloguekirreact redundantly during embryonic muscle development inDrosophila. Development 2001; 128:4229-39. [PMID: 11684659 DOI: 10.1242/dev.128.21.4229] [Citation(s) in RCA: 155] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The polynucleate myotubes of vertebrates and invertebrates form by fusion of myoblasts. We report the involvement of the Drosophila melanogaster Roughest (Rst) protein as a new membrane-spanning component in this process. Rst is strongly expressed in mesodermal tissues during embryogenesis, but rst null mutants display only subtle embryonic phenotypes. Evidence is presented that this is due to functional redundancy between Rst and its paralogue Kirre. Both are highly related single-pass transmembrane proteins with five extracellular immunoglobulin domains and three conserved motifs in the intracellular domain. The expression patterns of kirre and rst overlap during embryonic development in muscle founder cells. Simultaneous deletion of both genes causes an almost complete failure of fusion between muscle founder cells and fusion-competent myoblasts. This defect can be rescued by one copy of either gene. Moreover, Rst, like Kirre is a myoblast attractant.
Collapse
Affiliation(s)
- M Strünkelnberg
- Institut für Biologie III, Schänzlestr.1, Albert-Ludwigs-Universität, D-79104 Freiburg im Breisgau, Germany
| | | | | | | | | | | | | |
Collapse
|
74
|
Dworak HA, Charles MA, Pellerano LB, Sink H. Characterization ofDrosophila hibris, a gene related to human nephrin. Development 2001; 128:4265-76. [PMID: 11684662 DOI: 10.1242/dev.128.21.4265] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Hibris encodes a protein that is a newly identified member of the immunoglobulin superfamily and has homology to vertebrate Nephrins and Drosophila Sticks-and-Stones. The Hibris protein has eight Ig-like domains, a fibronectin domain and a 160 amino acid cytoplasmic tail. The hibris transcript is expressed in a broad range of tissues and across life stages. In the embryo, hibris transcript is present in the mesectoderm, then in a group of cells at the developing CNS midline and in a subset of glia. In the periphery, hibris is expressed by fusion competent myoblasts and the epidermal muscle attachment site cells. Deletion analyses show that loss of hibris does not visibly affect embryonic CNS or somatic muscle development. However overexpressing hibris in the somatic mesoderm disrupts myoblast fusion. Furthermore, when overexpressed in the epidermis, Hibris causes comprehensive derangement of muscle insertion locations. A similar myoblast fusion defect is observed when the Drosophila homologs of DM-GRASP/BEN/SC1 (irregular chiasm-roughest and dumbfounded) are deleted together. Our S2 cell aggregation assays have revealed a heterotypic interaction between Hibris and Dumbfounded, but not between Hibris and Irregular Chiasm-Roughest. We propose that Hibris is an extracellular partner for Dumbfounded and potentially mediates the response of myoblasts to this attractant.
Collapse
Affiliation(s)
- H A Dworak
- Skirball Institute of Biomolecular Medicine and Department of Pharmacology, New York University Medical School, 540 First Avenue, New York, NY 10016, USA
| | | | | | | |
Collapse
|
75
|
Schubert W, Kaprielian Z. Identification and characterization of a cell surface marker for embryonic rat spinal accessory motor neurons. J Comp Neurol 2001; 439:368-83. [PMID: 11596060 DOI: 10.1002/cne.1356] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The developing mammalian spinal cord contains distinct populations of motor neurons that can be distinguished by their cell body positions, by the expression of specific combinations of regulatory genes, and by the paths that their axons take to exit the central nervous system (CNS). Subclasses of spinal motor neurons are also thought to express specific cell surface proteins that function as receptors which control the guidance of their axons. We identified monoclonal antibody (mAb) SAC1 in a screen aimed at generating markers for specific subsets of neurons/axons in the developing rat spinal cord. During early embryogenesis, mAb SAC1 selectively labels a small subset of Isl1-positive motor neurons located exclusively within cervical segments of the spinal cord. Strikingly, these neurons extend mAb SAC1-positive axons along a dorsally directed trajectory toward the lateral exit points. Consistent with the finding that mAb SAC1 also labels spinal accessory nerves, these observations identify mAb SAC1 as a specific marker of spinal accessory motor neurons/axons. During later stages of embryogenesis, mAb SAC1 is transiently expressed on both dorsally and ventrally projecting spinal motor neurons/axons. Interestingly, mAb SAC1 also labels the notochord and floor plate during most stages of spinal cord development. The mAb SAC1 antigen is a 100-kD glycoprotein that is likely to be the rat homolog of SC1/BEN/DM-GRASP, a homophilic adhesion molecule that mediates axon outgrowth and fasciculation.
