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










Publication year range
1.
Nat Commun ; 8(1): 1848, 2017 11 29.
Article in English | MEDLINE | ID: mdl-29184052

ABSTRACT

Differentiation abnormalities are a hallmark of tuberous sclerosis complex (TSC) manifestations; however, the genesis of these abnormalities remains unclear. Here we report on mechanisms controlling the multi-lineage, early neuronal progenitor and neural stem-like cell characteristics of lymphangioleiomyomatosis (LAM) and angiomyolipoma cells. These mechanisms include the activation of a previously unreported Rheb-Notch-Rheb regulatory loop, in which the cyclic binding of Notch1 to the Notch-responsive elements (NREs) on the Rheb promoter is a key event. This binding induces the transactivation of Rheb. The identified NRE2 and NRE3 on the Rheb promoter are important to Notch-dependent promoter activity. Notch cooperates with Rheb to block cell differentiation via similar mechanisms in mouse models of TSC. Cell-specific loss of Tsc1 within nestin-expressing cells in adult mice leads to the formation of kidney cysts, renal intraepithelial neoplasia, and invasive papillary renal carcinoma.


Subject(s)
Angiomyolipoma/pathology , Lung Neoplasms/pathology , Lymphangioleiomyomatosis/pathology , Ras Homolog Enriched in Brain Protein/metabolism , Receptor, Notch1/metabolism , Angiomyolipoma/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Female , Humans , Lung Neoplasms/metabolism , Lymphangioleiomyomatosis/metabolism , Male , Mice, SCID , Mice, Transgenic , Neural Crest/metabolism , Neural Crest/pathology , Promoter Regions, Genetic , Ras Homolog Enriched in Brain Protein/genetics , Receptor, Notch1/genetics , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Tuberous Sclerosis/metabolism , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
2.
Mol Biol Cell ; 27(18): 2857-66, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27466320

ABSTRACT

The Notch signaling pathway plays essential roles in both animal development and human disease. Regulation of Notch receptor levels in membrane compartments has been shown to affect signaling in a variety of contexts. Here we used steady-state and pulse-labeling techniques to follow Notch receptors in sensory organ precursor cells in Drosophila. We find that the endosomal adaptor protein Numb regulates levels of Notch receptor trafficking to Rab7-labeled late endosomes but not early endosomes. Using an assay we developed that labels different pools of Notch receptors as they move through the endocytic system, we show that Numb specifically suppresses a recycled Notch receptor subpopulation and that excess Notch signaling in numb mutants requires the recycling endosome GTPase Rab11 activity. Our data therefore suggest that Numb controls the balance between Notch receptor recycling and receptor targeting to late endosomes to regulate signaling output after asymmetric cell division in Drosophila neural progenitors.


Subject(s)
Drosophila Proteins/metabolism , Juvenile Hormones/metabolism , Neural Stem Cells/metabolism , Receptors, Notch/metabolism , Animals , Cell Differentiation/physiology , Cell Membrane/metabolism , Drosophila melanogaster/metabolism , Endocytosis/physiology , Endosomes/metabolism , Endosomes/physiology , Protein Transport , Receptors, Notch/physiology , Signal Transduction , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
3.
J Cell Biol ; 201(3): 439-48, 2013 Apr 29.
Article in English | MEDLINE | ID: mdl-23609534

ABSTRACT

In Drosophila peripheral neurogenesis, Notch controls cell fates in sensory organ precursor (SOP) cells. SOPs undergo asymmetric cell division by segregating Numb, which inhibits Notch signaling, into the pIIb daughter cell after cytokinesis. In contrast, in the pIIa daughter cell, Notch is activated and requires Sanpodo, but its mechanism of action has not been elucidated. As Sanpodo is present in both pIIa and pIIb cells, a second role for Sanpodo in regulating Notch signaling in the low-Notch pIIb cell has been proposed. Here we demonstrate that Sanpodo regulates Notch signaling levels in both pIIa and pIIb cells via distinct mechanisms. The interaction of Sanpodo with Presenilin, a component of the γ-secretase complex, was required for Notch activation and pIIa cell fate. In contrast, Sanpodo suppresses Notch signaling in the pIIb cell by driving Notch receptor internalization. Together, these results demonstrate that a single protein can regulate Notch signaling through distinct mechanisms to either promote or suppress signaling depending on the local cellular context.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Receptors, Notch/metabolism , Amino Acid Motifs , Cell Line , Endocytosis , Mechanoreceptors/physiology , Microfilament Proteins/physiology , Presenilins/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Subunits/metabolism , Protein Transport , Stem Cells/physiology
4.
PLoS One ; 7(11): e48720, 2012.
Article in English | MEDLINE | ID: mdl-23144943

