Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Cell ; 162(2): 425-440, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26186194

ABSTRACT

Protein interactions form a network whose structure drives cellular function and whose organization informs biological inquiry. Using high-throughput affinity-purification mass spectrometry, we identify interacting partners for 2,594 human proteins in HEK293T cells. The resulting network (BioPlex) contains 23,744 interactions among 7,668 proteins with 86% previously undocumented. BioPlex accurately depicts known complexes, attaining 80%-100% coverage for most CORUM complexes. The network readily subdivides into communities that correspond to complexes or clusters of functionally related proteins. More generally, network architecture reflects cellular localization, biological process, and molecular function, enabling functional characterization of thousands of proteins. Network structure also reveals associations among thousands of protein domains, suggesting a basis for examining structurally related proteins. Finally, BioPlex, in combination with other approaches, can be used to reveal interactions of biological or clinical significance. For example, mutations in the membrane protein VAPB implicated in familial amyotrophic lateral sclerosis perturb a defined community of interactors.


Subject(s)
Protein Interaction Maps , Proteomics/methods , Amyotrophic Lateral Sclerosis/genetics , Humans , Mass Spectrometry , Protein Interaction Mapping , Proteins/chemistry , Proteins/isolation & purification , Proteins/metabolism
2.
Proc Natl Acad Sci U S A ; 121(24): e2400732121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38838021

ABSTRACT

Cytoplasmic mislocalization and aggregation of TDP-43 protein are hallmarks of amyotrophic lateral sclerosis (ALS) and are observed in the vast majority of both familial and sporadic cases. How these two interconnected processes are regulated on a molecular level, however, remains enigmatic. Genome-wide screens for modifiers of the ALS-associated genes TDP-43 and FUS have identified the phospholipase D (Pld) pathway as a key regulator of ALS-related phenotypes in the fruit fly Drosophila melanogaster [M. W. Kankel et al., Genetics 215, 747-766 (2020)]. Here, we report the results of our search for downstream targets of the enzymatic product of Pld, phosphatidic acid. We identify two conserved negative regulators of the cAMP/PKA signaling pathway, the phosphodiesterase dunce and the inhibitory subunit PKA-R2, as modifiers of pathogenic phenotypes resulting from overexpression of the Drosophila TDP-43 ortholog TBPH. We show that knockdown of either of these genes results in a mitigation of both TBPH aggregation and mislocalization in larval motor neuron cell bodies, as well as an amelioration of adult-onset motor defects and shortened lifespan induced by TBPH. We determine that PKA kinase activity is downstream of both TBPH and Pld and that overexpression of the PKA target CrebA can rescue TBPH mislocalization. These findings suggest a model whereby increasing cAMP/PKA signaling can ameliorate the molecular and functional effects of pathological TDP-43.


Subject(s)
Cyclic AMP-Dependent Protein Kinases , Cyclic AMP , DNA-Binding Proteins , Drosophila Proteins , Drosophila melanogaster , Signal Transduction , Animals , Cyclic AMP/metabolism , Drosophila melanogaster/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/genetics , Humans , Motor Neurons/metabolism
3.
Cell ; 147(3): 690-703, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22036573

ABSTRACT

Determining the composition of protein complexes is an essential step toward understanding the cell as an integrated system. Using coaffinity purification coupled to mass spectrometry analysis, we examined protein associations involving nearly 5,000 individual, FLAG-HA epitope-tagged Drosophila proteins. Stringent analysis of these data, based on a statistical framework designed to define individual protein-protein interactions, led to the generation of a Drosophila protein interaction map (DPiM) encompassing 556 protein complexes. The high quality of the DPiM and its usefulness as a paradigm for metazoan proteomes are apparent from the recovery of many known complexes, significant enrichment for shared functional attributes, and validation in human cells. The DPiM defines potential novel members for several important protein complexes and assigns functional links to 586 protein-coding genes lacking previous experimental annotation. The DPiM represents, to our knowledge, the largest metazoan protein complex map and provides a valuable resource for analysis of protein complex evolution.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Protein Interaction Mapping , Animals , Drosophila Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Proteomics , SNARE Proteins/metabolism
4.
Nature ; 545(7655): 505-509, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28514442