Collapse
Affiliation(s)
- W Schubert
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | |
Collapse
|
76
|
Abstract
A requirement for nitric oxide (NO) in visual system development has been demonstrated in many model systems, but the role of potential downstream effector molecules has not been established. Developing Drosophila photoreceptors express an NO-sensitive soluble guanylate cyclase (sGC), whereas the optic lobe targets express NO synthase. Both of these molecules are expressed after photoreceptor outgrowth to the optic lobe, when retinal growth cones are actively selecting their postsynaptic partners. We have previously shown that inhibition of the NO-cGMP pathway in vitro leads to overgrowth of retinal axons. Here we examined flies mutant for the alpha subunit gene of the Drosophila sGC (Gcalpha1). This mutation severely reduced but did not abolish GCalpha1 protein levels and NO-stimulated sGC activity in the developing photoreceptors. Although few mutant individuals possessed a disorganized retinal projection pattern, pharmacological NOS inhibition during metamorphosis increased this disorganization in mutants to a greater degree than in the wild type. Adult mutants lacked phototactic behavior, and the off-transient component of electroretinograms was frequently absent or greatly reduced in amplitude. Normal phototaxis and off-transient amplitude were restored by heat shock-mediated Gcalpha1 expression applied during metamorphosis but not in the adult. We propose that diminished sGC activity in the visual system during development causes inappropriate or inadequate formation of first-order retinal synapses, leading to defects in visual system function and visually mediated behavior.
Collapse
|
77
|
Donoviel DB, Freed DD, Vogel H, Potter DG, Hawkins E, Barrish JP, Mathur BN, Turner CA, Geske R, Montgomery CA, Starbuck M, Brandt M, Gupta A, Ramirez-Solis R, Zambrowicz BP, Powell DR. Proteinuria and perinatal lethality in mice lacking NEPH1, a novel protein with homology to NEPHRIN. Mol Cell Biol 2001; 21:4829-36. [PMID: 11416156 PMCID: PMC87176 DOI: 10.1128/mcb.21.14.4829-4836.2001] [Citation(s) in RCA: 317] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
A high-throughput, retrovirus-mediated mutagenesis method based on gene trapping in embryonic stem cells was used to identify a novel mouse gene. The human ortholog encodes a transmembrane protein containing five extracellular immunoglobulin-like domains that is structurally related to human NEPHRIN, a protein associated with congenital nephrotic syndrome. Northern analysis revealed wide expression in humans and mice, with highest expression in kidney. Based on similarity to NEPHRIN and abundant expression in kidney, this protein was designated NEPH1 and embryonic stem cells containing the retroviral insertion in the Neph1 locus were used to generate mutant mice. Analysis of kidney RNA from Neph1(-/-) mice showed that the retroviral insertion disrupted expression of Neph1 transcripts. Neph1(-/-) pups were represented at the expected normal Mendelian ratios at 1 to 3 days of age but at only 10% of the expected frequency at 10 to 12 days after birth, suggesting an early postnatal lethality. The Neph1(-/-) animals that survived beyond the first week of life were sickly and small but without edema, and all died between 3 and 8 weeks of age. Proteinuria ranging from 300 to 2,000 mg/dl was present in all Neph1(-/-) mice. Electron microscopy demonstrated NEPH1 expression in glomerular podocytes and revealed effacement of podocyte foot processes in Neph1(-/-) mice. These findings suggest that NEPH1, like NEPHRIN, may play an important role in maintaining the structure of the filtration barrier that prevents proteins from freely entering the glomerular urinary space.
Collapse
Affiliation(s)
- D B Donoviel
- Pharmaceutical Biology, Lexicon Genetics, Inc., The Woodlands, Texas 77381, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Ott H, Diekmann H, Stuermer CA, Bastmeyer M. Function of Neurolin (DM-GRASP/SC-1) in guidance of motor axons during zebrafish development. Dev Biol 2001; 235:86-97. [PMID: 11412029 DOI: 10.1006/dbio.2001.0278] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neurolin (zf DM-GRASP), a transmembrane protein with five extracellular immunoglobulin domains, is expressed by secondary but not primary motoneurons during zebrafish development. The spatiotemporally restricted expression pattern suggests that Neurolin plays a role in motor axon growth and guidance. To test this hypothesis, we injected zebrafish embryos with function-blocking Neurolin antibodies. In injected embryos, secondary motor axons form a broadened bundle along the common path and ectopic branches leave the common path at right angles. Moreover, the formation of the ventral and the rostral projection of secondary motor axons is inhibited during the second day of development. Pathfinding errors, resulting in secondary motor axons growing through ectopic regions of the somites, occur along the common path and in the dorsal and rostral projection. Our data are compatible with the view that Neurolin is involved in the recognition of guidance cues and acts as a receptor on secondary motor axons. Consistent with this idea is the binding pattern of a soluble Neurolin-Fc construct showing that putative ligands are distributed along the common path, the ventral projection, and in the area where the rostral projection develops.
