Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
Int Microbiol ; 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39172274

ABSTRACT

The honeybee (Apis mellifera) is a key pollinator critical to global agriculture, facing threats from various stressors, including the ectoparasitic Varroa mite (Varroa destructor). Previous studies have identified shared bacteria between Varroa mites and honeybees, yet it remains unclear if these bacteria assemble similarly in both species. This study builds on existing knowledge by investigating co-occurrence patterns in the microbiomes of both Varroa mites and honeybees, shedding light on potential interactions. Leveraging 16S rRNA datasets, we conducted co-occurrence network analyses, explored Core Association Networks (CAN) and assess network robustness. Comparative network analyses revealed structural differences between honeybee and mite microbiomes, along with shared core features and microbial motifs. The mite network exhibited lower robustness, suggesting less resistance to taxa extension compared to honeybees. Furthermore, analyses of predicted functional profiling and taxa contribution revealed that common central pathways in the metabolic networks have different taxa contributing to Varroa mites and honeybee microbiomes. The results show that while both microbial systems exhibit functional redundancy, in which different taxa contribute to the functional stability and resilience of the ecosystem, there is evidence for niche specialization resulting in unique contributions to specific pathways in each part of this host-parasite system. The specificity of taxa contribution to key pathways offers targeted approaches to Varroa microbiome management and preserving honeybee microbiome. Our findings provide valuable insights into microbial interactions, aiding farmers and beekeepers in maintaining healthy and resilient bee colonies amid increasing Varroa mite infestations.

2.
J Insect Physiol ; 152: 104583, 2024 01.
Article in English | MEDLINE | ID: mdl-37979771

ABSTRACT

Deformed wing virus (DWV) transmitted by the parasitic mite Varroa destructor is one of the most significant factors contributing to massive losses of managed colonies of western honey bee (Apis mellifera) subspecies of European origin reported worldwide in recent decades. Despite this fact, no antiviral treatment against honey bee viruses is currently available for practical applications and the level of viral infection can only be controlled indirectly by reducing the number of Varroa mites in honey bee colonies. In this study, we investigated the antiviral potential of the gypsy mushroom (Cortinarius caperatus) to reduce DWV infection in honey bees. Our results indicate that the alcohol extract of C. caperatus prevented the development of DWV infection in cage experiments as well as after direct application to honey bee colonies in a field experiment. The applied doses did not shorten the lifespan of honey bees. The reduced levels of DWV in C. caperatus-treated honey bees in cage experiments were accompanied by significant changes in the gene expression of Tep7, Bap1, and Vago. The C. caperatus treatment was not effective against the trypanosomatid Lotmaria passim. No residues of C.caperatus were found in honey harvested in the spring from colonies supplemented with the mushroom extract for their winter feeding. These findings suggest that C. caperatus alcohol extract could be a potential natural remedy to treat DWV infection in honey bees.


Subject(s)
Agaricales , RNA Viruses , Roma , Varroidae , Bees , Animals , Humans , RNA Viruses/genetics
3.
Microbiol Res ; 274: 127418, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37315341