ABSTRACT

The physiology of a cell can be viewed as the product of thousands of proteins acting in concert to shape the cellular response. Coordination is achieved in part through networks of protein-protein interactions that assemble functionally related proteins into complexes, organelles, and signal transduction pathways. Understanding the architecture of the human proteome has the potential to inform cellular, structural, and evolutionary mechanisms and is critical to elucidating how genome variation contributes to disease. Here we present BioPlex 2.0 (Biophysical Interactions of ORFeome-derived complexes), which uses robust affinity purification-mass spectrometry methodology to elucidate protein interaction networks and co-complexes nucleated by more than 25% of protein-coding genes from the human genome, and constitutes, to our knowledge, the largest such network so far. With more than 56,000 candidate interactions, BioPlex 2.0 contains more than 29,000 previously unknown co-associations and provides functional insights into hundreds of poorly characterized proteins while enhancing network-based analyses of domain associations, subcellular localization, and co-complex formation. Unsupervised Markov clustering of interacting proteins identified more than 1,300 protein communities representing diverse cellular activities. Genes essential for cell fitness are enriched within 53 communities representing central cellular functions. Moreover, we identified 442 communities associated with more than 2,000 disease annotations, placing numerous candidate disease genes into a cellular framework. BioPlex 2.0 exceeds previous experimentally derived interaction networks in depth and breadth, and will be a valuable resource for exploring the biology of incompletely characterized proteins and for elucidating larger-scale patterns of proteome organization.


Subject(s)
Databases, Protein , Disease , Protein Interaction Mapping , Protein Interaction Maps , Proteome/metabolism , Cell Physiological Phenomena/genetics , Genome, Human , Humans , Intracellular Space/metabolism , Markov Chains , Mass Spectrometry , Molecular Sequence Annotation , Open Reading Frames , Proteome/analysis , Proteome/chemistry , Proteome/genetics
5.
Genome Res ; 25(11): 1692-702, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26294687

ABSTRACT

In eukaryotic cells, RNAs exist as ribonucleoprotein particles (RNPs). Despite the importance of these complexes in many biological processes, including splicing, polyadenylation, stability, transportation, localization, and translation, their compositions are largely unknown. We affinity-purified 20 distinct RNA-binding proteins (RBPs) from cultured Drosophila melanogaster cells under native conditions and identified both the RNA and protein compositions of these RNP complexes. We identified "high occupancy target" (HOT) RNAs that interact with the majority of the RBPs we surveyed. HOT RNAs encode components of the nonsense-mediated decay and splicing machinery, as well as RNA-binding and translation initiation proteins. The RNP complexes contain proteins and mRNAs involved in RNA binding and post-transcriptional regulation. Genes with the capacity to produce hundreds of mRNA isoforms, ultracomplex genes, interact extensively with heterogeneous nuclear ribonuclear proteins (hnRNPs). Our data are consistent with a model in which subsets of RNPs include mRNA and protein products from the same gene, indicating the widespread existence of auto-regulatory RNPs. From the simultaneous acquisition and integrative analysis of protein and RNA constituents of RNPs, we identify extensive cross-regulatory and hierarchical interactions in post-transcriptional control.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation , RNA-Binding Proteins/metabolism , Animals , Drosophila Proteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Sequence Analysis, RNA , Transfection
6.
Nat Rev Genet ; 13(9): 654-66, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22868267

ABSTRACT

Notch signalling links the fate of one cell to that of an immediate neighbour and consequently controls differentiation, proliferation and apoptotic events in multiple metazoan tissues. Perturbations in this pathway activity have been linked to several human genetic disorders and cancers. Recent genome-scale studies in Drosophila melanogaster have revealed an extraordinarily complex network of genes that can affect Notch activity. This highly interconnected network contrasts our traditional view of the Notch pathway as a simple linear sequence of events. Although we now have an unprecedented insight into the way in which such a fundamental signalling mechanism is controlled by the genome, we are faced with serious challenges in analysing the underlying molecular mechanisms of Notch signal control.


Subject(s)
Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Gene Dosage , Humans , Male , Proteomics , Receptors, Notch/genetics , Signal Transduction/genetics , Systems Biology
7.
Adv Exp Med Biol ; 1066: 125-140, 2018.
Article in English | MEDLINE | ID: mdl-30030825

ABSTRACT

The Notch pathway controls a very broad spectrum of cell fates in metazoans during development, influencing proliferation, differentiation and cell death. Given its central role in normal development and homeostasis, misregulation of Notch signals can lead to various disorders including cancer. How the Notch pathway mediates such pleiotropic and differential effects is of fundamental importance. It is becoming increasingly clear through a number of large-scale genetic and proteomic studies that Notch interacts with a staggeringly large number of other genes and pathways in a context-dependent, complex, and highly regulated network, which determines the ultimate biological outcome. How best to interpret and analyze the continuously increasing wealth of data on Notch interactors remains a challenge. Here we review the current state of genetic and proteomic data related to the Notch interactome.


