Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Exp Cell Res ; 389(2): 111913, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32084392

ABSTRACT

Since epigenetic modifications are a key driver for cellular differentiation, the regulation of these modifications is tightly controlled. Interestingly, recent studies have revealed metabolic regulation for epigenetic modifications in pluripotent cells. As metabolic differences are prominent between naive (pre-implantation) and primed (post-implantation) pluripotent cells, the epigenetic changes regulated by metabolites has become an interesting topic of analysis. In this review we discuss how combinatorial metabolic activities drive the developmental progression through early pluripotent stages.


Subject(s)
Blastocyst/cytology , Chromatin/genetics , Embryonic Stem Cells/cytology , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Metabolome , Pluripotent Stem Cells/cytology , Animals , Blastocyst/metabolism , Cell Differentiation , Embryonic Stem Cells/metabolism , Humans , Pluripotent Stem Cells/metabolism
2.
Nat Commun ; 10(1): 632, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30733432

ABSTRACT

To reveal how cells exit human pluripotency, we designed a CRISPR-Cas9 screen exploiting the metabolic and epigenetic differences between naïve and primed pluripotent cells. We identify the tumor suppressor, Folliculin(FLCN) as a critical gene required for the exit from human pluripotency. Here we show that FLCN Knock-out (KO) hESCs maintain the naïve pluripotent state but cannot exit the state since the critical transcription factor TFE3 remains active in the nucleus. TFE3 targets up-regulated in FLCN KO exit assay are members of Wnt pathway and ESRRB. Treatment of FLCN KO hESC with a Wnt inhibitor, but not ESRRB/FLCN double mutant, rescues the cells, allowing the exit from the naïve state. Using co-immunoprecipitation and mass spectrometry analysis we identify unique FLCN binding partners. The interactions of FLCN with components of the mTOR pathway (mTORC1 and mTORC2) reveal a mechanism of FLCN function during exit from naïve pluripotency.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Wnt Signaling Pathway/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , CRISPR-Cas Systems/genetics , CRISPR-Cas Systems/physiology , Cell Line , Estrone/genetics , Estrone/metabolism , Humans , Immunoprecipitation , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 2/genetics , Proteomics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Wnt Signaling Pathway/genetics
3.
Oncogene ; 36(22): 3119-3136, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28092677

ABSTRACT

Aberrant regulation of WNT/ß-catenin signaling has a crucial role in the onset and progression of cancers, where the effects are not always predictable depending on tumor context. In melanoma, for example, models of the disease predict differing effects of the WNT/ß-catenin pathway on metastatic progression. Understanding the processes that underpin the highly context-dependent nature of WNT/ß-catenin signaling in tumors is essential to achieve maximal therapeutic benefit from WNT inhibitory compounds. In this study, we have found that expression of the tumor suppressor, phosphatase and tensin homolog deleted on chromosome 10 (PTEN), alters the invasive potential of melanoma cells in response to WNT/ß-catenin signaling, correlating with differing metabolic profiles. This alters the bioenergetic potential and mitochondrial activity of melanoma cells, triggered through regulation of pro-survival autophagy. Thus, WNT/ß-catenin signaling is a regulator of catabolic processes in cancer cells, which varies depending on the metabolic requirements of tumors.


Subject(s)
Melanoma/metabolism , Mitochondria/metabolism , PTEN Phosphohydrolase/genetics , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Humans , Male , Melanoma/enzymology , Melanoma/genetics , Mice , Mice, Inbred NOD , Mice, SCID , PTEN Phosphohydrolase/metabolism
4.
Curr Top Dev Biol ; 121: 83-109, 2017.
Article in English | MEDLINE | ID: mdl-28057309

ABSTRACT

Drosophila melanogaster provides a powerful platform with which researchers can dissect complex genetic questions and biochemical pathways relevant to a vast array of human diseases and disorders. Of particular interest, much work has been done with flies to elucidate the molecular mechanisms underlying muscle degeneration diseases. The fly is particularly useful for modeling muscle degeneration disorders because there are no identified satellite muscle cells to repair adult muscle following injury. This allows for the identification of endogenous processes of muscle degeneration as discrete events, distinguishable from phenotypes due to the lack of stem cell-based regeneration. In this review, we will discuss the ways in which the fruit fly provides a powerful platform with which to study human muscle degeneration disorders.