ABSTRACT

The spread of the parasite Varroa destructor and associated viruses has resulted in massive honey bee colony losses with considerable economic and ecological impact. The gut microbiota has a major role in shaping honey bees tolerance and resistance to parasite infestation and viral infection, but the contribution of viruses to the assembly of the host microbiota in the context of varroa resistance and susceptibility remains unclear. Here, we used a network approach including viral and bacterial nodes to characterize the impact of five viruses, Apis Rhabdovirus-1 (ARV-1), Black Queen Cell virus (BQCV), Lake Sinai virus (LSV), Sacbrood virus (SBV) and Deformed wing virus (DWV) on the gut microbiota assembly of varroa-susceptible and Gotland varroa-surviving honey bees. We found that microbiota assembly was different in varroa-surviving and varroa-susceptible honey bees with the network of the latter having a whole module not present in the network of the former. Four viruses, ARV-1, BQCV, LSV, and SBV, were tightly associated with bacterial nodes of the core microbiota of varroa-susceptible honey bees, while only two viruses BQCV and LSV, appeared correlated with bacterial nodes in varroa-surviving honey bees. In silico removal of viral nodes caused major re-arrangement of microbial networks with changes in nodes centrality and significant reduction of the networks' robustness in varroa-susceptible, but not in varroa-surviving honey bees. Comparison of predicted functional pathways in bacterial communities using PICRUSt2 showed the superpathway for heme b biosynthesis from uroporphyrinogen-III and a pathway for arginine, proline, and ornithine interconversion as significantly increased in varroa-surviving honey bees. Notably, heme and its reduction products biliverdin and bilirubin have been reported as antiviral agents. These findings show that viral pathogens are differentially nested in the bacterial communities of varroa-surviving and varroa-susceptible honey bees. These results suggest that Gotland honey bees are associated with minimally-assembled and reduced bacterial communities that exclude viral pathogens and are resilient to viral nodes removal, which, together with the production of antiviral compounds, may explain the resiliency of Gotland honey bees to viral infections. In contrast, the intertwined virus-bacterium interactions in varroa-susceptible networks suggest that the complex assembly of microbial communities in this honey bee strain favor viral infections, which may explain viral persistence in this honey bee strain. Further understanding of protective mechanisms mediated by the microbiota could help developing novel ways to control devastating viral infections affecting honey bees worldwide.


Subject(s)
Gastrointestinal Microbiome , RNA Viruses , Varroidae , Virus Diseases , Viruses , Animals , Bees
4.
Trends Parasitol ; 39(6): 487-488, 2023 06.
Article in English | MEDLINE | ID: mdl-36967341
5.
iScience ; 25(11): 105445, 2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36388978

ABSTRACT

Some bee species use wax to build their nests. They store honey and raise their brood in cells made entirely from wax. How can the bee brood breathe and develop properly when sealed in wax cells? We compared the chemical composition and structural properties of the honey cappings and worker brood cappings of the honeybee Apis mellifera carnica, measured the worker brood respiration, and calculated the CO2 gradients across the two types of cappings. We identified microscopic pores present in the brood cappings that allow efficient gas exchange of the developing brood. In contrary, honey cappings are nearly gas impermeable to protect honey from fermenting. Similar principles apply in bumble bees. Our data suggest the control of gas exchange of cappings as a selective pressure in the evolution of wax-building bees that drives their adaptation for using wax in two highly contrasting biological contexts.

6.
Sci Rep ; 10(1): 4401, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32157127

ABSTRACT

Mitochondrial dysfunctions belong amongst the most common metabolic diseases but the signalling networks that lead to the manifestation of a disease phenotype are often not well understood. We identified the subunits of respiratory complex I, III and IV as mediators of major signalling changes during Drosophila wing disc development. Their downregulation in larval wing disc leads to robust stimulation of TOR activity, which in turn orchestrates a complex downstream signalling network. Specifically, after downregulation of the complex I subunit ND-49 (mammalian NDUFS2), TOR activates JNK to induce cell death and ROS production essential for the stimulation of compensatory apoptosis-induced proliferation within the tissue. Additionally, TOR upregulates Notch and JAK/STAT signalling and it directs glycolytic switch of the target tissue. Our results highlight the central role of TOR signalling in mediating the complex response to mitochondrial respiratory dysfunction and they provide a rationale why the disease symptoms associated with respiratory dysfunctions are often alleviated by mTOR inhibitors.


