Article

Ghiglione C, Carraway III KL, Amundadottir LT, Boswell RE, Perrimon N, Duffy JBThe transmembrane molecule kekkon 1 acts in a feedback loop to negatively regulate the activity of the Drosophila EGF receptor during oogenesis. Cell 96: 847-856

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract

We have identified the Drosophila transmembrane molecule kekkon 1 (kek1) as an inhibitor of the epidermal growth factor receptor (EGFR) and demonstrate that it acts in a negative feedback loop to modulate the activity of the EGFR tyrosine kinase. During oogenesis, kek1 is expressed in response to the Gurken/EGFR signaling pathway, and loss of kek1 activity is associated with an increase in EGFR signaling. Consistent with our loss-of-function studies, we demonstrate that ectopic overexpression of kek1 mimics a loss of EGFR activity. We show that the extracellular and transmembrane domains of Kek1 can inhibit and physically associate with the EGFR, suggesting potential models for this inhibitory mechanism.

No full-text available

Request Full-text Paper PDF

To read the full-text of this research,
you can request a copy directly from the authors.

... The LRIG gene family was discovered when searching for paralogous genes in the human genome, based on research previously performed on Drosophila and mice, where it was discovered that this gene was regulated downstream by epidermal growth factor receptor (EGFR) [12,13]. The gene encoded a transmembrane protein containing a cell adhesion molecule with leucine-rich repeats and immunoglobulin-like domain. ...
... The gene encoded a transmembrane protein containing a cell adhesion molecule with leucine-rich repeats and immunoglobulin-like domain. In Drosophila, it was established that this protein is produced by epidermal growth factor (EGF) stimulation, where it binds to EGFR, which, in turn regulates its activity through a negative feedback loop [12]. The search for a paralogous gene in humans led to the discovery of the LRIG gene family, namely genes LRIG1, LRIG2, and LRIG3 [14][15][16]. ...
Article
Full-text available
Cell-free DNA (cfDNA) has recently emerged as a promising minimally invasive diagnostic biomarker for various cancers. In this study, our aim was to identify cfDNA biomarkers by investigating genes that displayed significant differences between glioma patients and their corresponding controls. To accomplish this, we utilized publicly available data from the Gene Expression Omnibus, focusing on 5-hydroxymethylcytosine (5hmC) profiles in both cfDNA and genomic DNA (gDNA) from glioma patients and healthy individuals. The intersection of gene lists derived from these comparative analyses unveiled LRIG1 and ZNF703 as the two genes with elevated 5hmC levels in both the cfDNA of glioma patients and gDNA of glioma tissue compared to their respective controls. The gene expression data revealed both genes were upregulated in glioma tissue compared to normal brain tissue. Integration of 5hmC data revealed a strong positive correlation in the glioma tissue group between 5hmC and the gene expression of the LRIG1 gene. Furthermore, exploration using the AmiCa web tool indicated that LRIG1 gene expression was elevated compared to 17 other cancers included in the database, emphasizing its potential as a distinctive biomarker across multiple cancer types.
... In CIN-wing discs, the pERK signal was increased in the whole transgene-expressing compartment both apically and basally ( Figures 5B and 5B 0 ; see also Figures S6O and S6Q), and this increase was reduced upon expression of a dominant negative version of the EGFR receptor that lacks the cytoplasmic kinase domain (EGFR DN ; Figure 5C). We also observed the upregulation of kekkon-1 (a gene regulated by EGFR, (Ghiglione et al., 1999; see also Figure S6A) in CIN-tissues ( Figures 5H and 5I) and in cells invading the V compartment (red arrows in Figure 5I 0 ). Interestingly, the disorganization of the DV compartment boundary and invasive behavior of CIN-induced aneuploid cells were both largely rescued by EGFR DN expression (Figures 5J and 5K; quantification in Figure 5P). ...
... The levels of JNK activity, monitored by the expression of MMP1, and MyoII activation (pSqh) were unaffected by EGFR DN (Figures S6J and S6K). Interestingly, depletion of kekkon-1, which encodes for a transmembrane protein that acts as a potent repressor of EGFR (Ghiglione et al., 1999), enhanced CIN-induced migration (Figures 5P and 5Q). Taken together, these results indicate that the ectopic activation of EGFR contributes to CIN-induced tissue overgrowth and to the invasive behavior of aneuploid cells. ...
Article
Most sporadic carcinomas with high metastatic activity show an increased rate of changes in chromosome structure and number, known as chromosomal instability (CIN). However, the role of CIN in driving invasiveness remains unclear. Using an epithelial model in Drosophila, we present evidence that CIN promotes a rapid and general invasive behavior. Cells with an abnormal number of chromosomes delaminate from the epithelium, extend actin-based cellular protrusions, form membrane blebs, and invade neighboring tissues. This behavior is governed by the activation of non-muscle Myosin II by Rho kinase and by the expression of the secreted EGF/Spitz ligand. We unravel fundamental roles of the mitogen-activated protein kinase pathways mediated by the Fos proto-oncogene and the Capicua tumor suppressor gene in the invasive behavior of CIN-induced aneuploid cells. Our results support the proposal that the simple production of unbalanced karyotypes contributes to CIN-induced metastatic progression.
... BuGZ depletion in ISCs resulted in a significant decrease of dpERK signial in ISCs (Supplementary Fig. 7i), while YT521-B RNAi restored the down-regulation of dpERK in ISCs caused by BuGZ depletion (Supplementary Fig. 7i). We also investigated the impact of BuGZ and YT521-B on the mRNA expressions of Argos (aos), Kekkon 1 (kek1), and Sprouty (sty) in sorted esg + cells, all of which were reported to act as downstream targets of MAPK signaling pathway 37,[40][41][42] . We found that the overexpression of BuGZ FL augmented the mRNA levels of kek1, sty, and aos (Supplementary Fig. 7j). ...
Article
Full-text available
Exploring the role of phase separation in intracellular compartment formation is an active area of research. However, the associations of phase separation with intestinal stem cell (ISC)-dependent regeneration and aging remain unclear. Here, we demonstrate that BuGZ, a coacervating mitotic effector, shows age- and injury-associated condensation in Drosophila ISC nuclei during interphase. BuGZ condensation promotes ISC proliferation, affecting Drosophila gut repair and longevity. Moreover, m⁶A reader YT521-B acts as the transcriptional and functional downstream of BuGZ. The binding of YT521-B promotor or m⁶A writer Ime4/ Mettl14 to BuGZ controls its coacervation, indicating that the promotor may accelerate the phase transition of its binding transcription factor. Hence, we propose that phase separation and m⁶A regulators may be critical for ameliorating ISC-dependent gut regeneration and aging and requires further study.
... oogenesis and involves a physical and direct association between the extracellular or transmembrane domains of both proteins [3][4] . Further study revealed that the 6 LRRs of Kekkon-1 are necessary for the recognition of EGFR and for consequent inhibition of activation by growth factors [4][5] . ...
Article
Objective Leucine-rich repeats and immunoglobulin-like domains 1 ( LRIG1 ) is a newly identified human gene that inhibits the epidermal growth factor receptor (EGFR), which on combining with a ligand, can drive tumor growth. This study investigated the interaction between human LRIG1 and EGFR and attempted to delineate the functions of as well as the mechanisms used by the extracellular (ECD) and cytoplasmic (CPD) domains of the human LRIG1 protein to downregulate human EGFR signaling activity. Methods Two constructed chimeric eukaryotic expression vectors, pIRES2-EGFP-3XFLAG-LRIG1-ET and p3FLAG-LRIG1-TC, encoding the extracellular and transmembrane regions (LRIG1-ET) and the transmembrane and cytoplasmic regions (LRIG1-TC), respectively, and the plasmid p3XFLAG-CMV-9-LRIG1 encoding full-length LRIG1 (LRIG1-FL) were transfected into the human glioma cell line U251 or primary astrocytoma cells by using liposomes. The number and affinity of cell surface EGFR on transfected cells was determined by ¹²⁵ I-EGF binding assay. Results The dissociation constant (KD) values for EGFR were higher, and the maximum increase was observed in the cells transfected into LRIG1-ET (1.36 folds). The number of maximal binding sites (Bmax) of the receptors was decreased in all transfected cells; the maximum decrease was noted in the cells transfected into LRIG1-FL (40.05%). Conclusion Both the ECD and CPD of LRIG1 are important to negate EGFR signaling. The ECD may interfere with the binding between EGFR and its ligand and facilitate the functions of CPD. The CPD may, when brought in proximity to EGFR, enhance receptor degradation. These two mechanisms can contribute to the downregulation of EGFR-mediated signaling by LRIG1.
... Negative feedback loops are present at multiple levels of the EGFR signaling pathway including ligand sequestration, direct association with EGFR, blockade of Ras binding to receptor complexes, and the activation of MAPK or downstream transcription factors [71][72][73][74][75]. It is within this framework of extensive negative feedback regulation where the function of EGFRAP can be better understood. ...
Article
Full-text available
Activation of Ras signaling occurs in ~30% of human cancers. However, activated Ras alone is insufficient to produce malignancy. Thus, it is imperative to identify those genes cooperating with activated Ras in driving tumoral growth. In this work, we have identified a novel EGFR inhibitor, which we have named EGFRAP , for EGFR a daptor p rotein. Elimination of EGFRAP potentiates activated Ras-induced overgrowth in the Drosophila wing imaginal disc. We show that EGFRAP interacts physically with the phosphorylated form of EGFR via its SH2 domain. EGFRAP is expressed at high levels in regions of maximal EGFR/Ras pathway activity, such as at the presumptive wing margin. In addition, EGFRAP expression is up-regulated in conditions of oncogenic EGFR/Ras activation. Normal and oncogenic EGFR/Ras-mediated upregulation of EGRAP levels depend on the Notch pathway. We also find that elimination of EGFRAP does not affect overall organogenesis or viability. However, simultaneous downregulation of EGFRAP and its ortholog PVRAP results in defects associated with increased EGFR function. Based on these results, we propose that EGFRAP is a new negative regulator of the EGFR/Ras pathway, which, while being required redundantly for normal morphogenesis, behaves as an important modulator of EGFR/Ras-driven tissue hyperplasia. We suggest that the ability of EGFRAP to functionally inhibit the EGFR pathway in oncogenic cells results from the activation of a feedback loop leading to increase EGFRAP expression. This could act as a surveillance mechanism to prevent excessive EGFR activity and uncontrolled cell growth.
... For example, homologs of three of the highly differentiated genes in the Waresley Wood population-obg-like ATPase, transmembrane protein 214, and liprin-alpha-show elevated expression in the ovaries of fruit flies (Chintapalli et al. 2013) and other arthropods (Cao and Jiang 2017), suggesting a plausible role in regulating egg production. Another gene, kekkon1, is a transmembrane protein known to regulate the activity of the epidermal growth factor receptor during oogenesis in Drosophila (Ghiglione et al. 1999;Wittes and Schüpbach 2019). Finally, we asked whether genes involved in oogenesis generally showed elevated levels of differentiation in each population, in comparison with the rest of the genome. ...
Article
Full-text available
Models of “plasticity‐first” evolution are attractive because they explain the rapid evolution of new complex adaptations. Nevertheless, it is unclear whether plasticity can facilitate rapid microevolutionary change between diverging populations. Here, we show how plasticity may have generated adaptive differences in fecundity between neighboring wild populations of burying beetles Nicrophorus vespilloides. These populations occupy distinct Cambridgeshire woodlands that are just 2.5 km apart and that probably originated from a common ancestral population about 1000‐4000 years ago. We find that populations are divergently adapted to breed on differently sized carrion. Adaptive differences in clutch size and egg size are associated with divergence at loci connected with oogenesis. The populations differ specifically in the elevation of the reaction norm linking clutch size to carrion size (i.e., genetic accommodation), and in the likelihood that surplus offspring will be lost after hatching. We suggest that these two processes may have facilitated rapid local adaptation on a fine‐grained spatial scale.
... Deux autres protéines peuvent réguler l'activation de la voie. Tout d'abord Kekkon, qui est une protéine transmembranaire qui forme des complexes inactifs avec EGFR (Ghiglione et al., 1999). Sprouty, elle, est une protéine cytoplasmique capable d'inhiber la voie canonique RAS/MAPK à plusieurs niveaux, à l'instar du rôle de ses orthologues chez l'humain (Casci et al., 1999). ...
Thesis
Full-text available
L’étude d’échantillons humains montre que les voies de signalisation RAS/MAPK et PI3K/AKT/mTOR sont fréquemment activées de manière aberrante dans les tumeurs de la prostate, d’autant plus dans les phases de résistance aux traitements. Ces deux voies de signalisation sont sensibles aux facteurs de croissances et impliquées dans la régulation de processus cellulaires fondamentaux tels que la prolifération, la croissance ou encore la différenciation cellulaire. Ces données suggèrent qu’elles ont un rôle essentiel dans la tumorigenèse prostatique. Cependant, le rôle respectif de chacune de ces voies dans la carcinogenèse prostatique, particulièrement dans les phases précoces, n’est pas clairement établit. L’objectif de ma thèse est donc de définir le rôle possible de ces deux voies dans l’initiation et la progression du cancer de la prostate, ainsi que les mécanismes impliqués dans leur co-dérégulation. Cette étude est réalisée dans un modèle in vivo alternatif, la drosophile, qui possèdent un équivalent fonctionnel de la prostate : les glandes accessoires. La première partie des travaux réalisés montre que seule la suractivation de la voie RAS/MAPK dans la glande accessoire conduit à un processus de tumorigenèse, avec la production de masses cellulaires récapitulant de nombreuses caractéristiques cancéreuses : croissance cellulaire et prolifération incontrôlée, expression de métalloprotéases, perte de l’expression de marqueurs épithéliaux et formation de nouvelles trachées. Cependant, les deux voies de signalisation sont nécessaires à la tumorigenèse, mais avec des rôles différents : la voie RAS/MAPK est activée précocement et est capable de recruter la voie PI3K/AKT/TOR grâce à la mise en place de deux boucles autocrines de régulation. La première dépend de spitz (dEGF) et du récepteur EGFR pour amplifier l’activation de la voie RAS/MAPK. La seconde dépend de l’activation d’ILP6 (dIGF1), produit suite à l’activation de la voie RAS/MAPK, et permet le recrutement de la voie PI3K/AKT/TOR par l’intermédiaire du récepteur à l’insuline InR. La deuxième partie des travaux réalisés montre que l’activation de la voie RAS/MAPK conduit à la production de MMP1 dans les cellules qui seront à l’origine des tumeurs avant leur extravasation hors de l’épithélium. Cette expression temporelle contrôlée correspond à une étape où une réorganisation du cytosquelette a lieu et où le microenvironnement est altéré. Ces données placent donc la dérégulation de la voie RAS/MAPK comme un évènement précoce de la tumorigenèse prostatique, capable de recruter la voie PI3K/AKT/TOR et d’entrainer la production de MMP1, pour in fine conduire à l’extravasation des cellules et à la formation de tumeurs.
