Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Development ; 128(23): 4923-33, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11731471

ABSTRACT

During development of the Drosophila tracheal (respiratory) system, the cell bodies and apical and basal surfaces of the tracheal epithelium normally move in concert as new branches bud and grow out to form tubes. We show that mutations in the Drosophila ribbon (rib) gene disrupt this coupling: the basal surface continues to extend towards its normal targets, but movement and morphogenesis of the tracheal cell bodies and apical surface is severely impaired, resulting in long basal membrane protrusions but little net movement or branch formation. rib mutant tracheal cells are still responsive to the Branchless fibroblast growth factor (FGF) that guides branch outgrowth, and they express apical membrane markers normally. This suggests that the defect lies either in transmission of the FGF signal from the basal surface to the rest of the cell or in the apical cell migration and tubulogenesis machinery. rib encodes a nuclear protein with a BTB/POZ domain and Pipsqueak DNA-binding motif. It is expressed in the developing tracheal system and other morphogenetically active epithelia, many of which are also affected in rib mutants. We propose that Rib is a key regulator of epithelial morphogenesis that promotes migration and morphogenesis of the tracheal cell bodies and apical surface and other morphogenetic movements.


Subject(s)
Cytoskeletal Proteins/genetics , Drosophila Proteins/genetics , Drosophila/embryology , Drosophila/genetics , Genes, Insect , Nuclear Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Cytoskeletal Proteins/chemistry , Drosophila/metabolism , Drosophila Proteins/chemistry , Epithelium/growth & development , Epithelium/metabolism , Gene Expression Regulation, Developmental , In Situ Hybridization , Molecular Sequence Data , Morphogenesis/genetics , Mutation , Nuclear Proteins/chemistry , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Trachea/embryology , Trachea/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics
2.
Nat Cell Biol ; 3(9): 848-51, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11533666

ABSTRACT

Nuclear lamins are intermediate filaments that compose the nuclear lamina--the filamentous meshwork underlying the inner nuclear membrane--and are required for nuclear assembly, organization and maintenance. Here we present evidence that a nuclear lamin is also required for cytoplasmic organization in two highly polarized cell types. Zygotic loss-of-function mutations in the Drosophila gene encoding the principal lamin (Dm(0)) disrupt the directed outgrowth of cytoplasmic extensions from terminal cells of the tracheal system. Germline mutant clones disrupt dorsal-ventral polarity of the oocyte. In mutant oocytes, transcripts of the dorsal determinant Gurken, a transforming growth factor-alpha homologue, fail to localize properly around the anterodorsal surface of the oocyte nucleus; their ventral spread results in dorsalized eggs that resemble those of the classical dorsalizing mutations squid and fs(1)K10. The requirement of a nuclear lamin for cytoplasmic as well as nuclear organization has important implications for both the cellular functions of lamins and the pathogenesis of human diseases caused by lamin mutations.


Subject(s)
Cell Nucleus/physiology , Drosophila Proteins , Drosophila/genetics , Drosophila/physiology , Morphogenesis/physiology , Nuclear Proteins/genetics , Oocytes/cytology , Oocytes/physiology , Transforming Growth Factor alpha , Animals , Base Sequence , Cell Division , Cell Nucleus/ultrastructure , Cell Polarity , Cytoplasm/physiology , Cytoplasm/ultrastructure , Drosophila/embryology , Female , Gene Expression Regulation , Germ-Line Mutation , Humans , Insect Hormones/genetics , Insect Proteins/genetics , Lamins , Molecular Sequence Data , Nuclear Proteins/metabolism , RNA-Binding Proteins/genetics , Transcription, Genetic , Transforming Growth Factors/genetics , Zygote/cytology , Zygote/physiology
3.
J Biol Chem ; 276(6): 4128-33, 2001 Feb 09.
Article in English | MEDLINE | ID: mdl-11053436