Subject(s)
Disease Models, Animal , Drosophila melanogaster/physiology , Muscle, Skeletal/pathology , Muscular Diseases/pathology , Animals , Humans , Muscular Diseases/therapy , Signal Transduction
5.
Curr Mol Med ; 13(5): 757-64, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23642057

ABSTRACT

There are currently 1527 known microRNAs (miRNAs) in human, each of which may regulate hundreds or thousands of target genes. miRNA expression levels vary between cell types; for example, miR- 302 and miR-290 families are highly enriched in embryonic stem cells, while miR-1 is a muscle specific miRNA. miRNA biosynthesis and function are highly regulated and this regulation may be cell type specific. The processing enzymes and factors that recognize features in sequence and secondary structure of the miRNA play key roles in regulating the production of mature miRNA. Mature miRNA enriched in stem cells control stem cell self-renewal as well as their differentiation. Though specific miRNAs have been shown to control differentiation towards various lineages such as neural or skin cells, some of the most well characterized miRNAs have been found in promoting the formation of cardiac cells. In addition, miRNAs also play a critical role in cardiomyocyte hypertrophy, especially in a pathological context. Such miRNAs are predicted to be therapeutic targets for treating cardiovascular diseases. In this review we will discuss how miRNAs act to maintain the stem cell state and also explore the current knowledge of the mechanisms that regulate miRNAs. Furthermore, we will discuss the emerging roles of miRNAs using cardiomyocyte differentiation and maturation as a paradigm. Emphasis will also be given on some of the less ventured areas such as the role of miRNAs in the physiological maturation of cardiomyocytes. These potentially beneficial miRNAs are likely to improve cardiac function in both in vivo and in vitro settings and thus provide additional strategy to treat heart diseases and more importantly serve as a good model for understanding cardiomyocyte maturation in vitro.


Subject(s)
Cardiomegaly/pathology , Embryonic Stem Cells/physiology , MicroRNAs/genetics , Myocardium/cytology , Myocytes, Cardiac/physiology , Animals , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cell Differentiation , Cell Size , Embryonic Stem Cells/cytology , Heart/growth & development , Humans , MicroRNAs/metabolism , Myocardium/metabolism , Myocytes, Cardiac/cytology , RNA Interference
6.
BMC Dev Biol ; 9: 18, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19250553

ABSTRACT

BACKGROUND: Dystroglycan (Dg) is a transmembrane protein that is a part of the Dystrophin Glycoprotein Complex (DGC) which connects the extracellular matrix to the actin cytoskeleton. The C-terminal end of Dg contains a number of putative SH3, SH2 and WW domain binding sites. The most C-terminal PPXY motif has been established as a binding site for Dystrophin (Dys) WW-domain. However, our previous studies indicate that both Dystroglycan PPXY motives, WWbsI and WWbsII can bind Dystrophin protein in vitro. RESULTS: We now find that both WW binding sites are important for maintaining full Dg function in the establishment of oocyte polarity in Drosophila. If either WW binding site is mutated, the Dg protein can still be active. However, simultaneous mutations in both WW binding sites abolish the Dg activities in both overexpression and loss-of-function oocyte polarity assays in vivo. Additionally, sequence comparisons of WW binding sites in 12 species of Drosophila, as well as in humans, reveal a high level of conservation. This preservation throughout evolution supports the idea that both WW binding sites are functionally required. CONCLUSION: Based on the obtained results we propose that the presence of the two WW binding sites in Dystroglycan secures the essential interaction between Dg and Dys and might further provide additional regulation for the cytoskeletal interactions of this complex.


Subject(s)
Drosophila Proteins/chemistry , Dystroglycans/chemistry , Amino Acid Sequence , Animals , Animals, Genetically Modified , Binding Sites , Cell Polarity , Conserved Sequence , Cytoskeleton/metabolism , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Dystroglycans/genetics , Dystroglycans/metabolism , Dystrophin/chemistry , Dystrophin/metabolism , Humans , Molecular Sequence Data , Mutation , Oocytes/cytology , Phylogeny
7.
Cell Cycle ; 5(2): 172-5, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16357538

ABSTRACT

One of the key characteristics of stem cells is their capacity for self-renewal for long periods of time. In this respect, stem cells are similar to cancer cells, which also have the ability to escape cell cycle stop signals. Therefore, a critical question in stem cell and cancer biology is how cell division is regulated in these cell types. In this review, we summarize recent progress and describe future challenges to understanding the role the microRNA pathway plays in regulating mechanisms controlling stem cell division.


