Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters










Publication year range
1.
Curr Biol ; 33(5): 807-816.e4, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36706752

ABSTRACT

Germline mutations upregulating RAS signaling are associated with multiple developmental disorders. A hallmark of these conditions is that the same mutation may present vastly different phenotypes in different individuals, even in monozygotic twins. Here, we demonstrate how the origins of such largely unexplained phenotypic variations may be dissected using highly controlled studies in Drosophila that have been gene edited to carry activating variants of MEK, a core enzyme in the RAS pathway. This allowed us to measure the small but consistent increase in signaling output of such alleles in vivo. The fraction of mutation carriers reaching adulthood was strongly reduced, but most surviving animals had normal RAS-dependent structures. We rationalize these results using a stochastic signaling model and support it by quantifying cell fate specification errors in bilaterally symmetric larval trachea, a RAS-dependent structure that allows us to isolate the effects of mutations from potential contributions of genetic modifiers and environmental differences. We propose that the small increase in signaling output shifts the distribution of phenotypes into a regime, where stochastic variation causes defects in some individuals, but not in others. Our findings shed light on phenotypic heterogeneity of developmental disorders caused by deregulated RAS signaling and offer a framework for investigating causal effects of other pathogenic alleles and mild mutations in general.


Subject(s)
Signal Transduction , ras Proteins , Animals , ras Proteins/genetics , ras Proteins/metabolism , Signal Transduction/genetics , Mutation , Drosophila/genetics , Drosophila/metabolism , Phenotype
2.
Mol Biol Cell ; 32(9): 974-983, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33476180

ABSTRACT

Terminal regions of Drosophila embryos are patterned by signaling through ERK, which is genetically deregulated in multiple human diseases. Quantitative studies of terminal patterning have been recently used to investigate gain-of-function variants of human MEK1, encoding the MEK kinase that directly activates ERK by dual phosphorylation. Unexpectedly, several mutations reduced ERK activation by extracellular signals, possibly through a negative feedback triggered by signal-independent activity of the mutant variants. Here we present experimental evidence supporting this model. Using a MEK variant that combines a mutation within the negative regulatory region with alanine substitutions in the activation loop, we prove that pathogenic variants indeed acquire signal-independent kinase activity. We also demonstrate that signal-dependent activation of these variants is independent of kinase suppressor of Ras, a conserved adaptor that is indispensable for activation of normal MEK. Finally, we show that attenuation of ERK activation by extracellular signals stems from transcriptional induction of Mkp3, a dual specificity phosphatase that deactivates ERK by dephosphorylation. These findings in the Drosophila embryo highlight its power for investigating diverse effects of human disease mutations.


Subject(s)
MAP Kinase Kinase 1/genetics , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Dual-Specificity Phosphatases , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Mutation , Phosphorylation/drug effects , Signal Transduction
3.
Curr Top Dev Biol ; 140: 55-86, 2020.
Article in English | MEDLINE | ID: mdl-32591083

ABSTRACT

In Drosophila, specification of the embryonic body axes requires signaling between the germline and the somatic follicle cells. These signaling events are necessary to properly localize embryonic patterning determinants in the egg or eggshell during oogenesis. There are three maternal patterning systems that specify the anterior-posterior axis, and one that establishes the dorsal-ventral axis. We will first review oogenesis, focusing on the establishment of the oocyte and nurse cells and patterning of the follicle cells into different subpopulations. We then describe how two coordinated signaling events between the oocyte and follicle cells establish polarity of the oocyte and localize the anterior determinant bicoid, the posterior determinant oskar, and Gurken/epidermal growth factor (EGF), which breaks symmetry to initiate dorsal-ventral axis establishment. Next, we review how dorsal-ventral asymmetry of the follicle cells is transmitted to the embryo. This process also involves Gurken-EGF receptor (EGFR) signaling between the oocyte and follicle cells, leading to ventrally-restricted expression of the sulfotransferase Pipe. These events promote the ventral processing of Spaetzle, a ligand for Toll, which ultimately sets up the embryonic dorsal-ventral axis. We then describe the activation of the terminal patterning system by specialized polar follicle cells. Finally, we present open questions regarding soma-germline signaling during Drosophila oogenesis required for cell identity and embryonic axis formation.