ABSTRACT

Sprouty negatively modulates branching morphogenesis in the Drosophila tracheal system. To address the role of mammalian Sprouty homologues in angiogenesis, another form of branching morphogenesis, a recombinant adenovirus engineered to express murine Sprouty-4 selectively in endothelial cells, was injected into the sinus venosus of embryonic day 9.0 cultured mouse embryos. Sprouty-4 expression inhibited branching and sprouting of small vessels, resulting in abnormal embryonic development. In vitro, Sprouty-4 inhibited fibroblast growth factor and vascular endothelial cell growth factor-mediated cell proliferation and migration and prevented basic fibroblast growth factor and vascular endothelial cell growth factor-induced MAPK phosphorylation in endothelial cells, indicating inhibition of tyrosine kinase-mediated signaling pathways. The ability of constitutively activated mutant Ras(L61) to rescue Sprouty-4 inhibition of MAPK phosphorylation suggests that Sprouty inhibits receptor tyrosine kinase signaling upstream of Ras. Thus, Sprouty may regulate angiogenesis in normal and disease processes by modulating signaling by endothelial tyrosine kinases.


Subject(s)
Drosophila Proteins , Insect Proteins/physiology , Membrane Proteins , Neovascularization, Physiologic/physiology , Animals , Cell Cycle/physiology , Cell Division/physiology , Endothelium, Vascular/cytology , Flow Cytometry , Insect Proteins/genetics , Mice , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Transgenes
4.
Development ; 127(15): 3271-82, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10887083

ABSTRACT

The proper size of epithelial tubes is critical for the function of the lung, kidney, vascular system and other organs, but the genetic and cellular mechanisms that control epithelial tube size are unknown. We investigated tube size control in the embryonic and larval tracheal (respiratory) system of Drosophila. A morphometric analysis showed that primary tracheal branches have characteristic sizes that undergo programmed changes during development. Branches grow at different rates and their diameters and lengths are regulated independently: tube length increases gradually throughout development, whereas tube diameter increases abruptly at discrete times in development. Cellular analysis and manipulation of tracheal cell number using cell-cycle mutations demonstrated that tube size is not dictated by the specific number or shape of the tracheal cells that constitute it. Rather, tube size appears to be controlled by coordinately regulating the apical (lumenal) surface of tracheal cells. Genetic analysis showed that tube sizes are specified early by branch identity genes, and the subsequent enlargement of branches to their mature sizes and maintenance of the expanded tubes involves a new set of genes described here, which we call tube expansion genes. This work establishes a genetic system for investigating tube size regulation, and provides an outline of the genetic program and cellular events underlying tracheal tube size control.


Subject(s)
Drosophila/embryology , Animals , Cell Size , Drosophila/anatomy & histology , Drosophila/genetics , Larva , Trachea/anatomy & histology , Trachea/embryology
5.
Science ; 287(5461): 2218-20, 2000 Mar 24.
Article in English | MEDLINE | ID: mdl-10731136

ABSTRACT

The 120-megabase euchromatic portion of the Drosophila melanogaster genome has been sequenced. Because the genome is compact and many genetic tools are available, and because fly cell biology and development have much in common with mammals, this sequence may be the Rosetta stone for deciphering the human genome.


Subject(s)
Biology , Drosophila melanogaster/genetics , Genetics, Medical , Genome, Human , Genome , Sequence Analysis, DNA , Animals , Cloning, Molecular , DNA Transposable Elements , Drosophila melanogaster/physiology , Genes, Insect , Humans , Mutation , Physical Chromosome Mapping
6.
Cell ; 99(2): 211-20, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10535739

ABSTRACT

The Drosophila tracheal (respiratory) system is a tubular epithelial network that delivers oxygen to internal tissues. Sprouting of the major tracheal branches is stereotyped and controlled by hard-wired developmental cues. Here we show that ramification of the fine terminal branches is variable and regulated by oxygen, and that this process is controlled by a local signal or signals produced by oxygen-starved cells. We provide evidence that the critical signal is Branchless (Bnl) FGF, the same growth factor that patterns the major branches during embryogenesis. During larval life, oxygen deprivation stimulates expression of Bnl, and the secreted growth factor functions as a chemoattractant that guides new terminal branches to the expressing cells. Thus, a single growth factor is reiteratively used to pattern each level of airway branching, and the change in branch patterning results from a switch from developmental to physiological control of its expression.