Subject(s)
Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Humans , Receptors, Notch/genetics
8.
EMBO J ; 31(13): 2895-907, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22580825

ABSTRACT

Genetic analyses in Drosophila revealed a synergy between Notch and the pleiotropic transcription factor Mef2 (myocyte enhancer factor 2), which profoundly influences proliferation and metastasis. We show that these hyperproliferative and invasive Drosophila phenotypes are attributed to upregulation of eiger, a member of the tumour necrosis factor superfamily of ligands, and the consequent activation of Jun N-terminal kinase signalling, which in turn triggers the expression of the invasive marker MMP1. Expression studies in human breast tumour samples demonstrate correlation between Notch and Mef2 paralogues and support the notion that Notch-MEF2 synergy may be significant for modulating human mammary oncogenesis.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation , Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , MAP Kinase Signaling System/physiology , Myogenic Regulatory Factors/physiology , Receptors, Notch/physiology , Animals , Breast Neoplasms/metabolism , Drosophila Proteins/biosynthesis , Female , Gene Expression Profiling , Humans , MEF2 Transcription Factors , Male , Matrix Metalloproteinase 1/metabolism , Membrane Proteins/biosynthesis , Myogenic Regulatory Factors/metabolism , Neoplasm Metastasis , Up-Regulation
9.
Development ; 140(9): 2039-49, 2013 May.
Article in English | MEDLINE | ID: mdl-23571220

ABSTRACT

Cell-to-cell communication via the Notch pathway is mediated between the membrane-bound Notch receptor and either of its canonical membrane-bound ligands Delta or Serrate. Notch ligands mediate receptor transactivation between cells and also mediate receptor cis-inhibition when Notch and ligand are co-expressed on the same cell. We demonstrate in Drosophila that removal of any of the EGF-like repeats (ELRs) 4, 5 or 6 results in a Serrate molecule capable of transactivating Notch but exhibiting little or no Notch cis-inhibition capacity. These forms of Serrate require Epsin (Liquid facets) to transduce a signal, suggesting that ELR 4-6-deficient ligands still require endocytosis for Notch activation. We also demonstrate that ELRs 4-6 are responsible for the dominant-negative effects of Serrate ligand forms that lack the intracellular domain and are therefore incapable of endocytosis in the ligand-expressing cell. We find that ELRs 4-6 of Serrate are conserved across species but do not appear to be conserved in Delta homologs.


Subject(s)
Calcium-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Calcium-Binding Proteins/genetics , Cell Membrane/genetics , Cell Membrane/metabolism , Conserved Sequence , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Endocytosis , Female , Gene Deletion , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Ligands , Male , Membrane Proteins/genetics , Protein Binding , Receptors, Notch/genetics , Serrate-Jagged Proteins , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , Transfection , Transgenes , Wings, Animal/cytology , Wings, Animal/metabolism
10.
Proc Natl Acad Sci U S A ; 110(26): E2371-80, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23757500

ABSTRACT

The clinical severity of the neurodegenerative disorder spinal muscular atrophy (SMA) is dependent on the levels of functional Survival Motor Neuron (SMN) protein. Consequently, current strategies for developing treatments for SMA generally focus on augmenting SMN levels. To identify additional potential therapeutic avenues and achieve a greater understanding of SMN, we applied in vivo, in vitro, and in silico approaches to identify genetic and biochemical interactors of the Drosophila SMN homolog. We identified more than 300 candidate genes that alter an Smn-dependent phenotype in vivo. Integrating the results from our genetic screens, large-scale protein interaction studies, and bioinformatic analysis, we define a unique interactome for SMN that provides a knowledge base for a better understanding of SMA.


