Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters











Publication year range
1.
Development ; 150(11)2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37294169

ABSTRACT

Active Notch signalling is elicited through receptor-ligand interactions that result in release of the Notch intracellular domain (NICD), which translocates into the nucleus. NICD activates transcription at target genes, forming a complex with the DNA-binding transcription factor CSL [CBF1/Su(H)/LAG-1] and co-activator Mastermind. However, CSL lacks its own nuclear localisation sequence, and it remains unclear where the tripartite complex is formed. To probe the mechanisms involved, we designed an optogenetic approach to control NICD release (OptIC-Notch) and monitored the subsequent complex formation and target gene activation. Strikingly, we observed that, when uncleaved, OptIC-Notch sequestered CSL in the cytoplasm. Hypothesising that exposure of a juxta membrane ΦWΦP motif is key to sequestration, we masked this motif with a second light-sensitive domain (OptIC-Notch{ω}), which was sufficient to prevent CSL sequestration. Furthermore, NICD produced by light-induced cleavage of OptIC-Notch or OptIC-Notch{ω} chaperoned CSL into the nucleus and induced target gene expression, showing efficient light-controlled activation. Our results demonstrate that exposure of the ΦWΦP motif leads to CSL recruitment and suggest this can occur in the cytoplasm prior to nuclear entry.


Subject(s)
Receptors, Notch , Transcription Factors , Receptors, Notch/metabolism , Transcription Factors/metabolism , Gene Expression Regulation , Transcriptional Activation , Signal Transduction/physiology
2.
Development ; 149(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-35005772

ABSTRACT

Aggressive neoplastic growth can be initiated by a limited number of genetic alterations, such as the well-established cooperation between loss of cell architecture and hyperactive signaling pathways. However, our understanding of how these different alterations interact and influence each other remains very incomplete. Using Drosophila paradigms of imaginal wing disc epithelial growth, we have monitored the changes in Notch pathway activity according to the polarity status of cells (scrib mutant). We show that the scrib mutation impacts the direct transcriptional output of the Notch pathway, without altering the global distribution of Su(H), the Notch-dedicated transcription factor. The Notch-dependent neoplasms require, however, the action of a group of transcription factors, similar to those previously identified for Ras/scrib neoplasm (namely AP-1, Stat92E, Ftz-F1 and basic leucine zipper factors), further suggesting the importance of this transcription factor network during neoplastic growth. Finally, our work highlights some Notch/scrib specificities, in particular the role of the PAR domain-containing basic leucine zipper transcription factor and Notch direct target Pdp1 for neoplastic growth.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Receptors, Notch/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Carcinogenesis , Drosophila/growth & development , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Larva/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Neoplasms/metabolism , Neoplasms/pathology , RNA Interference , Signal Transduction , Wings, Animal/metabolism
3.
Development ; 148(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34486648

ABSTRACT

The Notch pathway mediates cell-to-cell communication in a variety of tissues, developmental stages and organisms. Pathway activation relies on the interaction between transmembrane ligands and receptors on adjacent cells. As such, pathway activity could be influenced by the size, composition or dynamics of contacts between membranes. The initiation of Notch signalling in the Drosophila embryo occurs during cellularization, when lateral cell membranes and adherens junctions are first being deposited, allowing us to investigate the importance of membrane architecture and specific junctional domains for signalling. By measuring Notch-dependent transcription in live embryos, we established that it initiates while lateral membranes are growing and that signalling onset correlates with a specific phase in their formation. However, the length of the lateral membranes per se was not limiting. Rather, the adherens junctions, which assemble concurrently with membrane deposition, contributed to the high levels of signalling required for transcription, as indicated by the consequences of α-Catenin depletion. Together, these results demonstrate that the establishment of lateral membrane contacts can be limiting for Notch trans-activation and suggest that adherens junctions play an important role in modulating Notch activity.


Subject(s)
Adherens Junctions/metabolism , Drosophila Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction , Adherens Junctions/ultrastructure , Animals , Drosophila melanogaster , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , alpha Catenin/metabolism
4.
EMBO Rep ; 22(10): e52729, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34347930

ABSTRACT

Accurate Notch signalling is critical for development and homeostasis. Fine-tuning of Notch-ligand interactions has substantial impact on signalling outputs. Recent structural studies have identified a conserved N-terminal C2 domain in human Notch ligands which confers phospholipid binding in vitro. Here, we show that Drosophila ligands Delta and Serrate adopt the same C2 domain structure with analogous variations in the loop regions, including the so-called ß1-2 loop that is involved in phospholipid binding. Mutations in the ß1-2 loop of the Delta C2 domain retain Notch binding but have impaired ability to interact with phospholipids in vitro. To investigate its role in vivo, we deleted five residues within the ß1-2 loop of endogenous Delta. Strikingly, this change compromises ligand function. The modified Delta enhances phenotypes produced by Delta loss-of-function alleles and suppresses that of Notch alleles. As the modified protein is present on the cell surface in normal amounts, these results argue that C2 domain phospholipid binding is necessary for robust signalling in vivo fine-tuning the balance of trans and cis ligand-receptor interactions.


Subject(s)
Drosophila Proteins , Receptors, Notch , C2 Domains , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Humans , Ligands , Membrane Proteins , Phospholipids , Receptors, Notch/genetics
5.
Neural Dev ; 15(1): 10, 2020 08 02.
Article in English | MEDLINE | ID: mdl-32741363

ABSTRACT

BACKGROUND: Drosophila neuroblasts (NBs) are neural stem cells whose maintenance relies on Notch activity. NBs proliferate throughout larval stages to generate a large number of adult neurons. Their proliferation is protected under conditions of nutrition restriction but the mechanisms responsible are not fully understood. As amino acid transporters (Solute Carrier transporters, SLCs), such as SLC36, have important roles in coupling nutrition inputs to growth pathways, they may have a role in this process. For example, an SLC36 family transporter Pathetic (Path) that supports body size and neural dendrite growth in Drosophila, was identified as a putative Notch target in genome-wide studies. However, its role in sustaining stem cell proliferation and maintenance has not been investigated. This study aimed to investigate the function of Path in the larval NBs and to determine whether it is involved in protecting them from nutrient deprivation. METHODS: The expression and regulation of Path in the Drosophila larval brain was analysed using a GFP knock-in allele and reporter genes containing putative Notch regulated enhancers. Path function in NB proliferation and overall brain growth was investigated under different nutrition conditions by depleting it from specific cell types in the CNS, using mitotic recombination to generate mutant clones or by directed RNA-interference. RESULTS: Path is expressed in both NBs and glial cells in the Drosophila CNS. In NBs, path is directly targeted by Notch signalling via Su(H) binding at an intronic enhancer, PathNRE. This enhancer is responsive to Notch regulation both in cell lines and in vivo. Loss of path in neural stem cells delayed proliferation, consistent with it having a role in NB maintenance. Expression from pathNRE was compromised in conditions of amino acid deprivation although other Notch regulated enhancers are unaffected. However, NB-expressed Path was not required for brain sparing under amino acid deprivation. Instead, it appears that Path is important in glial cells to help protect brain growth under conditions of nutrient restriction. CONCLUSIONS: We identify a novel Notch target gene path that is required in NBs for neural stem cell proliferation, while in glia it protects brain growth under nutrition restriction.


Subject(s)
Animal Nutritional Physiological Phenomena/physiology , Brain/growth & development , Cell Proliferation/physiology , Drosophila Proteins/physiology , Gene Expression Regulation, Developmental/physiology , Larva/growth & development , Neural Stem Cells/physiology , Animals , Brain/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , Larva/metabolism
6.
Dev Growth Differ ; 62(1): 4-14, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31886523

ABSTRACT

Notch signalling controls many key cellular processes which differ according to the context where the pathway is deployed due to the transcriptional activation of specific sets of genes. The pathway is unusual in its lack of amplification, also raising the question of how it can efficiently activate transcription with limited amounts of nuclear activity. Here, we focus on mechanisms that enable Notch to produce appropriate transcriptional responses and speculate on models that could explain the current gaps in knowledge.


Subject(s)
Receptors, Notch/metabolism , Signal Transduction/physiology , Transcriptional Activation/physiology , Animals , Humans , Receptors, Notch/genetics
7.
Sci Rep ; 9(1): 18628, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31819141

ABSTRACT

In all metazoans, a small number of evolutionarily conserved signaling pathways are reiteratively used during development to orchestrate critical patterning and morphogenetic processes. Among these, Notch (N) signaling is essential for most aspects of tissue patterning where it mediates the communication between adjacent cells to control cell fate specification. In Drosophila, Notch signaling is required for several features of eye development, including the R3/R4 cell fate choice and R7 specification. Here we show that hypomorphic alleles of Notch, belonging to the Nfacet class, reveal a novel phenotype: while photoreceptor specification in the mutant ommatidia is largely normal, defects are observed in ommatidial rotation (OR), a planar cell polarity (PCP)-mediated cell motility process. We demonstrate that during OR Notch signaling is specifically required in the R4 photoreceptor to upregulate the transcription of argos (aos), an inhibitory ligand to the epidermal growth factor receptor (EGFR), to fine-tune the activity of EGFR signaling. Consistently, the loss-of-function defects of Nfacet alleles and EGFR-signaling pathway mutants are largely indistinguishable. A Notch-regulated aos enhancer confers R4 specific expression arguing that aos is directly regulated by Notch signaling in this context via Su(H)-Mam-dependent transcription.


Subject(s)
Drosophila Proteins/genetics , ErbB Receptors/genetics , Eye Proteins/genetics , Nerve Tissue Proteins/genetics , Receptors, Invertebrate Peptide/genetics , Receptors, Notch/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Polarity/genetics , Drosophila melanogaster/genetics , Eye/metabolism , Frizzled Receptors/genetics , Morphogenesis/genetics , Photoreceptor Cells, Invertebrate/metabolism
8.
Dev Cell ; 50(4): 411-425.e8, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31378591

ABSTRACT

Information from developmental signaling pathways must be accurately decoded to generate transcriptional outcomes. In the case of Notch, the intracellular domain (NICD) transduces the signal directly to the nucleus. How enhancers decipher NICD in the real time of developmental decisions is not known. Using the MS2-MCP system to visualize nascent transcripts in single cells in Drosophila embryos, we reveal how two target enhancers read Notch activity to produce synchronized and sustained profiles of transcription. By manipulating the levels of NICD and altering specific motifs within the enhancers, we uncover two key principles. First, increased NICD levels alter transcription by increasing duration rather than frequency of transcriptional bursts. Second, priming of enhancers by tissue-specific transcription factors is required for NICD to confer synchronized and sustained activity; in their absence, transcription is stochastic and bursty. The dynamic response of an individual enhancer to NICD thus differs depending on the cellular context.


Subject(s)
Drosophila Proteins/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , Receptors, Notch/genetics , Transcription Factors/genetics , Transcription, Genetic , Twist-Related Protein 1/genetics , Animals , Animals, Genetically Modified/genetics , Binding Sites/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/physiology , Embryonic Development/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Regulatory Sequences, Nucleic Acid/genetics , Signal Transduction/genetics , Transcriptional Activation/genetics
9.
EMBO Rep ; 20(5)2019 05.
Article in English | MEDLINE | ID: mdl-30914409

ABSTRACT

Notch signaling plays a key role in many cell fate decisions during development by directing different gene expression programs via the transcription factor CSL, known as Su(H) in Drosophila Which target genes are responsive to Notch signaling is influenced by the chromatin state of enhancers, yet how this is regulated is not fully known. Detecting a specific increase in the histone variant H3.3 in response to Notch signaling, we tested which chromatin remodelers or histone chaperones are required for the changes in enhancer accessibility to Su(H) binding. We show a crucial role for the Brahma SWI/SNF chromatin remodeling complex, including the actin-related BAP55 subunit, in conferring enhancer accessibility and enabling the transcriptional response to Notch activity. The Notch-responsive regions have high levels of nucleosome turnover which depend on the Brahma complex, increase in magnitude with Notch signaling, and primarily involve histone H3.3. Together these results highlight the importance of SWI/SNF-mediated nucleosome turnover in rendering enhancers responsive to Notch.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromatin/genetics , Drosophila Proteins/genetics , Receptors, Notch/genetics , Regulatory Sequences, Nucleic Acid/genetics , Acetylation , Animals , Chromosomal Proteins, Non-Histone/genetics , Drosophila/genetics , Gene Expression Regulation , Histones/genetics , Nucleosomes/genetics , Transcription Factors/genetics , Transcription, Genetic/genetics
10.
Dev Cell ; 44(5): 611-623.e7, 2018 03 12.
Article in English | MEDLINE | ID: mdl-29478922

ABSTRACT

A key feature of Notch signaling is that it directs immediate changes in transcription via the DNA-binding factor CSL, switching it from repression to activation. How Notch generates both a sensitive and accurate response-in the absence of any amplification step-remains to be elucidated. To address this question, we developed real-time analysis of CSL dynamics including single-molecule tracking in vivo. In Notch-OFF nuclei, a small proportion of CSL molecules transiently binds DNA, while in Notch-ON conditions CSL recruitment increases dramatically at target loci, where complexes have longer dwell times conferred by the Notch co-activator Mastermind. Surprisingly, recruitment of CSL-related corepressors also increases in Notch-ON conditions, revealing that Notch induces cooperative or "assisted" loading by promoting local increase in chromatin accessibility. Thus, in vivo Notch activity triggers changes in CSL dwell times and chromatin accessibility, which we propose confer sensitivity to small input changes and facilitate timely shut-down.


Subject(s)
Cell Nucleus/genetics , DNA-Binding Proteins/metabolism , DNA/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Receptors, Notch/metabolism , Animals , Cell Nucleus/metabolism , DNA/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Models, Molecular , Protein Binding , Receptors, Notch/genetics , Signal Transduction , Transcriptional Activation
11.
Curr Opin Cell Biol ; 51: 103-109, 2018 04.
Article in English | MEDLINE | ID: mdl-29289895

ABSTRACT

The discovery that Notch activation involves a proteolytic cleavage to release the intracellular domain (NICD) revolutionized the field of Notch signaling. It resulted in a simple model whereby the cleaved NICD enters the nucleus and activates expression of genes by forming a DNA bound complex with CSL. However, is it really this simple? The realization that the outcome from activating Notch varies greatly from cell to cell raised many questions about what governs the target gene selections in different cell types. Insights have come from recent genome-wide studies, which highlight the importance of tissue-specific transcription factors and epigenetics. Co-factors also have been identified that participate in the regulation of enhancers. Finally, it is generally assumed that once cleaved, NICD goes on to do its job, but with a burgeoning number of post-translations, it may not be that simple.


Subject(s)
Receptors, Notch/metabolism , Transcription Factors/metabolism , Humans , Signal Transduction
12.
PLoS Genet ; 13(11): e1007096, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29155828

ABSTRACT

Repressors are frequently deployed to limit the transcriptional response to signalling pathways. For example, several co-repressors interact directly with the DNA-binding protein CSL and are proposed to keep target genes silenced in the absence of Notch activity. However, the scope of their contributions remains unclear. To investigate co-repressor activity in the context of this well defined signalling pathway, we have analysed the genome-wide binding profile of the best-characterized CSL co-repressor in Drosophila, Hairless, and of a second CSL interacting repressor, SMRTER. As predicted there was significant overlap between Hairless and its CSL DNA-binding partner, both in Kc cells and in wing discs, where they were predominantly found in chromatin with active enhancer marks. However, while the Hairless complex was widely present at some Notch regulated enhancers in the wing disc, no binding was detected at others, indicating that it is not essential for silencing per se. Further analysis of target enhancers confirmed differential requirements for Hairless. SMRTER binding significantly overlapped with Hairless, rather than complementing it, and many enhancers were apparently co-bound by both factors. Our analysis indicates that the actions of Hairless and SMRTER gate enhancers to Notch activity and to Ecdysone signalling respectively, to ensure that the appropriate levels and timing of target gene expression are achieved.


Subject(s)
Drosophila Proteins/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Transcription Factors/genetics , Animals , Binding Sites , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , DNA-Binding Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Ecdysone/metabolism , Gene Expression Regulation, Developmental/genetics , Genomics , Protein Binding , Regulatory Sequences, Nucleic Acid/genetics , Repressor Proteins/genetics , Signal Transduction , Transcription Factors/metabolism
13.
Dis Model Mech ; 10(4): 399-407, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28237966

ABSTRACT

Myeloproliferative neoplasms (MPNs) of the Philadelphia-negative class comprise polycythaemia vera, essential thrombocythaemia and primary myelofibrosis (PMF). They are associated with aberrant numbers of myeloid lineage cells in the blood, and in the case of overt PMF, with development of myelofibrosis in the bone marrow and failure to produce normal blood cells. These diseases are usually caused by gain-of-function mutations in the kinase JAK2. Here, we use Drosophila to investigate the consequences of activation of the JAK2 orthologue in haematopoiesis. We have identified maturing haemocytes in the lymph gland, the major haematopoietic organ in the fly, as the cell population susceptible to induce hypertrophy upon targeted overexpression of JAK. We show that JAK activates a feed-forward loop, including the cytokine-like ligand Upd3 and its receptor, Domeless, which are required to induce lymph gland hypertrophy. Moreover, we present evidence that p38 MAPK signalling plays a key role in this process by inducing expression of the ligand Upd3. Interestingly, we also show that forced activation of the p38 MAPK pathway in maturing haemocytes suffices to generate hypertrophic organs and the appearance of melanotic tumours. Our results illustrate a novel pro-tumourigenic crosstalk between the p38 MAPK pathway and JAK signalling in a Drosophila model of MPNs. Based on the shared molecular mechanisms underlying MPNs in flies and humans, the interplay between Drosophila JAK and p38 signalling pathways unravelled in this work might have translational relevance for human MPNs.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Janus Kinases/metabolism , MAP Kinase Signaling System , Myeloproliferative Disorders/pathology , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Disease Models, Animal , Hemocytes/metabolism , Hypertrophy , Lymph Nodes/metabolism
14.
Nat Rev Mol Cell Biol ; 17(11): 722-735, 2016 11.
Article in English | MEDLINE | ID: mdl-27507209

ABSTRACT

The highly conserved Notch signalling pathway functions in many different developmental and homeostatic processes, which raises the question of how this pathway can achieve such diverse outcomes. With a direct route from the membrane to the nucleus, the Notch pathway has fewer opportunities for regulation than do many other signalling pathways, yet it generates exquisitely patterned structures, including sensory hair cells and branched arterial networks. More confusingly, its activity promotes tissue growth and cancers in some circumstances but cell death and tumour suppression in others. Many different regulatory mechanisms help to shape the activity of the Notch pathway, generating functional outputs that are appropriate for each context. These mechanisms include the receptor-ligand landscape, the tissue topology, the nuclear environment and the connectivity of the regulatory networks.


Subject(s)
Receptors, Notch/physiology , Signal Transduction , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Gene Expression Regulation , Humans , Morphogenesis , Neovascularization, Physiologic
15.
Open Biol ; 6(2): 150155, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26887408

ABSTRACT

Glycolytic shift is a characteristic feature of rapidly proliferating cells, such as cells during development and during immune response or cancer cells, as well as of stem cells. It results in increased glycolysis uncoupled from mitochondrial respiration, also known as the Warburg effect. Notch signalling is active in contexts where cells undergo glycolytic shift. We decided to test whether metabolic genes are direct transcriptional targets of Notch signalling and whether upregulation of metabolic genes can help Notch to induce tissue growth under physiological conditions and in conditions of Notch-induced hyperplasia. We show that genes mediating cellular metabolic changes towards the Warburg effect are direct transcriptional targets of Notch signalling. They include genes encoding proteins involved in glucose uptake, glycolysis, lactate to pyruvate conversion and repression of the tricarboxylic acid cycle. The direct transcriptional upregulation of metabolic genes is PI3K/Akt independent and occurs not only in cells with overactivated Notch but also in cells with endogenous levels of Notch signalling and in vivo. Even a short pulse of Notch activity is able to elicit long-lasting metabolic changes resembling the Warburg effect. Loss of Notch signalling in Drosophila wing discs as well as in human microvascular cells leads to downregulation of glycolytic genes. Notch-driven tissue overgrowth can be rescued by downregulation of genes for glucose metabolism. Notch activity is able to support growth of wing during nutrient-deprivation conditions, independent of the growth of the rest of the body. Notch is active in situations that involve metabolic reprogramming, and the direct regulation of metabolic genes may be a common mechanism that helps Notch to exert its effects in target tissues.


Subject(s)
Citric Acid Cycle/genetics , Gene Expression Regulation , Glycolysis/genetics , Receptors, Notch/metabolism , Animals , Binding Sites , Cell Line , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Energy Metabolism/genetics , Gene Expression , Genes, Reporter , Humans , Models, Biological , Promoter Regions, Genetic , Protein Binding , Receptors, Notch/genetics , Regulatory Sequences, Nucleic Acid , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcriptional Activation
16.
Development ; 143(2): 219-31, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26657768

ABSTRACT

Notch signalling is involved in a multitude of developmental decisions and its aberrant activation is linked to many diseases, including cancers. One example is the neural stem cell tumours that arise from constitutive Notch activity in Drosophila neuroblasts. To investigate how hyperactivation of Notch in larval neuroblasts leads to tumours, we combined results from profiling the upregulated mRNAs and mapping the regions bound by the core Notch pathway transcription factor Su(H). This identified 246 putative direct Notch targets. These genes were highly enriched for transcription factors and overlapped significantly with a previously identified regulatory programme dependent on the proneural transcription factor Asense. Included were genes associated with the neuroblast maintenance and self-renewal programme that we validated as Notch regulated in vivo. Another group were the so-called temporal transcription factors, which have been implicated in neuroblast maturation. Normally expressed in specific time windows, several temporal transcription factors were ectopically expressed in the stem cell tumours, suggesting that Notch had reprogrammed their normal temporal regulation. Indeed, the Notch-induced hyperplasia was reduced by mutations affecting two of the temporal factors, which, conversely, were sufficient to induce mild hyperplasia on their own. Altogether, the results suggest that Notch induces neuroblast tumours by directly promoting the expression of genes that contribute to stem cell identity and by reprogramming the expression of factors that could regulate maturity.


Subject(s)
Drosophila Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Receptors, Notch/metabolism , Animals , Drosophila , Drosophila Proteins/genetics , Drosophila melanogaster , Receptors, Notch/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
17.
J Cell Biol ; 210(2): 303-18, 2015 Jul 20.
Article in English | MEDLINE | ID: mdl-26169355

ABSTRACT

Notch signaling is a major regulator of cell fate, proliferation, and differentiation. Like other signaling pathways, its activity is strongly influenced by intracellular trafficking. Besides contributing to signal activation and down-regulation, differential fluxes between trafficking routes can cause aberrant Notch pathway activation. Investigating the function of the retromer-associated DNAJ protein Rme-8 in vivo, we demonstrate a critical role in regulating Notch receptor recycling. In the absence of Rme-8, Notch accumulated in enlarged tubulated Rab4-positive endosomes, and as a consequence, signaling was compromised. Strikingly, when the retromer component Vps26 was depleted at the same time, Notch no longer accumulated and instead was ectopically activated. Likewise, depletion of ESCRT-0 components Hrs or Stam in combination with Rme-8 also led to high levels of ectopic Notch activity. Together, these results highlight the importance of Rme-8 in coordinating normal endocytic recycling route and reveal that its absence predisposes toward conditions in which pathological Notch signaling can occur.


Subject(s)
Drosophila Proteins/metabolism , Receptors, Notch/metabolism , Animals , Disintegrins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Gene Knockdown Techniques , Metalloendopeptidases/metabolism , Protein Transport , Signal Transduction , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , Wnt1 Protein/metabolism
18.
EMBO J ; 34(14): 1889-904, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26069324

ABSTRACT

The conserved Notch pathway functions in diverse developmental and disease-related processes, requiring mechanisms to ensure appropriate target selection and gene activation in each context. To investigate the influence of chromatin organisation and dynamics on the response to Notch signalling, we partitioned Drosophila chromatin using histone modifications and established the preferred chromatin conditions for binding of Su(H), the Notch pathway transcription factor. By manipulating activity of a co-operating factor, Lozenge/Runx, we showed that it can help facilitate these conditions. While many histone modifications were unchanged by Su(H) binding or Notch activation, we detected rapid changes in acetylation of H3K56 at Notch-regulated enhancers. This modification extended over large regions, required the histone acetyl-transferase CBP and was independent of transcription. Such rapid changes in H3K56 acetylation appear to be a conserved indicator of enhancer activation as they also occurred at the mammalian Notch-regulated Hey1 gene and at Drosophila ecdysone-regulated genes. This intriguing example of a core histone modification increasing over short timescales may therefore underpin changes in chromatin accessibility needed to promote transcription following signalling activation.


Subject(s)
Chromatin/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Enhancer Elements, Genetic , Histones/metabolism , Receptors, Notch/metabolism , Repressor Proteins/metabolism , Acetylation , Animals , Cell Cycle Proteins/genetics , DNA, Intergenic , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Ecdysone/metabolism , Gene Expression Regulation , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Histones/genetics , Receptors, Notch/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
19.
Development ; 142(6): 1102-12, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25725070

ABSTRACT

Morphogenesis is crucial during development to generate organs and tissues of the correct size and shape. During Drosophila late eye development, interommatidial cells (IOCs) rearrange to generate the highly organized pupal lattice, in which hexagonal ommatidial units pack tightly. This process involves the fine regulation of adherens junctions (AJs) and of adhesive E-Cadherin (E-Cad) complexes. Localized accumulation of Bazooka (Baz), the Drosophila PAR3 homolog, has emerged as a critical step to specify where new E-Cad complexes should be deposited during junction remodeling. However, the mechanisms controlling the correct localization of Baz are still only partly understood. We show here that Drosophila Magi, the sole fly homolog of the mammalian MAGI scaffolds, is an upstream regulator of E-Cad-based AJs during cell rearrangements, and that Magi mutant IOCs fail to reach their correct position. We uncover a direct physical interaction between Magi and the Ras association domain protein RASSF8 through a WW domain-PPxY motif binding, and show that apical Magi recruits the RASSF8-ASPP complex during AJ remodeling in IOCs. We further show that this Magi complex is required for the cortical recruitment of Baz and of the E-Cad-associated proteins α- and ß-catenin. We propose that, by controlling the proper localization of Baz to remodeling junctions, Magi and the RASSF8-ASPP complex promote the recruitment or stabilization of E-Cad complexes at junction sites.


Subject(s)
Adherens Junctions/physiology , Cadherins/metabolism , Carrier Proteins/metabolism , Drosophila Proteins/metabolism , Eye/embryology , Morphogenesis/physiology , Nucleoside-Phosphate Kinase/metabolism , Adherens Junctions/metabolism , Animals , Blotting, Western , Drosophila , Immunohistochemistry , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/metabolism , Plasmids/genetics , Two-Hybrid System Techniques
20.
Nucleic Acids Res ; 42(16): 10550-63, 2014.
Article in English | MEDLINE | ID: mdl-25114055

ABSTRACT

Regulation of transcription is fundamental to development and physiology, and occurs through binding of transcription factors to specific DNA sequences in the genome. CSL (CBF1/Suppressor of Hairless/LAG-1), a core component of the Notch signaling pathway, is one such transcription factor that acts in concert with co-activators or co-repressors to control the activity of associated target genes. One fundamental question is how CSL can recognize and select among different DNA sequences available in vivo and whether variations between selected sequences can influence its function. We have therefore investigated CSL-DNA recognition using computational approaches to analyze the energetics of CSL bound to different DNAs and tested the in silico predictions with in vitro and in vivo assays. Our results reveal novel aspects of CSL binding that may help explain the range of binding observed in vivo. In addition, using molecular dynamics simulations, we show that domain-domain correlations within CSL differ significantly depending on the DNA sequence bound, suggesting that different DNA sequences may directly influence CSL function. Taken together, our results, based on computational chemistry approaches, provide valuable insights into transcription factor-DNA binding, in this particular case increasing our understanding of CSL-DNA interactions and how these may impact on its transcriptional control.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Regulatory Elements, Transcriptional , Binding Sites , Computer Simulation , Consensus Sequence , Cytosine/analysis , DNA/chemistry , DNA/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Molecular Dynamics Simulation , Nucleotide Motifs , Protein Binding , Software
SELECTION OF CITATIONS
SEARCH DETAIL