Subject(s)
Body Patterning/genetics , Drosophila/genetics , Embryo, Nonmammalian/metabolism , Oocytes/metabolism , Oogenesis/genetics , Signal Transduction/genetics , Animals , Drosophila/embryology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryo, Nonmammalian/embryology , Female , Gene Expression Regulation, Developmental , Oocytes/cytology
4.
Annu Rev Genet ; 53: 1-18, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31794267

ABSTRACT

In Drosophila development, the axes of the egg and future embryo are established during oogenesis. To learn about the underlying genetic and molecular pathways that lead to axis formation, I conducted a large-scale genetic screen at the beginning of my independent career. This led to the eventual understanding that both anterior-posterior and dorsal-ventral pattern information is transmitted from the oocyte to the surrounding follicle cells and in turn from the follicle cells back to the oocyte. How I came to conduct this screen and what further insights were gained by studying the mutants isolated in the screen are the topics of this autobiographical article.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Genetics/history , Ovum/physiology , Animals , Body Patterning/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryo, Nonmammalian , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Developmental , History, 20th Century , History, 21st Century , Male , Oocytes/physiology , Ovary/growth & development , Ovary/physiology , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism , Sex Determination Analysis , Sex Determination Processes , United States
5.
G3 (Bethesda) ; 9(1): 47-60, 2019 01 09.
Article in English | MEDLINE | ID: mdl-30385460

ABSTRACT

The Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) and epidermal growth factor receptor (EGFR) signaling pathways are conserved regulators of tissue patterning, morphogenesis, and other cell biological processes. During Drosophila oogenesis, these pathways determine the fates of epithelial follicle cells (FCs). JAK/STAT and EGFR together specify a population of cells called the posterior follicle cells (PFCs), which signal to the oocyte to establish the embryonic axes. In this study, whole genome expression analysis was performed to identify genes activated by JAK/STAT and/or EGFR. We observed that 317 genes were transcriptionally upregulated in egg chambers with ectopic JAK/STAT and EGFR activity in the FCs. The list was enriched for genes encoding extracellular matrix (ECM) components and ECM-associated proteins. We tested 69 candidates for a role in axis establishment using RNAi knockdown in the FCs. We report that the signaling protein Semaphorin 1b becomes enriched in the PFCs in response to JAK/STAT and EGFR. We also identified ADAM metallopeptidase with thrombospondin type 1 motif A (AdamTS-A) as a novel target of JAK/STAT in the FCs that regulates egg chamber shape. AdamTS-A mRNA becomes enriched at the anterior and posterior poles of the egg chamber at stages 6 to 7 and is regulated by JAK/STAT. Altering AdamTS-A expression in the poles or middle of the egg chamber produces rounder egg chambers. We propose that AdamTS-A regulates egg shape by remodeling the basement membrane.


Subject(s)
ADAMTS1 Protein/genetics , Drosophila Proteins/genetics , ErbB Receptors/genetics , Morphogenesis/genetics , Oogenesis/genetics , Receptors, Invertebrate Peptide/genetics , Animals , Cell Polarity/genetics , Drosophila melanogaster/genetics , Female , Gene Expression Regulation, Developmental , Genome, Insect/genetics , Janus Kinases/genetics , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Ovum/growth & development , Ovum/metabolism , STAT Transcription Factors/genetics , Signal Transduction/genetics
6.
Dev Biol ; 442(1): 80-86, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30026122

ABSTRACT

Receptor tyrosine kinases (RTKs) control a wide range of developmental processes, from the first stages of embryogenesis to postnatal growth and neurocognitive development in the adult. A significant share of our knowledge about RTKs comes from genetic screens in model organisms, which provided numerous examples demonstrating how specific cell fates and morphologies are abolished when RTK activation is either abrogated or significantly reduced. Aberrant activation of such pathways has also been recognized in many forms of cancer. More recently, studies of human developmental syndromes established that excessive activation of RTKs and their downstream signaling effectors, most notably the Ras signaling pathway, can also lead to structural and functional defects. Given that both insufficient and excessive pathway activation can lead to abnormalities, mechanistic analysis of developmental RTK signaling must address quantitative questions about its regulation and function. Patterning events controlled by the RTK Torso in the early Drosophila embryo are well-suited for this purpose. This mini review summarizes current state of knowledge about Torso-dependent Ras activation and discusses its potential to serve as a quantitative model for studying the general principles of Ras signaling in development and disease.