... In response to EGFR activity, the FCs on the dorsal side express MAP Kinase target genes, such as kekkon (kek), pointed and spitz (spi) (Ghiglione et al., 1999;Peri, B€ okel, & Roth, 1999;Ruohola-Baker et al., 1993;Sapir, Schweitzer, & Shilo, 1998;Wasserman & Freeman, 1998;Yakoby et al., 2008). EGFR activity also leads to the repression of genes with general activity in the FCs such as Chorion factor 2 (Cf2) and capicua (cic) (Atkey, Boisclair Lachance, Walczak, Rebello, & Nilson, 2006;Goff, Nilson, & Morisato, 2001;Jim enez, Guichet, Ephrussi, & Casanova, 2000;Mantrova & Hsu, 2008). ...
Chapter
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.
... The ERK signaling pathway components also showed significant changes from the bulk scrib tumor time-series transcriptomic data ( Figure 4A). Moreover, when we ordered genes by the percentage change of cells expressing the specific gene in the single-cell transcriptomic data, we noticed that kekkon1 (kek1), a well-established ERK signaling target (Ghiglione et al., 2003;Ghiglione et al., 1999), was ranked at the top with few kek1+ cells existing in the early scrib tumors and majority of cells being kek1+ in the late scrib tumors ( Figure 4B and Figure S11). Using a kek1-lacZ line (Musacchio and Perrimon, 1996), we confirmed that the late scrib tumors indeed harbored much more kek1+ cells than the early scrib tumors ( Figure 4C-E). ...
Article
Full-text available
Human tumors exhibit plasticity and evolving capacity over time. It is difficult to study the mechanisms of how tumors change over time in human patients, in particular during the early stages when a few oncogenic cells are barely detectable. Here, we used a Drosophila tumor model caused by loss of scribble (scrib), a highly conserved apicobasal cell polarity gene, to investigate the spatial-temporal dynamics of early tumorigenesis events. The fly scrib mutant tumors have been successfully used to model many aspects of tumorigenesis processes. However, it is still unknown whether Drosophila scrib mutant tumors exhibit plasticity and evolvability along the temporal axis. We found that scrib mutant tumors displayed different growth rates and cell cycle profiles over time, indicative of a growth arrest-to-proliferation transition as the scrib mutant tumors progress. Longitudinal bulk and single-cell transcriptomic analysis of scrib mutant tumors revealed that the MAPK pathway, including JNK and ERK signaling activities, showed quantitative changes over time. We found that high JNK signaling activity caused G2/M cell cycle arrest in early scrib mutant tumors. In addition, JNK signaling activity displayed a radial polarity with the JNKhigh cells located at the periphery of scrib mutant tumors, providing an inherent mechanism that leads to an overall decrease in JNK signaling activity over time. We also found that ERK signaling activity, in contrast to JNK activity, increased over time and promoted growth in late-stage scrib mutant tumors. Furthermore, high JNK signaling activity repressed ERK signaling activity in early scrib mutant tumors. Together, these data demonstrate that dynamic MAPK signaling activity, fueled by intratumor heterogeneity derived from tissue topological differences, drives a growth arrest-to-proliferation transition in scrib mutant tumors. This article has an associated First Person interview with the joint first authors of the paper.
... We next analyzed the transcriptional profile of wing disc cells subjected to a 3 day exposure to RasV12 expression and compared it with the profile of wildtype wing disc cells. RasV12-expressing cells were enriched in genes previously identified to be regulated by the canonical Ras/Raf/MAP kinase cascade ( Figure S1I; Table S1), such as the ETS domain transcription factors Ets21C and Pointed (Jin et al., 2015), the nuclear protein Phyllopod (Dickson et al., 1995), or the EGFR/ERK antagonists Sprouty (Casci et al., 1999), Argos (Golembo et al., 1996), and Kekkon-1 (Ghiglione et al., 1999). The transcriptome dataset thus contains genes known to contribute to mediating or modulating RTK/ERK signaling. ...
Article
Full-text available
Several oncogenes induce untimely entry into S phase and alter replication timing and progression, thereby generating replicative stress, a well-known source of genomic instability and a hallmark of cancer. Using an epithelial model in Drosophila, we show that the RAS oncogene, which triggers G1/S transition, induces DNA damage and, at the same time, silences the DNA damage response pathway. RAS compromises ATR-mediated phosphorylation of the histone variant H2Av and ATR-mediated cell-cycle arrest in G2 and blocks, through ERK, Dp53-dependent induction of cell death. We found that ERK is also activated in normal tissues by an exogenous source of damage and that this activation is necessary to dampen the pro-apoptotic role of Dp53. We exploit the pro-survival role of ERK activation upon endogenous and exogenous sources of DNA damage to present evidence that its genetic or chemical inhibition can be used as a therapeutic opportunity to selectively eliminate RAS-malignant tissues. : RAS oncoproteins are virtually “undruggable” cancer targets. Murcia et al. take advantage of the inherent induction of DNA damage and additional blockade of the DDR by the RAS oncogene to show that genetic or chemical inhibition of ERK coupled to radiation can selectively eliminate RAS-malignant tumors once they are formed. Keywords: Dp53, ERK, genomic instability, DNA damage, ATR, cancer, radiotherapy, cell death, malignancy
... Kekkon Sprouty and Yan for example are activated in some cells receiving EGF activation and act within these cells. Kekkon is a transmembrane protein that binds to the extracellular domain of EGFR and attenuates receptor dimerization (Ghiglione et al., 1999). Sprouty is an intracellular protein, which inhibits the Ras1/MAP kinase pathway. ...
Thesis
Full-text available
Das Nervensystem von Drosophila melanogaster entwickelt sich aus dem Neuroectoderm entlang der anterior-posterioren Axe des Embryos. Dieses Gewebe unterteilt sich in drei Expressionsdomänen, die von ventral nach dorsal durch Expression der drei Homöobox-Transkriptionsfaktoren vnd, ind und msh charakterisiert sind. Vnd und Ind wurden als notwendig und ausreichend beschrieben, um die Zellidentitäten ihrer jeweiligen Gewebe zu determinieren. Um zu untersuchen, wie diese Transkriptionsfaktoren agieren, um ihre jeweiligen Zelltypen hervorzubringen, habe ich die genomweiten Genexpressionsmuster dieser verwandten Gewebe in Wildtyp-Embryos und in einer vnd-Mutante bestimmt. Ich fand heraus, dass anstelle eines Gewebes, das der IC-Domäne ähnelt, die Vnd-Mutante ein Gewebe hervorbringt, das neuronale Charakteristika zum grossen Teil verloren hat. Ich habe gefunden, dass der Transkiptionsfaktor "eyeless" bei ektopischer Expression in der VC den Vnd-Nervensystemphänotyp phänokopiert und ein ähnliches DNA-Bindemuster aufweist wie Vnd. Unsere Daten lassen vermuten, das Vnd sowohl als direkter Aktivator von VC-spezifischen Genen als auch als direkter Repressor von nicht-neuronalen Genen wirkt. Um die zeitlich-örtliche Auflösung weiter zu erhöhen, habe ich mit Nikolaos Karaiskos kollaboriert, um Einzelzell-Transkriptome von Embryos einer einzigen embryonalen Stufe zu generieren und einen Genexpressions-Atlas des frühen Drosophila-Embryos mit Einzelzellauflösung zu erstellen der die nahezu genomweite Genexpression fast jeder Zelle zu rekapitulieren. Um die Nützlichkeit der Plattform zu demonstrieren, untersuchten wir Expressionsmuster von Genen, die an der Hippo-Signalkaskade beteiligt sind. Wir prognostizierten Hippo-Signalaktivität in bestimmten Bereichen des Embryos und zeigten, dass der Hippo-Signalweg die synchronisierte Zellteilung in diesen Bereichen unterbricht.
... The PI3K/Akt and AKT/mTOR pathways are critical for cellular proliferation, growth, survival, and mobility [48]. Dorso-ventral axis formation, during Drosophila embryogenesis, is required for the establishment of dorsoventral cell fates, determination of segmental identity, maintenance of amnioserosa and ventral neuroectodermal cells, germ band retraction, and production of cuticle [49]. MAPKs are serine-threonine protein kinases that play an important role in the regulation of many cellular processes including cell growth and proliferation, differentiation, and apoptosis [50]. ...
Article
Full-text available
The larval segment formation and secondary loss in echiurans is a special phenomenon, which is considered to be one of the important characteristics in the evolutionary relationship between the Echiura and Annelida. To better understand the molecular mechanism of this phenomenon, we revealed the larval transcriptome profile of the echiuran worm Urechis unicinctus using RNA-Seq technology. Twelve cDNA libraries of U. unicinctus larvae, late-trochophore (LT), early-segmentation larva (ES), segmentation larva (SL), and worm-shaped larva (WL) were constructed. Totally 243,381 unigenes were assembled with an average length of 1125 bp and N50 of 1836 bp, and 149,488 unigenes (61.42%) were annotated. We obtained 70,517 differentially expressed genes (DEGs) by pairwise comparison of the larval transcriptome data at different developmental stages and clustered them into 20 gene expression profiles using STEM software. Based on the typical profiles during the larval segment formation and secondary loss, eight signaling pathways were enriched, and five of which, mTOR, PI3K-AKT, TGF-β, MAPK, and Dorso-ventral axis formation signaling pathway, were proposed for the first time to be involved in the segment formation. Furthermore, we identified 119 unigenes related to the segment formation of annelids, arthropods, and chordates, in which 101 genes were identified in Drosophila and annelids. The function of most segment polarity gene homologs (hedgehog, wingless, engrailed, etc.) was conserved in echiurans, annelids, and arthropods based on their expression profiles, while the gap and pair-rule gene homologs were not. Finally, we verified that strong positive signals of Hedgehog were indeed located on the boundary of larval segments using immunofluorescence. Data in this study provide molecular evidence for the understanding of larval segment development in echiurans and may serve as a blueprint for segmented ancestors in future research.
... Since grk mRNA is a known orb regulatory target, one explanation for the repolarization defects is that the PFCs are not properly specified when orb The translational regulator, Orb, functions with Par proteins to polarize the Drosophila oocyte activity is compromised. To test this possibility we examined the expression of an EGFR dependent enhancer trap, kekkon-lacZ, that is activated in follicle cells by grk signaling [68][69][70][71][72]. As illustrated for two stage 7 egg chambers in S4A and S4B Fig we found that kekkon-lacZ expression in PFCs in orb mel /orb 343 egg chambers resembles that in control egg chambers. ...
Article
Full-text available
orb is a founding member of the CPEB family of translational regulators and is required at multiple steps during Drosophila oogenesis. Previous studies showed that orb is required during mid-oogenesis for the translation of the posterior/germline determinant oskar mRNA and the dorsal-ventral determinant gurken mRNA. Here, we report that orb also functions upstream of these axes determinants in the polarization of the microtubule network (MT). Prior to oskar and gurken translational activation, the oocyte MT network is repolarized. The MT organizing center at the oocyte posterior is disassembled, and a new MT network is established at the oocyte anterior. Repolarization depends upon cross-regulatory interactions between anterior (apical) and posterior (basal) Par proteins. We show that repolarization of the oocyte also requires orb and that orb is needed for the proper functioning of the Par proteins. orb interacts genetically with aPKC and cdc42 and in egg chambers compromised for orb activity, Par-1 and aPKC protein and aPKC mRNA are mislocalized. Moreover, like cdc42⁻, the defects in Par protein localization appear to be connected to abnormalities in the cortical actin cytoskeleton. These abnormalities also disrupt the localization of the spectraplakin Shot and the microtubule minus-end binding protein Patronin. These two proteins play a critical role in the repolarization of the MT network.
... Feedback regulators play a major role in fine-tuning these variables by attenuating or amplifying the signaling output. They can be already present and act prior to or immediately after receptor activation (early attenuators and amplifiers) (Haglund et al., 2003;Thien and Langdon, 2001) or can be transcriptionally induced by the pathways on which they eventually act (late attenuators and amplifiers) http (Table 1) Fiorini et al., 2002;Ghiglione et al., 1999;Golembo et al., 1996;Korsensky and Ron, 2016;Tsang and Dawid, 2004). ...
Article
Precise regulation of the amplitude and duration of receptor tyrosine kinase (RTK) signaling is critical for the execution of cellular programs and behaviors. Understanding these control mechanisms has important implications for the field of developmental biology, and in recent years, the question of how augmentation or attenuation of RTK signaling via feedback loops modulates development has become of increasing interest. RTK feedback regulation is also important for human disease research; for example, germline mutations in genes that encode RTK signaling pathway components cause numerous human congenital syndromes, and somatic alterations contribute to the pathogenesis of diseases such as cancers. In this review, we survey regulators of RTK signaling that tune receptor activity and intracellular transduction cascades, with a focus on the roles of these genes in the developing embryo. We detail the diverse inhibitory mechanisms utilized by negative feedback regulators that, when lost or perturbed, lead to aberrant increases in RTK signaling. We also discuss recent biochemical and genetic insights into positive regulators of RTK signaling and how these proteins function in tandem with negative regulators to guide embryonic development.