Collapse
Affiliation(s)
- H Ott
- Department of Biology, University of Konstanz, Fach M626, Konstanz, D-78457, Germany
| | | | | | | |
Collapse
|
79
|
Callaerts P, Leng S, Clements J, Benassayag C, Cribbs D, Kang YY, Walldorf U, Fischbach KF, Strauss R. Drosophila Pax-6/eyeless is essential for normal adult brain structure and function. JOURNAL OF NEUROBIOLOGY 2001; 46:73-88. [PMID: 11153010 DOI: 10.1002/1097-4695(20010205)46:2<73::aid-neu10>3.0.co;2-n] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
A role for the Pax-6 homologue eyeless in adult Drosophila brain development and function is described. eyeless expression is detected in neurons, but not glial cells, of the mushroom bodies, the medullar cortex, the lateral horn, and the pars intercerebralis. Furthermore, severe defects in adult brain structures essential for vision, olfaction, and for the coordination of locomotion are provoked by two newly isolated mutations of Pax-6/eyeless that result in truncated proteins. Consistent with the morphological lesions, we observe defective walking behavior for these eyeless mutants. The implications of these data for understanding postembryonic brain development and function in Drosophila are discussed.
Collapse
Affiliation(s)
- P Callaerts
- Department of Biology and Biochemistry, University of Houston, Houston, Texas 77204-5513, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Hiesinger PR, Scholz M, Meinertzhagen IA, Fischbach KF, Obermayer K. Visualization of synaptic markers in the optic neuropils of Drosophila using a new constrained deconvolution method. J Comp Neurol 2001; 429:277-88. [PMID: 11116220 DOI: 10.1002/1096-9861(20000108)429:2<277::aid-cne8>3.0.co;2-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The fruitfly Drosophila melanogaster offers compelling genetic advantages for the analysis of its nervous system, but cell size precludes immunocytochemical analysis of wild-type structure and mutant phenotypes beyond the level of neuronal arborizations. For many antibodies, especially when immunoelectron microscopy is not feasible, it would therefore be desirable to extend the resolution limit of confocal microscopy as far as possible. Because high-resolution confocal microscopy suffers from considerable blurring, so-called deconvolution algorithms are needed to remove, at least partially, the blur introduced by the microscope and by the specimen itself. Here, we present the establishment and application of a new deconvolution method to visualize synaptic markers in Drosophila optic neuropils at the resolution limit of light. We ascertained all necessary parameters experimentally and verified them by deconvolving injected fluorescent microspheres in immunostained optic lobe tissue. The resulting deconvolution method was used to analyze colocalization between the synaptic vesicle marker neuronal synaptobrevin and synaptic and putative synaptic markers in photoreceptor terminals. We report differential localization of these near the resolution limit of light, which could not be distinguished without deconvolution.
Collapse
Affiliation(s)
- P R Hiesinger
- Institute for Biology III, University of Freiburg, D-79104 Freiburg, Germany.
| | | | | | | | | |
Collapse
|
81
|
Abstract
Selective cell death provides developing tissues with the means to precisely sculpt emerging structures. By imposing patterned cell death across a tissue, boundaries can be created and tightened. As such, programmed cell death is becoming recognized as a major mechanism for patterning of a variety of complex structures. Typically, cell types are initially organized into a fairly loose pattern; selective death then removes cells between pattern elements to create correct structures. In this review, we examine the role of selective cell death across the course of Drosophila development, including the tightening of embryonic segmental boundaries, head maturation, refining adult structures such as the eye and the wing, and the ability of cell death to correct for pattern defects introduced by gene mutation. We also review what is currently known of the relationship between signals at the cell surface that are responsible for tissue patterning and the basal cell death machinery, an issue that remains poorly understood.
Collapse
Affiliation(s)
- J C Rusconi
- Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, Missouri, MO 63110, USA
| | | | | |
Collapse
|
82
|
Gorski SM, Brachmann CB, Tanenbaum SB, Cagan RL. Delta and notch promote correct localization of irreC-rst. Cell Death Differ 2000; 7:1011-3. [PMID: 11324581 DOI: 10.1038/sj.cdd.4400742] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
83
|
Tanenbaum SB, Gorski SM, Rusconi JC, Cagan RL. A screen for dominant modifiers of the irreC-rst cell death phenotype in the developing Drosophila retina. Genetics 2000; 156:205-17. [PMID: 10978286 PMCID: PMC1461222 DOI: 10.1093/genetics/156.1.205] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Programmed cell death (PCD) in the Drosophila retina requires activity of the irregular chiasmC-roughest (irreC-rst) gene. Loss-of-function mutations in irreC-rst block PCD during retinal development and lead to a rough eye phenotype in the adult. To identify genes that interact with irreC-rst and may be involved in PCD, we conducted a genetic screen for dominant enhancers and suppressors of the adult rough eye phenotype. We screened 150,000 mutagenized flies and recovered 170 dominant modifiers that localized primarily to the second and third chromosomes. At least two allelic groups correspond to previously identified death regulators, Delta and dRas1. Examination of retinae from homozygous viable mutants indicated two major phenotypic classes. One class exhibited pleiotropic defects while the other class exhibited defects specific to the cell population that normally undergoes PCD.