Subject(s)
Body Patterning/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/physiology , Animals , Body Patterning/genetics , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/genetics
7.
RNA ; 23(7): 1097-1109, 2017 07.
Article in English | MEDLINE | ID: mdl-28420675

ABSTRACT

Piwi-interacting RNAs (piRNAs) are central components of the piRNA pathway, which directs transposon silencing and guarantees genome integrity in the germ cells of several metazoans. In Drosophila, piRNAs are produced from discrete regions of the genome termed piRNA clusters, whose expression relies on the RDC complex comprised of the core proteins Rhino, Deadlock, and Cutoff. To date, the RDC complex has been exclusively implicated in the regulation of the piRNA loci. Here we further elucidate the function of Cutoff and the RDC complex by performing genome-wide ChIP-seq and RNA-seq assays in the Drosophila ovaries and analyzing these data together with other publicly available data sets. In agreement with previous studies, we confirm that Cutoff is involved in the transcriptional regulation of piRNA clusters and in the repression of transposable elements in germ cells. Surprisingly, however, we find that Cutoff is enriched at and affects the expression of other noncoding RNAs, including spliceosomal RNAs (snRNAs) and small nucleolar RNAs (snoRNAs). At least in some instances, Cutoff appears to act at a transcriptional level in concert with Rhino and perhaps Deadlock. Finally, we show that mutations in Cutoff result in the deregulation of hundreds of protein-coding genes in germ cells. Our study uncovers a broader function for the RDC complex in the Drosophila germline development.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Ovary/growth & development , RNA, Small Interfering/metabolism , RNA, Untranslated/metabolism , RNA-Binding Proteins/metabolism , Animals , Chromatin Immunoprecipitation , DNA Transposable Elements , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Female , Gene Expression Regulation , Gene Expression Regulation, Developmental , Mutation , Ovary/chemistry , RNA-Binding Proteins/genetics , Sequence Analysis, RNA/methods
8.
Cell Rep ; 18(8): 1831-1839, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228250

ABSTRACT

The basement membrane (BM), a sheet of extracellular matrix lining the basal side of epithelia, is essential for epithelial cell function and integrity, yet the mechanisms that control the basal restriction of BM proteins are poorly understood. In epithelial cells, a specialized pathway is dedicated to restrict the deposition of BM proteins basally. Here, we report the identification of a factor in this pathway, a homolog of the mammalian guanine nucleotide exchange factor (GEF) Mss4, which we have named Stratum. The loss of Stratum leads to the missecretion of BM proteins at the apical side of the cells, forming aberrant layers in close contact with the plasma membrane. We found that Rab8GTPase acts downstream of Stratum in this process. Altogether, our results uncover the importance of this GEF/Rab complex in specifically coordinating the basal restriction of BM proteins, a critical process for the establishment and maintenance of epithelial cell polarity.


Subject(s)
Basement Membrane/metabolism , Cell Membrane/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Epithelial Cells/metabolism , GTP Phosphohydrolases/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Membrane Proteins/metabolism , Animals , Animals, Genetically Modified/metabolism , Cell Polarity/physiology , Humans
9.
Nat Genet ; 49(3): 465-469, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28166211

ABSTRACT

Germline mutations in Ras pathway components are associated with a large class of human developmental abnormalities, known as RASopathies, that are characterized by a range of structural and functional phenotypes, including cardiac defects and neurocognitive delays. Although it is generally believed that RASopathies are caused by altered levels of pathway activation, the signaling changes in developing tissues remain largely unknown. We used assays with spatiotemporal resolution in Drosophila melanogaster (fruit fly) and Danio rerio (zebrafish) to quantify signaling changes caused by mutations in MAP2K1 (encoding MEK), a core component of the Ras pathway that is mutated in both RASopathies and cancers in humans. Surprisingly, we discovered that intrinsically active MEK variants can both increase and reduce the levels of pathway activation in vivo. The sign of the effect depends on cellular context, implying that some of the emerging phenotypes in RASopathies may be caused by increased, as well as attenuated, levels of Ras signaling.