... This is thought to modulate kinase-signaling levels by TrkB [64]. Similarly, in Drosophila, Kek-5 is an inhibitor of BMP signaling [26] and Kek-1 an inhibitor of EGFR signaling [65]. Thus, each Kek could function as a specific inhibitor of different signaling pathways. ...
Article
Full-text available
Neurotrophism, structural plasticity, learning and long-term memory in mammals critically depend on neurotrophins binding Trk receptors to activate tyrosine kinase (TyrK) signaling, but Drosophila lacks full-length Trks, raising the question of how these processes occur in the fly. Paradoxically, truncated Trk isoforms lacking the TyrK predominate in the adult human brain, but whether they have neuronal functions independently of full-length Trks is unknown. Drosophila has TyrK-less Trk-family receptors, encoded by the kekkon (kek) genes, suggesting that evolutionarily conserved functions for this receptor class may exist. Here, we asked whether Keks function together with Drosophila neurotrophins (DNTs) at the larval glutamatergic neuromuscular junction (NMJ). We tested the eleven LRR and Ig-containing (LIG) proteins encoded in the Drosophila genome for expression in the central nervous system (CNS) and potential interaction with DNTs. Kek-6 is expressed in the CNS, interacts genetically with DNTs and can bind DNT2 in signaling assays and co-immunoprecipitations. Ligand binding is promiscuous, as Kek-6 can also bind DNT1, and Kek-2 and Kek-5 can also bind DNT2. In vivo, Kek-6 is found presynaptically in motoneurons, and DNT2 is produced by the muscle to function as a retrograde factor at the NMJ. Kek-6 and DNT2 regulate NMJ growth and synaptic structure. Evidence indicates that Kek-6 does not antagonise the alternative DNT2 receptor Toll-6. Instead, Kek-6 and Toll-6 interact physically, and together regulate structural synaptic plasticity and homeostasis. Using pull-down assays, we identified and validated CaMKII and VAP33A as intracellular partners of Kek-6, and show that they regulate NMJ growth and active zone formation downstream of DNT2 and Kek-6. The synaptic functions of Kek-6 could be evolutionarily conserved. This raises the intriguing possibility that a novel mechanism of structural synaptic plasticity involving truncated Trk-family receptors independently of TyrK signaling may also operate in the human brain.
... In addition to changes in the cis-regulatory region of br, which appears to be the key node in the appendage patterning network, it is important to keep in mind the mechanisms that rely on the intracellular modulation of inductive signals. In particular, GRK induces several negative feedback loops that modulate the signals sensed by the enhancers of the genes within the patterning network, changing the 2D patterns of br expression and affecting eggshell morphology (15,(48)(49)(50)(51)(52). Changing the induction thresholds of these negative feedback regulators provides another degree of flexibility for the evolution of eggshell patterning (18,28). ...
Article
Full-text available
A common path to the formation of complex 3D structures starts with an epithelial sheet that is patterned by inductive cues that control the spatiotemporal activities of transcription factors. These activities are then interpreted by the cis-regulatory regions of the genes involved in cell differentiation and tissue morphogenesis. Although this general strategy has been documented in multiple developmental contexts, the range of experimental models in which each of the steps can be examined in detail and evaluated in its effect on the final structure remains very limited. Studies of the Drosophila eggshell patterning provide unique insights into the multiscale mechanisms that connect gene regulation and 3D epithelial morphogenesis. Here we review the current understanding of this system, emphasizing how the recent identification of cis-regulatory regions of genes within the eggshell patterning network enables mechanistic analysis of its spatiotemporal dynamics and evolutionary diversification. It appears that cis-regulatory changes can account for only some aspects of the morphological diversity of Drosophila eggshells, such as the prominent differences in the number of the respiratory dorsal appendages. Other changes, such as the appearance of the respiratory eggshell ridges, are caused by changes in the spatial distribution of inductive signals. Both types of mechanisms are at play in this rapidly evolving system, which provides an excellent model of developmental patterning and morphogenesis.
... Because dAtg1 is required for wg expression in AiP, we tested if dAtg1 was also sufficient for expression of AiP markers including wg, dpp, and kekkon1 (kek), the latter being a marker of EGFR activity [51,52,[54][55][56]70]. However, while expression of hid in the DE ts > hid model is sufficient to induce wg, dpp, and kek expression (Additional file 4: Figure S4A-B' , D-E' , G-H'), expression of dAtg1 alone under the same conditions (DE ts > Atg1) is not (Additional file 4: Figure S4C, C' , F, F' , I, I'). ...
Article
Full-text available
BACKGROUND: ATG1 belongs to the Uncoordinated-51-like kinase protein family. Members of this family are best characterized for roles in macroautophagy and neuronal development. Apoptosis-induced proliferation (AiP) is a caspase-directed and JNK-dependent process which is involved in tissue repair and regeneration after massive stress-induced apoptotic cell loss. Under certain conditions, AiP can cause tissue overgrowth with implications for cancer. RESULTS: Here, we show that Atg1 in Drosophila (dAtg1) has a previously unrecognized function for both regenerative and overgrowth-promoting AiP in eye and wing imaginal discs. dAtg1 acts genetically downstream of and is transcriptionally induced by JNK activity, and it is required for JNK-dependent production of mitogens such as Wingless for AiP. Interestingly, this function of dAtg1 in AiP is independent of its roles in autophagy and in neuronal development. CONCLUSION: In addition to a role of dAtg1 in autophagy and neuronal development, we report a third function of dAtg1 for AiP.
... Argos is a secreted protein that sequesters the Drosophila EGFR (dEGFR) ligand Spitz, functioning as a ligand sink (Klein et al. 2004). Kekkon-1 (Kek1), a transmembrane glycoprotein in Drosophila, was found to modulate dEGFR signaling through an inhibitory feedback loop (Ghiglione et al. 1999). Genetic studies suggest that the extracellular region of Kek1 is required for its dEGFR inhibitory effect, but it is unclear whether there is direct interaction between dEGFR and Kek1 ( Figure 5.3). ...
Article
Receptor tyrosine kinases (RTKs) are a family of 58 transmembrane proteins in humans that play crucial roles in many biological processes and diseases. Different RTKs utilize subtly (but importantly) distinct molecular mechanisms for transmembrane signaling, and understanding these differences is crucial for devising new ways to intervene pharmacologically when aberrant RTK signaling causes cancer and other diseases. In this thesis, I focus on three RTK families: the ALK/LTK family, the Wnt-binding RTKs, and the EGF receptor – where I concentrate on efforts to understand its C-terminal regulatory region. My studies of ALK, for anaplastic lymphoma kinase, were motivated by the fact that this RTK sub-group has a unique domain architecture in its extracellular region. Little is known about the mechanisms of ligand binding to – and activation of – ALK, and the nature of its ligand(s) is(are) still not completely clear. Using biochemical, biophysical and structural biology approaches, I characterized the low-resolution structure of the ALK extracellular region. I further identified the binding mode of ALK binding to heparin, a recently discovered modulatory ligand for ALK. Based on a low-resolution structural analysis of ALK/heparin complex, I propose a model for ligand-induced ALK dimerization and activation. Ryk is one of the five RTKs that are now known to be Wnt receptors. In this thesis, I studied the Drosophila homolog of Ryk, Derailed (Drl), and its binding to ligand DWnt5. We were able to express and purify milligrams of active DWnt5 – thus overcoming a major obstacle in this field. We further characterized Drl/DWnt5 interactions. Using hydrogen/deuterium exchange approaches, I identified the DWnt5-binding interface on Drl. My efforts to understand the molecular mechanisms of Drl/DWnt5 binding using experimental and computational approaches suggest that DWnt5 may interact with Drl through a binding mode that differs from Wnt binding to other receptors. Across the RTK family, many receptors contain a long carboxy-terminal tail (C-tail) that harbors autophosphorylation sites for docking of downstream signaling molecules. This region is generally considered to be intrinsically disordered. I studied the dynamics of the EGFR C-tail, and showed that it is highly unstructured – but contains some somewhat ‘structured’ regions. I also showed that phosphorylation of the EGFR C-tail promotes receptor dimerization. Using hydrogen exchange, I identified possible C-tail docking sites on the kinase domain that may be responsible for this effect. I also studied binding of downstream SH2 domain-containing molecules to the EGFR C-tail, with results that indicate that not all features of SH2 domain binding to the C-tail can be recapitulated by simple phosphopeptides; binding of SH2 domains to the C-tail exhibits binding affinities and stoichiometries that are not captured by simple peptide-level studies. Moreover, my binding competition assays suggest that there may be cooperativity in binding of multiple SH2 domains to a single phosphorylated C-tail.
... Because dAtg1 is required for wg expression in AiP, we tested if dAtg1 was also sufficient for expression of AiP markers including wg, dpp, and kekkon1 (kek), the latter being a marker of EGFR activity [51,52,[54][55][56]70]. However, while expression of hid in the DE ts > hid model is sufficient to induce wg, dpp, and kek expression (Additional file 4: Figure S4A-B' , D-E' , G-H'), expression of dAtg1 alone under the same conditions (DE ts > Atg1) is not (Additional file 4: Figure S4C, C' , F, F' , I, I'). ...
Article
Full-text available
BackgroundATG1 belongs to the Uncoordinated-51-like kinase protein family. Members of this family are best characterized for roles in macroautophagy and neuronal development. Apoptosis-induced proliferation (AiP) is a caspase-directed and JNK-dependent process which is involved in tissue repair and regeneration after massive stress-induced apoptotic cell loss. Under certain conditions, AiP can cause tissue overgrowth with implications for cancer. ResultsHere, we show that Atg1 in Drosophila (dAtg1) has a previously unrecognized function for both regenerative and overgrowth-promoting AiP in eye and wing imaginal discs. dAtg1 acts genetically downstream of and is transcriptionally induced by JNK activity, and it is required for JNK-dependent production of mitogens such as Wingless for AiP. Interestingly, this function of dAtg1 in AiP is independent of its roles in autophagy and in neuronal development. Conclusion In addition to a role of dAtg1 in autophagy and neuronal development, we report a third function of dAtg1 for AiP.
... For instance, rho-1 and step are target genes forming positive feedback loops in the Drosophila egg and wing disc, respectively [66,102]. EGFR activity also controls the expression of many inhibitors of the pathway, such as Argos [103,104], Kekkon [76,105], Sprouty [81] and Fas2 [80]. In contrast, some inhibitors like Ed do not appear to be regulated transcriptionally by EGFR signalling. ...
Article
Full-text available
EGFR signalling is a well-conserved signalling pathway playing major roles during development and cancers. This review explores what studying the EGFR pathway during Drosophila eye development has taught us in terms of the diversity of its regulatory mechanisms. This model system has allowed the identification of numerous positive and negative regulators acting at specific time and place, thus participating to the tight control of signalling. EGFR signalling regulation is achieved by a variety of mechanisms, including the control of ligand processing, the availability of the receptor itself and the transduction of the cascade in the cytoplasm. Ultimately, the transcriptional responses contribute to the establishment of positive and negative feedback loops. The combination of these multiple mechanisms employed to regulate the EGFR pathway leads to specific cellular outcomes involved in functions as diverse as the acquisition of cell fate, proliferation, survival, adherens junction remodelling and morphogenesis.
... 4,10,17,18 The gene is the human homolog of mouse Lrig1 28 and encodes a transmembrane protein which, in the extracellular part, shows structural similarities to the drosophila cell surface protein Kekkon-1. 29 The extracellular part of Kekkon-1 functions in drosophila as an inhibitor of EGFR- 30 As far as we know, the case is similar in humans, though more complicated. ...
Article
Full-text available
Somatic mutations, which are associated with a certain rate of response to targeted therapies, are ubiquitously found in human non-small cell lung cancer (NSCLC). However, it is largely unknown which group of patients may benefit from the respective treatments targeting different somatic mutations. Therefore, more effective prognostic and predictive markers are desperately needed for the treatment of NSCLC harboring different somatic mutations. The leucine-rich repeats and immunoglobulin-like domains (LRIG)-1 is a tumor suppressor gene that belongs to the LRIG family. LRIG1 expression has prognostic significance in various human cancers. In this study, we first used the quantitative polymerase chain reaction (qPCR) and immunohistochemical analysis of 36 and 182 NSCLC patient tissues to analyze the LRIG1 expression respectively. To investigate the prognostic value of LRIG1 in NSCLC, we examined the correlation between clinical features and overall survival (OS) with Cox proportional hazard regression. We also compared the sensitivity and specificity of LRIG1 in NSCLC prognosis by logistic regression to further evaluate the prognostic efficiency of LRIG1 in NSCLC. We found that the LRIG1 expression was associated with pathological type, differentiation status, and stage of NSCLC. The result showed that LRIG1 was an independent prognostic factor for OS of NSCLC patients. LRIG1 in combination with other clinicopathological risk factors was a stronger prognostic model than clinicopathological risk factors alone. Thus, the LRIG1 expression potentially offered a significant clinical value in directing personal treatment for NSCLC patients.
... Sprouty est activée par la voie EGFR mais doit aussiêtre phosphorylée indépendamment de la voie EGFR, ce qui permet certainement de gagner en spécificité(Hanafusa et al. 2002). L'inhibiteur Kekkon est une protéine transmembranaire qui forme des hétérodimères avec le récepteur EGFR(Ghiglione et al. 1999)(Fig. 1.17).Enfin, uneétape de régulation négative se produit au niveau de la cellule envoyant le signal en limitant la production du ligand. ...