ABSTRACT

In Drosophila, the pattern of adult pigmentation is initiated during late pupal stages by the production of catecholamines DOPA and dopamine, which are converted to melanin. The pattern and degree of melanin deposition is controlled by the expression of genes such as ebony and yellow as well as by the enzymes involved in catecholamine biosynthesis. In this study, we show that the conserved TSC/TORC1 cell growth pathway controls catecholamine biosynthesis in Drosophila during pigmentation. We find that high levels of Rheb, an activator of the TORC1 complex, promote premature pigmentation in the mechanosensory bristles during pupal stages, and alter pigmentation in the cuticle of the adult fly. Disrupting either melanin synthesis by RNAi knockdown of melanogenic enzymes such as tyrosine hydroxylase (TH), or downregulating TORC1 activity by Raptor knockdown, suppresses the Rheb-dependent pigmentation phenotype in vivo. Increased Rheb activity drives pigmentation by increasing levels of TH in epidermal cells. Our findings indicate that control of pigmentation is linked to the cellular nutrient-sensing pathway by regulating levels of a critical enzyme in melanogenesis, providing further evidence that inappropriate activation of TORC1, a hallmark of the human tuberous sclerosis complex tumor syndrome disorder, can alter metabolic and differentiation pathways in unexpected ways.


Subject(s)
Catecholamines/biosynthesis , Cell Cycle Proteins/physiology , Drosophila Proteins/physiology , Drosophila/metabolism , Pigmentation/genetics , Transcription Factors/physiology , Animals , Catecholamines/genetics , Cell Cycle Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Epidermis/metabolism , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/genetics , Melanins/metabolism , Monomeric GTP-Binding Proteins/physiology , Neuropeptides/physiology , Pupa/metabolism , RNA Interference , Ras Homolog Enriched in Brain Protein , Ribosomal Protein S6 Kinases/genetics , Ribosomal Protein S6 Kinases/metabolism , Transcription Factors/metabolism
5.
Curr Opin Cell Biol ; 24(4): 534-40, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22818956

ABSTRACT

The Notch signaling pathway controls patterning and cell fate decisions during development in metazoans, and is associated with human diseases such as cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and certain cancers. Studies over the last several years have revealed sophisticated regulation of both the membrane-bound Notch receptor and its ligands by vesicle trafficking. This is perhaps most evident in neural progenitor cells in Drosophila, which divide asymmetrically to segregate Numb, an endocytic adaptor protein that acts as a Notch pathway inhibitor, to one daughter cell. Here, we discuss recent findings addressing how receptor and ligand trafficking to specific membrane compartments control activation of the Notch pathway in asymmetrically dividing cells and other tissues.


Subject(s)
Endocytosis , Receptors, Notch/metabolism , Signal Transduction , Animals , Asymmetric Cell Division , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Humans , Juvenile Hormones/metabolism , Ligands , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Protein Transport
6.
J Cell Biol ; 196(1): 65-83, 2012 Jan 09.
Article in English | MEDLINE | ID: mdl-22213802