Subject(s)
Germ-Line Mutation/genetics , Mitogen-Activated Protein Kinases/genetics , Signal Transduction/genetics , ras Proteins/genetics , Animals , Drosophila melanogaster/genetics , Heart Diseases/genetics , Humans , Neoplasms/genetics , Neurocognitive Disorders/genetics , Phenotype , Zebrafish/genetics
10.
Dev Cell ; 40(2): 185-192, 2017 01 23.
Article in English | MEDLINE | ID: mdl-28118601

ABSTRACT

Animal development is characterized by signaling events that occur at precise locations and times within the embryo, but determining when and where such precision is needed for proper embryogenesis has been a long-standing challenge. Here we address this question for extracellular signal regulated kinase (Erk) signaling, a key developmental patterning cue. We describe an optogenetic system for activating Erk with high spatiotemporal precision in vivo. Implementing this system in Drosophila, we find that embryogenesis is remarkably robust to ectopic Erk signaling, except from 1 to 4 hr post-fertilization, when perturbing the spatial extent of Erk pathway activation leads to dramatic disruptions of patterning and morphogenesis. Later in development, the effects of ectopic signaling are buffered, at least in part, by combinatorial mechanisms. Our approach can be used to systematically probe the differential contributions of the Ras/Erk pathway and concurrent signals, leading to a more quantitative understanding of developmental signaling.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/enzymology , Embryonic Development , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Animals , Drosophila melanogaster/genetics , Embryonic Development/genetics , Embryonic Development/radiation effects , Enzyme Activation/radiation effects , Gene Expression Regulation, Developmental/radiation effects , Light , Optogenetics , Time Factors , Transcription, Genetic/radiation effects , ras Proteins/metabolism
11.
Proc Natl Acad Sci U S A ; 114(3): 510-515, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28049852

ABSTRACT

Germ-line mutations in components of the Ras/MAPK pathway result in developmental disorders called RASopathies, affecting about 1/1,000 human births. Rapid advances in genome sequencing make it possible to identify multiple disease-related mutations, but there is currently no systematic framework for translating this information into patient-specific predictions of disease progression. As a first step toward addressing this issue, we developed a quantitative, inexpensive, and rapid framework that relies on the early zebrafish embryo to assess mutational effects on a common scale. Using this assay, we assessed 16 mutations reported in MEK1, a MAPK kinase, and provide a robust ranking of these mutations. We find that mutations found in cancer are more severe than those found in both RASopathies and cancer, which, in turn, are generally more severe than those found only in RASopathies. Moreover, this rank is conserved in other zebrafish embryonic assays and Drosophila-specific embryonic and adult assays, suggesting that our ranking reflects the intrinsic property of the mutant molecule. Furthermore, this rank is predictive of the drug dose needed to correct the defects. This assay can be readily used to test the strengths of existing and newly found mutations in MEK1 and other pathway components, providing the first step in the development of rational guidelines for patient-specific diagnostics and treatment of RASopathies.


Subject(s)
Developmental Disabilities/genetics , ras Proteins/genetics , Animals , Animals, Genetically Modified , Developmental Disabilities/drug therapy , Developmental Disabilities/metabolism , Dose-Response Relationship, Drug , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mutation , Phenotype , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
Dev Biol ; 414(2): 193-206, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27130192