Subject(s)
MicroRNAs/genetics , MicroRNAs/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Division , G1 Phase/physiology , S Phase/physiology
8.
Nature ; 435(7044): 974-8, 2005 Jun 16.
Article in English | MEDLINE | ID: mdl-15944714

ABSTRACT

One of the key characteristics of stem cells is their capacity to divide for long periods of time in an environment where most of the cells are quiescent. Therefore, a critical question in stem cell biology is how stem cells escape cell division stop signals. Here, we report the necessity of the microRNA (miRNA) pathway for proper control of germline stem cell (GSC) division in Drosophila melanogaster. Analysis of GSCs mutant for dicer-1 (dcr-1), the double-stranded RNaseIII essential for miRNA biogenesis, revealed a marked reduction in the rate of germline cyst production. These dcr-1 mutant GSCs exhibit normal identity but are defective in cell cycle control. On the basis of cell cycle markers and genetic interactions, we conclude that dcr-1 mutant GSCs are delayed in the G1 to S transition, which is dependent on the cyclin-dependent kinase inhibitor Dacapo, suggesting that miRNAs are required for stem cells to bypass the normal G1/S checkpoint. Hence, the miRNA pathway might be part of a mechanism that makes stem cells insensitive to environmental signals that normally stop the cell cycle at the G1/S transition.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , MicroRNAs/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Division , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , G1 Phase , Gene Deletion , Genome , MicroRNAs/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , S Phase , Stem Cells/enzymology
9.
Development ; 128(23): 4737-46, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11731454

ABSTRACT

In many developmental processes, polyploid cells are generated by a variation of the normal cell cycle called the endocycle in which cells increase their genomic content without dividing. How the transition from the normal mitotic cycle to endocycle is regulated is poorly understood. We show that the transition from mitotic cycle to endocycle in the Drosophila follicle cell epithelium is regulated by the Notch pathway. Loss of Notch function in follicle cells or its ligand Delta function in the underlying germline disrupts the normal transition of the follicle cells from mitotic cycle to endocycle, mitotic cycling continues, leading to overproliferation of these cells. The regulation is at the transcriptional level, as Su(H), a downstream transcription factor in the pathway, is also required cell autonomously in follicle cells for proper transitioning to the endocycle. One target of Notch and Su(H) is likely to be the G2/M cell cycle regulator String, a phosphatase that activates Cdc2 by dephosphorylation. String is normally repressed in the follicle cells just before the endocycle transition, but is expressed when Notch is inactivated. Analysis of the activity of String enhancer elements in follicle cells reveals the presence of an element that promotes expression of String until just before the onset of polyploidy in wild-type follicle cells but well beyond this stage in Notch mutant follicle cells. This suggests that it may be the target of the endocycle promoting activity of the Notch pathway. A second element that is insensitive to Notch regulation promotes String expression earlier in follicle cell development, which explains why Notch, while active at both stages, represses String only at the mitotic cycle-endocycle transition.


Subject(s)
Drosophila Proteins , Drosophila melanogaster/cytology , Membrane Proteins/metabolism , Ovarian Follicle/cytology , Protein Tyrosine Phosphatases , Animals , Cell Cycle , Cell Cycle Proteins , Cell Differentiation , Cell Polarity , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Female , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Mitosis , Mutation , Oogenesis , Ovarian Follicle/metabolism , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Polyploidy , Receptors, Notch , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction
10.
Dev Genes Evol ; 211(1): 44-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11277405

ABSTRACT

The factors that determine intracellular polarity are largely unknown. In Drosophila oocytes one of the earliest polar events is the positioning of the microtubule-organizing center (MTOC). Here we present data that are consistent with the hypothesis that maelstrom is required for posterior positioning of the MTOC.