ABSTRACT

Notch signaling governs binary cell fate determination in asymmetrically dividing cells. Through a forward genetic screen we identified the fly homologue of Eps15 homology domain containing protein-binding protein 1 (dEHBP1) as a novel regulator of Notch signaling in asymmetrically dividing cells. dEHBP1 is enriched basally and at the actin-rich interface of pII cells of the external mechanosensory organs, where Notch signaling occurs. Loss of function of dEHBP1 leads to up-regulation of Sanpodo, a regulator of Notch signaling, and aberrant trafficking of the Notch ligand, Delta. Furthermore, Sec15 and Rab11, which have been previously shown to regulate the localization of Delta, physically interact with dEHBP1. We propose that dEHBP1 functions as an adaptor molecule for the exocytosis and recycling of Delta, thereby affecting cell fate decisions in asymmetrically dividing cells.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Asymmetric Cell Division/physiology , Drosophila Proteins/physiology , Drosophila/cytology , Exocytosis/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation , Microfilament Proteins/metabolism , Signal Transduction , Vesicular Transport Proteins/metabolism , rab GTP-Binding Proteins/metabolism
7.
J Cell Sci ; 124(Pt 23): 4001-13, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22159415

ABSTRACT

Adherens junctions (AJs) in epithelial cells are constantly turning over to modulate adhesion properties under various physiological and developmental contexts, but how such AJ dynamics are regulated during the apical-basal polarization of primary epithelia remains unclear. Here, we used new and genetically validated GFP markers of Drosophila E-cadherin (DE-cadherin, hereafter referred to as DE-Cad) and ß-catenin (Armadillo, Arm) to quantitatively assay the in vivo dynamics of biosynthetic turnover and membrane redistribution by fluorescence recovery after photobleaching (FRAP) assays. Our data showed that membrane DE-Cad and Arm in AJs of polarizing epithelial cells had much faster biosynthetic turnover than in polarized cells. Fast biosynthetic turnover of membrane DE-Cad is independent of actin- and dynamin-based trafficking, but is microtubule-dependent. Furthermore, Arm in AJs of polarizing cells showed a faster and diffusion-based membrane redistribution that was both quantitatively and qualitatively different from the slower and exchange-based DE-Cad membrane distribution, indicating that the association of Arm with DE-Cad is more dynamic in polarizing cells, and only becomes stable in polarized epithelial cells. Consistently, biochemical assays showed that the binding of Arm to DE-Cad is weaker in polarizing cells than in polarized cells. Our data revealed that the molecular interaction between DE-Cad and Arm is modulated during apical-basal polarization, suggesting a new mechanism that might be crucial for establishing apical-basal polarity through regulating the AJ dynamics.


Subject(s)
Adherens Junctions/physiology , Armadillo Domain Proteins/chemistry , Cadherins/chemistry , Cell Polarity , Drosophila Proteins/chemistry , Epithelial Cells/physiology , Transcription Factors/chemistry , Adherens Junctions/chemistry , Animals , Drosophila/chemistry , Drosophila/genetics , Embryo, Nonmammalian/chemistry , Embryo, Nonmammalian/physiology , Epithelial Cells/chemistry , Epithelial Cells/cytology , Fluorescence Recovery After Photobleaching , Green Fluorescent Proteins/chemistry , Immunoprecipitation , Membranes/chemistry , Membranes/physiology , Multiprotein Complexes/chemistry , Protein Binding , Protein Stability , Protein Transport
8.
J Vis Exp ; (51)2011 May 27.
Article in English | MEDLINE | ID: mdl-21654627