Article
In metazoan, the successful development of many organs requires the elimination of supernumerary cells by apoptosis. For example, the elimination of about two thousand interommatidial cells (IOCs) during Drosophila eye development allows the precise rearrangement of ommatidia in a perfect hexagonal array. Maximal apoptosis occurs during pupal life and the remaining IOCs differentiate into secondary and tertiary pigment cells. The precise removal of unwanted IOCs requires coordinated activation of Notch (pro-death) and EGF (pro-survival) pathways. IOCs undergoing apoptosis express the IAP inhibitor Hid, which leads to the activation of initiator and effector caspases. However, the mechanisms that coordinate the death and survival pathways for timed and precise IOC removal are poorly understood.Here, we report that spen encodes a nuclear protein expressed in the pupal eye that is required for IOC survival. We showed that the inhibition of spen, by either RNAi or in spen mutant clones resulted in disorganized ommatidia with missing IOCs. Moreover, overexpression of spen leads to extra IOCs. These results indicate that spen expression promotes IOC survival during eye development. Importantly blocking apoptosis prevents the loss of IOC in a spen mutant retina. Spen is a protein known to be ubiquitous in tissue during development. Indeed, we have shown using an enhancer trap line that spen is expressed in all the cells in the eye pupal disk. To better understand where spen is acting from in this tissue to regulate cell death, we performed a clonal analysis. We found that the inactivation of spen in the cone cells was causing the loss of IOC, indicating that spen is required non-autonomously in cone cell for IOC survival. In parallel we have shown that the inactivation of spen was disrupting eye bristles morphology. Even if studies discuss the role of bristles in the regulation of developmental apoptosis in this context, our clonal analysis excluded this possibility. Furthermore, we found that spitz, the EGFR ligand, accumulate in cone cells upon spen inactivation. Our current hypothesis is that spen is likely to be required for the release of Spitz from the cone cells in order to active the survival signaling pathway EGFR in the IOCs. Also, we examined the protective role of spen in a chemical model of Parkinson disease (paraquat treatment). We showed that the glial expression of spen is protective in this context, which suggest against that spen acts non-autonomously. Interestingly we found that the inactivation of spen in glia downregulates the Notch signaling pathway. Spen is likely to be a key factor integrating cues from different signaling pathways to promote cell survival.
... This may explain why simply increasing EGF receptor activity does not normally cause R8 twinning. For example, mutations of other negative regulators of Egfr (argos, sprouty, kekkon I) do not show R8 twinning, despite raising levels of Egfr signalling (Casci et al., 1999;Ghiglione et al., 1999;Baonza et al., 2001;Yang and Baker, 2001). Neither does increased expression of Spi ligand (this paper). ...
Article
EGF receptor signalling plays diverse inductive roles during development. To achieve this, its activity must be carefully regulated in a variety of ways to control the time, pattern, intensity and duration of signalling. We show that the cell surface protein Echinoid is required to moderate Egfr signalling during R8 photoreceptor selection by the proneural gene atonal during Drosophila eye development. In echinoid mutants, Egfr signalling is increased during R8 formation, and this causes isolated R8 cells to be replaced by groups of two or three cells. This mutant phenotype resembles the normal inductive function of Egfr in other developmental contexts, particularly during atonal-controlled neural recruitment of chordotonal sense organ precursors. We suggest that echinoid acts to prevent a similar inductive outcome of Egfr signalling during R8 selection.
... There are at least three types of follicle cells that can be distinguished by their gene expression pattern, by the eggshell structures they secrete and by the function they fulfil Introduction for embryonic axis induction along the DV axis of the egg chamber. These are the dorsal cells that express kekkon (kek1) (Ghiglione et al., 1999) and are responsible for dorsal chorion structures including the dorsal appendages, the ventral cells that express pipe (pip) and contribute the cues for the embryonic DV axis (Sen et al., 1998) (Nilson and Schupbach, 1998) (Fig.4B) and the lateral cells that neither express kek1 nor pip. Grk signaling induces secondary signaling cascades that define these expression domains, as in the patterning of the DAs. ...
... Therefore, we looked in fc for activity readout of these pathways when germline growth was genetically manipulated. EGFR-ras transcriptional readout is already described as expressed in specific populations of fc and at specific stages, suggesting that it cannot be generally controlled in all cells by the coordination process (Ghiglione et al., 1999). The expression of Expanded-lacZ, a transgene used as a reporter for Hippo pathway activity, is not modified in a follicle with a germline clone for Akt compared with a WT follicle, confirming that this pathway is not involved in the coordination between the germline and the soma ( Figure 4A). ...
Article
Full-text available
Organs often need to coordinate the growth of distinct tissues during their development. Here, we analyzed the coordination between germline cysts and the surrounding follicular epithelium during Drosophila oogenesis. Genetic manipulations of the growth rate of both germline and somatic cells influence the growth of the other tissue accordingly. Growth coordination is therefore ensured by a precise, two-way, intrinsic communication. This coordination tends to maintain constant epithelial cell shape, ensuring tissue homeostasis. Moreover, this intrinsic growth coordination mechanism also provides cell differentiation synchronization. Among growth regulators, PI3-kinase and TORC1 also influence differentiation timing cell-autonomously. However, these two pathways are not regulated by the growth of the adjacent tissue, indicating that their function reflects an extrinsic and systemic influence. Altogether, our results reveal an integrated and particularly robust mechanism ensuring the spatial and temporal coordination of tissue size, cell size, and cell differentiation for the proper development of two adjacent tissues.
... Sty is an intracellular inhibitor, which inhibits Ras signaling, thereby inhibiting EGF signaling (46). Kek-1 is a transmembrane protein that interacts directly with the EGF receptor to inhibit ligand binding (47,48). ...
... Argos acts in an autocrine/paracrine fashion by binding and sequestering the DERactivating ligand Spitz, thereby inhibiting Spitz-induced receptor signaling (Klein et al., 2004). Kekkon-1 is a transmembrane protein, which extracellular domain physically interacts with DER and thereby inhibits ligand binding and receptor dimerization (Ghiglione et al., 1999). Sprouty, finally, is a promiscuous inhibitor of several RTK, which was originally cloned as a negative feedback inhibitor of the dFGF receptor (Hacohen et al., 1998). ...
... 47 These observations raised the question of how pipe expression is repressed in the lateral regions of the follicular epithelium in the absence of Torpedo activation. Despite the failure to visualize phospho-MAP kinase in these regions, subsequent studies utilizing kekkon, a target of Torpedo signaling, 50 indicated that Gurken functions as a long-range morphogen that activates Torpedo all along the DV axis of the follicle layer in a graded manner. kekkon is normally expressed only in dorsal follicle cells. ...
Article
Unlabelled: The pathway that generates the dorsal-ventral (DV) axis of the Drosophila embryo has been the subject of intense investigation over the previous three decades. The initial asymmetric signal originates during oogenesis by the movement of the oocyte nucleus to an anterior corner of the oocyte, which establishes DV polarity within the follicle through signaling between Gurken, the Drosophila Transforming Growth Factor (TGF)-α homologue secreted from the oocyte, and the Drosophila Epidermal Growth Factor Receptor (EGFR) that is expressed by the follicular epithelium cells that envelop the oocyte. Follicle cells that are not exposed to Gurken follow a ventral fate and express Pipe, a sulfotransferase that enzymatically modifies components of the inner vitelline membrane layer of the eggshell, thereby transferring DV spatial information from the follicle to the egg. These ventrally sulfated eggshell proteins comprise a localized cue that directs the ventrally restricted formation of the active Spätzle ligand within the perivitelline space between the eggshell and the embryonic membrane. Spätzle activates Toll, a transmembrane receptor in the embryonic membrane. Transmission of the Toll signal into the embryo leads to the formation of a ventral-to-dorsal gradient of the transcription factor Dorsal within the nuclei of the syncytial blastoderm stage embryo. Dorsal controls the spatially specific expression of a large constellation of zygotic target genes, the Dorsal gene regulatory network, along the embryonic DV circumference. This article reviews classic studies and integrates them with the details of more recent work that has advanced our understanding of the complex pathway that establishes Drosophila embryo DV polarity. For further resources related to this article, please visit the WIREs website. Conflict of interest: The authors have declared no conflicts of interest for this article.
Article
Full-text available
The rapid transmission of action potentials is an important ability that enables efficient communication within the nervous system. Glial cells influence conduction velocity along axons by regulating the radial axonal diameter, providing electrical insulation as well as affecting the distribution of voltage-gated ion channels. Differentiation of these wrapping glial cells requires a complex set of neuron–glia interactions involving three basic mechanistic features. The glia must recognize the axon, grow around it, and eventually arrest its growth to form single or multiple axon wraps. This likely depends on the integration of numerous evolutionary conserved signaling and adhesion systems. Here, we summarize the mechanisms and underlying signaling pathways that control glial wrapping in Drosophila and compare those to the mechanisms that control glial differentiation in mammals. This analysis shows that Drosophila is a beneficial model to study the development of even complex structures like myelin.
Article
Leucine-rich repeat and immunoglobulin domain containing protein (LRR-IG) family is an important class of immune molecules in invertebrates. Herein, a novel LRR-IG, named as EsLRR-IG5, was identified from Eriocheir sinensis. It contained typical structures of LRR-IG including an N-terminal LRR region and three IG domains. EsLRR-IG5 was ubiquitously expressed in all the tested tissues, and its transcriptional levels increased after being challenged with Staphylococcus aureus and Vibrio parahaemolyticus. Recombinant proteins of LRR and IG domains from the EsLRR-IG5 (named as rEsLRR5 and rEsIG5) were successfully obtained. rEsLRR5 and rEsIG5 could bind to both gram-positive bacteria and gram-negative bacteria as well as lipopolysaccharide (LPS) and peptidoglycan (PGN). Moreover, rEsLRR5 and rEsIG5 exhibited antibacterial activities against V. parahaemolyticus and V. alginolyticus and displayed bacterial agglutination activities against S. aureus, Corynebacterium glutamicum, Micrococcus lysodeikticus, V. parahaemolyticus and V. alginolyticus. The scanning electron microscopy (SEM) observation revealed that the membrane integrity of V. parahaemolyticus and V. alginolyticus was destroyed by rEsLRR5 and rEsIG5, which may lead to the leakage of cell contents and death. This study provided clues for further studies on the immune defense mechanism mediated by LRR-IG in crustaceans and provided candidate antibacterial agents for prevention and control of diseases in aquaculture.
Article
Full-text available
The Epidermal Growth Factor Receptor (EGFR) signaling pathway plays a critical role in regulating tissue patterning. Drosophila EGFR signaling achieves specificity through multiple ligands and feedback loops to finetune signaling outcomes spatiotemporally. The principal Drosophila EGF ligand, cleaved Spitz, and the negative feedback regulator, Argos are diffusible and can act both in a cell autonomous and non-autonomous manner. The expression dose of Spitz and Argos early in photoreceptor cell fate determination has been shown to be critical in patterning the Drosophila eye, but the exact identity of the cells expressing these genes in the larval eye disc has been elusive. Using single molecule RNA Fluorescence in situ Hybridization (smFISH), we reveal an intriguing differential expression of spitz and argos mRNA in the Drosophila third instar eye imaginal disc indicative of directional non-autonomous EGFR signaling. By genetically tuning EGFR signaling, we show that rather than absolute levels of expression, the ratio of expression of spitz-to-argos to be a critical determinant of the final adult eye phenotype. Proximate effects on EGFR signaling in terms of cell cycle and differentiation markers are affected differently in the different perturbations. Proper ommatidial patterning is robust to thresholds around a tightly maintained wildtype spitz-to-argos ratio, and breaks down beyond. This provides a powerful instance of developmental buffering against gene expression fluctuations.
Article
Full-text available
High levels of microvessel density (MVD) indicate poor prognosis in patients with malignant glioma. Leucine-rich repeats and immunoglobulin-like domains (LRIG) 3, a potential tumor suppressor, plays an important role in tumor progression and may serve as a biomarker in many human cancers. However, its role and underlying mechanism of action in glioma angiogenesis remain unclear. In the present study, we used loss- and gain-of-function assays to show that LRIG3 significantly suppressed glioma-induced angiogenesis, both in vitro and in vivo. Mechanistically, LRIG3 inhibited activation of the PI3K/AKT signaling pathway, downregulating vascular endothelial growth factor A (VEGFA) in glioma cells, thereby inhibiting angiogenesis. Notably, LRIG3 had a significant negative correlation with VEGFA expression in glioma tissues. Taken together, our results suggest that LRIG3 is a novel regulator of glioma angiogenesis and may be a promising option for developing anti-angiogenic therapy.
Article
Full-text available
LRIG1, leucine-rich repeats and immunoglobulin-like domains protein 1, was discovered more than 20 years ago and has been shown to be downregulated or lost, and to function as a tumor suppressor in several cancers. Another well-reported biological function of LRIG1 is to regulate and help enforce the quiescence of adult stem cells (SCs). In both contexts, LRIG1 regulates SC quiescence and represses tumor growth via, primarily, antagonizing the expression and activities of ERBB and other receptor tyrosine kinases (RTKs). We have recently reported that in treatment-naïve human prostate cancer (PCa), LRIG1 is primarily regulated by androgen receptor (AR) and is prominently overexpressed. In castration-resistant PCa (CRPC), both LRIG1 and AR expression becomes heterogeneous and, frequently, discordant. Importantly, in both androgen-dependent PCa and CRPC models, LRIG1 exhibits tumor-suppressive functions. Moreover, LRIG1 induction inhibits the growth of pre-established AR⁺ and AR- PCa. Here, upon a brief introduction of the LRIG1 and the LRIG family, we provide an updated overview on LRIG1 functions in regulating SC quiescence and repressing tumor development. We further highlight the expression, regulation and functions of LRIG1 in treatment-naïve PCa and CRPC. We conclude by offering the perspectives of identifying novel cancer-specific LRIG1-interacting signaling partners and developing LRIG1-based anti-cancer therapeutics and diagnostic/prognostic biomarkers.
Preprint
Full-text available
To understand the brain, molecular details need to be overlaid onto neural wiring diagrams so that synaptic mode, neuromodulation and critical signaling operations can be considered. Single-cell transcriptomics provide a unique opportunity to collect this information. Here we present an initial analysis of thousands of individual cells from Drosophila midbrain, that were acquired using Drop-Seq. A number of approaches permitted the assignment of transcriptional profiles to several major brain regions and cell-types. Expression of biosynthetic enzymes and reuptake mechanisms allows all the neurons to be typed according to the neurotransmitter or neuromodulator that they produce and presumably release. Some neuropeptides are preferentially co-expressed in neurons using a particular fast-acting transmitter, or monoamine. Neuromodulatory and neurotransmitter receptor subunit expression illustrates the potential of these molecules in generating complexity in neural circuit function. This cell atlas dataset provides an important resource to link molecular operations to brain regions and complex neural processes.
Preprint
Full-text available
Models of ‘plasticity-first’ evolution are attractive because they explain the rapid evolution of new complex adaptations. Nevertheless, it is unclear whether plasticity can still facilitate rapid evolution when diverging populations are connected by gene flow. Here we show how plasticity has generated adaptive divergence in fecundity in wild populations of burying beetles Nicrophorus vespilloides , which are still connected by gene flow, which occupy distinct Cambridgeshire woodlands that are just 2.5km apart and which diverged from a common ancestral population c. 1000-4000 years ago. We show that adaptive divergence is due to the coupling of an evolved increase in the elevation of the reaction norm linking clutch size to carrion size (i.e. genetic accommodation) with plastic secondary elimination of surplus offspring. Working in combination, these two processes have facilitated rapid adaptation to fine-scale environmental differences, despite ongoing gene flow.
Article
Genomes of metazoan organisms possess a large number of genes encoding cell surface and secreted (CSS) proteins that carry out crucial functions in cell adhesion and communication, signal transduction, extracellular matrix establishment, nutrient digestion and uptake, immunity, and developmental processes. We developed the FlyXCDB database (http://prodata.swmed.edu/FlyXCDB) that provides a comprehensive resource to investigate extracellular (XC) domains in CSS proteins of Drosophila melanogaster, the most studied insect model organism in various aspects of animal biology. More than three hundred Drosophila XC domains were discovered in Drosophila CSS proteins encoded by over 2,500 genes through analyses of computational predictions of signal peptide, transmembrane (TM) segment, and GPI-anchor signal sequence, profile-based sequence similarity searches, gene ontology, and literature. These domains were classified into six classes mainly based on their molecular functions, including protein-protein interactions (class P), signaling molecules (class S), binding of non-protein molecules or groups (class B), enzyme homologs (class E), enzyme regulation and inhibition (class R), and unknown molecular function (class U). Main cellular functions such as cell adhesion, cell signaling and extracellular matrix composition were described for the most abundant domains in each functional class. We assigned cell membrane topology categories (E - secreted; S - type I/III single-pass TM; T - type II single-pass TM; M - multi-pass TM; and G - GPI-anchored) to the products of genes with XC domains and investigated their regulation by mechanisms such as alternative splicing and stop codon readthrough.
Article
Full-text available
To understand the brain, molecular details need to be overlaid onto neural wiring diagrams so that synaptic mode, neuromodulation and critical signaling operations can be considered. Single-cell transcriptomics provide a unique opportunity to collect this information. Here we present an initial analysis of thousands of individual cells from Drosophila midbrain, that were acquired using Drop-Seq. A number of approaches permitted the assignment of transcriptional profiles to several major brain regions and cell-types. Expression of biosynthetic enzymes and reuptake mechanisms allows all the neurons to be typed according to the neurotransmitter or neuromodulator that they produce and presumably release. Some neuropeptides are preferentially co-expressed in neurons using a particular fast-acting transmitter, or monoamine. Neuromodulatory and neurotransmitter receptor subunit expression illustrates the potential of these molecules in generating complexity in neural circuit function. This cell atlas dataset provides an important resource to link molecular operations to brain regions and complex neural processes.
Article
Understanding the mechanisms driving tissue and organ formation requires knowledge across scales. How do signaling pathways specify distinct tissue types? How does the patterning system control morphogenesis? How do these processes evolve? The Drosophila egg chamber, where EGF and BMP signaling intersect to specify unique cell types that construct epithelial tubes for specialized eggshell structures, has provided a tractable system to ask these questions. Work there has elucidated connections between scales of development, including across evolutionary scales, and fostered the development of quantitative modeling tools. These tools and general principles can be applied to the understanding of other developmental processes across organisms.
Article
Full-text available
The specification of distinct neural cell types in central nervous system development crucially depends on positional cues conferred to neural stem cells in the neuroectoderm. Here, we investigate the regulation and function of the epidermal growth factor receptor (EGFR) signalling pathway in early development of the Drosophila brain. We find that localized EGFR signalling in the brain neuroectoderm relies on a neuromere-specific deployment of activating (Spitz, Vein) and inhibiting (Argos) ligands. Activated EGFR controls the spatially restricted expression of all dorsoventral (DV) patterning genes in a gene- and neuromere-specific manner. Further, we reveal a novel role of DV genes—ventral nervous system defective (vnd), intermediate neuroblast defective (ind), Nkx6—in regulating the expression of vein and argos, which feed back on EGFR, indicating that EGFR signalling stands not strictly atop the DV patterning genes. Within this network of genetic interactions, Vnd acts as a positive EGFR feedback regulator. Further, we show that EGFR signalling becomes dependent on single-minded-expressing midline cells in the posterior brain (tritocerebrum), but remains midline-independent in the anterior brain (deuto- and protocerebrum). Finally, we demonstrate that activated EGFR controls the proper formation of brain neuroblasts by regulating the number, survival and proneural gene expression of neuroectodermal progenitor cells. These data demonstrate that EGFR signalling is crucially important for patterning and early neurogenesis of the brain.
Chapter
In 1997 we wrote a review entitled "A thousand and one roles for the Drosophila epidermal growth factor (EGF) receptor (DER/EGFR)." We are not there yet in terms of the number of developmental roles assigned to this receptor in Drosophila. Nevertheless, DER has certainly emerged as one of the key players in development, since it is used repeatedly to direct cell fate choices, cell division, cell survival, and migration. A battery of activating ligands and an inhibitory ligand achieves this versatility. For the ligands that are produced as membrane-bound precursors, trafficking and processing are the key regulatory steps, determining the eventual temporal and spatial pattern of receptor activation. In most cases DER is activated at a short range, in the cells adjacent to the ones producing the active ligand. This activation dictates a binary choice. In some instances DER is also activated over a longer range, and multiple cell fate choices may be induced, according to its level of activation. A battery of negative feedback loops assures the limited range of DER induction. The distinct responses to DER activation in the different tissues depend upon combinatorial interactions with other signaling pathways and tissue-specific factors, at the level of target-gene regulation.
Article
Receptor tyrosine kinases were first isolated from the Drosophila genome in 1985 (Livneh et al. Cell 40(3):599–607, 1985; Wadsworth et al. Nature 314(6007):178–80, 1985), shortly after the isolation of the vertebrate receptors. Following the initial structural studies of these receptors, additional components in their signaling pathways were identified. Genetic epistasis studies have aided further in determining the linear order of signaling components within the pathway. Importantly, in genetically tractable organisms like Drosophila and C. elegans, the different roles of each receptor during development were deciphered. This chapter focuses on the insights obtained by studying RTKs in invertebrate organisms. One important observation is that RTK pathways guiding developmental decisions appear to be linear, rather than branched. A second observation is that there is no temporal overlap between distinct RTK pathways within the same cells. Since the activation of MAPK represents a common node to all RTK cascades, it was possible to examine this notion directly. Antibodies that specifically recognize the active, double phosphorylated form of MAPK allow to trace immunohistochemically the dynamic pattern of activation by all RTKs (Gabay et al. Development 124(18):3535–41, 1997). Each aspect of this pattern could be completely removed in a genetic background that specifically eliminates signaling by only one RTK pathway. This implies that the activation of MAPK per se does not provide information on the identity of the activated receptor, and it is the time and tissue context of MAPK activation that defines the target genes that will be induced. The time and place of the activation of each RTK pathway are tightly regulated by the expression of the respective ligands or ligand-processing components. The range of activation is defined by the level of ligand that is secreted and by negative feedback circuits that restrict the range of pathway stimulation.
Article
Regulation of epidermal growth factor receptor (EGFR) signaling requires the concerted action of both positive and negative factors. While the existence of numerous molecules that stimulate EGFR activity has been well documented, direct biological inhibitors appear to be more limited in number and phylogenetic distribution. Kekkon1 (Kek1) represents one such inhibitor. Kek1 was initially identified in Drosophila melanogaster and appears to be absent from vertebrates and the invertebrate Caenorhabditis. To further investigate Kek1's function and evolution, we identified kek1 orthologs within dipterans. In D. melanogaster, kek1 is a transcriptional target of EGFR signaling during oogenesis, where it acts to attenuate receptor activity through an inhibitory feedback loop. The extracellular and transmembrane portion of Kek1 is sufficient for its inhibitory activity in D. melanogaster. Consistent with conservation of its role in EGFR signaling, interspecies comparisons indicate a high degree of identity throughout these regions. During formation of the dorsal-ventral axis Kek1 is expressed in dorsal follicle cells in a pattern that reflects the profile of receptor activation. D. virilis Kek1 (DvKek1) is also expressed dynamically in the dorsal follicle cells, supporting a conserved role in EGFR signaling. Confirming this, biochemical and transgenic assays indicate that DvKek1 is functionally interchangeable with DmKek1. Strikingly, we find that the cytoplasmic domain contains a region with the highest degree of conservation, which we have implicated in EGFR inhibition and dubbed the Kek tail (KT) box.
Article
In Drosophila, signaling by the epidermal growth factor receptor (EGFR) is required for a diverse array of developmental decisions. Essential to these decisions is the precise regulation of the receptor's activity by both stimulatory and inhibitory molecules. To better understand the regulation of EGFR activity we investigated inhibition of EGFR by the transmembrane protein Kekkon1 (Kek1). Kek1 encodes a molecule containing leucine-rich repeats (LRR) and an immunoglobulin (Ig) domain and is the founding member of the Drosophila Kekkon family. Here we demonstrate with a series of Kek1-Kek2 chimeras that while the LRRs suffice for EGFR binding, inhibition in vivo requires the Kek1 juxta/transmembrane region. We demonstrate directly, and using a series of Kek1-EGFR chimeras, that Kek1 is not a phosphorylation substrate for the receptor in vivo. In addition, we show that EGFR inhibition is unique to Kek1 among Kek family members and that this function is not ligand or tissue specific. Finally, we have identified a unique class of EGFR alleles that specifically disrupt Kek1 binding and inhibition, but preserve receptor activation. Interestingly, these alleles map to domain V of the Drosophila EGFR, a region absent from the vertebrate receptors. Together, our results support a model in which the LRRs of Kek1 in conjunction with its juxta/transmembrane region direct association and inhibition of the Drosophila EGFR through interactions with receptor domain V.
Article
We have recently identified and cloned the human LRIG1 gene (formerly LIG1). LRIG1 is a predicted integral membrane protein with a domain organization reminiscent of the Drosophila epidermal growth factor (EGF)-receptor antagonist Kekkon-1. We have searched for additional members of the human LRIG family and identified LRIG2 (KIAA0806). The LRIG2 gene was localized to chromosome 1p13 and had an open reading frame of 1065 amino acids. The LRIG2 protein was predicted to have the same domain organization as LRIG1 with a signal peptide, an extracellular part containing15 leucine-rich repeats and three immunoglobulin-like domains, a transmembrane domain, and a cytoplasmic tail. The LRIG2 amino acid sequence was 47% identical to human LRIG1 and mouse Lrig1 (also known as Lig-1). Northern blotting and RT-PCR revealed LRIG2 transcripts in all tissues analyzed. Quantitative real-time RT-PCR showed the most prominent RNA expression in skin, uterus, ovary, kidney, brain, small intestine, adrenal gland, and stomach. Immunoblotting of COS-7 cell lysates demonstrated that heterologously expressed human LRIG2 had an apparent molecular weight of 132 kDa under reducing gel-running conditions. N-glycosidase F treatment resulted in a reduction of the apparent molecular weight to 107 kDa, showing that LRIG2 was a glycoprotein carrying N-linked oligosaccharides. Cell surface biotinylation experiments and confocal fluorescence laser microscopy demonstrated expression of LRIG2 both at the cell surface and in the cytoplasm. LRIG2 was detected in tissue lysates from stomach, prostate, lung, and fetal brain by immunoblotting. In conclusion, LRIG2 was found to be a glycoprotein which was encoded by a gene on human chromosome 1p13 and its mRNA was present in all tissues analyzed.
Article
Tissue homeostasis, accomplished through the self-renewal and differentiation of resident stem cells, is critical for the maintenance of adult tissues throughout an animal's lifetime. Adult Drosophila Malpighian tubules (MTs or fly kidney) are maintained by renal and nephric stem cells (RNSCs) via self-renewing divisions, however, it is unclear how RNSC proliferation and differentiation are regulated. Here we show that EGFR/MAPK signaling is dispensable for RNSC maintenance, but required for RNSC proliferation in vivo. Inactivation of the EGFR/MAPK pathway blocks or greatly retards RNSC cell cycle progression; conversely, over-activation of EGFR/MAPK signaling results in RNSC over-proliferation and disrupts the normal differentiation of renablasts (RBs), the immediate daughters of RNSC divisions. Our data further suggest that EGFR/MAPK signaling functions independently of JAK/STAT signaling and that dMyc and CycE partially mediate EGFR/MAPK signaling in MTs. Together, our data suggest a principal role of EGFR/MAPK signaling in regulating RNSC proliferation, which may provide important clues for understanding mammalian kidney repair and regeneration following injury. Copyright © 2014 Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, and Genetics Society of China. Published by Elsevier Ltd. All rights reserved.
Article
Oligodendrocyte differentiation is negatively regulated by LINGO-1 and positively regulated by the ErbB2 receptor tyrosine kinase. In wild-type oligodendrocytes, inhibition of ErbB2 blocks differentiation, whereas activation of ErbB2 promotes differentiation. In LINGO-1−/− oligodendrocytes, inhibition of ErbB2 blocks oligodendrocyte differentiation; whereas activation of ErbB2 has no effect on differentiation. We present biological and biochemical evidence showing that LINGO-1 can directly bind to ErbB2, block ErbB2 translocation into lipid rafts, and inhibit its phosphorylation for activation. The study demonstrates a novel regulatory mechanism of ErbB2 function whereby LINGO-1 suppresses oligodendrocyte differentiation by inhibiting ErbB2 translocation and activation in lipid rafts.
Article
The Mitogen-Activated Protein Kinase (MAPK) pathway represents one of the most conserved signaling cascades in multicellular organisms, since its cytoplasmic components can also be found in single-celled eukaryotic organisms such as yeast. With this broad view in mind, we can ask ourselves not only what are the seminal features and functions of the pathway in Drosophila, but also what have the studies in Drosophila taught us about the pathway in the wide range of organisms where it functions. We discuss the linearity of the MAPK signaling pathway in developmental decisions, and the ability of the developing organism to discriminate between different receptor tyrosine kinases that converge on the common MAPK pathway. Diverse modes of regulating the dynamics and level of MAPK signaling are presented. Finally, the convergence of MAPK signaling with other pathways is reviewed.
Article
Full-text available
The torpedo (top) locus of Drosophila encodes the fruitfly homolog of the vertebrate epidermal growth factor receptor gene and the neu proto-oncogene. We have isolated 13 top alleles in a screen for mutations failing to complement the female sterility of top, a recessive maternal effect allele that disrupts the establishment of the dorsoventral pattern of the egg shell and embryo. Several alleles recovered in this screen are zygotic lethal mutations; genetic analysis of these alleles has demonstrated that top is allelic to the embryonic lethal locus faint little ball. The 13 mutations recovered in our screens and 19 previously isolated top alleles have been genetically characterized through complementation tests with a series of hypomorphic and amorphic alleles. Nearly every top allele fails to complement the maternal effect sterility of top. Complementation tests show that the gene is required not only for oogenesis and embryogenesis, but also for pupal viability, for the growth of certain imaginal discs and for the patterning of specific ectodermal derivatives of the imaginal discs. Complementation analysis further demonstrates that the top lesions can be divided into general phenotypic categories: alleles affecting all gene activities in a coordinate manner, alleles preferentially affecting embryogenesis, alleles preferentially retaining oogenesis activity and alleles differentially affecting the development of specific imaginal disc derivatives. Correlations observed between the various developmental defects produced by top lesions suggest that the gene possesses several differentially, though not independently, mutable activities.
Article
Full-text available
In Drosophila, the dorsoventral asymmetry of the egg chamber depends on a dorsalizing signal that emanates from the oocyte. This signal is supplied by the TGFa-like gurken protein whose RNA is localized to the dorsalanterior corner of the oocyte. gurken protein is the potential ligand of the Drosophila EGF receptor homolog (torpedo), which is expressed in the follicular epithelium surrounding the oocyte. Here, we describe how changes in the dorsalizing germ-line signal affect the embryonic dorsoventral pattern. A reduction in strength of the germline signal as produced by mutations in gurken or torpedo does not change the slope of the embryonic dorsoventral morphogen gradient, but causes a splitting of the gradient ventrally. This leads to embryos with two partial dorsoventral axes. A change in distribution of the germ-line signal as caused by fs(1)K10, squid and orb mutations leads to a shift in the orientation of the embryonic dorsoventral axis relative to the anterior-posterior axis. In extreme cases, this results in embryos with a dorsoventral axis almost parallel to the anterior-posterior axis. These results imply that gurken, unlike other localized cytoplasmic determinants, is not directly responsible for the establishment of cell fates along a body axis, but that it restricts and orients an active axis-forming process which occurs later in the follicular epithelium or in the early embryo.
Article
Full-text available
The torpedo gene of Drosophila melanogaster is in-volved in the establishment of the dorsoventral pat-tern of eggshell and embryo. We have isolated new al-leles of torpedo and have found that torpedo is allelic to the zygotic embryonic lethal faint lift/e ball. We have shown that torpedo resides in subdivision 57F on the second chromosome-at the same location as the Drosophila homolog of the EGF receptor (DER). Using a cosmid that contains most of the DER coding region as a hybridization probe, we have shown that a cyto-logically small deficiency that eliminates torpedo ac-tivity also removes the DER gene, and that an inver-sion that was isolated as a strong torpedo allele breaks the coding region of the DER gene. We con-clude that torpedo is the DER gene.
Article
Full-text available
Reactive oxygen species have been implicated both in the ageing process and in degenerative diseases, including arthritis and cancer. Bacteria adapt to the lethal effects of oxidants such as hydrogen peroxide by inducing the expression of protective stress genes. Analogous responses have been identified in human cells. For example, haem oxygenase is a major stress protein in human cells treated with oxidants, and reactive oxygen intermediates activate NF-kappa B, a transcriptional regulator of genes involved in inflammatory and acute-phase responses. We report here the isolation and characterization of a novel complementary DNA (CL100) corresponding to a messenger RNA that is highly inducible by oxidative stress and heat shock in human skin cells. The cDNA contains an open reading frame specifying a protein of M(r) 39.3K with the structural features of a non-receptor-type protein-tyrosine phosphatase and which has significant amino-acid sequence similarity to a Tyr/Ser-protein phosphatase encoded by the late gene H1 of vaccinia virus. The purified protein encoded by the CL100 open reading frame expressed in bacteria has intrinsic phosphatase activity. Given the relationship between the levels of protein-tyrosine phosphorylation, receptor activity, cellular proliferation and cell-cycle control, the induction of this gene may play an important regulatory role in the human cellular response to environmental stress.
Article
Full-text available
We describe the molecular characterization of the Drosophila gene spitz (spi), which encodes a putative 26-kD, EGF-like transmembrane protein that is structurally similar to TGF-alpha. Temporal and spatial expression patterns of spi transcripts indicate that spi is expressed throughout the embryo. Examination of mutant embryos reveals that spi is involved in a number of unrelated developmental choices, for example, dorsal-ventral axis formation, glial migration, sensory organ determination, and muscle development. We propose that spi may act as a ligand for cell-specific receptors, possibly rhomboid and/or the Drosophila EGF receptor homolog.
Article
Full-text available
To generate cell- and tissue-specific expression patterns of the reporter gene lacZ in Drosophila, we have generated and characterized 1,426 independent insertion strains using four different P-element constructs. These four transposons carry a lacZ gene driven either by the weak promoter of the P-element transposase gene or by partial promoters from the even-skipped, fushi-tarazu, or engrailed genes. The tissue-specific patterns of beta-galactosidase expression that we are able to generate depend on the promoter utilized. We describe in detail 13 strains that can be used to follow specific cell lineages and demonstrate their utility in analyzing the phenotypes of developmental mutants. Insertion strains generated with P-elements that carry various sequences upstream of the lacZ gene exhibit an increased variety of expression patterns that can be used to study Drosophila development.
Article
Full-text available
The genetic interval 35C to 36A on chromosome arm 2L of Drosophila melanogaster has been saturated for mutations with visible or lethal phenotypes. 38 loci have been characterized, including several maternal-effect lethals (vasa, Bic-C, chiffon, cactus and cornichon) and several early embryonic lethals, including snail and fizzy. About 130 deletions have been used to order these loci. Complex interactions between mutant alleles have been uncovered in the immediate genetic environs of the snail gene, as has further evidence for an interaction between this region and that including the nearby genes no-ocelli and elbow.
Article
Full-text available
We have developed a non-radioactive in situ hybridization technique for the localization of RNA in whole mount Drosophila embryos. After fixation, whole embryos are hybridized in situ with a DNA probe which has been labeled with digoxygenin. The hybridization products are detected by using a phosphatase-coupled antibody against digoxygenin. In parallel experiments, embryos can be treated with an antibody directed against the corresponding protein product to allow the detection of its distribution using standard immunochemical techniques. We have used this approach to compare the spatial and temporal distribution patterns of the RNA and protein products of the segmentation gene hunchback (hb) during the early stages of embryogenesis. This comparison revealed translational control of the maternally derived hb mRNA, which was difficult to detect by conventional techniques. The non-radioactive in situ hybridization method is as sensitive as conventional methods, but is faster and easier to perform. This may make it a useful tool for a variety of other systems.
Article
Full-text available
The torpedo (top) locus of Drosophila encodes the fruitfly homolog of the vertebrate epidermal growth factor receptor gene and the neu proto-oncogene. We have isolated 13 top alleles in a screen for mutations failing to complement the female sterility of top, a recessive maternal effect allele that disrupts the establishment of the dorsoventral pattern of the egg shell and embryo. Several alleles recovered in this screen are zygotic lethal mutations; genetic analysis of these alleles has demonstrated that top is allelic to the embryonic lethal locus faint little ball. The 13 mutations recovered in our screens and 19 previously isolated top alleles have been genetically characterized through complementation tests with a series of hypomorphic and amorphic alleles. Nearly every top allele fails to complement the maternal effect sterility of top. Complementation tests show that the gene is required not only for oogenesis and embryogenesis, but also for pupal viability, for the growth of certain imaginal discs and for the patterning of specific ectodermal derivatives of the imaginal discs. Complementation analysis further demonstrates that the top lesions can be divided into general phenotypic categories: alleles affecting all gene activities in a coordinate manner, alleles preferentially affecting embryogenesis, alleles preferentially retaining oogenesis activity and alleles differentially affecting the development of specific imaginal disc derivatives. Correlations observed between the various developmental defects produced by top lesions suggest that the gene possesses several differentially, though not independently, mutable activities.
Article
Full-text available
Mutations in gurken and torpedo cause a ventralization in the follicle cell epithelium during Drosophila oogenesis and in the pattern of the embryo that develops in the resultant egg. Both genes lie midway in an epistatic series between fs(1)K10 and dorsal; the mutations block the dorsalization normally observed in K10 eggs but have no effect on the phenotype of embryos derived from dorsal mothers. Analysis of germ-line mosaics demonstrates that both ovarian and embryonic phenotypes will be produced when either the gurken+ gene is removed from the germ line or torpedo+ is removed from the soma. This shows that the dorsoventral pattern of the Drosophila egg chamber depends on the transfer of spatial information from the germ line to the somatic follicle cells, and from somatic cells to the oocyte.
Article
Full-text available
The twist gene is involved in the establishment of germ layers in Drosophila embryos: twist homozygous mutant embryos fail to form the ventral furrow at gastrulation and lack mesoderm and all internal organs. We have determined the sequence of the twist gene, that contains 'CAX' repeats in its 5' moiety, and codes for a protein of 490 amino acids. We have raised anti-twist antibodies that were used to study the distribution of the twist protein in whole mounts and tissue sections of wild-type embryos. Twist protein appears to be a nuclear protein at all developmental stages. It is present over both poles and in the midventral region (endoderm and mesoderm anlagen) at cellular blastoderm stage; later in development, it is detected within the mesodermal layer until its differentiation into somatopleura and splanchnopleura in which some cells are still labelled by anti-twist antibodies.
Article
Full-text available
Biologists require genetic as well as molecular tools to decipher genomic information and ultimately to understand gene function. The Berkeley Drosophila Genome Project is addressing these needs with a massive gene disruption project that uses individual, genetically engineered P transposable elements to target open reading frames throughout the Drosophila genome. DNA flanking the insertions is sequenced, thereby placing an extensive series of genetic markers on the physical genomic map and associating insertions with specific open reading frames and genes. Insertions from the collection now lie within or near most Drosophila genes, greatly reducing the time required to identify new mutations and analyze gene functions. Information revealed from these studies about P element site specificity is being used to target the remaining open reading frames.
Article
Full-text available
The spitz gene encoding a TGF-alpha homolog, has been shown to affect a subset of developmental processes that are similar to those regulated by DER, the Drosophila EGF receptor homolog. This work demonstrates that Spitz triggers the DER signaling cascade. Addition of a secreted, but not the membrane-associated form of Spitz to S2 Drosophila cells expressing DER gives rise to a rapid tyrosine autophosphorylation of DER. Following autophosphorylation, DER associates with the Drk adapter protein. Consequently, activation of MAP kinase is observed. The profile of MAP kinase activation provides a quantitative assay for DER activation. A dose response between the levels of Spitz and MAP kinase activity was observed. The secreted Spitz protein was expressed in embryos to assess its biological activity. An alteration in cell fates was observed in the ventral ectoderm, such that lateral cells acquired the ventral-most fates. The result indicates that graded activation of the DER pathway may normally give rise to a repertoire of discrete cell fates in the ventral ectoderm. Spatially restricted processing of Spitz may be responsible for this graded activation. The Rhomboid (Rho) and Star proteins were suggested, on the basis of genetic interactions, to act as modulators of DER signaling. No alteration in DER autophosphorylation or the pattern of MAP kinase activation by secreted Spitz was observed when the Rho and Star proteins were coexpressed with DER in S2 cells. In embryos mutant for rho or Star the ventralizing effect of secreted Spitz is epistatic, suggesting that Rho and Star may normally facilitate processing of the Spitz precursor.
Article
Full-text available
Cell-cell interactions in the Drosophila ovary play a crucial role in the establishment of dorsoventral polarity of both the egg shell and the future embryo. Torpedo/DER (top/DER), a homolog of the vertebrate epidermal growth factor receptor, is required for this signaling process in the somatic cells of the ovary. In contrast, gurken (grk), which also functions in this pathway, is required in the germline. We cloned the grk gene and found that it encodes a TGF alpha-like protein. Grk is, therefore, likely to be a ligand of top/DER, activating the receptor during oogenesis. During oogenesis, the grk transcript becomes asymmetrically localized to the dorsal corner of the oocyte. We propose that the dorsal localization of grk RNA results in a spatially restricted ligand that asymmetrically activates the receptor.
Article
Full-text available
In mammals, many cytokines and growth factors stimulate members of the Janus kinase (JAK) family to transduce signals for the proliferation and differentiation of various cell types, particularly in hematopoietic lineages. Mutations in the Drosophila hopscotch (hop) gene, which encodes a JAK, also cause proliferative defects. Loss-of-function alleles result in lethality and underproliferation of diploid tissues of the larva. A dominant gain-of-function allele, Tumorous-lethal (hopTum-l), leads to formation of melanotic tumors and hypertrophy of the larval lymph glands, the hematopoietic organs. We show that a single amino acid change in Hop is associated with the hopTum-l mutation. Overexpression of either wild-type hop or hopTum-l in the larval lymph glands causes melanotic tumors and lymph gland hypertrophy indistinguishable from the original hopTum-l mutation. In addition, overexpression of Hop in other tissues of the larva leads to pattern defects in the adult or to lethality. Finally, overexpression of either hop or hopTum-l in Drosophila cell culture results in tyrosine phosphorylation of Hop protein. However, overexpression of hopTum-l results in greater phosphorylation than overexpression of the wild-type. We conclude that hopTum-l encodes a hyperactive Hop kinase and that overactivity of Hop in lymph glands causes malignant neoplasia of Drosophila blood cells.
Article
Full-text available
In Drosophila, the dorsoventral asymmetry of the egg chamber depends on a dorsalizing signal that emanates from the oocyte. This signal is supplied by the TGF alpha-like gurken protein whose RNA is localized to the dorsal-anterior corner of the oocyte, gurken protein is the potential ligand of the Drosophila EGF receptor homolog (torpedo), which is expressed in the follicular epithelium surrounding the oocyte. Here, we describe how changes in the dorsalizing germ-line signal affect the embryonic dorsoventral pattern. A reduction in strength of the germ-line signal as produced by mutations in gurken or torpedo does not change the slope of the embryonic dorsoventral morphogen gradient, but causes a splitting of the gradient ventrally. This leads to embryos with two partial dorsoventral axes. A change in distribution of the germ-line signal as caused by fs(1)K10, squid and orb mutations leads to a shift in the orientation of the embryonic dorsoventral axis relative to the anterior-posterior axis. In extreme cases, this results in embryos with a dorsoventral axis almost parallel to the anterior-posterior axis. These results imply that gurken, unlike other localized cytoplasmic determinants, is not directly responsible for the establishment of cell fates along a body axis, but that it restricts and orients an active axis-forming process which occurs later in the follicular epithelium or in the early embryo.
Article
Full-text available
Internalization of ligand-receptor complexes is a well-documented mechanism for limiting the duration and magnitude of a signaling event. In the case of the EGF-Receptor (EGF-R), exposure to EGF or TGF-alpha results in internalization of up to 95% of the surface receptor pool within 5 minutes of exposure to ligand. In this report, we show that levels of Drosophila Egf-r mRNA are strongly down-regulated in epidermal cells likely to have recently undergone high levels of EGF-R signaling. The cells in which Egf-r mRNA levels are down-regulated express the rhomboid gene, which is thought to locally amplify EGF-R signaling. Widespread Egf-r mRNA down-regulation can be induced by ubiquitous expression of rhomboid or by eliminating the Gap1 gene. These results suggest that cells engaged in intense EGF-R/RAS signaling limit the duration of the signal through a combination of short-acting negative feedback mechanisms such as receptor internalization followed by a longer lasting reduction in receptor transcript levels. Control of Egf-r mRNA levels by altering transcription or mRNA stability is a new tier of regulation to be considered in analysis of EGF-R signaling during development.
Article
Full-text available
The Drosophila gene gurken participates in a signaling process that occurs between the germ line and the somatic cells (follicle cells) of the ovary. This process is required for correct patterning of the dorsoventral axis of both the egg and the embryo. gurken produces a spatially localized transcript which encodes a TGF-alpha-like molecule (Neuman-Silberberg and Schupbach, Cell 75, 165-174, 1993). Mutations in gurken cause a ventralized phenotype in egg and embryo. To determine whether the gurken gene product plays an instructive role in dorsoventral patterning, we constructed females containing extra copies of a gurken transgene. Such females produce dorsalized eggs and embryos, which is expected if gurken acts as a limiting factor in the dorsoventral patterning process. In addition, the expression pattern of the gene rhomboid in the follicle cells is altered in ovaries of females containing extra copies of gurken. Our results indicate that changing gurken dosage in otherwise wild-type ovaries is sufficient to alter the number of somatic follicle cells directed to the dorsal fate. Therefore the gurken-torpedo signaling process plays an instructive role in oogenesis. It induces dorsal cell fates in the follicle cell epithelium and it controls the production of maternal components that will direct the embryonic dorsoventral pattern after fertilization.
Article
Full-text available
Argos is a secreted molecule with an atypical EGF motif. It was recently shown to function as an inhibitor of the signaling triggered by the Drosophila EGF receptor (DER). In this work, we determine the contribution of Argos to the establishment of cell fates in the embryonic ventral ectoderm. Graded activation of DER is essential for patterning the ventral ectoderm. argos mutant embryos show expansion of ventral cell fates suggesting hyperactivation of the DER pathway. In the embryonic ventral ectoderm, argos is expressed in the ventralmost row of cells. We show that argos expression in the ventral ectoderm is induced by the DER pathway: argos is not expressed in DER mutant embryos, while it is ectopically expressed in the entire ventral ectoderm following ubiquitous activation of the DER pathway. argos expression appears to be triggered directly by the DER pathway, since induction can also be observed in cell culture, following activation of DER by its ligand, Spitz. Argos therefore functions in a sequential manner, to restrict the duration and level of DER signaling. This type of inhibitory feedback loop may represent a general paradigm for signaling pathways inducing diverse cell fates within a population of non-committed cells.
Article
Full-text available
A cDNA encoding a protein designated as LIG-1 has been cloned and characterized. A fragment of this cDNA was found previously in a screen for genes up-regulated during neural differentiation in mouse P19 embryonal carcinoma cells. Comparative sequence analysis revealed LIG-1 to be a novel integral membrane glycoprotein (1091 amino acids) containing an extracellular region (794 amino acids) with a potential signal peptide, 15 leucine-rich repeats, 3 immnunoglobulin-like domains, and 7 potential N-glycosylation sites, a transmembrane region of 23 amino acids, and a cytoplasmic region of 274 amino acids. This protein, therefore, is a new member of both the leucine-rich repeat and the immunoglobulin superfamilies. Furthermore, Northern blot and in situ hybridization analyses showed LIG-1 gene expression to be predominantly in the brain, restricted to a small subset of glial cells such as Bergmann glial cells of the cerebellum and glial cells in the nerve fiber layer of the olfactory bulb. On the basis of its structural features and expression pattern, we propose that LIG-1 functions as a cell type-specific adhesion molecule or receptor at the glial cell surface, and plays a role in the nervous system in for example neuroglial differentiation, development, and/or maintenance of neural functions where it is expressed.
Article
Full-text available
Phosphotyrosine phosphatases are critical negative or positive regulators in the intracellular signalling pathways that result in growth-factor-specific cell responses such as mitosis, differentiation, migration, survival, transformation or death. The SH2-domain-containing phosphotyrosine phosphatase SHP-2 is a positive signal transducer for several receptor tyrosine kinases (RTKs) and cytokine receptors. To investigate its mechanism of action we purified a tyrosine-phosphorylated glycoprotein which in different cell types associates tightly with SHP-2 and appears to serve as its substrate. Peptide sequencing in conjunction with complementary DNA cloning revealed a new gene family of at least fifteen members designated signal-regulatory proteins (SIRPs). They consist of two subtypes distinguished by the presence or absence of a cytoplasmic SHP-2-binding domain. The transmembrane polypeptide SIRP alpha1 is a substrate of activated RTKs and in its tyrosine-phosphorylated form binds SHP-2 through SH2 interactions and acts as its substrate. It also binds SHP-1 and Grb2 in vitro and has negative regulatory effects on cellular responses induced by growth factors, oncogenes or insulin. Our findings indicate that proteins belonging to the SIRP family generally regulate signals defining different physiological and pathological processes.
Article
Drosophila oocytes develop within cysts containing 16 cells that are interconnected by cytoplasmic bridges. Although the cysts are syncytial, the 16 cells differentiate to form a single oocyte and 15 nurse cells, and several mRNAs that are synthesized in the nurse cells accumulate specifically in the oocyte. To gain insight into the mechanisms that generate the cytoplasmic asymmetry within these cysts, we have examined cytoskeletal organization during oocyte differentiation. Shortly after formation of the 16 cell cysts, a prominent microtubule organizing center (MTOC) is established within the syncytial cytoplasm, and at the time the oocyte is determined, a single microtubule cytoskeleton connects the oocyte with the remaining 15 cells of each cyst. Recessive mutations at the Bicaudal-D (Bic-D) and egalitarian (egl) loci, which block oocyte differentiation, disrupt formation and maintenance of this polarized microtubule cytoskeleton. Microtubule assembly-inhibitors phenocopy these mutations, and prevent oocyte-specific accumulation of oskar, cyclin B and 65F mRNAs. We propose that formation of the polarized microtubule cytoskeleton is required for oocyte differentiation, and that this structure mediates the asymmetric accumulation of mRNAs within the syncytial cysts.
Article
The oriented peptide library technique was used to investigate the peptide-binding specificities of nine PDZ domains. Each PDZ domain selected peptides with hydrophobic residues at the carboxyl terminus. Individual PDZ domains selected unique optimal motifs defined primarily by the carboxyl terminal three to seven residues of the peptides. One family of PDZ domains, including those of the Discs Large protein, selected peptides with the consensus motif Glu-(Ser/Thr)-Xxx-(Val/Ile) (where Xxx represents any amino acid) at the carboxyl terminus. In contrast, another family of PDZ domains, including those of LIN-2, p55, and Tiam-1, selected peptides with hydrophobic or aromatic side chains at the carboxyl terminal three residues. On the basis of crystal structures of the PSD-95-3 PDZ domain, the specificities observed with the peptide library can be rationalized.
Article
even-skipped represses wingless and transforms cells that would normally secrete naked cuticle into denticle secreting cells. The GAL4 system can thus be used to study regulatory interactions during embryonic devel- opment. In adults, targeted expression can be used to generate dominant phenotypes for use in genetic screens. We have directed expression of an activated form of the Dras2 protein, resulting in dominant eye and wing defects that can be used in screens to identify other members of the Dras2 signal transduction path- way. SUMMARY
Article
The argos gene encodes a protein that is required for viability and that regulates the determination of cells in the Drosophila eye. A developmental analysis of argos mutant eyes indicates that the mystery cells, which are usually nonneuronal, are transformed into extra photoreceptors, and that supernumerary cone cells and pigment cells are also recruited. Clonal analysis indicates that argos acts nonautonomously and can diffuse over the range of several cell diameters. Conceptual translation of the argos gene suggests that it encodes a secreted protein.
Article
Guidelines for submitting commentsPolicy: Comments that contribute to the discussion of the article will be posted within approximately three business days. We do not accept anonymous comments. Please include your email address; the address will not be displayed in the posted comment. Cell Press Editors will screen the comments to ensure that they are relevant and appropriate but comments will not be edited. The ultimate decision on publication of an online comment is at the Editors' discretion. Formatting: Please include a title for the comment and your affiliation. Note that symbols (e.g. Greek letters) may not transmit properly in this form due to potential software compatibility issues. Please spell out the words in place of the symbols (e.g. replace “α” with “alpha”). Comments should be no more than 8,000 characters (including spaces ) in length. References may be included when necessary but should be kept to a minimum. Be careful if copying and pasting from a Word document. Smart quotes can cause problems in the form. If you experience difficulties, please convert to a plain text file and then copy and paste into the form.
Article
The dorsoventral axis of the Drosophila embryo is determined by a morphogen gradient established by the action of 12 maternal-effect genes: the dorsal group genes and cactus. One of the dorsal group genes, dorsal (dl), encodes the putative morphogen. Although no overall asymmetry in the distribution of dorsal protein is observed, a gradient of nuclear concentration of dl protein is established during cleavage stages, with a maximum at the ventral side of the egg. At the dorsal side of the egg, the protein remains in the cytoplasm. Nuclear localization of the dl protein, and hence gradient formation, is blocked in dorsalizing alleles of all of the other dorsal group genes, while in ventralizing mutants nuclear localization extends to the dorsal side of the egg. A correlation between dl protein distribution and embryonic pattern in mutant embryos indicates that the nuclear concentration of the dl protein determines pattern along the dorsoventral axis.
Article
A single P element insert in Drosophila melanogaster, called P[ry+ delta 2-3](99B), is described that caused mobilization of other elements at unusually high frequencies, yet is itself remarkably stable. Its transposase activity is higher than that of an entire P strain, but it rarely undergoes internal deletion, excision or transposition. This element was constructed by F. Laski, D. Rio and G. Rubin for other purposes, but we have found it to be useful for experiments involving P elements. We demonstrate that together with a chromosome bearing numerous nonautonomous elements it can be used for P element mutagenesis. It can also substitute efficiently for "helper" plasmids in P element mediated transformation, and can be used to move transformed elements around the genome.
Article
The Drosophila homologue of the mammalian epidermal growth factor (EGF) receptor (DER) is a receptor tyrosine kinase involved in many stages of fly development, including photoreceptor determination, and wing-vein formation. Its primary activating ligand is the Spitz protein, which is similar to mammalian TGF-alpha. Argos is a secreted protein that, like Spitz, contains a single EGF motif. It is a repressor of cell determination in the eye, and acts in other tissues, including the wing. Because Argos has the opposite effects to DER in the eye (the former blocks photoreceptor determination, the latter promotes it) we have tested whether it acts by blocking the DER pathway. We show that Argos does indeed repress this pathway in vivo and find that, in vitro, Argos protein can inhibit the activation of DER by Spitz. Thus the determination of cells by the DER pathway is regulated by a balance between extracellular activating and inhibiting signals. This is the first in vivo example of an extracellular inhibitor of a receptor tyrosine kinase.
Article
In Drosophila, as in mammalian cells, the Raf serine/threonine kinase appears to act as a common transducer of signals from several different receptor tyrosine kinases. We describe a new role for Raf in Drosophila development, showing that Raf acts in the somatic follicle cells to specify the dorsoventral polarity of the egg. Targeted expression of activated Raf (Rafgof) within follicle cells is sufficient to dorsalize both the eggshell and the embryo, whereas reduced Raf activity ventralizes the eggshell. We show that Raf functions downstream of the EGF receptor to instruct the dorsal follicle cell fate. In this assay, human and Drosophila Rafgof are functionally similar, in that either can induce ventral follicle cells to assume a dorsal fate.
Article
MEK, a dual specificity threonine/tyrosine kinase, has been postulated to be a convergent point for signaling from receptor protein tyrosine kinases (RTKs) and G-protein-coupled receptors. In contrast to yeast and mammalian cells where several MEKs have been isolated, only one Drosophila MEK (D-Mek) has been characterized to date. Previous studies have shown that D-Mek acts in the Torso RTK signaling pathway. To demonstrate that D-Mek also operates downstream of other RTKs, we generated a temperature-sensitive allele of D-mek (D-mekts) by site-directed mutagenesis based on the amino acid change of a yeast cdc2ts mutation. Using D-mekts, we show that in addition to its role in Torso signaling, D-Mek operates in the Sevenless and in the Drosophila epidermal growth factor RTK pathways. Because loss-of-function mutations in D-mek and the upstream receptors give rise to similar phenotypes, it suggests that D-mek is the only MEK activated by Drosophila RTKs. In addition, we demonstrate that different RTK pathways respond differently to alteration in D-Mek activity.
Article
Drosophila oocytes develop within cysts containing 16 cells that are interconnected by cytoplasmic bridges. Although the cysts are syncytial, the 16 cells differentiate to form a single oocyte and 15 nurse cells, and several mRNAs that are synthesized in the nurse cells accumulate specifically in the oocyte. To gain insight into the mechanisms that generate the cytoplasmic asymmetry within these cysts, we have examined cytoskeletal organization during oocyte differentiation. Shortly after formation of the 16 cell cysts, a prominent microtubule organizing center (MTOC) is established within the syncytial cytoplasm, and at the time the oocyte is determined, a single microtubule cytoskeleton connects the oocyte with the remaining 15 cells of each cyst. Recessive mutations at the Bicaudal-D (Bic-D) and egalitarian (egl) loci, which block oocyte differentiation, disrupt formation and maintenance of this polarized microtubule cytoskeleton. Microtubule assembly-inhibitors phenocopy these mutations, and prevent oocyte-specific accumulation of oskar, cyclin B and 65F mRNAs. We propose that formation of the polarized microtubule cytoskeleton is required for oocyte differentiation, and that this structure mediates the asymmetric accumulation of mRNAs within the syncytial cysts.
Article
P element enhancer trapping has become an indispensable tool in the analysis of the Drosophila melanogaster genome. However, there is great variation in the mutability of loci by these elements such that some loci are relatively refractory to insertion. We have developed the hobo transposable element for use in enhancer trapping and we describe the results of a hobo enhancer trap screen. In addition, we present evidence that a hobo enhancer trap element has a pattern of insertion into the genome that is different from the distribution of P elements in the available database. Hence, hobo insertion may facilitate access to genes resistant to P element insertion.
Article
Formation of the tail region of the Drosophila larva requires the activities of the terminal class genes. Genetic and molecular analyses of these genes suggests that localized activation of the receptor tyrosine kinase torso at the posterior egg pole triggers a signal transduction pathway. This pathway, mediated through the serine/threonine protein kinase D-raf and the protein tyrosine phosphatase corkscrew, controls the domains of expression of the transcription factors tailless and huckebein. In this paper, we report the molecular and developmental characterization of mutations in the D-raf gene. We show that mutations that alter conserved residues known to be necessary for kinase activity are associated with a null phenotype, demonstrating that D-raf kinase activity is required for its role in torso signaling. Another mutation, D-rafPB26, which prematurely truncates the kinase domain shows a weaker maternal effect phenotype that is strikingly similar to the corkscrew maternal effect phenotype, suggesting that a lower amount of kinase activity decreases the terminal signaling pathway. Finally, molecular and developmental characterization of two mutations that affect the late D-raf zygotic function(s) implies a novel role for D-raf in cell fate establishment in the eye. One of these mutations, D-rafC110, is associated with a single amino acid change within the putative D-raf regulatory region, while the other, D-rafHM-7, most likely reduces the wild-type amount of D-raf protein.
Article
The establishment of dorsal-ventral asymmetry of the Drosophila embryo requires a group of genes that act maternally. None of the previously identified dorsal-ventral axis genes are known to produce asymmetrically localized gene products during oogenesis. We show that rhomboid (rho), a novel member of this group, encodes a protein that is localized on the apical surface of the dorsal-anterior follicle cells surrounding the oocyte. Loss of rho function causes ventralization of the eggshell and the embryo, whereas ectopic expression leads to dorsalization of both structures. Thus, spatially restricted rho is necessary and sufficient for dorsal-ventral axis formation. We propose, based on these observations and double mutant experiments, that the spatially restricted rho protein leads to selective activation of the epidermal growth factor receptor in the dorsal follicle cells and subsequently the specification of the dorsal follicle cells.
Article
Signals released by the Spemann organizer of the amphibian gastrula can directly induce neural tissue from ectoderm and can dorsalize ventral mesoderm to form muscle. The secreted polypeptide noggin mimics these activities and is expressed at the appropriate time and place to participate in the organizer signal. Neural induction and mesoderm dorsalization are antagonized by bone morphogenetic proteins (BMPs), which induce epidermis and ventral mesoderm instead. Here we report that noggin protein binds BMP4 with high affinity and can abolish BMP4 activity by blocking binding to cognate cell-surface receptors. These data suggest that noggin secreted by the organizer patterns the embryo by interrupting BMP signaling.
Article
We have identified two members of a novel class of genes in Drosophila that encode putative transmembrane proteins with six leucine-rich repeats and a single immunoglobulin loop. These two molecules, Kek1 and Kek2, show striking conservation in their extracellular domains and have large and more divergent intracellular regions. Both genes are expressed in neurons as they differentiate in the embryonic central nervous system (CNS). kek1 is also expressed in other patterned epithelia, such as the follicle cells of the developing egg chamber, where it is found in a dorsal-ventral gradient around the oocyte. The homology of the kek genes to other known adhesion and signaling molecules, together with their expression patterns, suggests that both genes are involved in interactions at the cell surface. Genetic analysis reveals that deletion of the kek1 gene causes no obvious developmental defects. The coexpression of kek2 in the CNS leads us to suggest that Kek1 is part of a family of cell surface proteins with redundant function.
Article
The activation signal from tyrosine kinase receptors, such as the epidermal growth factor receptor (EGFR), is relayed via a highly conserved intracellular pathway involving Ras, Raf, and MAPK. In Drosophila, the EGFR and components of the intracellular pathway are broadly expressed, yet receptor activation evokes tissue-specific cell responses. Extracellular events that lead to receptor activation are one mechanism by which signaling is modulated. Here we show molecular and genetic evidence that Drosophila vein (vn) encodes a candidate EGFR ligand and that vn expression is spatially restricted. Consequently, vn may promote tissue-specific receptor activation. Unlike two other ligands, Gurken (Grk) and Spitz (Spi), which are transforming growth factor alpha-like proteins, Vn has both an immunoglobulin-like and an EGF-like domain. This combination of domains mirrors those in the vertebrate neuregulins that bind EGFR relatives.
Article
The dorsal-ventral pattern of the Drosophila embryo is established by three sequential signaling pathways. Each pathway transmits spatial information by localizing the activity of an extracellular signal, which acts as a ligand for a broadly distributed transmembrane receptor. The components of the first two pathways are encoded by maternal effect genes, while the third pathway is specified by genes expressed in the zygote. During oogenesis, the oocyte transmits a signal to the surrounding follicle cells by the gurken-torpedo pathway. After fertilization, the initial asymmetry of the egg chamber is used by the spätzle-Toll pathway to generate within the embryo a nuclear gradient of the transcription factor Dorsal, which regulates the regional expression of a set of zygotic genes. On the dorsal side of the embryo, the decapentaplegic-punt/thick veins pathway then establishes patterning of the amnioserosa and dorsal ectoderm. Each pathway uses a distinct strategy to achieve spatial localization of signaling activity.
Article
The Drosophila eye has contributed much to our knowledge of cell differentiation. This understanding has primarily come from the study of the R7 photoreceptor; much less is known about the development of the other classes of photoreceptor or the nonneuronal cone or pigment cells. I have used a dominant-negative form of the Drosophila epidermal growth factor receptor (DER) to show that this receptor tyrosine kinase (RTK) is required for the differentiation of all these cell types, and I have also shown that DER is sufficient to trigger their development. DER is even required in R7, where it can replace Sevenless, another RTK. These results broaden our view of eye development to include the whole ommatidium and suggest that reiterative activation of DER is critical for triggering the differentiation of all cell types.
Article
The Drosophila EGF receptor (DER) is activated by secreted Spitz to induce different cell fates in the ventral ectoderm. Processing of the precursor transmembrane Spitz to generate the secreted form was shown to be the limiting event, but the cells in which processing takes place and the mechanism that may generate a gradient of secreted Spitz in the ectoderm were not known. The ectodermal defects in single minded (sim) mutant embryos, in which the midline fails to develop, suggested that the midline cells contribute to patterning of the ventral ectoderm. This work shows that the midline provides the site for Spitz expression and processing. The Rhomboid and Star proteins are also expressed and required in the midline. The ectodermal defects of spitz, rho or Star mutant embryos could be rescued by inducing the expression of the respective normal genes only in the midline cells. Rho and Star thus function non-autonomously, and may be required for the production or processing of the Spitz precursor. Secreted Spitz is the only sim-dependent contribution of the midline to patterning the ectoderm, since the ventral defects observed in sim mutant embryos can be overcome by expression of secreted Spitz in the ectoderm. While ectopic expression of secreted Spitz in the ectoderm or mesoderm gave rise to ventralization of the embryo, increased expression of secreted Spitz in the midline did not lead to alterations in ectoderm patterning. A mechanism for adjustment to variable levels of secreted Spitz emanating from the midline may be provided by Argos, which forms an inhibitory feedback loop for DER activation. The production of secreted Spitz in the midline, may provide a stable source for graded DER activation in the ventral ectoderm.
Article
The oriented peptide library technique was used to investigate the peptide-binding specificities of nine PDZ domains. Each PDZ domain selected peptides with hydrophobic residues at the carboxyl terminus. Individual PDZ domains selected unique optimal motifs defined primarily by the carboxyl terminal three to seven residues of the peptides. One family of PDZ domains, including those of the Discs Large protein, selected peptides with the consensus motif Glu-(Ser/Thr)-Xxx-(Val/Ile) (where Xxx represents any amino acid) at the carboxyl terminus. In contrast, another family of PDZ domains, including those of LIN-2, p55, and Tiam-1, selected peptides with hydrophobic or aromatic side chains at the carboxyl terminal three residues. On the basis of crystal structures of the PSD-95-3 PDZ domain, the specificities observed with the peptide library can be rationalized.
Article
In Drosophila, the Ras1 gene is required downstream of receptor tyrosine kinases for correct eye development, embryonic patterning, wing vein formation, and border cell migration. Here we characterize a P-element allele of Ras1, Ras1(5703), that affects viability, eye morphogenesis, and early and late stages of oogenesis. Flies transheterozgyous for Ras1(5703) and existing EMS-induced Ras1 alleles are viable and exhibit a range of eye and eggshell defects. Differences in the severity of these phenotypes in different tissues suggest that there are allele-specific effects of Ras1 in development. Analysis of rescue constructs demonstrates that these differential phenotypes are due to loss of function in Ras1 alone and not due to effects on neighboring genes. Females mutant at the Ras1 locus lay eggs with reduced or missing dorsal eggshell structures. We observe dominant interactions between Ras1 mutants and other dorsoventral pathway mutants, including Egfr(top) and gurken. Ras1 is also epistatic to K10. Unlike Egfr(top) and gurken mutants, however, Ras1 females are moderately fertile, laying eggs with ventralized eggshells that can hatch normal larvae. These results suggest that Ras1 may have a different requirement in the patterning of the eggshell axis than in the patterning of the embryonic axis during oogenesis.
Article
We have discussed some of the mechanisms that can regulate the activity of the Egfr in Drosophila. However, from studies of this receptor in other species, additional mechanisms that modulate the activity of this protein have been identified and may also play a role in Egfr regulation. Among these are the regulation of RTK activity by endocytosis, control of receptor turnover, subcellular localization of the RTK within the membrane, and cross-talk with other signaling pathways.A detailed understanding of RTK regulatory mechanisms may have important therapeutic applications. Many cancers are caused by misregulation of RTK pathways, and some of the strategies to design drugs that cure malignancies have focused on targeting drugs against components of the RTK conserved signaling cassette, such as p21ras. However, because these molecules are shared by multiple RTKs, it may be difficult to achieve specific therapeutic effects. An alternative strategy is the design of drugs that interfere with the activities of molecules, such as Aos, Rho, and S, that function in modulating specific RTK signaling pathways.