Subject(s)
Cell Polarity , Drosophila Proteins , Drosophila melanogaster/physiology , Insect Proteins/metabolism , Microtubule-Organizing Center/metabolism , Oocytes/physiology , Animals , Genes, Reporter , Insect Proteins/genetics , Kinesins , Microscopy, Confocal , Microtubule Proteins/metabolism , Microtubules/metabolism , Oocytes/growth & development , Oocytes/ultrastructure , Recombinant Fusion Proteins/metabolism
11.
Curr Biol ; 10(11): 683-6, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10837250

ABSTRACT

The establishment of the anterior-posterior (AP) axis in Drosophila melanogaster requires signaling between the oocyte and surrounding somatic follicle cells during oogenesis [1] [2]. First, a signal from the oocyte (Gurken (Grk), a transforming growth factor-alpha (TGFalpha) homolog) is received by predetermined terminal follicle cells in which the epidermal growth factor receptor (EGFR) pathway is activated and a posterior fate is induced [2] [3] [4]. Later, the posterior follicle cells send an unidentified signal back to the oocyte, which leads to the reorganization of its cytoskeletal polarity. This reorganization is required for proper localization of maternal determinants, such as oskar (osk) and bicoid (bcd) mRNAs, that determine the AP polarity of the oocyte and the subsequent embryo [2]. We show here that when the gene lanA, which encodes the extracellular matrix component laminin A, is mutated in posterior follicle cells, localization of AP determinants is disrupted in the underlying oocyte. Posterior follicle-cell differentiation and follicle cell apical-basal polarity are unaffected in the lanA mutant cells, suggesting that laminin A is required for correct signaling from the posterior follicle cells that polarizes the oocyte. This is the first evidence that the extracellular matrix is involved in the establishment of a major body axis.


Subject(s)
Drosophila Proteins , Drosophila melanogaster/physiology , Embryo, Nonmammalian/physiology , Laminin/physiology , Oocytes/physiology , Oogenesis , Ovarian Follicle/physiology , Animals , Body Patterning , Drosophila melanogaster/embryology , Female , Homeodomain Proteins/genetics , Insect Proteins/genetics , Signal Transduction , Trans-Activators/genetics , Transcription, Genetic
12.
Nat Genet ; 24(4): 429-33, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10742112

ABSTRACT

Recent studies in vertebrates and Drosophila melanogaster have revealed that Fringe-mediated activation of the Notch pathway has a role in patterning cell layers during organogenesis. In these processes, a homeobox-containing transcription factor is responsible for spatially regulating fringe (fng) expression and thus directing activation of the Notch pathway along the fng expression border. Here we show that this may be a general mechanism for patterning epithelial cell layers. At three stages in Drosophila oogenesis, mirror (mirr) and fng have complementary expression patterns in the follicle-cell epithelial layer, and at all three stages loss of mirr enlarges, and ectopic expression of mirr restricts, fng expression, with consequences for follicle-cell patterning. These morphological changes are similar to those caused by Notch mutations. Ectopic expression of mirr in the posterior follicle cells induces a stripe of rhomboid (rho) expression and represses pipe (pip), a gene with a role in the establishment of the dorsal-ventral axis, at a distance. Ectopic Notch activation has a similar long-range effect on pip. Our results suggest that Mirror and Notch induce secretion of diffusible morphogens and we have identified TGF-beta (encoded by dpp) as such a molecule in germarium. We also found that mirr expression in dorsal follicle cells is induced by the EGF-receptor (EGFR) pathway and that mirr then represses pip expression in all but the ventral follicle cells, connecting EGFR activation in the dorsal follicle cells to repression of pip in the dorsal and lateral follicle cells. Our results suggest that the differentiation of ventral follicle cells is not a direct consequence of germline signalling, but depends on long-range signals from dorsal follicle cells, and provide a link between early and late events in Drosophila embryonic dorsal-ventral axis formation.


Subject(s)
Body Patterning/genetics , Drosophila Proteins , Embryo, Nonmammalian/embryology , Epidermal Growth Factor/metabolism , Eye Proteins/metabolism , Genes, Homeobox/genetics , Homeodomain Proteins/metabolism , Membrane Proteins/metabolism , N-Acetylglucosaminyltransferases , Transcription Factors , Animals , Crosses, Genetic , Drosophila melanogaster/embryology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Eye Proteins/genetics , Female , Homeodomain Proteins/genetics , In Situ Hybridization , Insect Proteins/metabolism , Male , Oogenesis/genetics , Ovum/cytology , Ovum/metabolism , Receptors, Notch , Signal Transduction/genetics
13.
Proc Natl Acad Sci U S A ; 96(10): 5559-64, 1999 May 11.
Article in English | MEDLINE | ID: mdl-10318923