ABSTRACT

Since the discovery of Green Fluorescent Protein (GFP), there has been a revolutionary change in the use of live-cell imaging as a tool for understanding fundamental biological mechanisms. Striking progress has been particularly evident in Drosophila, whose extensive toolkit of mutants and transgenic lines provides a convenient model to study evolutionarily-conserved developmental and cell biological mechanisms. We are interested in understanding the mechanisms that control cell fate specification in the adult peripheral nervous system (PNS) in Drosophila. Bristles that cover the head, thorax, abdomen, legs and wings of the adult fly are individual mechanosensory organs, and have been studied as a model system for understanding mechanisms of Notch-dependent cell fate decisions. Sensory organ precursor (SOP) cells of the microchaetes (or small bristles), are distributed throughout the epithelium of the pupal thorax, and are specified during the first 12 hours after the onset of pupariation. After specification, the SOP cells begin to divide, segregating the cell fate determinant Numb to one daughter cell during mitosis. Numb functions as a cell-autonomous inhibitor of the Notch signaling pathway. Here, we show a method to follow protein dynamics in SOP cell and its progeny within the intact pupal thorax using a combination of tissue-specific Gal4 drivers and GFP-tagged fusion proteins. This technique has the advantage over fixed tissue or cultured explants because it allows us to follow the entire development of an organ from specification of the neural precursor to growth and terminal differentiation of the organ. We can therefore directly correlate changes in cell behavior to changes in terminal differentiation. Moreover, we can combine the live imaging technique with mosaic analysis with a repressible cell marker (MARCM) system to assess the dynamics of tagged proteins in mitotic SOPs under mutant or wildtype conditions. Using this technique, we and others have revealed novel insights into regulation of asymmetric cell division and the control of Notch signaling activation in SOP cells (examples include references 1-6, 7, 8).


Subject(s)
Cytological Techniques/methods , Drosophila/cytology , Peripheral Nervous System/cytology , Sense Organs/cytology , Animals , Drosophila/growth & development , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/metabolism , Image Processing, Computer-Assisted/methods , Peripheral Nervous System/growth & development , Pupa , Sense Organs/growth & development
9.
PLoS One ; 5(8): e12369, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20808771

ABSTRACT

Mammalian Cas proteins regulate cell migration, division and survival, and are often deregulated in cancer. However, the presence of four paralogous Cas family members in mammals (BCAR1/p130Cas, EFS/Sin1, NEDD9/HEF1/Cas-L, and CASS4/HEPL) has limited their analysis in development. We deleted the single Drosophila Cas gene, Dcas, to probe the developmental function of Dcas. Loss of Dcas had limited effect on embryonal development. However, we found that Dcas is an important modulator of the severity of the developmental phenotypes of mutations affecting integrins (If and mew) and their downstream effectors Fak56D or Src42A. Strikingly, embryonic lethal Fak56D-Dcas double mutant embryos had extensive cell polarity defects, including mislocalization and reduced expression of E-cadherin. Further genetic analysis established that loss of Dcas modified the embryonal lethal phenotypes of embryos with mutations in E-cadherin (Shg) or its signaling partners p120- and beta-catenin (Arm). These results support an important role for Cas proteins in cell-cell adhesion signaling in development.


Subject(s)
Cell Polarity , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Alleles , Animals , Cadherins/metabolism , Cell Adhesion , Cytoskeleton/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Epithelial Cells/cytology , Female , Gene Deletion , Integrins/metabolism , Intercellular Junctions/metabolism , Male , Mesoderm/cytology , Mutation , Phenotype , Protein Transport , Substrate Specificity
10.
Mol Biol Cell ; 21(5): 802-10, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20053677

ABSTRACT

In Drosophila, mitotic neural progenitor cells asymmetrically segregate the cell fate determinant Numb in order to block Notch signaling in only one of the two daughter cells. Sanpodo, a membrane protein required for Notch signaling in asymmetrically dividing cells, is sequestered from the plasma membrane to intracellular vesicles in a Numb-dependent way after neural progenitor cell mitosis. However, the significance of Numb-dependent Sanpodo regulation is unclear. In this study, we conducted a structure-function analysis to identify the determinants of Sanpodo targeting in vivo. We identified an NPAF motif in the amino-terminal cytoplasmic tail of Sanpodo, which is conserved among insect Sanpodo homologues. The Sanpodo NPAF motif is predicted to bind directly to the Numb phosphotyrosine-binding domain and is critical for Numb binding in vitro. Deletion or mutation of the NPAF motif results in accumulation of Sanpodo at the plasma membrane in Numb-positive cells in vivo. Genetic analysis of Sanpodo NPAF mutants shows that Numb-dependent Sanpodo endocytic targeting can be uncoupled from Notch signaling regulation. Our findings demonstrate that Sanpodo contains an evolutionarily conserved motif that has been linked to Numb-dependent regulation in vertebrates and further support the model that Numb regulates Notch signaling independently of Sanpodo membrane trafficking in neural progenitor cells.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Juvenile Hormones/metabolism , Neurons/physiology , Receptors, Notch/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Microfilament Proteins/metabolism , Mitosis , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid
11.
J Clin Invest ; 120(1): 93-102, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20038815