Subject(s)
Drosophila Proteins/genetics , Genes, Insect , RNA-Binding Proteins/genetics , Animals , Animals, Genetically Modified , Gene Regulatory Networks , Humans , Knowledge Bases , Neuromuscular Junction/genetics , Phenotype , RNA Interference , Species Specificity , Spinal Muscular Atrophies of Childhood/genetics
11.
J Cell Sci ; 126(Pt 10): 2135-40, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23729744

ABSTRACT

Cell-cell interactions define a quintessential aspect of multicellular development. Metazoan morphogenesis depends on a handful of fundamental, conserved cellular interaction mechanisms, one of which is defined by the Notch signaling pathway. Signals transmitted through the Notch surface receptor have a unique developmental role: Notch signaling links the fate of one cell with that of a cellular neighbor through physical interactions between the Notch receptor and the membrane-bound ligands that are expressed in an apposing cell. The developmental outcome of Notch signals is strictly dependent on the cellular context and can influence differentiation, proliferation and apoptotic cell fates. The Notch pathway is conserved across species (Artavanis-Tsakonas et al., 1999; Bray, 2006; Kopan and Ilagan, 2009). In humans, Notch malfunction has been associated with a diverse range of diseases linked to changes in cell fate and cell proliferation including cancer (Louvi and Artavanis-Tsakonas, 2012). In this Cell Science at a Glance article and the accompanying poster we summarize the molecular biology of Notch signaling, its role in development and its relevance to disease.


Subject(s)
Cell Communication/physiology , Receptors, Notch/metabolism , Signal Transduction , Animals , Cellular Microenvironment , Embryonic Development , Humans , Morphogenesis
12.
Semin Cell Dev Biol ; 23(4): 473-80, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22373641

ABSTRACT

Signals through the Notch receptors are used throughout development to control cellular fate choices. Our intention here is to provide an overview of the involvement of Notch signaling in human disease, which, keeping pace with the known biology of the pathway, manifests itself in a pleiotropic fashion. A pathway with such broad action in normal development, a profound involvement in the biology of adult stem cells and intricate and complex controls governing its activity, poses numerous challenges. We provide an overview of Notch related pathologies identified thus far and emphasize aspects that have been modeled in experimental systems in order to understand the underlying pathobiology and, hopefully, help the definition of rational therapeutic avenues.


Subject(s)
Mutation , Receptors, Notch/genetics , Signal Transduction/genetics , Alagille Syndrome/genetics , Alagille Syndrome/metabolism , Animals , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/metabolism , CADASIL/genetics , CADASIL/metabolism , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Genetic Pleiotropy , Humans , Neoplasms/genetics , Oncogenes , Receptors, Notch/metabolism , Receptors, Notch/physiology
13.
Curr Opin Cell Biol ; 19(2): 166-75, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17317139

ABSTRACT

The development of complex and diverse metazoan morphologies is coordinated by a surprisingly small number of evolutionarily conserved signaling mechanisms. These signals can act in parallel but often appear to function as an integrated hyper-network. The nodes defining this complex molecular circuitry are poorly understood, but the biological significance of pathway cross-talk is profound. The importance of such large-scale signal integration is exemplified by Notch and its ability to cross-talk with all the major pathways to influence cell differentiation, proliferation, survival and migration. The Notch pathway is, thus, a useful paradigm to illustrate the complexity of pathway cross-talk: its pervasiveness, context dependency, and importance in development and disease.


Subject(s)
Receptor Cross-Talk/physiology , Receptors, Notch/metabolism , Signal Transduction , Animals , Apoptosis , Cell Movement , Cell Proliferation , Humans , Models, Biological , Morphogenesis , Neoplasms/metabolism , Receptors, Cell Surface , Stem Cells/metabolism
14.
Proc Natl Acad Sci U S A ; 108(21): E128-35, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21555590

ABSTRACT

The most common monogenic cause of small-vessel disease leading to ischemic stroke and vascular dementia is the neurodegenerative syndrome cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), which is associated with mutations in the Notch 3 receptor. CADASIL pathology is characterized by vascular smooth muscle cell degeneration and accumulation of diagnostic granular osmiophilic material (GOM) in vessels. The functional nature of the Notch 3 mutations causing CADASIL and their mechanistic connection to small-vessel disease and GOM accumulation remain enigmatic. To gain insight into how Notch 3 function is linked to CADASIL pathophysiology, we studied two phenotypically distinct mutations, C455R and R1031C, respectively associated with early and late onset of stroke, by using hemodynamic analyses in transgenic mouse models, receptor activity assays in cell culture, and proteomic examination of postmortem human tissue. We demonstrate that the C455R and R1031C mutations define different hypomorphic activity states of Notch 3, a property linked to ischemic stroke susceptibility in mouse models we generated. Importantly, these mice develop osmiophilic deposits and other age-dependent phenotypes that parallel remarkably the human condition. Proteomic analysis of human brain vessels, carrying the same CADASIL mutations, identified clusterin and collagen 18 α1/endostatin as GOM components. Our findings link loss of Notch signaling with ischemic cerebral small-vessel disease, a prevalent human condition. We determine that CADASIL pathophysiology is associated with hypomorphic Notch 3 function in vascular smooth muscle cells and implicate the accumulation of clusterin and collagen 18 α1/endostatin in brain vessel pathology.