Collapse
Affiliation(s)
- S B Tanenbaum
- Department of Molecular Biology and Pharmacology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
84
|
Moda L, Machado RC, Ramos RG. Ubiquitous overexpression of a transgene encoding the extracellular portion of the Drosophila roughest-irregular chiasm C protein induces early embryonic lethality. AN ACAD BRAS CIENC 2000; 72:381-8. [PMID: 11028102 DOI: 10.1590/s0001-37652000000300011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The cell adhesion molecule Rst-irreC is a transmembrane glycoprotein of the immunoglobulin superfamily involved in several important developmental processes in Drosophila, including axonal pathfinding in the optic lobe and programmed cell death and pigment cell differentiation in the pupal retina. As an initial step towards the "in vivo" functional analysis of this protein we have generated transgenic fly stocks carrying a truncated cDNA construct encoding only the extracellular domain of Rst-IrreC under the transcriptional control of the heat shock inducible promoter hsp70. We show that heat-shocking embryos bearing the transgene during the first 8hs of development lead to a 3-4 fold reduction in their viability compared to wild type controls. The embryonic lethality can already be produced by applying the heat pulse in the first 3hs of embryonic development, does not seem to be suppressed in the absence of wildtype product and is progressively reduced as the heat treatment is applied later in embryogenesis. These results are compatible with the hypothesis of the lethal phenotype being primarily due to heterophilic interactions between Rst-IrreC extracellular domain and an yet unknown ligand.
Collapse
Affiliation(s)
- L Moda
- Departamento de Morfologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | | | | |
Collapse
|
85
|
Hoyle HD, Turner FR, Raff EC. A transient specialization of the microtubule cytoskeleton is required for differentiation of the Drosophila visual system. Dev Biol 2000; 221:375-89. [PMID: 10790333 DOI: 10.1006/dbio.2000.9674] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Drosophila beta3-tubulin is an essential isoform expressed during differentiation of many cell types in embryos and pupae. We report here that during pupal development transient beta3 expression demarcates a unique subset of neurons in the developing adult visual system. beta3 is coassembled into microtubules with beta1, the sole beta-tubulin isoform in the permanent microtubule cytoskeleton of the adult eye and brain. Examination of beta3 mutant phenotypes showed that beta3 is required for axonal patterning and connectivity and for spatial positioning within the optic lobe. Comparison of the phenotypes of beta3 mutations with those that result from disruption of the Hedgehog signaling pathway shows that beta3 functions early in the establishment of the adult visual system. Our data support the hypothesis that beta3 confers specialized properties on the microtubules into which it is incorporated. Thus a transient specialization of the microtubule cytoskeleton during differentiation of a specific subset of the neurons has permanent consequences for later cell function.
Collapse
Affiliation(s)
- H D Hoyle
- Department of Biology and Institute for Molecular Biology, Indiana University, Bloomington, Indiana, 47405, USA.
| | | | | |
Collapse
|
86
|
Meinertzhagen IA, Piper ST, Sun XJ, Fröhlich A. Neurite morphogenesis of identified visual interneurons and its relationship to photoreceptor synaptogenesis in the flies, Musca domestica and Drosophila melanogaster. Eur J Neurosci 2000; 12:1342-56. [PMID: 10762363 DOI: 10.1046/j.1460-9568.2000.00033.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The first neuropile, or lamina, of the fly's optic lobe comprises a model set of identified neurons that are arrayed in cylindrical modules, called cartridges. The cartridge acquires adult form only in the second half of the fly's pupal life. All cells are by then correctly located within each of the lamina's cartridges (Drosophila, Musca), becoming invested by glial cells after 75% of pupal development (P + 75%). In adult cartridges, two lamina cells, L1 and L2, receive input from photoreceptor terminals R1-R6, at so-called tetrad synapses that form in the pupa when these cells' dendrites contact R1-R6. Single-section electron microscopy (EM, Drosophila) and serial-EM reconstructions of L1 and L2 (Musca) reveal relationships between the morphogenesis of L1/L2 dendrites and the formation of tetrads. Neurite outgrowth is initially (P + 55%) random and neurites are unbranched; many neurites invaginate surrounding terminals of R1-R6 but, later, embrace the outer surfaces of these. The maximum profusion of neurites at P + 74% coincides with peak numbers of nascent tetrads; neurites then branch vertically, in the lamina's depth. Later, neurites failing to reach R1-R6's outer surfaces regress. Down the length of their axons, L1 and L2's neurites initially form a random sequence, L1 partnering L1 as often as L2, etc., but beginning at P + 74%, L1 partners L2, and L2 partners L1, with progressive strictness. L1 has more neurites overall than L2. These observations are consistent with the following hypotheses: a neurite only survives if it contacts a presynaptic site; a synapse only survives if it progressively acquires the appropriate number and combination of postsynaptic neurites, culminating in a tetrad; an interaction exists between the neurites of L1 and L2, so that the growth of one respects the pattern of growth of the other.