ABSTRACT

Axis formation in Drosophila depends on correct patterning of the follicular epithelium and on signaling between the germ line and soma during oogenesis. We describe a method for identifying genes expressed in the follicle cells with potential roles in axis formation. Follicle cells are purified from whole ovaries by enzymatic digestion, filtration, and fluorescence-activated cell sorting (FACS). Two strategies are used to obtain complementary cell groups. In the first strategy, spatially restricted subpopulations are marked for FACS selection using a green fluorescent protein (GFP) reporter. In the second, cells are purified from animals mutant for the epidermal growth factor receptor ligand gurken (grk) and from their wild-type siblings. cDNA from these samples of spatially restricted or genetically mutant follicle cells is used in differential expression screens employing PCR-based differential display or hybridization to a cDNA microarray. Positives are confirmed by in situ hybridization to whole mounts. These methods are found to be capable of identifying both spatially restricted and grk-dependent transcripts. Results from our pilot screens include (i) the identification of a homologue of the immunophilin FKBP-12 with dorsal anterior expression in egg chambers, (ii) the discovery that the ecdysone-inducible nuclear hormone receptor gene E78 is regulated by grk during oogenesis and is required for proper dorsal appendage formation, and (iii) the identification of a Drosophila homologue of the human SET-binding factor gene SBF1 with elevated transcription in grk mutant egg chambers.


Subject(s)
Drosophila Proteins , Drosophila/embryology , Gene Expression Regulation, Developmental/genetics , Genes, Insect , Transforming Growth Factor alpha , Animals , Drosophila/genetics , Flow Cytometry , Green Fluorescent Proteins , Immunophilins/genetics , In Situ Hybridization , Insect Proteins/genetics , Luminescent Proteins/genetics , Molecular Sequence Data , Mutation , Reproduction/genetics , Signal Transduction/genetics , Tacrolimus Binding Proteins , Transforming Growth Factors/genetics
14.
Genetics ; 151(2): 739-48, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9927465

ABSTRACT

The fates of two small subgroups of the ovarian follicle cells appear to be linked: mutations in Notch, Delta, fs(1)Yb, or hedgehog cause simultaneous defects in the specification of stalk cells and polar cells. Both of these subgroups are determined in the germarium, and both cease division early in oogenesis. To test the possibility that these subgroups are related by lineage, we generated dominantly marked mitotic clones in ovaries. Small, restricted clones in stalk cells and polar cells were found adjacent to each other at a frequency much too high to be explained by independent induction. We therefore propose a model in which stalk cells and polar cells are derived from a precursor population that is distinct from the precursors for other follicle cells. We support and extend this model by characterization of mutants that affect stalk and polar cell formation. We find that ectopic expression of Hedgehog can induce both polar and stalk cell fate, presumably by acting on the precursor stage. In contrast, we find that stall affects neither the induction of the precursors nor the decision between the stalk cell and polar cell fate but, rather, some later differentiation step of stalk cells. In addition, we show that ectopic polar and stalk cells disturb the anterior-posterior polarity of the underlying oocyte.


Subject(s)
Cell Lineage/genetics , Drosophila Proteins , Drosophila/genetics , Gene Expression Regulation, Developmental , Genes, Insect , Insect Proteins/genetics , Ovary/cytology , Animals , Cell Differentiation/genetics , Drosophila/cytology , Female , Hedgehog Proteins
15.
Dev Genes Evol ; 209(5): 301-11, 1999 May.
Article in English | MEDLINE | ID: mdl-11252183