Subject(s)
Alleles , Arterioles/pathology , Cerebrovascular Disorders/etiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Brain/blood supply , Disease Models, Animal , Humans , Ischemia , Mice , Mutation, Missense , Receptor, Notch3 , Receptors, Notch/genetics , Transgenes
15.
Dev Cell ; 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38944040

ABSTRACT

We describe a next-generation Drosophila protein interaction map-"DPIM2"-established from affinity purification-mass spectrometry of 5,805 baits, covering the largest fraction of the Drosophila proteome. The network contains 32,668 interactions among 3,644 proteins, organized into 632 clusters representing putative functional modules. Our analysis expands the pool of known protein interactions in Drosophila, provides annotation for poorly studied genes, and postulates previously undescribed protein interaction relationships. The predictive power and functional relevance of this network are probed through the lens of the Notch signaling pathway, and we find that newly identified members of complexes that include known Notch modifiers can also modulate Notch signaling. DPIM2 allows direct comparisons with a recently published human protein interaction network, defining the existence of functional interactions conserved across species. Thus, DPIM2 defines a valuable resource for predicting protein co-complex memberships and functional associations as well as generates functional hypotheses regarding specific protein interactions.

16.
J Cell Sci ; 124(Pt 7): 1067-76, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21402876

ABSTRACT

Notch receptors and their ligands have crucial roles in development and tumorigenesis. We present evidence demonstrating the existence of an antagonistic relationship between Notch 4 and Trp53, which is controlled by the Mdm2-dependent ubiquitylation and degradation of the Notch receptor. We show that this signal-controlling mechanism is mediated by physical interactions between Mdm2 and Notch 4 and suggest the existence of a trimeric complex between Trp53, Notch 4 and Mdm2, which ultimately regulates Notch activity. Functional studies indicate that Trp53 can suppress NICD4-induced anchorage-independent growth in mammary epithelial cells and present evidence showing that Trp53 has a pivotal role in the suppression of Notch-associated tumorigenesis in the mammary gland.


Subject(s)
Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Epithelial Cells/chemistry , Epithelial Cells/metabolism , Humans , Mice , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Receptor, Notch4 , Receptors, Notch/chemistry , Receptors, Notch/genetics , Tumor Suppressor Protein p53/genetics
17.
PLoS Genet ; 6(10): e1001172, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-21124729

ABSTRACT

Spinal Muscular Atrophy (SMA) is caused by diminished function of the Survival of Motor Neuron (SMN) protein, but the molecular pathways critical for SMA pathology remain elusive. We have used genetic approaches in invertebrate models to identify conserved SMN loss of function modifier genes. Drosophila melanogaster and Caenorhabditis elegans each have a single gene encoding a protein orthologous to human SMN; diminished function of these invertebrate genes causes lethality and neuromuscular defects. To find genes that modulate SMN function defects across species, two approaches were used. First, a genome-wide RNAi screen for C. elegans SMN modifier genes was undertaken, yielding four genes. Second, we tested the conservation of modifier gene function across species; genes identified in one invertebrate model were tested for function in the other invertebrate model. Drosophila orthologs of two genes, which were identified originally in C. elegans, modified Drosophila SMN loss of function defects. C. elegans orthologs of twelve genes, which were originally identified in a previous Drosophila screen, modified C. elegans SMN loss of function defects. Bioinformatic analysis of the conserved, cross-species, modifier genes suggests that conserved cellular pathways, specifically endocytosis and mRNA regulation, act as critical genetic modifiers of SMN loss of function defects across species.