Collapse
Affiliation(s)
- I A Meinertzhagen
- Life Sciences Centre, Dalhousie University, Halifax, Nova Scotia, Canada.
| | | | | | | |
Collapse
|
87
|
Bonini NM, Fortini ME. Surviving Drosophila eye development: integrating cell death with differentiation during formation of a neural structure. Bioessays 1999; 21:991-1003. [PMID: 10580984 DOI: 10.1002/(sici)1521-1878(199912)22:1<991::aid-bies3>3.0.co;2-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Normal differentiation requires an appropriately orchestrated sequence of developmental events. Regulation of cell survival and cell death is integrated with these events to achieve proper cell number, cell type, and tissue structure. Here we review regulation of cell survival in the context of a precisely patterned neural structure: the Drosophila compound eye. Numerous mutations lead to altered differentiation and are frequently accompanied by altered patterns of cell death. We discuss various critical times of normal eye development, highlighting how inappropriate regulation of cell death contributes to different mutant phenotypes associated with genes that specify the entire eye primordia, others that pattern the retina, and those that eliminate extraneous cells to refine the precise pigment cell lattice. Finally, we address how the Drosophila eye may allow identification of additional mechanisms that contribute to the normal integration of cell survival with appropriate events of cellular differentiation.
Collapse
Affiliation(s)
- N M Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | | |
Collapse
|
88
|
Neuropil pattern formation and regulation of cell adhesion molecules in Drosophila optic lobe development depend on synaptobrevin. J Neurosci 1999. [PMID: 10460261 DOI: 10.1523/jneurosci.19-17-07548.1999] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
To investigate a possible involvement of synaptic machinery in Drosophila visual system development, we studied the effects of a loss of function of neuronal synaptobrevin, a protein required for synaptic vesicle release. Expression of tetanus toxin light chain (which cleaves neuronal synaptobrevin) and genetic mosaics were used to analyze neuropil pattern formation and levels of selected neural adhesion molecules in the optic lobe. We show that targeted toxin expression in the developing optic lobe results in disturbances of the columnar organization of visual neuropils and of photoreceptor terminal morphology. IrreC-rst immunoreactivity in neuropils is increased after widespread expression of toxin. In photoreceptors, targeted toxin expression results in increased Fasciclin II and chaoptin but not IrreC-rst immunoreactivity. Axonal pathfinding and programmed cell death are not affected. In genetic mosaics, patches of photoreceptors that lack neuronal synaptobrevin exhibit the same phenotypes observed after photoreceptor-specific toxin expression. Our results demonstrate the requirement of neuronal synaptobrevin for regulation of cell adhesion molecules and development of the fine structure of the optic lobe. A possible causal link to fine-tuning processes that may include synaptic plasticity in the development of the Drosophila CNS is discussed.
Collapse
|
89
|
Abstract
Cell surface adhesion molecules are thought to play a necessary role in axon guidance and fasciculation in the developing nervous system. We have studied a potential adhesion molecule using the zn-5 monoclonal antibody, which recognizes the surfaces of zebrafish spinal motoneurons. We show that zn-5 recognizes zebrafish DM-GRASP. DM-GRASP is a cell adhesion molecule of the immunoglobulin superfamily that mediates homophilic adhesion and neurite outgrowth in vitro. It is necessary for correct axon routing and fasciculation in the Drosophila visual system. In zebrafish, primary motoneurons pioneer the peripheral motor nerve pathways, and the axons of secondary motoneurons follow the routes established by the primary motoneuron axons. We show that, of the two classes of zebrafish spinal motoneurons, only the later growing secondary motoneurons express DM-GRASP. The secondary motoneurons restrict DM-GRASP protein to their cell bodies and fasciculated segments of their axons. Expression of DM-GRASP is transient: The protein is present during the period of axonal growth and disappears after axons have reached their muscle targets. Thus, homophilic adhesion mediated by DM-GRASP may play a role in fasciculation of secondary motoneuron axons but not in pathfinding by the pioneer axons of the primary motoneurons or in guidance of secondary motoneuron axons to their targets.
Collapse
Affiliation(s)
- D Fashena
- Institute of Neuroscience, University of Oregon, Eugene 97403, USA
| | | |
Collapse
|
90
|
Lekven AC, Tepass U, Keshmeshian M, Hartenstein V. faint sausage encodes a novel extracellular protein of the immunoglobulin superfamily required for cell migration and the establishment of normal axonal pathways in the Drosophila nervous system. Development 1998; 125:2747-58. [PMID: 9636088 DOI: 10.1242/dev.125.14.2747] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We examined the structure of the nervous system in Drosophila embryos homozygous for a null mutation in the faint sausage (fas) gene. In the peripheral nervous system (PNS) of fas mutants, neurons fail to delaminate from the ectodermal epithelium; in the central nervous system (CNS), the positions of neuronal cell bodies and glial cells are abnormal and normal axonal pathways do not form. Sequence analysis of fas cDNAs revealed that the fas protein product has characteristics of an extracellular protein and that it is a novel member of the immunoglobulin (Ig) superfamily. In situ hybridization demonstrated that fas transcripts are expressed throughout the embryo but they are in relatively high concentrations in the lateral ectoderm, from which the peripheral nervous system delaminates and in the CNS. Antiserum directed against Fas protein was found to stain neurons but not glia in the CNS. We conclude that fas encodes a protein that, in the developing nervous system, is present on the surface of neurons and is essential for nerve cell migration and the establishment of axonal pathways.
Collapse
Affiliation(s)
- A C Lekven
- Department of Molecular, Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, CA 90095-1606 USA
| | | | | | | |
Collapse
|
91
|
Abstract
Young axons of new retinal ganglion cells (RGCs) in the continuously growing goldfish retina fasciculate with one another and their immediate forerunners on their path toward the optic disk and along the optic nerve. They express the immunoglobulin superfamily cell adhesion molecules (CAMs) neurolin (DM-GRASP) and the L1-like E587 antigen. Repeated injections of Fab fragments from polyclonal antisera against neurolin (neurolin Fabs) into the eye of 3. 4-cm-long and rapidly growing goldfish caused highly aberrant pathways of young RGC axon subfascicles in the dorsal retina. Many axons grew in circles and failed to reach the optic disk. In contrast, E587 Fabs, used in parallel experiments, disrupted the fascicles but did not interfere with the disk-directed growth. Neurolin Fabs also disturbed axonal fasciculation in vivo as well as in vitro but less severely than E587 Fabs. Coinjections of both Fabs increased defasciculation of the dorsal axons in both aberrant and disk-directed routes. They also disrupted the order of young RGC axons in the optic nerve more severely than E587 Fabs alone. This demonstrates that the development of tight and orderly fascicles in the dorsal retina and in the optic nerve requires both E587 antigen and neurolin. More importantly, our results suggest an involvement of neurolin in RGC axonal guidance from the retinal periphery to the optic disk. Because disrupted fascicles and errant axon routes were found only in the dorsal retinal half, a cooperation with so-called positional markers may be conceived.
Collapse
|
92
|
Yamamoto D, Fujitani K, Usui K, Ito H, Nakano Y. From behavior to development: genes for sexual behavior define the neuronal sexual switch in Drosophila. Mech Dev 1998; 73:135-46. [PMID: 9622612 DOI: 10.1016/s0925-4773(98)00042-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The isolation and analysis of Drosophila mutants with altered sexual orientation lead to the identification of novel branches in the sex-determination cascade which govern the sexually dimorphic development of the nervous system. One such example is the fruitless (fru) gene, the mutation of which induces male-to-male courtship and malformation of a male-specific muscle, the muscle of Lawrence (MOL). Since the MOL is formed in wild-type flies when the innervating nerve is male, regardless of the sex of the MOL itself, the primary site of Fru function is likely to be the motoneurons controlling the MOL. The fru gene produces multiple transcripts including sex-specific ones. A female-specific mRNA from the fru locus has a putative Transformer (Tra) binding site in its 5' untranslated region, suggesting that fru is a direct target of Tra. The fru transcripts encode a set of proteins similar to the BTB (Bric à brac, Tramtrack and Broad-complex)-Zn finger family of transcription factors. Mutations in the dissatisfaction (dsf) gene result in male-to-male courtship and reduced sexual receptivity of females. The dsf mutations also give rise to poor curling of the abdomen in males during copulation and failure of egg-laying by females. The latter phenotypes are ascribable to aberrant innervation of the relevant muscles. A genetic analysis reveals that expression of the dsf phenotypes depends on Tra but not on Doublesex (Dsx) or Fru, suggesting that dsf represents another target of Tra. Taken together, these findings suggest that the sex-determination protein Tra has at least three different targets, dsx, fru and dsf, each of which represents the first gene in a branch of the sex-determination hierarchy functioning in a mutually-exclusive set of neuronal cells in the Drosophila central nervous system.