Subject(s)
Drosophila Proteins , Drosophila melanogaster/physiology , Insect Proteins/physiology , Oxygen/physiology , Respiratory Mucosa/physiology , Respiratory Physiological Phenomena , Animals , Animals, Genetically Modified , Cues , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/genetics , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/physiology , Gene Expression Regulation, Developmental , Insect Proteins/genetics , Larva , Membrane Proteins/genetics , Membrane Proteins/physiology , Mosaicism , Mutagenesis , R-SNARE Proteins , Respiratory Mucosa/anatomy & histology , Respiratory System/anatomy & histology
7.
Development ; 126(20): 4465-75, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10498682

ABSTRACT

The Drosophila sprouty gene encodes an antagonist of FGF and EGF signaling whose expression is induced by the signaling pathways that it inhibits. Here we describe a family of vertebrate Sprouty homologs and demonstrate that the regulatory relationship with FGF pathways has been conserved. In both mouse and chick embryos, Sprouty genes are expressed in intimate association with FGF signaling centers. Gain- and loss-of-function experiments demonstrate that FGF signaling induces Sprouty gene expression in various tissues. Sprouty overexpression obtained by infecting the prospective wing territory of the chick embryo with a retrovirus containing a mouse Sprouty gene causes a reduction in limb bud outgrowth and other effects consistent with reduced FGF signaling from the apical ectodermal ridge. At later stages of development in the infected limbs there was a dramatic reduction in skeletal element length due to an inhibition of chondrocyte differentiation. The results provide evidence that vertebrate Sprouty proteins function as FGF-induced feedback inhibitors, and suggest a possible role for Sprouty genes in the pathogenesis of specific human chondrodysplasias caused by activating mutations in Fgfr3.


Subject(s)
Drosophila Proteins , Fibroblast Growth Factors/metabolism , Insect Proteins/genetics , Membrane Proteins , Osteochondrodysplasias/embryology , Osteochondrodysplasias/genetics , Amino Acid Sequence , Animals , Chick Embryo , Cloning, Molecular , Drosophila/embryology , Drosophila/genetics , Evolution, Molecular , Extremities/embryology , Female , Gene Expression Regulation, Developmental , Humans , Male , Mice , Mice, Knockout , Molecular Sequence Data , Mutation , Pregnancy , Sequence Homology, Amino Acid , Signal Transduction , Species Specificity
8.
Science ; 284(5420): 1635-9, 1999 Jun 04.
Article in English | MEDLINE | ID: mdl-10383344

ABSTRACT

The genetic programs that direct formation of the treelike branching structures of two animal organs have begun to be elucidated. In both the developing Drosophila tracheal (respiratory) system and mammalian lung, a fibroblast growth factor (FGF) signaling pathway is reiteratively used to pattern successive rounds of branching. The initial pattern of signaling appears to be established by early, more global embryonic patterning systems. The FGF pathway is then modified at each stage of branching by genetic feedback controls and other signals to give distinct branching outcomes. The reiterative use of a signaling pathway by both insects and mammals suggests a general scheme for patterning branching morphogenesis.


Subject(s)
Body Patterning/genetics , Drosophila/embryology , Fibroblast Growth Factors/physiology , Lung/embryology , Animals , Drosophila/anatomy & histology , Drosophila/genetics , Epithelium/metabolism , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental , Larva/growth & development , Lung/anatomy & histology , Mesoderm/metabolism , Morphogenesis/genetics , Signal Transduction , Trachea/anatomy & histology , Trachea/embryology
9.
Genetics ; 152(1): 307-18, 1999 May.
Article in English | MEDLINE | ID: mdl-10224263