Subject(s)
Drosophila Proteins/metabolism , Electron Transport Complex I/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Wings, Animal/growth & development , Animals , Down-Regulation , Drosophila , Drosophila Proteins/genetics , Electron Transport Complex I/metabolism , Janus Kinases/metabolism , Reactive Oxygen Species/metabolism , Receptors, Notch/metabolism , STAT Transcription Factors/metabolism , Wings, Animal/metabolism
7.
Dev Biol ; 452(2): 83-94, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31085193

ABSTRACT

The proliferation, differentiation and function of immune cells in vertebrates, as well as in the invertebrates, is regulated by distinct signalling pathways and crosstalk with systemic and cellular metabolism. We have identified the Lime gene (Linking Immunity and Metabolism, CG18446) as one such connecting factor, linking hemocyte development with systemic metabolism in Drosophila. Lime is expressed in larval plasmatocytes and the fat body and regulates immune cell type and number by influencing the size of hemocyte progenitor populations in the lymph gland and in circulation. Lime mutant larvae exhibit low levels of glycogen and trehalose energy reserves and they develop low number of hemocytes. The low number of hemocytes in Lime mutants can be rescued by Lime overexpression in the fat body. It is well known that immune cell metabolism is tightly regulated with the progress of infection and it must be supported by systemic metabolic changes. Here we demonstrate that Lime mutants fails to induce such systemic metabolic changes essential for the larval immune response. Indeed, Lime mutants are not able to sustain high numbers of circulating hemocytes and are compromised in the number of lamellocytes produced during immune system challenge, using a parasitic wasp infection model. We therefore propose the Lime gene as a novel functional link between systemic metabolism and Drosophila immunity.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/immunology , Drosophila melanogaster/metabolism , Immunity , Nuclear Proteins/metabolism , Animals , Cell Differentiation , Energy Metabolism , Fat Body/metabolism , Hemocytes/cytology , Hemocytes/metabolism , Larva/metabolism , Lymphoid Tissue/metabolism , Mutation/genetics
8.
Development ; 144(18): 3193-3198, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28928279

ABSTRACT

Despite the fact that metabolic studies played a prominent role in the early history of developmental biology research, the field of developmental metabolism was largely ignored following the advent of modern molecular biology. Metabolism, however, has recently re-emerged as a focal point of biomedical studies and, as a result, developmental biologists are once again exploring the chemical and energetic forces that shape growth, development and maturation. In May 2017, a diverse group of scientists assembled at the EMBO/EMBL Symposium 'Metabolism in Time and Space' to discuss how metabolism influences cellular and developmental processes. The speakers not only described how metabolic flux adapts to the energetic needs of a developing organism, but also emphasized that metabolism can directly regulate developmental progression. Overall, and as we review here, this interdisciplinary meeting provided a valuable forum to explore the interface between developmental biology and metabolism.


Subject(s)
Developmental Biology , Metabolism , Animals , Cell Cycle , Humans , Immunity , Plant Development , Signal Transduction
9.
Biochem J ; 473(22): 4129-4143, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27623778

ABSTRACT

The silent information regulator 1 (Sirt1) has been shown to have negative effects on the Notch pathway in several contexts. We bring evidence that Sirt1 has a positive effect on Notch activation in Drosophila, in the context of sensory organ precursor specification and during wing development. The phenotype of Sirt1 mutant resembles weak Notch loss-of-function phenotypes, and genetic interactions of Sirt1 with the components of the Notch pathway also suggest a positive role for Sirt1 in Notch signalling. Sirt1 is necessary for the efficient activation of enhancer of split [E(spl)] genes by Notch in S2N cells. Additionally, the Notch-dependent response of several E(spl) genes is sensitive to metabolic stress caused by 2-deoxy-d-glucose treatment, in a Sirt1-dependent manner. We found Sirt1 associated with several proteins involved in Notch repression as well as activation, including the cofactor exchange factor Ebi (TBL1), the RLAF/LAF histone chaperone complex and the Tip60 acetylation complex. Moreover, Sirt1 participates in the deacetylation of the CSL transcription factor Suppressor of Hairless. The role of Sirt1 in Notch signalling is, therefore, more complex than previously recognized, and its diverse effects may be explained by a plethora of Sirt1 substrates involved in the regulation of Notch signalling.