Collapse
Affiliation(s)
- D Yamamoto
- ERATO Yamamoto Behavior Genes Project, JST, Mitsubishi Kasei Institute of Life Sciences, Machida, Tokyo 194-8511, Japan.
| | | | | | | | | |
Collapse
|
93
|
Abstract
Numerous in vitro assays and in vivo perturbation studies have led to a model of neural development in which selective fasciculation helps to define accurate axonal projections. Genetic analysis in vivo confirms the hypothesis that axonal fasciculation and defasciculation are controlled by adhesion mechanisms, but also suggests that, in many cases, adhesion and guidance are separable phenomena. In addition, receptors that control the level of tyrosine phosphorylation may play an important role in fasciculation, suggesting that complex intracellular pathways lie just beneath the surface.
Collapse
Affiliation(s)
- D Van Vactor
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA.
| |
Collapse
|
94
|
Abstract
The avian basilar papilla is composed of hair and supporting cells arranged in a regular pattern in which the hair cells are surrounded and isolated from each other by supporting cell processes. This arrangement of cells, in which the apical borders of hair cells do not contact one another, may be generated by contact-mediated lateral inhibition. Little is known, however, about the way in which hair and supporting cells are organized during development. Whole mounts double-labeled with antibodies to the 275 kDa hair-cell antigen and the tight junction protein cingulin were therefore used to examine the development of cell patterns in the basilar papilla. Hair cells that contact each other at their apical borders are seen during early development, especially on embryonic days (E) 8 and 9, but are no longer observed after E12. Hair and supporting cell patterns were analyzed in three different areas of the papilla at E9 and E12. In two of these regions between E9 and E12, the ratio of supporting cells to hair cells does not change significantly, whereas there is an increase in both the number of supporting cells around each hair cell and the number of hair cells that each supporting cell contacts. In the third region examined, there is a dramatic rise in the number of supporting cells around each hair cell, which although accompanied by a small, significant increase in the ratio of supporting cells to hair cells cannot be accounted for by an increase in supporting cell numbers. These data show that a rearrangement of hair and supporting cells with respect to one another may be a fundamental process underlying the development of a regular pattern in the basilar papilla.
Collapse
|
95
|
Iwai Y, Usui T, Hirano S, Steward R, Takeichi M, Uemura T. Axon patterning requires DN-cadherin, a novel neuronal adhesion receptor, in the Drosophila embryonic CNS. Neuron 1997; 19:77-89. [PMID: 9247265 DOI: 10.1016/s0896-6273(00)80349-9] [Citation(s) in RCA: 251] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We identified DN-cadherin, a novel Drosophila cadherin that is expressed in axons and in the mesoderm. Although DN-cadherin has diverged from vertebrate classic cadherins in terms of its extracellular structure, it still can form a complex with catenins and induce cell aggregation, as do the vertebrate molecules. Loss-of-function mutations of the gene resulted in either embryonic lethality or uncoordinated locomotion of adults. In the central nervous system of null mutant embryos, subsets of ipsilateral axons displayed a variety of aberrant trajectories including failure of position shifts, defective bundling, and errors in directional migration of growth cones. These results suggest that processes of axon patterning critically depend on DN-cadherin-mediated axon-axon interactions.
Collapse
Affiliation(s)
- Y Iwai
- Department of Biophysics, Faculty of Science, Kyoto University, Kitashirakawa, Sakyo-ku, Japan
| | | | | | | | | | | |
Collapse
|
96
|
Butler SJ, Ray S, Hiromi Y. klingon, a novel member of the Drosophila immunoglobulin superfamily, is required for the development of the R7 photoreceptor neuron. Development 1997; 124:781-92. [PMID: 9043060 DOI: 10.1242/dev.124.4.781] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
klingon is a member of the Immunoglobulin superfamily and is expressed in a restricted pattern of neurons during embryonic neurogenesis and in the R7 photoreceptor precursor throughout its development. Starting from the H214 enhancer trap line, we identified a transcription unit, klingon, that encodes a putative protein of 528 amino acids and contains three C2-type Immunoglobulin-like domains followed by one fibronectin type III repeat. When Klingon is expressed in S2 tissue culture cells, it is associated with the cell membrane by a glycosyl-phosphatidylinositol linkage and can mediate homophilic adhesion. Genetic analysis has revealed that klingon is an essential gene that participates in the development of the R7 neuron. Ectopic expression of klingon in all neurons in a sevenless background can alter the position of the R8 rhabdomere.