ABSTRACT

Fibroblast growth factors (FGFs) bind to FGF receptors, transmembrane tyrosine kinases that activate mitogenic, motogenic, and differentiative responses in different tissues. While there has been substantial progress in elucidating the Ras-MAP kinase pathway that mediates the differentiative responses, the signal transduction pathways that lead to directed cell migrations are not well defined. Here we describe a Drosophila gene called stumps that is required for FGF-dependent migrations of tracheal and mesodermal cells. These migrations are controlled by different FGF ligands and receptors, and they occur by different cellular mechanisms: the tracheal migrations occur as part of an epithelium whereas the mesodermal migrations are fibroblast-like. In the stumps mutant, tracheal cells fail to move out from the epithelial sacs, and only rudimentary tracheal branches form. Mesodermal cells fail in their dorsal migrations after gastrulation. The stumps mutation does not block all FGF signaling effects in these tissues: both random cell migrations and Ras-MAP kinase-mediated induction of FGF-specific effector genes occurred upon ectopic expression of the ligand or upon expression of a constitutively activated Ras protein in the migrating cells. The results suggest that stumps function promotes FGF-directed cell migrations, either by potentiating the FGF signaling process or by coupling the signal to the cellular machinery required for directed cell movement.


Subject(s)
Cell Movement/physiology , Drosophila Proteins , Drosophila melanogaster/genetics , Fibroblast Growth Factors/genetics , Mesoderm/physiology , Trachea/physiology , Animals , Chromosome Mapping , Dose-Response Relationship, Drug , Gene Expression Regulation , Immunohistochemistry , Insect Proteins/metabolism , Mesoderm/cytology , Models, Biological , Phenotype , Signal Transduction , Trachea/cytology , ras Proteins/metabolism
10.
Development ; 126(11): 2515-25, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10226010

ABSTRACT

Extracellular factors such as FGF and EGF control various aspects of morphogenesis, patterning and cellular proliferation in both invertebrates and vertebrates. In most systems, it is primarily the distribution of these factors that controls the differential behavior of the responding cells. Here we describe the role of Sprouty in eye development. Sprouty is an extracellular protein that has been shown to antagonize FGF signaling during tracheal branching in Drosophila. It is a novel type of protein with a highly conserved cysteine-rich region. In addition to the embryonic tracheal system, sprouty is also expressed in other tissues including the developing eye imaginal disc, embryonic chordotonal organ precursors and the midline glia. In each of these tissues, EGF receptor signaling is known to participate in the control of the correct number of neurons or glia. We show that, in all three tissues, the loss of sprouty results in supernumerary neurons or glia, respectively. Furthermore, overexpression of sprouty in wing veins and ovarian follicle cells, two other tissues where EGF signaling is required for patterning, results in phenotypes that resemble the loss-of-function phenotypes of Egf receptor. These results suggest that Sprouty acts as an antagonist of EGF as well as FGF signaling pathways. These receptor tyrosine kinase-mediated pathways may share not only intracellular signaling components but also extracellular factors that modulate the strength of the signal.


Subject(s)
Drosophila Proteins , Drosophila/embryology , ErbB Receptors/antagonists & inhibitors , Fibroblast Growth Factors/antagonists & inhibitors , Insect Proteins/genetics , Membrane Proteins , Signal Transduction/genetics , Animals , Drosophila/genetics , Ethyl Methanesulfonate/pharmacology , Eye/embryology , Eye Proteins , Gene Expression Regulation, Developmental , Histocytochemistry , Insect Proteins/metabolism , Mutagenesis , Nerve Tissue Proteins , Nervous System/embryology , Phenotype , Receptor Protein-Tyrosine Kinases/metabolism , Wings, Animal/embryology , ras Proteins/genetics
11.
Development ; 126(7): 1505-14, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10068643