ABSTRACT

During Drosophila oogenesis the body axes are determined by signaling between the oocyte and the somatic follicle cells that surround the egg chamber. A key event in the establishment of oocyte anterior-posterior polarity is the differential patterning of the follicle cell epithelium along the anterior-posterior axis. Both the Notch and epithelial growth factor (EGF) receptor pathways are required for this patterning. To understand how these pathways act in the process we have analyzed markers for anterior and posterior follicle cells accompanying constitutive activation of the EGF receptor, loss of Notch function, and ectopic expression of Delta. We find that a constitutively active EGF receptor can induce posterior fate in anterior but not in lateral follicle cells, showing that the EGF receptor pathway can act only on predetermined terminal cells. Furthermore, Notch function is required at both termini for appropriate expression of anterior and posterior markers, while loss of both the EGF receptor and Notch pathways mimic the Notch loss-of-function phenotype. Ectopic expression of the Notch ligand, Delta, disturbs EGF receptor dependent posterior follicle cell differentiation and anterior-posterior polarity of the oocyte. Our data are consistent with a model in which the Notch pathway is required for early follicle cell differentiation at both termini, but is then repressed at the posterior for proper determination of the posterior follicle cells by the EGF receptor pathway.


Subject(s)
Cell Differentiation/physiology , Drosophila melanogaster/cytology , Membrane Proteins/physiology , Oogenesis/physiology , Ovarian Follicle/cytology , Animals , Drosophila Proteins , Drosophila melanogaster/physiology , ErbB Receptors/metabolism , Female , Immunohistochemistry , Receptors, Notch
16.
Development ; 124(22): 4661-71, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9409682

ABSTRACT

We describe a mutant, maelstrom, that disrupts a previously unobserved step in mRNA localization within the early oocyte, distinct from nurse-cell-to-oocyte RNA transport. Mutations in maelstrom disturb the localization of mRNAs for Gurken (a ligand for the Drosophila Egf receptor), Oskar and Bicoid at the posterior of the developing (stage 3-6) oocyte. maelstrom mutants display phenotypes detected in gurken loss-of-function mutants: posterior follicle cells with anterior cell fates, bicoid mRNA localization at both poles of the stage 8 oocyte and ventralization of the eggshell. These data are consistent with the suggestion that early posterior localization of gurken mRNA is essential for activation of the Egf receptor pathway in posterior follicle cells. Posterior localization of mRNA in stage 3-6 oocytes could therefore be one of the earliest known steps in the establishment of oocyte polarity. The maelstrom gene encodes a novel protein that has a punctate distribution in the cytoplasm of the nurse cells and the oocyte until the protein disappears in stage 7 of oogenesis.


Subject(s)
Drosophila Proteins , Drosophila melanogaster/growth & development , Drosophila melanogaster/genetics , Insect Proteins/genetics , Mutation , Oocytes/metabolism , Transforming Growth Factor alpha , Transforming Growth Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Polarity/genetics , Cloning, Molecular , DNA/genetics , Drosophila melanogaster/metabolism , ErbB Receptors/genetics , Female , Gene Expression Regulation, Developmental , Genes, Insect , Insect Proteins/metabolism , Microtubules/metabolism , Microtubules/ultrastructure , Molecular Sequence Data , Oocytes/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transforming Growth Factors/metabolism
17.
Development ; 122(12): 3745-54, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9012496

ABSTRACT

Spatially regulated activation of the Drosophila epidermal growth factor (EGF) receptor by its ligand, Gurken, is required for establishment of the dorsal/ventral axis of the oocyte and embryo. During mid-oogenesis, Gurken is concentrated at the dorsal-anterior of the oocyte and is thought to activate the EGF receptor pathway in adjacent follicle cells. In response to this signal, dorsal follicle cell fate is determined. These cells further differentiate into either appendage-producing or midline cells, resulting in patterning in the dorsal follicle cell layer. We show here that Pointed, an ETS transcription factor, is required in dorsal follicle cells for this patterning. Loss of pointed results in the loss of midline cells and an excess of appendage-forming cells, a phenotype associated with overactivation of the EGF receptor pathway in the dorsal region. Overexpression of pointed leads to a phenotype similar to that generated by loss of the EGF receptor pathway. This suggests that Pointed normally down-regulates EGF receptor signaling in the midline to generate patterning in the dorsal region. Interestingly, pointed expression is induced by the EGF receptor pathway. These data indicate a novel antagonistic function for Pointed in oogenesis; in response to activation of the EGF receptor, pointed is expressed and negatively regulates the EGF receptor pathway, possibly by integrating information from a second pathway.