Collapse
Affiliation(s)
- S J Butler
- Department of Molecular Biology, Princeton University, NJ 08544, USA
| | | | | |
Collapse
|
97
|
Cutforth T, Gaul U. The genetics of visual system development in Drosophila: specification, connectivity and asymmetry. Curr Opin Neurobiol 1997; 7:48-54. [PMID: 9039792 DOI: 10.1016/s0959-4388(97)80119-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Encoding visual information requires a complex neuronal network. Recently, genes regulating early tissue specification, the growth of retinal target structures, the connectivity of photoreceptor axons, and mirror-image retinal symmetry in Drosophila have been identified. The insights gained from studying visual system development in flies promise to inform our understanding of similar processes in vertebrates.
Collapse
Affiliation(s)
- T Cutforth
- Laboratory of Developmental Neurogenetics, Howard Hughes Medical Institute, College of Physicians and Surgeons, Columbia University, 701 West 168 Street, Room 1014, New York, New York 10032, USA.
| | | |
Collapse
|
98
|
Rachidi M, Lopes C, Benichou JC. Genetical analysis of visual system disorganizer (vid), a new gene involved in normal development of eye and optic lobe of the brain in Drosophila melanogaster. Genetica 1997; 99:31-45. [PMID: 9226435 DOI: 10.1007/bf02259496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A neuroanatomical screening of a collection of P-element mutagenized flies has been carried out with the aim of finding new mutants affecting the optic lobe of the adult brain in Drosophila melanogaster. We have identified a new gene that is involved in the development of the adult axon array in the optic ganglia and in the ommatidia assembly. We have named this locus visual system disorganizer (vid). Reversional mutagenesis demonstrated that the vid mutant was the result of a P-element insertion in the Drosophila genome and allowed us to generate independent alleles, some of which resulted in semilethality, like the vid original mutant, while the others were completely lethal. A genetic somatic mosaic analysis indicated that the vid gene is required in the eye for its normal development by inductive effects. This analysis also suggests an inductive effect of the vid gene on the distal portion of the optic lobe, particularly the lamina and the first optic chiasma. Moreover, the absence of mutant phenotype in the proximal region of the optic ganglia, including the medulla, the second optic chiasma, and the lobula complex underlying mosaic eyes, is suggestive of an autonomously acting mechanism of the vid gene in the optic lobe. The complete or partial lethality generated by different mutations at the vid locus suggests that this gene's role may not be limited to the visual system, but may also affect a vital function during Drosophila development.
Collapse
Affiliation(s)
- M Rachidi
- Department of Molecular Biology, Pasteur Institute, Paris, France
| | | | | |
Collapse
|
99
|
Koushika SP, Lisbin MJ, White K. ELAV, a Drosophila neuron-specific protein, mediates the generation of an alternatively spliced neural protein isoform. Curr Biol 1996; 6:1634-41. [PMID: 8994828 DOI: 10.1016/s0960-9822(02)70787-2] [Citation(s) in RCA: 135] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Tissue-specific alternative pre-mRNA splicing is a widely used mechanism for gene regulation and the generation of different protein isoforms, but relatively little is known about the factors and mechanisms that mediate this process. Tissue-specific RNA-binding proteins could mediate alternative pre-mRNA splicing. In Drosophila melanogaster, the RNA-binding protein encoded by the elav (embryonic lethal abnormal visual system) gene is a candidate for such a role. The ELAV protein is expressed exclusively in neurons, and is important for the formation and maintenance of the nervous system. RESULTS In this study, photoreceptor neurons genetically depleted of ELAV, and elav-null central nervous system neurons, were analyzed immunocytochemically for the expression of neural proteins. In both situations, the lack of ELAV corresponded with a decrease in the immunohistochemical signal of the neural-specific isoform of Neuroglian, which is generated by alternative splicing. Furthermore, when ELAV was expressed ectopically in cells that normally express only the non-neural isoform of Neuroglian, we observed the generation of the neural isoform of Neuroglian. CONCLUSIONS Drosophila ELAV promotes the generation of the neuron-specific isoform of Neuroglian by the regulation of pre-mRNA splicing. The findings reported in this paper demonstrate that ELAV is necessary, and the ectopic expression of ELAV in imaginal disc cells is sufficient, to mediate neuron-specific alternative splicing.
Collapse
Affiliation(s)
- S P Koushika
- Biology Department, Brandeis University, Waltham, Massachusetts 02254, USA
| | | | | |
Collapse
|
100
|
Abstract
Neuronal growth cones navigate over long distances along specific pathways to find their correct targets. The mechanisms and molecules that direct this pathfinding are the topics of this review. Growth cones appear to be guided by at least four different mechanisms: contact attraction, chemoattraction, contact repulsion, and chemorepulsion. Evidence is accumulating that these mechanisms act simultaneously and in a coordinated manner to direct pathfinding and that they are mediated by mechanistically and evolutionarily conserved ligand-receptor systems.
Collapse
Affiliation(s)
- M Tessier-Lavigne
- Department of Anatomy, Howard Hughes Medical Institute, University of California, San Francisco, CA 94143, USA
| | | |
Collapse
|