ABSTRACT

Neurons and glial cells provide guidance cues for migrating neurons. We show here that migrating epithelial cells also contact specific neurons and glia during their pathfinding, and we describe the first gene required in the process. In wild-type Drosophila embryos, the ganglionic tracheal branch navigates a remarkably complex path along specific neural and glial substrata, switching substrata five times before reaching its ultimate target in the CNS. In adrift mutants, ganglionic branches migrate normally along the intersegmental nerve, but sporadically fail to switch to the segmental nerve and enter the CNS; they wind up meandering along the ventral epidermis instead. adrift encodes a novel nuclear protein with an evolutionarily conserved motif. The gene is required in the trachea and is expressed in the leading cells of migrating ganglionic branches where it is induced by the branchless FGF pathway. We propose that Adrift regulates expression of tracheal genes required for pathfinding on the segmental nerve, and FGF induction of adrift expression in migrating tracheal cells promotes the switch from the intersegmental to the segmental nerve.


Subject(s)
Central Nervous System/embryology , Drosophila Proteins , Drosophila/genetics , Fibroblast Growth Factors , Genes, Insect , Insect Proteins/metabolism , Nuclear Proteins/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Movement , Cloning, Molecular , Drosophila/embryology , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/genetics , Molecular Sequence Data , Mutation/genetics , Nuclear Proteins/chemistry , Sequence Homology, Amino Acid , Trachea/embryology , Transcription Factors/chemistry
12.
Cell ; 92(2): 253-63, 1998 Jan 23.
Article in English | MEDLINE | ID: mdl-9458049

ABSTRACT

Antagonists of several growth factor signaling pathways play important roles in developmental patterning by limiting the range of the cognate inducer. Here, we describe an antagonist of FGF signaling that patterns apical branching of the Drosophila airways. In wild-type embryos, the Branchless FGF induces secondary branching by activating the Breathless FGF receptor near the tips of growing primary branches. In sprouty mutants, the FGF pathway is overactive and ectopic branches are induced on the stalks of primary branches. We show that FGF signaling induces sprouty expression in the nearby tip cells, and sprouty acts nonautonomously and in a competitive fashion to block signaling to the more distant stalk cells. sprouty encodes a novel cysteine-rich protein that defines a new family of putative signaling molecules that may similarly function as FGF antagonists in vertebrate development.


Subject(s)
Body Patterning/genetics , Drosophila Proteins , Drosophila/embryology , Fibroblast Growth Factors/physiology , Insect Proteins/genetics , Membrane Proteins , Signal Transduction/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Membrane/chemistry , DNA, Complementary/genetics , Drosophila/genetics , Gene Expression Regulation, Developmental , Insect Proteins/analysis , Insect Proteins/chemistry , Molecular Sequence Data , Mutation , Phenotype , Restriction Mapping , Sequence Analysis, DNA , Trachea/embryology
15.
Cell ; 87(6): 1091-101, 1996 Dec 13.
Article in English | MEDLINE | ID: mdl-8978613

ABSTRACT

The molecular basis for patterning of complex organ structures like the lung and insect tracheal system is unknown. Here, we describe the Drosophila gene branchless (bnl) and demonstrate that it is a key determinant of the tracheal branching pattern. bnl is required for tracheal branching and is expressed dynamically in clusters of cells surrounding the developing tracheal system at each position where a new branch will form and grow out. Localized misexpression of bnl can direct branch formation and outgrowth to new positions. Generalized misexpression activates later programs of tracheal gene expression and branching, resulting in massive networks of branches. bnl encodes a homolog of mammalian fibroblast growth factors (FGFs) and appears to function as a ligand for the breathless receptor tyrosine kinase, an FGF receptor homolog expressed on developing tracheal cells. The results suggest that this FGF pathway specifies the tracheal branching pattern by guiding tracheal cell migration during primary branch formation and then activating later programs of finer branching at the ends of growing primary branches.