Subject(s)
Body Patterning/physiology , Drosophila Proteins , ErbB Receptors/metabolism , Oogenesis/physiology , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Transforming Growth Factor alpha , Animals , DNA-Binding Proteins , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Drosophila melanogaster/growth & development , Female , Insect Hormones/metabolism , Models, Biological , Morphogenesis , Nerve Tissue Proteins , Signal Transduction , Tissue Distribution , Transforming Growth Factors/metabolism
18.
Development ; 122(11): 3639-50, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8951079

ABSTRACT

During early development, there are numerous instances where a bipotent progenitor divides to give rise to two progeny cells with different fates. The Notch gene of Drosophila and its homologues in other metazoans have been implicated in many of these cell fate decisions. It has been argued that the role of Notch in such instances may be to maintain cells in a precursor state susceptible to specific differentiating signals. This has been difficult to prove, however, due to a lack of definitive markers for precursor identity. We here perform molecular and morphological analyses of the roles of Notch in ovarian follicle cells during Drosophila oogenesis. These studies show directly that constitutively active Notch arrests cells at a precursor stage, while the loss of Notch function eliminates this stage. Expression of moderate levels of activated Notch leads to partial transformation of cell fates, as found in other systems, and we show that this milder phenotype correlates with a prolonged, but still transient, precursor stage. We also find that expression of constitutively active Notch in follicle cells at later stages leads to a defect in the anterior-posterior axis of the oocyte.


Subject(s)
Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Membrane Proteins/physiology , Oocytes/ultrastructure , Oogenesis , Ovary/physiology , Receptors, Cell Surface/physiology , Animals , Cell Compartmentation , Cytoplasm/physiology , Drosophila Proteins , Female , Phenotype , Receptors, Notch , Structure-Activity Relationship , Time Factors
19.
Curr Biol ; 4(4): 289-300, 1994 Apr 01.
Article in English | MEDLINE | ID: mdl-7922338

ABSTRACT

BACKGROUND: During oogenesis in Drosophila, determinants that will dictate abdomen and germline formation are localized to the 'polar plasm' in the posterior of the oocyte. Assembly of the polar plasm involves the sequential localization of several messenger RNAs and proteins to the posterior of the oocyte, beginning with the localization of oskar mRNA and Staufen protein during stages 8 and 9 of oogenesis. The mechanism by which these two early components accumulate at the posterior is not known. We have investigated whether directed transport along microtubules could be used to accomplish this localization. RESULTS: We have made a fusion protein composed of the bacterial beta-galactosidase enzyme as a reporter, joined to part of the plus-end-directed microtubule motor, kinesin, and have found that the fusion protein transiently localizes to the posterior of the oocyte during stages 8 and 9 of oogenesis. Treatment with the microtubule-depolymerizing agent colchicine prevents both the localization of the fusion protein and the posterior transport of oskar mRNA and Staufen protein. Furthermore, the fusion protein localizes normally in oocytes mutant for either oskar and staufen, but not in other mutants in which oskar mRNA and Staufen protein are mislocalized. CONCLUSIONS: Association with a plus-end-directed microtubule motor can promote posterior localization of a reporter protein during oogenesis. The genetic requirements for this localization and its sensitivity to colchicine, both of which are shared with the posterior transport of oskar mRNA and Staufen protein, suggest that similar mechanism may function in both processes.


Subject(s)
Kinesins/metabolism , Oocytes/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , Cell Polarity , Colchicine/pharmacology , Conserved Sequence , DNA/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Genes, Insect , Genes, Reporter , Kinesins/genetics , Models, Biological , Molecular Sequence Data , Mutation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , beta-Galactosidase/genetics
20.
Trends Genet ; 10(3): 89-94, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8178370

ABSTRACT

Establishment of the anteroposterior and dorsoventral axes of the fly originates during oogenesis and relies on signaling between the oocyte and the surrounding somatic follicle cells. Some genes originally identified as playing a role in signaling during embryonic development also mediate cell-cell communication during oogenesis. These genes have previously been grouped on the basis of their functions during embryogenesis, and this classification is largely maintained in oogenesis. The EGF receptor, the transmembrane protein rhomboid and proteins in the ras signal transduction pathway are required to initiate dorsoventral polarity, whereas the products of the neurogenic genes Notch and Delta are necessary for formation of the anteroposterior axis in the oocyte.


Subject(s)
Drosophila/embryology , Genes, Insect , Oogenesis/genetics , Animals , Cell Communication/genetics , Cell Polarity , Drosophila/genetics , Membrane Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...