Subject(s)
Genome-Wide Association Study/methods , Invertebrates/genetics , RNA Interference , SMN Complex Proteins/genetics , Analysis of Variance , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Evolution, Molecular , Female , Genome, Helminth/genetics , Genome, Insect/genetics , Humans , Invertebrates/growth & development , Male , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/physiopathology , Mutation , Pupa/genetics , Pupa/growth & development , SMN Complex Proteins/physiology
18.
Proc Natl Acad Sci U S A ; 107(11): 5012-7, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20194747

ABSTRACT

Aberrant activation of Notch receptors has been implicated in breast cancer; however, the mechanisms contributing to Notch-dependent transformation remain elusive because Notch displays dichotomous functional activities, promoting both proliferation and growth arrest. We investigated the cellular basis for the heterogeneous responses to Notch pathway activation in 3D cultures of MCF-10A mammary epithelial cells. Expression of a constitutively active Notch-1 intracellular domain (NICD) was found to induce two distinct types of 3D structures: large, hyperproliferative structures and small, growth-arrested structures with reduced cell-to-matrix adhesion. Interestingly, we found that these heterogeneous phenotypes reflect differences in Notch pathway activation levels; high Notch activity caused down-regulation of multiple matrix-adhesion genes and inhibition of proliferation, whereas low Notch activity maintained matrix adhesion and provoked a strong hyperproliferative response. Moreover, microarray analyses implicated NICD-induced p63 down-regulation in loss of matrix adhesion. In addition, a reverse-phase protein array-based analysis and subsequent loss-of-function studies identified STAT3 as a dominant downstream mediator of the NICD-induced outgrowth. These results indicate that the phenotypic responses to Notch are determined by the dose of pathway activation; and this dose affects the balance between growth-stimulative and growth-suppressive effects. This unique feature of Notch signaling provides insights into mechanisms that contribute to the dichotomous effects of Notch during development and tumorigenesis.


Subject(s)
Epithelial Cells/metabolism , Mammary Glands, Human/cytology , Receptor, Notch1/metabolism , Signal Transduction , Cell Adhesion , Cell Proliferation , Cells, Cultured , Epithelial Cells/cytology , Extracellular Matrix/metabolism , Female , Humans , Phenotype , Protein Structure, Tertiary , Receptor, Notch1/chemistry , STAT3 Transcription Factor/metabolism , Trans-Activators/metabolism , Transcription Factors , Tumor Suppressor Proteins/metabolism
19.
Nat Cell Biol ; 7(12): 1191-201, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16284625

ABSTRACT

Signalling activity of the Notch receptor, which plays a fundamental role in metazoan cell fate determination, is controlled at multiple levels. We uncovered a Notch signal-controlling mechanism that depends on the ability of the non-visual beta-arrestin, Kurtz (Krz), to influence the degradation and, consequently, the function of the Notch receptor. We identified Krz as a binding partner of a known Notch-pathway modulator, Deltex (Dx), and demonstrated the existence of a trimeric Notch-Dx-Krz protein complex. This complex mediates the degradation of the Notch receptor through a ubiquitination-dependent pathway. Our results establish a novel mode of regulation of Notch signalling and define a new function for non-visual beta-arrestins.


Subject(s)
Arrestins/physiology , Drosophila Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Arrestins/genetics , Arrestins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Female , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Multiprotein Complexes , Mutation/physiology , Two-Hybrid System Techniques , Ubiquitin/metabolism , Wings, Animal/growth & development , beta-Arrestins
20.
Proc Natl Acad Sci U S A ; 106(7): 2218-23, 2009 Feb 17.
Article in English | MEDLINE | ID: mdl-19171888

ABSTRACT

Metazoans use a handful of highly conserved signaling pathways to create a signaling backbone that governs development. How these few signals have such a versatile action likely depends upon the larger-scale network they form through integration, as exemplified by cross-talk between the Notch and receptor tyrosine kinase (RTK) pathways. We examined the transcriptional output of Notch-RTK cross-talk during Drosophila development and present in vivo data supporting a role for selected mutually regulated genes in signal integration. Interestingly, Notch-RTK integration did not lead to general antagonism of either pathway, as is commonly believed. Instead, integration had a combinatorial effect on specific cross-regulated targets, which unexpectedly included numerous core components of the RTK and other major signaling pathways (TGF-beta, Hh, Jak/Stat, nuclear receptor and Wnt). We find the majority of Ras-responsive genes are also Notch-responsive, suggesting Notch may function to specify the response to Ras activation.


Subject(s)
Gene Expression Regulation , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Notch/metabolism , ras Proteins/metabolism , Animals , Crosses, Genetic , Drosophila melanogaster , Homozygote , Janus Kinase 1/metabolism , Models, Biological , Models, Genetic , Oligonucleotide Array Sequence Analysis , Receptors, Notch/genetics , Signal Transduction , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL