Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 1627, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38388656

ABSTRACT

The number of embryonic primordial germ cells in Drosophila is determined by the quantity of germ plasm, whose assembly starts in the posterior region of the oocyte during oogenesis. Here, we report that extending JAK-STAT activity in the posterior somatic follicular epithelium leads to an excess of primordial germ cells in the future embryo. We show that JAK-STAT signaling is necessary for the differentiation of approximately 20 specialized follicle cells maintaining tight contact with the oocyte. These cells define, in the underlying posterior oocyte cortex, the anchoring of the germ cell determinant oskar mRNA. We reveal that the apical surface of these posterior anchoring cells extends long filopodia penetrating the oocyte. We identify two JAK-STAT targets in these cells that are each sufficient to extend the zone of contact with the oocyte, thereby leading to production of extra primordial germ cells. JAK-STAT signaling thus determines a fixed number of posterior anchoring cells required for anterior-posterior oocyte polarity and for the development of the future germline.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Oocytes/metabolism , Oogenesis/genetics , Germ Cells/metabolism , Cell Polarity , Drosophila melanogaster/metabolism
3.
Front Cell Dev Biol ; 10: 886312, 2022.
Article in English | MEDLINE | ID: mdl-36120588

ABSTRACT

The JAK-STAT pathway is evolutionary conserved. The simplicity of this signaling in Drosophila, due to the limited redundancy between pathway components, makes it an ideal model for investigation. In the Drosophila follicular epithelium, highly stereotyped functions of JAK-STAT signaling have been well characterized, but how signaling activity is regulated precisely to allow the different outcomes is not well understood. In this tissue, the ligand is secreted by the polar cells positioned at each follicle extremity, thus generating a gradient of JAK-STAT activity in adjacent cells. One way to control the delivered quantity of ligand is by regulating the number of polar cells, which is reduced by apoptosis to exactly two at each pole by mid-oogenesis. Hence, JAK-STAT activity is described as symmetrical between follicle anterior and posterior regions. Here, we show that JAK-STAT signaling activity is actually highly dynamic, resulting in asymmetry between poles by mid-oogenesis. Interestingly, we found similar temporal dynamics at follicle poles in the accumulation of the adherens junction E-cadherin protein. Remarkably, E-cadherin and JAK-STAT signaling not only display patterning overlaps but also share functions during oogenesis. In particular, we show that E-cadherin, like JAK-STAT signaling, regulates polar cell apoptosis non-cell-autonomously from follicle cells. Finally, our work reveals that E-cadherin is required for optimal JAK-STAT activity throughout oogenesis and that E-cadherin and Stat92E, the transcription factor of the pathway, form part of a physical complex in follicle cells. Taken together, our study establishes E-cadherin as a new positive regulator of JAK-STAT signaling during oogenesis.

4.
PLoS Genet ; 16(11): e1009128, 2020 11.
Article in English | MEDLINE | ID: mdl-33151937

ABSTRACT

Many studies have focused on the mechanisms of stem cell maintenance via their interaction with a particular niche or microenvironment in adult tissues, but how formation of a functional niche is initiated, including how stem cells within a niche are established, is less well understood. Adult Drosophila melanogaster ovary Germline Stem Cell (GSC) niches are comprised of somatic cells forming a stack called a Terminal Filament (TF) and associated Cap and Escort Cells (CCs and ECs, respectively), which are in direct contact with GSCs. In the adult ovary, the transcription factor Engrailed is specifically expressed in niche cells where it directly controls expression of the decapentaplegic (dpp) gene encoding a member of the Bone Morphogenetic Protein (BMP) family of secreted signaling molecules, which are key factors for GSC maintenance. In larval ovaries, in response to BMP signaling from newly formed niches, adjacent primordial germ cells become GSCs. The bric-à-brac paralogs (bab1 and bab2) encode BTB/POZ domain-containing transcription factors that are expressed in developing niches of larval ovaries. We show here that their functions are necessary specifically within precursor cells for TF formation during these stages. We also identify a new function for Bab1 and Bab2 within developing niches for GSC establishment in the larval ovary and for robust GSC maintenance in the adult. Moreover, we show that the presence of Bab proteins in niche cells is necessary for activation of transgenes reporting dpp expression as of larval stages in otherwise correctly specified Cap Cells, independently of Engrailed and its paralog Invected (En/Inv). Moreover, strong reduction of engrailed/invected expression during larval stages does not impair TF formation and only partially reduces GSC numbers. In the adult ovary, Bab proteins are also required for dpp reporter expression in CCs. Finally, when bab2 was overexpressed at this stage in somatic cells outside of the niche, there were no detectable levels of ectopic En/Inv, but ectopic expression of a dpp transgene was found in these cells and BMP signaling activation was induced in adjacent germ cells, which produced GSC-like tumors. Together, these results indicate that Bab transcription factors are positive regulators of BMP signaling in niche cells for establishment and homeostasis of GSCs in the Drosophila ovary.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Germ Cells/growth & development , Ovary/growth & development , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Count , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Larva/growth & development , Ovary/cytology , Signal Transduction/genetics , Stem Cell Niche/genetics , Transcription Factors/genetics
5.
Cell Death Dis ; 8(5): e2814, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28542149

ABSTRACT

Epithelial cell extrusion is crucial for proper development and tissue homeostasis. High-resolution 3D reconstruction and 4D imaging, combined with genetic analyis, have allowed us to reveal the highly-sterotyped morphogenetic events controlled by JAK/STAT signaling in a developmentally-programmed case of epithelial cell extrusion. Specialized somatic cells, Polar Cells (PCs), are produced in excess and then undergo apoptotic elimination from the follicular epithelium in the Drosophila ovary. We show that supernumerary PCs are first systematically enveloped by PC neighbors on all sides, first laterally, then apically in conjunction with highly-reinforced adherens junctions, and finally basally. The PC to be removed thus loses all contact with follicle cells, germline cells and the basement membrane in a process we have called cell 'monosis', for 'isolation' in Greek. PC monosis takes several hours, and always precedes, and is independent of, activation of apoptosis. JAK/STAT signaling is necessary within the surrounding follicular epithelium for PC monosis. Minutes after monosis is complete, PC apoptotic corpses are formed and extruded laterally within the epithelium, in contrast to the apical and basal extrusions described to date. These apoptotic corpses are engulfed and eliminated by surrounding follicle cells, which are thus acting as non-professional phagocytes. This study therefore shows the non cell-autonomous impact of an epithelium, via JAK/STAT signaling activation, on cell morphogenesis events leading to apoptotic extrusion. It is likely that the use of high-resolution 3D and 4D imaging, which allows for better spatio-temporal understanding of morphogenetic events, will reveal that cell monosis and lateral extrusion within an epithelium are pertinent for other cases of epithelial cell extrusion as well.


Subject(s)
Apoptosis , Drosophila melanogaster/cytology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Adherens Junctions/metabolism , Animals , Computer Systems , Drosophila melanogaster/metabolism , Female , Models, Biological , Ovarian Follicle/cytology , Ovarian Follicle/metabolism
6.
Development ; 140(1): 195-204, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23222440

ABSTRACT

During development, specific cells are eliminated by apoptosis to ensure that the correct number of cells is integrated in a given tissue or structure. How the apoptosis machinery is activated selectively in vivo in the context of a developing tissue is still poorly understood. In the Drosophila ovary, specialised follicle cells [polar cells (PCs)] are produced in excess during early oogenesis and reduced by apoptosis to exactly two cells per follicle extremity. PCs act as an organising centre during follicle maturation as they are the only source of the JAK/STAT pathway ligand Unpaired (Upd), the morphogen activity of which instructs distinct follicle cell fates. Here we show that reduction of Upd levels leads to prolonged survival of supernumerary PCs, downregulation of the pro-apoptotic factor Hid, upregulation of the anti-apoptotic factor Diap1 and inhibition of caspase activity. Upd-mediated activation of the JAK/STAT pathway occurs in PCs themselves, as well as in adjacent terminal follicle and interfollicular stalk cells, and inhibition of JAK/STAT signalling in any one of these cell populations protects PCs from apoptosis. Thus, a Stat-dependent unidentified relay signal is necessary for inducing supernumerary PC death. Finally, blocking apoptosis of PCs leads to specification of excess adjacent border cells via excessive Upd signalling. Our results therefore show that Upd and JAK/STAT signalling induce apoptosis of supernumerary PCs to control the size of the PC organising centre and thereby produce appropriate levels of Upd. This is the first example linking this highly conserved signalling pathway with developmental apoptosis in Drosophila.


Subject(s)
Apoptosis/physiology , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Janus Kinases/physiology , STAT Transcription Factors/physiology , Animals , Cell Count , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/biosynthesis , Drosophila Proteins/physiology , Drosophila melanogaster/enzymology , Female , Janus Kinases/antagonists & inhibitors , Ligands , Ovary/cytology , Ovary/enzymology , Ovary/metabolism , STAT Transcription Factors/antagonists & inhibitors , Signal Transduction/physiology , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis , Transcription Factors/physiology
7.
PLoS One ; 7(11): e49958, 2012.
Article in English | MEDLINE | ID: mdl-23185495

ABSTRACT

The potential to produce new cells during adult life depends on the number of stem cell niches and the capacity of stem cells to divide, and is therefore under the control of programs ensuring developmental homeostasis. However, it remains generally unknown how the number of stem cell niches is controlled. In the insect ovary, each germline stem cell (GSC) niche is embedded in a functional unit called an ovariole. The number of ovarioles, and thus the number of GSC niches, varies widely among species. In Drosophila, morphogenesis of ovarioles starts in larvae with the formation of terminal filaments (TFs), each made of 8-10 cells that pile up and sort in stacks. TFs constitute organizers of individual germline stem cell niches during larval and early pupal development. In the Drosophila melanogaster subgroup, the number of ovarioles varies interspecifically from 8 to 20. Here we show that pipsqueak, Trithorax-like, batman and the bric-à-brac (bab) locus, all encoding nuclear BTB/POZ factors of the Tramtrack Group, are involved in limiting the number of ovarioles in D. melanogaster. At least two different processes are differentially perturbed by reducing the function of these genes. We found that when the bab dose is reduced, sorting of TF cells into TFs was affected such that each TF contains fewer cells and more TFs are formed. In contrast, psq mutants exhibited a greater number of TF cells per ovary, with a normal number of cells per TF, thereby leading to formation of more TFs per ovary than in the wild type. Our results indicate that two parallel genetic pathways under the control of a network of nuclear BTB factors are combined in order to negatively control the number of germline stem cell niches.


Subject(s)
DNA-Binding Proteins , Drosophila Proteins , Nuclear Proteins , Stem Cell Niche/genetics , Transcription Factors , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Female , Gene Dosage/genetics , Gene Expression Regulation, Developmental , Germ Cells/cytology , Germ Cells/growth & development , Homeostasis/genetics , Homeostasis/physiology , Morphogenesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ovary/cytology , Ovary/growth & development , Stem Cell Niche/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
8.
PLoS One ; 7(3): e34310, 2012.
Article in English | MEDLINE | ID: mdl-22479597

ABSTRACT

The genome of the human immunodeficiency virus type 1 (HIV-1) encodes the canonical retroviral proteins, as well as additional accessory proteins that enhance the expression of viral genes, the infectivity of the virus and the production of virions. The accessory Viral Protein U (Vpu), in particular, enhances viral particle production, while also promoting apoptosis of HIV-infected human T lymphocytes. Some Vpu effects rely on its interaction with the ubiquitin-proteasome protein degradation system, but the mechanisms responsible for its pro-apoptotic effects in vivo are complex and remain largely to be elucidated.We took advantage of the Drosophila model to study the effects of Vpu activity in vivo. Expression of Vpu in the developing Drosophila wing provoked tissue loss due to caspase-dependent apoptosis. Moreover, Vpu induced expression of the pro-apoptotic gene reaper, known to down-regulate Inhibitor of Apoptosis Proteins (IAPs) which are caspase-antagonizing E3 ubiquitin ligases. Indeed, Vpu also reduced accumulation of Drosophila IAP1 (DIAP1). Though our results demonstrate a physical interaction between Vpu and the proteasome-addressing SLIMB/ß-TrCP protein, as in mammals, both SLIMB/ßTrCP-dependent and -independent Vpu effects were observed in the Drosophila wing. Lastly, the pro-apoptotic effect of Vpu in this tissue was abrogated upon inactivation of the c-Jun N-terminal Kinase (JNK) pathway. Our results in the fly thus provide the first functional evidence linking Vpu pro-apoptotic effects to activation of the conserved JNK pathway.


Subject(s)
Apoptosis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Viral , HIV-1/metabolism , Human Immunodeficiency Virus Proteins/metabolism , MAP Kinase Kinase 4/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Animals , Animals, Genetically Modified , Caspases/metabolism , Drosophila melanogaster , Enzyme Activation , Phenotype , Signal Transduction , Transgenes , Wings, Animal/metabolism
9.
PLoS One ; 5(11): e13946, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-21085656

ABSTRACT

BACKGROUND: Most human cancers originate from epithelial tissues and cell polarity and adhesion defects can lead to metastasis. The Polycomb-Group of chromatin factors were first characterized in Drosophila as repressors of homeotic genes during development, while studies in mammals indicate a conserved role in body plan organization, as well as an implication in other processes such as stem cell maintenance, cell proliferation, and tumorigenesis. We have analyzed the function of the Drosophila Polycomb-Group gene polyhomeotic in epithelial cells of two different organs, the ovary and the wing imaginal disc. RESULTS: Clonal analysis of loss and gain of function of polyhomeotic resulted in segregation between mutant and wild-type cells in both the follicular and wing imaginal disc epithelia, without excessive cell proliferation. Both basal and apical expulsion of mutant cells was observed, the former characterized by specific reorganization of cell adhesion and polarity proteins, the latter by complete cytoplasmic diffusion of these proteins. Among several candidate target genes tested, only the homeotic gene Abdominal-B was a target of PH in both ovarian and wing disc cells. Although overexpression of Abdominal-B was sufficient to cause cell segregation in the wing disc, epistatic analysis indicated that the presence of Abdominal-B is not necessary for expulsion of polyhomeotic mutant epithelial cells suggesting that additional polyhomeotic targets are implicated in this phenomenon. CONCLUSION: Our results indicate that polyhomeotic mutations have a direct effect on epithelial integrity that can be uncoupled from overproliferation. We show that cells in an epithelium expressing different levels of polyhomeotic sort out indicating differential adhesive properties between the cell populations. Interestingly, we found distinct modalities between apical and basal expulsion of ph mutant cells and further studies of this phenomenon should allow parallels to be made with the modified adhesive and polarity properties of different types of epithelial tumors.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Mutation , Nucleoproteins/genetics , Ovarian Follicle/metabolism , Wings, Animal/metabolism , Animals , Animals, Genetically Modified , Cell Adhesion , Cell Polarity , Cell Proliferation , Clone Cells/cytology , Clone Cells/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Epithelial Cells/metabolism , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Nucleoproteins/metabolism , Ovarian Follicle/cytology , Polycomb Repressive Complex 1 , Protein Binding , RNA Interference , Wings, Animal/cytology
10.
Mech Ageing Dev ; 130(8): 547-52, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19486910

ABSTRACT

The steroid hormone ecdysone influences Drosophila lifespan. Longevity is extended in mutants deficient for ecdysone synthesis or mutants of the ecdysone receptor (EcR). However, the underlying mechanisms remain unclear. Here we conditionally inactivated EcR by RNA interference or expression of dominant negative forms, using the RU486 inducible system. A mild ubiquitous inactivation of EcR during adulthood was sufficient to slow the aging of male flies, whereas a stronger EcR inactivation decreased longevity. Surprisingly, ubiquitous inactivation of EcR strongly decreased female lifespan. This deleterious effect was suppressed in sterile ovo(D1) mutant females, suggesting that EcR represses a negative signal for lifespan produced in ovaries. These results reveal a complex adult and sex-specific control of lifespan by steroid signalling in Drosophila.


Subject(s)
Drosophila/physiology , Gene Expression Regulation , Longevity , Receptors, Steroid/physiology , Animals , Female , Genes, Dominant , Male , Mifepristone/pharmacology , Models, Biological , RNA Interference , Receptors, Steroid/metabolism , Sex Factors , Signal Transduction , Steroids/metabolism
11.
Methods Mol Biol ; 397: 161-201, 2007.
Article in English | MEDLINE | ID: mdl-18025721

ABSTRACT

The GAL4/upstream activating sequence (UAS) system is one of the most powerful tools for targeted gene expression. It is based on the properties of the yeast GAL4 transcription factor which activates transcription of its target genes by binding to UAS cis-regulatory sites. In Drosophila, the two components are carried in separate lines allowing for numerous combinatorial possibilities. The driver lines provide tissue-specific GAL4 expression and the responder lines carry the coding sequence for the gene of interest under the control of UAS sites. In this chapter, the basic GAL4/UAS system and its extensions, namely those allowing precise temporal control and reversible expression, are described. In addition, a list of GAL4 and UAS lines and schematic maps of GAL4 and UAS vectors useful in the study of Hedgehog (Hh) signaling is given. Finally, uses of the GAL4/UAS system to resolve some of the questions addressed in the study of the Hh pathway are presented.


Subject(s)
Drosophila melanogaster/genetics , Gene Expression Regulation , Genetic Techniques , Hedgehog Proteins/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Animals , Body Patterning , DNA-Binding Proteins , Drosophila melanogaster/embryology , Embryo, Nonmammalian/metabolism , Genetic Vectors , Organ Specificity
12.
Mech Dev ; 123(3): 197-209, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16516445

ABSTRACT

The fused gene encodes a serine-threonine kinase that functions as a positive regulator of Hedgehog signal transduction in Drosophila embryogenesis, wing morphogenesis, and somatic cell development during oogenesis. Here, we have characterized the germline ovarian tumors present in adult ovaries of fused mutant females, a phenotype not observed upon deregulation of any other component of Hedgehog signaling. In the strongest fused mutant contexts, we found that tumorous ovarian follicles accumulate early spectrosome-containing germ cells corresponding to germline stem cells and/or early cystoblasts as evidenced by activated Dpp signal transduction and transcriptional repression of bag-of-marbles, encoding the cystoblast determination factor. These early germ cells are maintained far from their usual position in a specialized niche of somatic cells in the apical part of the germarium, which appears normal in size in fused mutant ovarioles. Therefore, these results indicate a novel function for fused in downregulation of Dpp signaling which is necessary for de-repression of bag-of-marbles and consequent cystoblast determination. The abnormal accumulation of these early germ cells seems to be due primarily to defects in differentiation since we show that germline stem cell proliferation in the germarium is not affected. A later block in germline development, at the 16-cell cyst stage before significant nurse cell and oocyte differentiation, was also observed in tumorous follicles when fused function was only partially lowered. Finally, fused mutant ovaries exhibit some germline cysts having undergone a supernumerary fifth mitotic division. Through clonal analysis, we provide evidence that fused regulates these cystocyte divisions cell autonomously, while the tumorous phenotype probably reflects both a somatic and germline requirement for fused for cyst and follicle development.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/physiology , Germ Cells/metabolism , Oogenesis , Ovarian Follicle , Ovarian Neoplasms/genetics , Protein Serine-Threonine Kinases/physiology , Animals , Animals, Genetically Modified , Cell Differentiation , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Female , Gene Expression Regulation, Developmental , Germ Cells/pathology , Immunohistochemistry , Mitosis , Oocytes/physiology , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Stem Cells
13.
Dev Dyn ; 234(2): 422-31, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16145667

ABSTRACT

The genetic analysis of Drosophila adult oogenesis has provided insights into the molecular mechanisms that control cell proliferation, differentiation, migration, and intercellular signaling. However, little is known about the larval and pupal cellular events leading to the formation of the highly organized adult ovary, which is composed of ovarioles each containing germline cells enveloped by specialized somatic cells. We describe here the presence of ovarioles devoid of any germ cells in adult females mutant for fused, which encodes a Hedgehog signal transducing serine/threonine kinase. We show that this phenotype corresponds to a requirement for fused function for the organization of germ cells with respect to ovarian somatic cells during ovariole formation specifically during pupal stages and provide some evidence by means of clonal analysis suggesting that fused function may be necessary in the germline. hedgehog is expressed specifically in somatic terminal filament cells in pupal ovaries, and females bearing hedgehog strong loss-of-function mutations also exhibit aberrant germ cell distribution and formation of agametic ovarioles. These results indicate a positive role for Fused in the transduction of somatic Hedgehog signaling instructing ovariole morphogenesis. We also provide evidence for the use of noncanonical Hedgehog signal transducer(s) within germline cells.


Subject(s)
Drosophila Proteins/physiology , Gene Expression Regulation, Developmental , Ovary/embryology , Protein Serine-Threonine Kinases/physiology , Alleles , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Crosses, Genetic , Drosophila , Drosophila Proteins/biosynthesis , Female , Germ Cells/metabolism , Hedgehog Proteins , Immunohistochemistry , Male , Microscopy, Fluorescence , Models, Genetic , Morphogenesis , Mutation , Ovary/pathology , Phenotype , Protein Serine-Threonine Kinases/biosynthesis , Recombination, Genetic , Signal Transduction , Time Factors , Transgenes
14.
Development ; 131(6): 1389-400, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14993188

ABSTRACT

The polyhomeotic (ph) gene of Drosophila is a member of the Polycomb group (Pc-G) genes, which are required for maintenance of a repressed state of homeotic gene transcription, which stabilizes cell identity throughout development. The ph gene was recovered in the course of a gain-of-function screen aimed at identifying genes with a role during ovarian follicle formation in Drosophila, a process that involves coordinated proliferation and differentiation of two cell lineages, somatic and germline. Subsequent analysis revealed that ph loss-of-function mutations lead to production of follicles with greater or fewer than the normal number of germ cells associated with reduced proliferation of somatic prefollicular cells, abnormal prefollicular cell encapsulation of germline cysts and an excess of both interfollicular stalk cells and polar cells. Clonal analysis showed that ph function for follicle formation resides specifically in somatic cells and not in the germline. This is thus the first time that a role has been shown for a Pc-G gene during Drosophila folliculogenesis. In addition, we tested mutations in a number of other Pc-G genes, and two of them, Sex combs extra (Sce) and Sex comb on midleg (Scm), also displayed ovarian defects similar to those observed for ph. Our results provide a new model system, the Drosophila ovary, in which the function of Pc-G genes, distinct from that of control of homeotic gene expression, can be explored.


Subject(s)
Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Nucleoproteins/metabolism , Ovarian Follicle/metabolism , Animals , Cell Division/physiology , Female , Polycomb Repressive Complex 1
15.
Development ; 130(5): 1017-27, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12538526

ABSTRACT

Polar cells have been described as pairs of specific follicular cells present at each pole of Drosophila egg chambers. They are required at different stages of oogenesis for egg chamber formation and establishment of both the anteroposterior and planar polarities of the follicular epithelium. We show that definition of polar cell pairs is a progressive process since early stage egg chambers contain a cluster of several polar cell marker-expressing cells at each pole, while as of stage 5, they contain invariantly two pairs of such cells. Using cell lineage analysis, we demonstrate that these pre-polar cell clusters have a polyclonal origin and derive specifically from the polar cell lineage, rather than from that giving rise to follicular cells. In addition, selection of two polar cells from groups of pre-polar cells occurs via an apoptosis-dependent mechanism and is required for correct patterning of the anterior follicular epithelium of vitellogenic egg chambers.


Subject(s)
Apoptosis/physiology , Cell Lineage , Drosophila melanogaster/physiology , Oogenesis/physiology , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Female , Genes, Insect , In Situ Nick-End Labeling , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Protein Serine-Threonine Kinases/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology
16.
Development ; 129(17): 4111-24, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12163413

ABSTRACT

The fused gene encodes a serine/threonine kinase involved in Hedgehog signal transduction during Drosophila embryo and larval imaginal disc development. Additionally, fused mutant females exhibit reduced fecundity that we report here to be associated with defects in three aspects of egg chamber formation: encapsulation of germline cysts by prefollicular cells in the germarium, interfollicular stalk morphogenesis and oocyte posterior positioning. Using clonal analysis we show that fused is required cell autonomously in prefollicular and pre-stalk cells to control their participation in these aspects of egg chamber formation. In contrast to what has been found for Hedgehog and other known components of Hedgehog signal transduction, we show that fused does not play a role in the regulation of somatic stem cell proliferation. However, genetic interaction studies, as well as the analysis of the effects of a partial reduction in Hedgehog signaling in the ovary, indicate that fused acts in the classical genetic pathway for Hedgehog signal transduction which is necessary for somatic cell differentiation during egg chamber formation. Therefore, we propose a model in which Hedgehog signals at least twice in germarial somatic cells: first, through a fused-independent pathway to control somatic stem cell proliferation; and second, through a classical fused-dependent pathway to regulate prefollicular cell differentiation.


Subject(s)
Drosophila Proteins/physiology , Drosophila/physiology , Protein Serine-Threonine Kinases/physiology , Signal Transduction/physiology , Animals , Cell Differentiation/physiology , Drosophila/embryology , Female , Hedgehog Proteins , Male , Mutation , Ovarian Follicle/cytology , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...