ABSTRACT

Cell migration plays crucial roles during development. An excellent model to study coordinated cell movements is provided by the migration of border cell clusters within a developing Drosophila egg chamber. In a mutagenesis screen, we isolated two alleles of the gene rickets (rk) encoding a G-protein-coupled receptor. The rk alleles result in border cell migration defects in a significant fraction of egg chambers. In rk mutants, border cells are properly specified and express the marker Slbo. Yet, analysis of both fixed as well as live samples revealed that some single border cells lag behind the main border cell cluster during migration, or, in other cases, the entire border cell cluster can remain tethered to the anterior epithelium as it migrates. These defects are observed significantly more often in mosaic border cell clusters, than in full mutant clusters. Reduction of the Rk ligand, Bursicon, in the border cell cluster also resulted in migration defects, strongly suggesting that Rk signaling is utilized for communication within the border cell cluster itself. The mutant border cell clusters show defects in localization of the adhesion protein E-cadherin, and apical polarity proteins during migration. E-cadherin mislocalization occurs in mosaic clusters, but not in full mutant clusters, correlating well with the rk border cell migration phenotype. Our work has identified a receptor with a previously unknown role in border cell migration that appears to regulate detachment and polarity of the border cell cluster coordinating processes within the cells of the cluster themselves.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/cytology , Oogenesis/physiology , Ovary/cytology , Receptors, G-Protein-Coupled/physiology , Alleles , Animals , CCAAT-Enhancer-Binding Proteins/physiology , Cadherins/physiology , Cell Adhesion , Cell Movement/physiology , Cell Polarity/physiology , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Epithelial Cells/cytology , Epithelial Cells/physiology , Female , Invertebrate Hormones/physiology , Mosaicism , Ovary/growth & development , Phenotype , RNA Interference , Receptors, G-Protein-Coupled/genetics , Sequence Deletion
13.
Curr Top Dev Biol ; 117: 319-30, 2016.
Article in English | MEDLINE | ID: mdl-26969986

ABSTRACT

The study of development involves many important techniques. Here I am trying to reflect on the strength of genetic analysis and its ability to uncover unexpected relationships and regulatory inputs from seemingly unrelated pathways.


Subject(s)
Developmental Biology , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Signal Transduction , Animals , Humans
14.
Curr Biol ; 25(13): 1784-90, 2015 Jun 29.
Article in English | MEDLINE | ID: mdl-26096970

ABSTRACT

Transient activation of the highly conserved extracellular-signal-regulated kinase (ERK) establishes precise patterns of cell fates in developing tissues. Quantitative parameters of these transients are essentially unknown, but a growing number of studies suggest that changes in these parameters can lead to a broad spectrum of developmental abnormalities. We provide a detailed quantitative picture of an ERK-dependent inductive signaling event in the early Drosophila embryo, an experimental system that offers unique opportunities for high-throughput studies of developmental signaling. Our analysis reveals a spatiotemporal pulse of ERK activation that is consistent with a model in which transient production of a short-ranged ligand feeds into a simple signal interpretation system. The pulse of ERK signaling acts as a switch in controlling the expression of the ERK target gene. The quantitative approach that led to this model, based on the integration of data from fixed embryos and live imaging, can be extended to other developmental systems patterned by transient inductive signals.


Subject(s)
Autocrine Communication/physiology , Drosophila/physiology , Enzyme Activation/physiology , Gene Expression Regulation, Developmental/physiology , MAP Kinase Signaling System/physiology , Models, Biological , Animals , Embryo, Nonmammalian/physiology , ErbB Receptors/metabolism , Image Processing, Computer-Assisted , In Situ Hybridization , Kinetics , Microscopy, Confocal , Time Factors
15.
Development ; 142(11): 1971-7, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25953345

ABSTRACT

The eggshells of drosophilid species provide a powerful model for studying the origins of morphological diversity. The dorsal appendages, or respiratory filaments, of these eggshells display a remarkable interspecies variation in number and shape, and the epithelial patterning underlying the formation of these structures is an area of active research. To extend the analysis of dorsal appendage formation to include morphogenesis, we developed an improved 3D image reconstruction approach. This approach revealed considerable interspecies variation in the cell shape changes and neighbor exchanges underlying appendage formation. Specifically, although the appendage floor in Drosophila melanogaster is formed through spatially ordered neighbor exchanges, the same structure in Scaptodrosophila pattersoni is formed through extreme changes in cell shape, whereas Drosophila funebris appears to display a combination of both cellular mechanisms. Furthermore, localization patterns of Par3/Bazooka suggest a self-organized, cell polarity-based origin for the variability of appendage number in S. pattersoni. Our results suggest that species deploy different combinations of apically and basally driven mechanisms to convert a two-dimensional primordium into a three-dimensional structure, and provide new directions for exploring the molecular origins of interspecies morphological variation.