Subject(s)
Drosophila Proteins/metabolism , Receptors, Notch/metabolism , Sirtuin 1/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Deoxyglucose/pharmacology , Drosophila , Drosophila Proteins/genetics , Immunoprecipitation , Mass Spectrometry , Protein Binding , RNA Interference/physiology , RNA, Messenger/antagonists & inhibitors , Receptors, Notch/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sirtuin 1/genetics
10.
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
11.
J Cell Sci ; 126(Pt 21): 4873-84, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23970418

ABSTRACT

It is firmly established that interactions between neurons and glia are fundamental across species for the correct establishment of a functional brain. Here, we found that the glia of the Drosophila larval brain display an essential non-autonomous role during the development of the optic lobe. The optic lobe develops from neuroepithelial cells that proliferate by dividing symmetrically until they switch to asymmetric/differentiative divisions that generate neuroblasts. The proneural gene lethal of scute (l'sc) is transiently activated by the epidermal growth factor receptor (EGFR)-Ras signal transduction pathway at the leading edge of a proneural wave that sweeps from medial to lateral neuroepithelium, promoting this switch. This process is tightly regulated by the tissue-autonomous function within the neuroepithelium of multiple signaling pathways, including EGFR-Ras and Notch. This study shows that the Notch ligand Serrate (Ser) is expressed in the glia and it forms a complex in vivo with Notch and Canoe, which colocalize at the adherens junctions of neuroepithelial cells. This complex is crucial for interactions between glia and neuroepithelial cells during optic lobe development. Ser is tissue-autonomously required in the glia where it activates Notch to regulate its proliferation, and non-autonomously in the neuroepithelium where Ser induces Notch signaling to avoid the premature activation of the EGFR-Ras pathway and hence of L'sc. Interestingly, different Notch activity reporters showed very different expression patterns in the glia and in the neuroepithelium, suggesting the existence of tissue-specific factors that promote the expression of particular Notch target genes or/and a reporter response dependent on different thresholds of Notch signaling.


Subject(s)
Calcium-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neuroepithelial Cells/metabolism , Neuroglia/metabolism , Optic Lobe, Nonmammalian/growth & development , Receptors, Notch/metabolism , Animals , Calcium-Binding Proteins/genetics , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Membrane Proteins/genetics , Optic Lobe, Nonmammalian/metabolism , Protein Binding , Receptors, Notch/genetics , Serrate-Jagged Proteins , Signal Transduction
12.
PLoS Genet ; 9(1): e1003162, 2013.
Article in English | MEDLINE | ID: mdl-23300480

ABSTRACT

Dynamic activity of signaling pathways, such as Notch, is vital to achieve correct development and homeostasis. However, most studies assess output many hours or days after initiation of signaling, once the outcome has been consolidated. Here we analyze genome-wide changes in transcript levels, binding of the Notch pathway transcription factor, CSL [Suppressor of Hairless, Su(H), in Drosophila], and RNA Polymerase II (Pol II) immediately following a short pulse of Notch stimulation. A total of 154 genes showed significant differential expression (DE) over time, and their expression profiles stratified into 14 clusters based on the timing, magnitude, and direction of DE. E(spl) genes were the most rapidly upregulated, with Su(H), Pol II, and transcript levels increasing within 5-10 minutes. Other genes had a more delayed response, the timing of which was largely unaffected by more prolonged Notch activation. Neither Su(H) binding nor poised Pol II could fully explain the differences between profiles. Instead, our data indicate that regulatory interactions, driven by the early-responding E(spl)bHLH genes, are required. Proposed cross-regulatory relationships were validated in vivo and in cell culture, supporting the view that feed-forward repression by E(spl)bHLH/Hes shapes the response of late-responding genes. Based on these data, we propose a model in which Hes genes are responsible for co-ordinating the Notch response of a wide spectrum of other targets, explaining the critical functions these key regulators play in many developmental and disease contexts.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Drosophila Proteins , Drosophila , Receptors, Notch , Repressor Proteins , Signal Transduction/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites/genetics , Conserved Sequence/genetics , DNA-Binding Proteins , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental , Humans , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription, Genetic , Transcriptional Activation
13.
Development ; 140(4): 926-37, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23325760