Subject(s)
Drosophila Proteins , Drosophila/genetics , Fibroblast Growth Factors/genetics , Insect Proteins/genetics , Animals , Base Sequence , Cell Movement/physiology , DNA, Complementary/genetics , Gene Expression Regulation, Developmental/physiology , Genes, Insect/physiology , Larva/genetics , Molecular Sequence Data , Phenotype , Receptors, Fibroblast Growth Factor/physiology , Sequence Homology, Amino Acid , Signal Transduction/physiology , Trachea/cytology , Trachea/embryology
16.
Development ; 122(11): 3531-6, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8951068

ABSTRACT

During development of tubular networks such as the mammalian vascular system, the kidney and the Drosophila tracheal system, epithelial tubes must fuse to each other to form a continuous network. Little is known of the cellular mechanisms or molecular control of epithelial tube fusion. We describe the cellular dynamics of a tracheal fusion event in Drosophila and identify a gene regulatory hierarchy that controls this extraordinary process. A tracheal cell located at the developing fusion point expresses a sequence of specific markers as it grows out and contacts a similar cell from another tube; the two cells adhere and form an intercellular junction, and they become doughnut-shaped cells with the lumen passing through them. The early fusion marker Fusion-1 is identified as the escargot gene. It lies near the top of the regulatory hierarchy, activating the expression of later fusion markers and repressing genes that promote branching. Ectopic expression of escargot activates the fusion process and suppresses branching throughout the tracheal system, leading to ectopic tracheal connections that resemble certain arteriovenous malformations in humans. This establishes a simple genetic system to study fusion of epithelial tubes.


Subject(s)
Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental , Insect Proteins/physiology , Trachea/embryology , Transcription Factors/physiology , Animals , Cell Adhesion , Cytoplasm/ultrastructure , Drosophila melanogaster/genetics , Epithelial Cells , Morphogenesis , Zinc Fingers
17.
Genes Dev ; 10(22): 2912-21, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8918892

ABSTRACT

Receptor tyrosine kinases (RTKs) are members of a diverse class of signaling molecules well known for their roles in cell fate specification, cell differentiation, and oncogenic transformation. Recently several RTKs have been implicated in cell and axon motility, and RTKs are known to mediate chemotactic guidance of tissue culture cells. We have investigated whether the Drosophila FGF receptor homolog, Breathless (BTL), whose activity is necessary for each phase of branching morphogenesis in the embryonic tracheal system, might play a role in guiding the directed migration of tracheal cells. We found that expression of a constitutively active receptor during tracheal development interfered with directed tracheal cell migration and led to extra secondary and terminal branch-forming cells. Reduction in endogenous BTL signaling enhanced the cell migration defects while suppressing the ectopic branching defects. These results are consistent with a model for tracheal development in which spatially regulated BTL activity guides tracheal cell migration and quantitatively regulated BTL activity determines the patterns of secondary and terminal branching cell fates.


Subject(s)
Cell Movement , Drosophila Proteins , Drosophila/embryology , Insect Proteins/physiology , Protein-Tyrosine Kinases , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Fibroblast Growth Factor/physiology , Respiratory System/embryology , Animals , Drosophila/anatomy & histology , Enzyme Activation , Gene Expression , Heat-Shock Response , Immunohistochemistry , Morphogenesis , Mutagenesis, Site-Directed , Mutation , Phosphorylation , Precipitin Tests , Respiratory System/anatomy & histology
18.
Development ; 122(9): 2589-97, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8787734

ABSTRACT

The adult Drosophila wing is formed by an epithelial sheet, which differentiates into two non-neural tissues, vein or intervein. A large number of genes, many of them encoding components of an EGF-receptor signaling pathway, have previously been shown to be required for differentiation of vein tissue. Much less is known about the molecular control of intervein differentiation. Here we report that the Drosophila homolog of the mammalian Serum Response Factor gene (DSRF), which encodes a MADS-box containing transcriptional regulator, is expressed in the future intervein tissue of wing imaginal discs. In adult flies carrying only one functional copy of the DSRF gene, additional vein tissue develops in the wing, indicating that DSRF is required to spatially restrict the formation of veins. In mitotic clones lacking DSRF, intervein tissue fails to differentiate and becomes vein-like in appearance. Genetic and molecular evidence demonstrates that DSRF is encoded by the blistered locus, which produces ectopic veins and blistered wings when mutant. Our results show that DSRF plays a dual role during wing differentiation. It acts in a dosage-dependent [correction of dosage-dependant] manner to suppress the formation of wing veins and is required cell-autonomously to promote the development of intervein cells. We propose that DSRF acts at a key step between regulatory genes that define the early positional values in the developing wing disc and the subsequent localized expression of intervein-specific structural genes.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila/growth & development , Genes, Insect , Nuclear Proteins/genetics , Wings, Animal/growth & development , Alleles , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation , Clone Cells , Cloning, Molecular , DNA Primers , DNA-Binding Proteins/physiology , Drosophila/genetics , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Genetic Markers , Molecular Sequence Data , Mutation , Nuclear Proteins/physiology , Phenotype , Serum Response Factor , Transcription, Genetic , Wings, Animal/cytology
19.
Development ; 122(5): 1353-62, 1996 May.
Article in English | MEDLINE | ID: mdl-8625824

ABSTRACT

We identified a Drosophila gene, pruned, that regulates formation of the terminal branches of the tracheal (respiratory) system. These branches arise by extension of long cytoplasmic processes from terminal tracheal cells towards oxygen-starved tissues, followed by formation of a lumen within the processes. The pruned gene is expressed in terminal cells throughout the period of terminal branching. pruned encodes the Drosophila homologue of serum response factor (SRF), which functions with an ETS domain ternary complex factor as a growth-factor-activated transcription complex in mammalian cells. In pruned loss of function mutants, terminal cells fail to extend cytoplasmic projections. A constitutively activated SRF drives formation of extra projections that grow out in an unregulated fashion. An activated ternary complex factor has a similar effect. We propose that the Drosophila SRF functions like mammalian SRF in an inducible transcription complex, and that activation of this complex by signals from target tissues induces expression of genes involved in cytoplasmic outgrowth.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila/genetics , Genes, Insect , Nuclear Proteins/genetics , Respiratory System/embryology , Animals , Base Sequence , Cytoplasm/physiology , Drosophila/embryology , Gene Expression Regulation, Developmental , Genetic Markers , Models, Genetic , Molecular Sequence Data , Morphogenesis , Respiratory System/anatomy & histology , Sequence Homology , Serum Response Factor , Signal Transduction , Transcription, Genetic
20.
Development ; 122(5): 1395-407, 1996 May.
Article in English | MEDLINE | ID: mdl-8625828

ABSTRACT

The tracheal (respiratory) system of Drosophila melanogaster is a branched network of epithelial tubes that ramifies throughout the body and transports oxygen to the tissues. It forms by a series of sequential branching events in each hemisegment from T2 to A8. Here we present a cellular and initial genetic analysis of the branching process. We show that although branching is sequential it is not iterative. The three levels of branching that we distinguish involve different cellular mechanisms of tube formation. Primary branches are multicellular tubes that arise by cell migration and intercalation; secondary branches are unicellular tubes formed by individual tracheal cells; terminal branches are subcellular tubes formed within long cytoplasmic extensions. Each level of branching is accompanied by expression of a different set of enhancer trap markers. These sets of markers are sequentially activated in progressively restricted domains and ultimately individual tracheal cells that are actively forming new branches. A clonal analysis demonstrates that branching fates are not assigned to tracheal cells until after cell division ceases and branching begins. We further show that the breathless FGF receptor, a tracheal gene required for primary branching, is also required to activate expression of markers involved in secondary branching and that the pointed ETS-domain transcription factor is required for secondary branching and also to activate expression of terminal branch markers. The combined morphological, marker expression and genetic data support a model in which successive branching events are mechanistically and genetically distinct but coupled through the action of a tracheal gene regulatory hierarchy.


Subject(s)
Drosophila melanogaster/embryology , Respiratory System/embryology , Animals , Antigens, Differentiation , Cell Count , Cell Lineage , Cell Movement , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Morphogenesis , Respiratory System/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...