Subject(s)
Epithelium/growth & development , Morphogenesis , Ovum/cytology , Animals , Cell Shape , Drosophila/cytology , Drosophila/growth & development , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Ovum/ultrastructure , Species Specificity
16.
Development ; 141(20): 3910-21, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25231760

ABSTRACT

In Drosophila melanogaster, the anteroposterior (AP) and dorsoventral (DV) axes of the oocyte and future embryo are established through the localization and translational regulation of gurken (grk) mRNA. This process involves binding of specific factors to the RNA during transport and a dynamic remodeling of the grk-containing ribonucleoprotein (RNP) complexes once they have reached their destination within the oocyte. In ovaries of spindle-class females, an activated DNA damage checkpoint causes inefficient Grk translation and ventralization of the oocyte. In a screen for modifiers of the oocyte DV patterning defects, we identified a mutation in the eIF1A gene as a dominant suppressor. We show that reducing the function of eIF1A in spnB ovaries suppresses the ventralized eggshell phenotype by restoring Grk expression. This suppression is not the result of more efficient DNA damage repair or of disrupted checkpoint activation, but is coupled to an increase in the amount of grk mRNA associated with polysomes. In spnB ovaries, the activated meiotic checkpoint blocks Grk translation by disrupting the accumulation of grk mRNA in a translationally competent RNP complex that contains the translational activator Oo18 RNA-binding protein (Orb); this regulation involves the translational repressor Squid (Sqd). We further propose that reduction of eIF1A allows more efficient Grk translation possibly because of the presence of specific structural features in the grk 5'UTR.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/embryology , Eukaryotic Initiation Factor-1/metabolism , Gene Expression Regulation, Developmental , Oogenesis , Transforming Growth Factor alpha/physiology , Animals , DNA Damage , Drosophila Proteins/metabolism , Egg Proteins/metabolism , Eukaryotic Initiation Factor-1/genetics , Female , Genotype , Male , Meiosis , Mutation , Oocytes/cytology , Ovary/metabolism , Phenotype , Polyribosomes/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Ribonucleoproteins/chemistry
17.
Proc Natl Acad Sci U S A ; 111(21): 7689-94, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24828534

ABSTRACT

The basement membrane (BM), a specialized sheet of the extracellular matrix contacting the basal side of epithelial tissues, plays an important role in the control of the polarized structure of epithelial cells. However, little is known about how BM proteins themselves achieve a polarized distribution. Here, we identify phosphatidylinositol 4,5-bisphosphate (PIP2) as a critical regulator of the polarized secretion of BM proteins. A decrease of PIP2 levels, in particular through mutations in Phosphatidylinositol synthase (Pis) and other members of the phosphoinositide pathway, leads to the aberrant accumulation of BM components at the apical side of the cell without primarily affecting the distribution of apical and basolateral polarity proteins. In addition, PIP2 controls the apical and lateral localization of Crag (Calmodulin-binding protein related to a Rab3 GDP/GTP exchange protein), a factor specifically required to prevent aberrant apical secretion of BM. We propose that PIP2, through the control of Crag's subcellular localization, restricts the secretion of BM proteins to the basal side.


Subject(s)
Basement Membrane/metabolism , CDP-Diacylglycerol-Inositol 3-Phosphatidyltransferase/metabolism , Cell Polarity/physiology , Cell Transformation, Neoplastic/genetics , Epithelial Cells/physiology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Animals , Drosophila , Epithelial Cells/metabolism , Female , Fluorescent Antibody Technique , Ovary/metabolism
18.
Chem Biol Drug Des ; 84(5): 505-12, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24751138

ABSTRACT

Inhibiting aggregation of the amyloid-beta (Aß) peptide may be an effective strategy for combating Alzheimer's disease. As the high-resolution structure of the toxic Aß aggregate is unknown, rational design of small molecule inhibitors is not possible, and inhibitors are best isolated by high-throughput screening. We applied high-throughput screening to a collection of 65,000 compounds to identify compound D737 as an inhibitor of Aß aggregation. D737 diminished the formation of oligomers and fibrils, and reduced Aß42-induced cytotoxicity. Most importantly, D737 increased the life span and locomotive ability of transgenic flies in a Drosophila melanogaster model of Alzheimer's disease (J Biol Chem, 287, 2012, 38992). To explore the chemical features that make D737 an effective inhibitor of Aß42 aggregation and toxicity, we tested a small collection of eleven analogues of D737. Overall, the ability of a compound to inhibit Aß aggregation was a good predictor of its efficacy in prolonging the life span and locomotive ability of transgenic flies expressing human Aß42 in the central nervous system. Two compounds (D744 and D830) with fluorine substitutions on an aromatic ring were effective inhibitors of Aß42 aggregation and increased the longevity of transgenic flies beyond that observed for the parent compound, D737.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Peptide Fragments/antagonists & inhibitors , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Aging/drug effects , Aging/genetics , Alzheimer Disease/drug therapy , Animals , Animals, Genetically Modified , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , High-Throughput Screening Assays/methods , Humans , Male , Rats , Toxicity Tests
19.
Dev Biol ; 386(2): 408-18, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24373956

ABSTRACT

The border cells of Drosophila are a model system for coordinated cell migration. Ecdysone signaling has been shown to act as the timing signal to initiate the migration process. Here we find that mutations in phantom (phm), encoding an enzyme in the ecdysone biosynthesis pathway, block border cell migration when the entire follicular epithelium of an egg chamber is mutant, even when the associated germline cells (nurse cells and oocyte) are wild-type. Conversely, mutant germline cells survive and do not affect border cell migration, as long as the surrounding follicle cells are wild-type. Interestingly, even small patches of wild-type follicle cells in a mosaic epithelium are sufficient to allow the production of above-threshold levels of ecdysone to promote border cell migration. The same phenotype is observed with mutations in shade (shd), encoding the last enzyme in the pathway that converts ecdysone to the active 20-hydroxyecdysone. Administration of high 20-hydroxyecdysone titers in the medium can also rescue the border cell migration phenotype in cultured egg chambers with an entirely phm mutant follicular epithelium. These results indicate that in normal oogenesis, the follicle cell epithelium of each individual egg chamber must supply sufficient ecdysone precursors, leading ultimately to high enough levels of mature 20-hydroxyecdysone to the border cells to initiate their migration. Neither the germline, nor the neighboring egg chambers, nor the surrounding hemolymph appear to provide threshold amounts of 20-hydroxyecdysone to do so. This "egg chamber autonomous" ecdysone synthesis constitutes a useful way to regulate the individual maturation of the asynchronous egg chambers present in the Drosophila ovary.


Subject(s)
Cell Movement/physiology , Drosophila Proteins/metabolism , Drosophila/embryology , Drosophila/enzymology , Ecdysone/biosynthesis , Ecdysterone/metabolism , Mixed Function Oxygenases/metabolism , Animals , Drosophila Proteins/genetics , Ecdysone/genetics , Microscopy, Fluorescence , Mixed Function Oxygenases/genetics , Mutation/genetics
20.
Mol Biol Cell ; 24(18): 2966-80, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23864715

ABSTRACT

Calcium-dependent cysteine proteases of the calpain family are modulatory proteases that cleave their substrates in a limited manner. Among their substrates, calpains target vertebrate and invertebrate IκB proteins. Because proteolysis by calpains potentially generates novel protein functions, it is important to understand how this affects NFκB activity. We investigate the action of Calpain A (CalpA) on the Drosophila melanogaster IκB homologue Cactus in vivo. CalpA alters the absolute amounts of Cactus protein. Our data indicate, however, that CalpA uses additional mechanisms to regulate NFκB function. We provide evidence that CalpA interacts physically with Cactus, recognizing a Cactus pool that is not bound to Dorsal, a fly NFκB/Rel homologue. We show that proteolytic cleavage by CalpA generates Cactus fragments lacking an N-terminal region required for Toll responsiveness. These fragments are generated in vivo and display properties distinct from those of full-length Cactus. We propose that CalpA targets free Cactus, which is incorporated into and modulates Toll-responsive complexes in the embryo and immune system.


Subject(s)
Calpain/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , I-kappa B Proteins/metabolism , Phosphoproteins/metabolism , Proteolysis , Toll-Like Receptors/metabolism , Animals , Body Patterning , Cell Line , DNA-Binding Proteins/chemistry , Drosophila Proteins/chemistry , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Drosophila melanogaster/immunology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Fat Body/cytology , Fat Body/metabolism , Immune System/metabolism , Larva/cytology , Larva/metabolism , Models, Biological , Mutation/genetics , NF-KappaB Inhibitor alpha , Phosphoproteins/chemistry , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...