ABSTRACT

Mutations in either of the genes encoding the tuberous sclerosis complex (TSC), TSC1 and TSC2, result in a multisystem tumor disorder characterized by lesions with unusual lineage expression patterns. How these unusual cell-fate determination patterns are generated is unclear. We therefore investigated the role of the TSC in the Drosophila external sensory organ (ESO), a classic model of asymmetric cell division. In normal development, the sensory organ precursor cell divides asymmetrically through differential regulation of Notch signaling to produce a pIIa and a pIIb cell. We report here that inactivation of Tsc1 and overexpression of the Ras homolog Rheb each resulted in duplication of the bristle and socket cells, progeny of the pIIa cell, and loss of the neuronal cell, a product of pIIb cell division. Live imaging of ESO development revealed this cell-fate switch occurred at the pIIa-pIIb 2-cell stage. In human angiomyolipomas, benign renal neoplasms often found in tuberous sclerosis patients, we found evidence of Notch receptor cleavage and Notch target gene activation. Further, an angiomyolipoma-derived cell line carrying biallelic TSC2 mutations exhibited TSC2- and Rheb-dependent Notch activation. Finally, inhibition of Notch signaling using a gamma-secretase inhibitor suppressed proliferation of Tsc2-null rat cells in a xenograft model. Together, these data indicate that the TSC and Rheb regulate Notch-dependent cell-fate decision in Drosophila and Notch activity in mammalian cells and that Notch dysregulation may underlie some of the distinctive clinical and pathologic features of TSC.


Subject(s)
Cell Cycle Proteins/physiology , Drosophila Proteins/physiology , Monomeric GTP-Binding Proteins/physiology , Neuropeptides/physiology , Receptors, Notch/physiology , Sense Organs/embryology , Signal Transduction/physiology , Angiomyolipoma/metabolism , Animals , Biological Evolution , Drosophila , Female , Humans , Intracellular Signaling Peptides and Proteins , Kidney/metabolism , Male , Membrane Proteins/physiology , Mice , Mice, SCID , Ras Homolog Enriched in Brain Protein , Rats , Tuberous Sclerosis/etiology
12.
Genetics ; 182(1): 407-10, 2009 May.
Article in English | MEDLINE | ID: mdl-19279324

ABSTRACT

Forty years ago, a high frequency of lethal giant larvae (lgl) alleles in wild populations of Drosophila melanogaster was reported. This locus has been intensively studied for its roles in epithelial polarity, asymmetric neural divisions, and restriction of tissue proliferation. Here, we identify a high frequency of lgl alleles in the Bloomington second chromosome deficiency kit and the University of California at Los Angeles Bruinfly FRT40A-lethal P collection. These unrecognized aberrations confound the use of these workhorse collections for phenotypic screening or genetic mapping. In addition, we determined that independent alleles of insensitive, reported to affect asymmetric cell divisions during sensory organ development, carry lgl deletions that are responsible for the observed phenotypes. Taken together, these results encourage the routine testing of second chromosome stocks for second-site alleles of lgl.


Subject(s)
Alleles , Chromosome Aberrations , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Genes, Lethal/genetics , Tumor Suppressor Proteins/genetics , Animals , Drosophila melanogaster/growth & development
13.
Mol Biol Cell ; 19(10): 4122-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18667536

ABSTRACT

In vertebrate neurons, axons have a uniform arrangement of microtubules with plus ends distal to the cell body (plus-end-out), and dendrites have equal numbers of plus- and minus-end-out microtubules. To determine whether microtubule orientation is a conserved feature of axons and dendrites, we analyzed microtubule orientation in invertebrate neurons. Using microtubule plus end dynamics, we mapped microtubule orientation in Drosophila sensory neurons, interneurons, and motor neurons. As expected, all axonal microtubules have plus-end-out orientation. However, in proximal dendrites of all classes of neuron, approximately 90% of dendritic microtubules were oriented with minus ends distal to the cell body. This result suggests that minus-end-out, rather than mixed orientation, microtubules are the signature of the dendritic microtubule cytoskeleton. Surprisingly, our map of microtubule orientation predicts that there are no tracks for direct cargo transport between the cell body and dendrites in unipolar neurons. We confirm this prediction, and validate the completeness of our map, by imaging endosome movements in motor neurons. As predicted by our map, endosomes travel smoothly between the cell body and axon, but they cannot move directly between the cell body and dendrites.


Subject(s)
Axons/metabolism , Dendrites/metabolism , Drosophila melanogaster/metabolism , Microtubules/metabolism , Neurons/metabolism , Animals , Animals, Genetically Modified , Cytoskeleton/metabolism , Endosomes/metabolism , Green Fluorescent Proteins/metabolism , Heterozygote , Luminescent Proteins/metabolism , Models, Biological , Transgenes , Red Fluorescent Protein
14.
Curr Opin Cell Biol ; 18(5): 507-15, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16919436

ABSTRACT

Adult metazoans represent the culmination of an intricate developmental process involving the temporally and spatially orchestrated division, migration, differentiation, attachment, polarization and death of individual cells. An elaborate infrastructure connecting the cell cycle and cell attachment machinery is essential for such exquisite integration of developmental processes. Integrin-, cadherin-, Merlin- and planar cell polarity (PCP)-dependent signaling cascades quantitatively and qualitatively program cell division during development. Proteins in this signaling infrastructure may represent an important source of cancer vulnerability in metazoans, as their dysfunction can pleiotropically promote the oncogenic process.


Subject(s)
Cell Adhesion/physiology , Cell Cycle/physiology , Animals , Cadherins/metabolism , Integrins/metabolism , Neurofibromin 2/metabolism , Signal Transduction/physiology
15.
J Cell Sci ; 118(Pt 11): 2393-404, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15923652

ABSTRACT

The mature ascidian oocyte is a large cell containing cytoplasmic and cortical domains polarized along a primary animal-vegetal (a-v) axis. The oocyte cortex is characterized by a gradient distribution of a submembrane monolayer of cortical rough endoplasmic reticulum (cER) and associated maternal postplasmic/PEM mRNAs (cER-mRNA domain). Between fertilization and first cleavage, this cER-mRNA domain is first concentrated vegetally and then relocated towards the posterior pole via microfilament-driven cortical contractions and spermaster-microtubule-driven translocations. The cER-mRNA domain further concentrates in a macroscopic cortical structure called the centrosome attracting body (CAB), which mediates a series of asymmetric divisions starting at the eight-cell stage. This results in the segregation of determinant mRNAs and their products in posterior cells of the embryo precursors of the muscle and germ line. Using two species of ascidians (Ciona intestinalis and Phallusia mammillata), we have pursued and amplified the work initiated in Halocynthia roretzi. We have analysed the cortical reorganizations in whole cells and in cortical fragments isolated from oocytes and from synchronously developing zygotes and embryos. After fertilization, we observe that a cortical patch rich in microfilaments encircles the cER-mRNA domain, concentrated into a cortical cap at the vegetal/contraction pole (indicating the future dorsal pole). Isolated cortices also retain microtubule asters rich in cER (indicating the future posterior pole). Before mitosis, parts of the cER-mRNA domain are detected, together with short microtubules, in isolated posterior (but not anterior) cortices. At the eight-cell stage, the posteriorly located cER-mRNA domain undergoes a cell-cycle-dependant compaction into the CAB. The CAB with embedded centrosomal microtubules can be isolated with cortical fragments from eight-cell-stage embryos. These and previous observations indicate that cytoskeleton-driven repositioning and compaction of a polarized cortical domain made of rough ER is a conserved mechanism used for polarization and segregation of cortical maternal mRNAs in embryos of evolutionarily distant species of ascidians.


Subject(s)
Blastomeres/metabolism , Oocytes/metabolism , RNA, Messenger, Stored/metabolism , Urochordata/embryology , Zygote/metabolism , Animals , Biological Evolution , Blastomeres/cytology , Centrosome/metabolism , Cytoskeleton/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Endoplasmic Reticulum, Rough/metabolism , Female , Oocytes/cytology , Zygote/cytology
16.
Mol Biol Cell ; 16(8): 3480-7, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15901829

ABSTRACT

In Drosophila, asymmetric division occurs during proliferation of neural precursors of the central and peripheral nervous system (PNS), where a membrane-associated protein, Numb, is asymmetrically localized during cell division and is segregated to one of the two daughter cells (the pIIb cell) after mitosis. numb has been shown genetically to function as an antagonist of Notch signaling and also as a negative regulator of the membrane localization of Sanpodo, a four-pass transmembrane protein required for Notch signaling during asymmetric cell division in the CNS. Previously, we identified lethal giant larvae (lgl) as a gene required for numb-mediated inhibition of Notch in the adult PNS. In this study we show that Sanpodo is expressed in asymmetrically dividing precursor cells of the PNS and that Sanpodo internalization in the pIIb cell is dependent cytoskeletally associated Lgl. Lgl specifically regulates internalization of Sanpodo, likely through endocytosis, but is not required for the endocytosis Delta, which is a required step in the Notch-mediated cell fate decision during asymmetric cell division. Conversely, the E3 ubiquitin ligase neuralized is required for both Delta endocytosis and the internalization of Sanpodo. This study identifies a hitherto unreported role for Lgl as a regulator of Sanpodo during asymmetric cell division in the adult PNS.


Subject(s)
Cell Membrane/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Sense Organs/cytology , Sense Organs/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Division , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Microfilament Proteins/metabolism , Peripheral Nervous System/cytology , Peripheral Nervous System/metabolism , Receptors, Notch/metabolism , Tumor Suppressor Proteins/genetics
17.
Development ; 132(10): 2319-32, 2005 May.
Article in English | MEDLINE | ID: mdl-15829515

ABSTRACT

The receptor Notch and its ligands of the Delta/Serrate/LAG2 (DSL) family are the central components in the Notch pathway, a fundamental cell signaling system that regulates pattern formation during animal development. Delta is directly ubiquitinated by Drosophila and Xenopus Neuralized, and by zebrafish Mind bomb, two unrelated RING-type E3 ubiquitin ligases with common abilities to promote Delta endocytosis and signaling activity. Although orthologs of both Neuralized and Mind bomb are found in most metazoan organisms, their relative contributions to Notch signaling in any single organism have not yet been assessed. We show here that a Drosophila ortholog of Mind bomb (D-mib) is a positive component of Notch signaling that is required for multiple Neuralized-independent, Notch-dependent developmental processes. Furthermore, we show that D-mib associates physically and functionally with both Serrate and Delta. We find that D-mib uses its ubiquitin ligase activity to promote DSL ligand activity, an activity that is correlated with its ability to induce the endocytosis and degradation of both Delta and Serrate (see also Le Borgne et al., 2005). We further demonstrate that D-mib can functionally replace Neuralized in multiple cell fate decisions that absolutely require endogenous Neuralized, a testament to the highly similar activities of these two unrelated ubiquitin ligases in regulating Notch signaling. We conclude that ubiquitination of Delta and Serrate by Neuralized and D-mib is an obligate feature of DSL ligand activation throughout Drosophila development.


Subject(s)
Body Patterning/physiology , Drosophila Proteins/metabolism , Drosophila/embryology , Gene Expression Regulation, Developmental , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Calcium-Binding Proteins , Cloning, Molecular , Crosses, Genetic , DNA Primers , Drosophila Proteins/genetics , Fluorescent Antibody Technique , Immunoprecipitation , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Jagged-1 Protein , Membrane Proteins/metabolism , Mutation/genetics , Receptors, Notch , Serrate-Jagged Proteins , Ubiquitin-Protein Ligases/genetics , Xenopus Proteins
18.
Curr Opin Cell Biol ; 16(2): 195-205, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15196564

ABSTRACT

Asymmetric cell division is a conserved mechanism for partitioning information during mitosis. Over the past several years, significant progress has been made in our understanding of how cells establish polarity during asymmetric cell division and how determinants, in the form of localized proteins and mRNAs, are segregated. In particular, genetic studies in Drosophila and Caenorhabditis elegans have linked cell polarity, G protein signaling and regulation of the cytoskeleton to coordination of mitotic spindle orientation and localization of determinants. Also, several new studies have furthered our understanding of how asymmetrically localized cell fate determinants, such as the Numb, a negative regulator Notch signaling, functions in biasing cell fates in the developing nervous system in Drosophila. In vertebrates, analysis of dividing neural progenitor cells by in vivo imaging has raised questions about the role of asymmetric cell divisions during neurogenesis.


Subject(s)
Cell Polarity/genetics , Cytoskeleton/metabolism , Signal Transduction/genetics , Spindle Apparatus/metabolism , Animals , Cell Division/physiology , Cytoskeleton/genetics , Drosophila Proteins , Humans , Juvenile Hormones/genetics , Juvenile Hormones/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Animal , Receptors, Notch , Spindle Apparatus/genetics
19.
Genes Dev ; 18(6): 623-8, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15075290

ABSTRACT

The Drosophila external sensory organ forms in a lineage elaborating from a single precursor cell via a stereotypical series of asymmetric divisions. HAMLET transcription factor expression demarcates the lineage branch that generates two internal cell types, the external sensory neuron and thecogen. In HAMLET mutant organs, these internal cells are converted to external cells via an unprecedented cousin-cousin cell-fate respecification event. Conversely, ectopic HAMLET expression in the external cell branch leads to internal cell production. The fate-determining signals NOTCH and PAX2 act at multiple stages of lineage elaboration and HAMLET acts to modulate their activity in a branch-specific manner.


Subject(s)
Cell Differentiation/physiology , Drosophila/physiology , Neuroglia/physiology , Neurons/physiology , Sensory Receptor Cells/physiology , Animals , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Female , Male , Nuclear Proteins/metabolism , PAX2 Transcription Factor , Sensory Receptor Cells/cytology , Transcription Factors/metabolism
20.
Curr Biol ; 13(9): 778-83, 2003 Apr 29.
Article in English | MEDLINE | ID: mdl-12725738

ABSTRACT

The tumor suppressor genes lethal giant larvae (lgl) and discs large (dlg) act together to maintain the apical basal polarity of epithelial cells in the Drosophila embryo. Neuroblasts that delaminate from the embryonic epithelium require lgl to promote formation of a basal Numb and Prospero crescent, which will be asymmetrically segregated to the basal daughter cell upon division to specify cell fate. Sensory organ precursors (SOPs) also segregate Numb asymmetrically at cell division. Numb functions to inhibit Notch signaling and to specify the fates of progenies of the SOP that constitute the cellular components of the adult sensory organ. We report here that, in contrast to the embryonic neuroblast, lgl is not required for asymmetric localization of Numb in the dividing SOP. Nevertheless, mosaic analysis reveals that lgl is required for cell fate specification within the SOP lineage; SOPs lacking Lgl fail to specify internal neurons and glia. Epistasis studies suggest that Lgl acts to inhibit Notch signaling by functioning downstream or in parallel with Numb. These findings uncover a previously unknown function of Lgl in the inhibition of Notch and reveal different modes of action by which Lgl can influence cell fate in the neuroblast and SOP lineages.


Subject(s)
Drosophila Proteins/physiology , Drosophila/embryology , Gene Silencing/physiology , Juvenile Hormones/physiology , Membrane Proteins/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Polarity/genetics , Cell Polarity/physiology , Drosophila/cytology , Epithelial Cells/physiology , Immunohistochemistry , Larva/physiology , Membrane Proteins/genetics , Receptors, Notch , Signal Transduction/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...