ABSTRACT

The diverse functions of Notch signalling imply that it must elicit context-specific programmes of gene expression. With the aim of investigating how Notch drives cells to differentiate, we have used a genome-wide approach to identify direct Notch targets in Drosophila haemocytes (blood cells), where Notch promotes crystal cell differentiation. Many of the identified Notch-regulated enhancers contain Runx and GATA motifs, and we demonstrate that binding of the Runx protein Lozenge (Lz) is required for enhancers to be competent to respond to Notch. Functional studies of targets, such as klumpfuss (ERG/WT1 family) and pebbled/hindsight (RREB1 homologue), show that Notch acts both to prevent the cells adopting alternate cell fates and to promote morphological characteristics associated with crystal cell differentiation. Inappropriate activity of Klumpfuss perturbs the differentiation programme, resulting in melanotic tumours. Thus, by acting as a master regulator, Lz directs Notch to activate selectively a combination of target genes that correctly locks cells into the differentiation programme.


Subject(s)
Cell Differentiation/physiology , Core Binding Factor alpha Subunits/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Hemocytes/physiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Chromatin Immunoprecipitation , Drosophila , Fluorescent Antibody Technique , Hemocytes/metabolism , Luciferases , Mutagenesis , Nuclear Proteins/metabolism , RNA Interference
14.
EMBO J ; 32(1): 60-71, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23232763

ABSTRACT

The outcome of the Notch pathway on proliferation depends on cellular context, being growth promotion in some, including several cancers, and growth inhibition in others. Such disparate outcomes are evident in Drosophila wing discs, where Notch overactivation causes hyperplasia despite having localized inhibitory effects on proliferation. To understand the underlying mechanisms, we have used genomic strategies to identify the Notch-CSL target genes directly activated during wing disc hyperplasia. Among them were genes involved in both autonomous and non-autonomous regulation of proliferation, growth and cell death, providing molecular explanations for many characteristics of Notch induced wing disc hyperplasia previously reported. The Notch targets exhibit different response patterns, which are shaped by both positive and negative feed-forward regulation between the Notch targets themselves. We propose, therefore, that both the characteristics of the direct Notch targets and their cross-regulatory relationships are important in coordinating the pattern of hyperplasia.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Hyperplasia/genetics , Receptors, Notch/genetics , Signal Transduction/physiology , Wings, Animal/embryology , Animals , Animals, Genetically Modified , Cell Division , Cell Proliferation , Chromatin Immunoprecipitation , Drosophila/embryology , Drosophila/growth & development , Drosophila/physiology , Drosophila Proteins/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genomics , Male , Oligonucleotide Array Sequence Analysis , Phenotype , Receptors, Notch/metabolism , Wings, Animal/growth & development , Wings, Animal/physiology
15.
Biochem Soc Trans ; 40(2): 311-23, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22435805

ABSTRACT

There is an intimate, yet poorly understood, link between cellular metabolic status, cell signalling and transcription. Central metabolic pathways are under the control of signalling pathways and, vice versa, the cellular metabolic profile influences cell signalling through the incorporation of various metabolic sensors into the signalling networks. Thus information about nutrients availability directly and crucially influences crucial cell decisions. In the present review, I summarize our current knowledge of various metabolic sensors and give some examples of the integration of metabolically derived inputs into the signalling system and the regulation of transcription. I also discuss the Warburg effect where the cross-talk between metabolism and signalling is used to orchestrate rapid cell growth and division. It is becoming clear that future research will concentrate on the collection of small-molecule metabolites, whose concentration fluctuates in response to cellular energy levels, searching for their sensors that connect them to the signalling and transcriptional networks.


Subject(s)
Cells/metabolism , Metabolic Networks and Pathways , Signal Transduction , Transcription, Genetic , Animals , Cell Proliferation , Cells/cytology , Energy Metabolism , Humans
16.
Nat Neurosci ; 15(2): 224-33, 2011 Dec 25.
Article in English | MEDLINE | ID: mdl-22197833

ABSTRACT

Neuronal-class diversification is central during neurogenesis. This requirement is exemplified in the olfactory system, which utilizes a large array of olfactory receptor neuron (ORN) classes. We discovered an epigenetic mechanism in which neuron diversity is maximized via locus-specific chromatin modifications that generate context-dependent responses from a single, generally used intracellular signal. Each ORN in Drosophila acquires one of three basic identities defined by the compound outcome of three iterated Notch signaling events during neurogenesis. Hamlet, the Drosophila Evi1 and Prdm16 proto-oncogene homolog, modifies cellular responses to these iteratively used Notch signals in a context-dependent manner, and controls odorant receptor gene choice and ORN axon targeting specificity. In nascent ORNs, Hamlet erases the Notch state inherited from the parental cell, enabling a modified response in a subsequent round of Notch signaling. Hamlet directs locus-specific modifications of histone methylation and histone density and controls accessibility of the DNA-binding protein Suppressor of Hairless at the Notch target promoter.


Subject(s)
Chromatin/physiology , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Olfactory Pathways/cytology , Olfactory Receptor Neurons/physiology , Receptors, Notch/metabolism , Receptors, Odorant/classification , Animals , Animals, Genetically Modified , Axons/physiology , Cell Differentiation/genetics , Cell Division/genetics , Cell Line , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Larva , Mutation/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Olfactory Pathways/growth & development , Olfactory Receptor Neurons/cytology , RNA, Messenger/metabolism , Receptors, Notch/genetics , Receptors, Odorant/genetics , Receptors, Odorant/metabolism , Signal Transduction/physiology , Statistics, Nonparametric , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
17.
J Immunol ; 186(6): 3373-82, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21307296

ABSTRACT

Plasticity within Th cell populations may play a role in enabling site-specific immune responses to infections while limiting tissue destruction. Epigenetic processes are fundamental to such plasticity; however, to date, most investigations have focused on in vitro-generated T cells. In this study, we have examined the molecular mechanisms underpinning murine Th17 plasticity in vivo by assessing H3K4 and H3K27 trimethylation marks at Tbx21, Rorc, Il17a, Ifng, and Il12rb2 loci in purified ex vivo-isolated and in vitro-generated Th17 cells. Although both populations had largely comparable epigenetic signatures, including bivalent marks at Tbx21, freshly isolated ex vivo Th17 cells displayed restricted expression from Il12rb2 due to the presence of repressive chromatin modifications. This receptor, however, could be upregulated on isolated ex vivo Th17 cells after in vitro activation or by in vivo immunization and was augmented by the presence of IFN-γ. Such activated cells could then be deviated toward a Th1-like profile. We show that IL-12 stimulation removes H3K27 trimethylation modifications at Tbx21/T-bet leading to enhanced T-bet expression with in vitro Th17 cells. Our study reveals important potential phenotypic differences between ex vivo- and in vitro-generated Th17 cells and provides mechanistic insight into Th17 cell plasticity.


Subject(s)
Cell Differentiation/immunology , Epigenesis, Genetic/immunology , Receptors, Interleukin-12/genetics , T-Box Domain Proteins/genetics , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Polarity/genetics , Cell Polarity/immunology , Cell Separation , Cells, Cultured , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/immunology , DNA Methylation/genetics , DNA Methylation/immunology , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Immunophenotyping , Interleukin-12/physiology , Mice , Mice, Inbred NOD , Receptors, Interleukin-12/biosynthesis , Receptors, Interleukin-12/physiology , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/physiology , Th17 Cells/cytology
18.
Development ; 137(16): 2633-42, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20610485

ABSTRACT

Cell-cell signalling mediated by Notch regulates many different developmental and physiological processes and is involved in a variety of human diseases. Activation of Notch impinges directly on gene expression through the Suppressor of Hairless [Su(H)] DNA-binding protein. A major question that remains to be elucidated is how the same Notch signalling pathway can result in different transcriptional responses depending on the cellular context and environment. Here, we have investigated the factors required to confer this specific response in Drosophila adult myogenic progenitor-related cells. Our analysis identifies Twist (Twi) as a crucial co-operating factor. Enhancers from several direct Notch targets require a combination of Twi and Notch activities for expression in vivo; neither alone is sufficient. Twi is bound at target enhancers prior to Notch activation and enhances Su(H) binding to these regulatory regions. To determine the breadth of the combinatorial regulation we mapped Twi occupancy genome-wide in DmD8 myogenic progenitor-related cells by chromatin immunoprecipitation. Comparing the sites bound by Su(H) and by Twi in these cells revealed a strong association, identifying a large spectrum of co-regulated genes. We conclude that Twi is an essential Notch co-regulator in myogenic progenitor cells and has the potential to confer specificity on Notch signalling at over 170 genes, showing that a single factor can have a profound effect on the output of the pathway.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Muscle, Skeletal/metabolism , Receptors, Notch/metabolism , Signal Transduction , Stem Cells/metabolism , Transcription, Genetic , Twist-Related Protein 1/metabolism , Aging , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression , Genome-Wide Association Study , Protein Binding , Receptors, Notch/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Twist-Related Protein 1/genetics
19.
Sci Signal ; 2(55): ra1, 2009 Jan 27.
Article in English | MEDLINE | ID: mdl-19176515

ABSTRACT

Notch is the receptor in one of a small group of conserved signaling pathways that are essential at multiple stages in development. Although the mechanism of transduction impinges directly on the nucleus to regulate transcription through the CSL [CBF-1/Su(H)/LAG-1] [corrected] DNA binding protein, there are few known direct target genes. Thus, relatively little is known about the immediate cellular consequences of Notch activation. We therefore set out to determine the genome-wide response to Notch activation by analyzing the changes in messenger RNA (mRNA) expression and the sites of CSL occupancy within 30 minutes of activating Notch in Drosophila cells. Through combining these data, we identify high-confidence direct targets of Notch that are implicated in the maintenance of adult muscle progenitors in vivo. These targets are enriched in cell morphogenesis genes and in components of other cell signaling pathways, especially the epidermal growth factor receptor (EGFR) pathway. Also evident are examples of incoherent network logic, where Notch stimulates the expression of both a gene and the repressor of that gene, which may result in a transient window of competence after Notch activation. Furthermore, because targets comprise both positive and negative regulators, cells become poised for both outcomes, suggesting one mechanism through which Notch activation can lead to opposite effects in different contexts.


Subject(s)
Receptors, Notch/metabolism , Signal Transduction , Animals , Cell Nucleus/metabolism , Chromatin Immunoprecipitation , Drosophila Proteins/metabolism , Drosophila melanogaster , ErbB Receptors/metabolism , Gene Expression Regulation , Genome , Lac Operon , Models, Biological , Muscles/metabolism , RNA, Messenger/metabolism
20.
J Cell Sci ; 121(Pt 6): 747-52, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18303052

ABSTRACT

Transcription factors of the Grainy head (Grh) family are required in epithelia to generate the impermeable apical layer that protects against the external environment. This function is conserved in vertebrates and invertebrates, despite the differing molecular composition of the protective barrier. Epithelial cells also have junctions that create a paracellular diffusion barrier (tight or septate junctions). To examine whether Grh has a role in regulating such characteristics, we used an epidermal layer in the Drosophila embryo that has no endogenous Grh and lacks septate junctions, the amnioserosa. Expression of Grh in the amnioserosa caused severe defects in dorsal closure, a process similar to wound closure, and induced robust expression of the septate junction proteins Coracle, Fasciclin 3 and Sinuous. Grh-binding sites are present within the genes encoding these proteins, consistent with them being direct targets. Removal of Grh from imaginal disc cells caused a reduction in Fasciclin 3 and Coracle levels, suggesting that Grh normally fine tunes their epithelial expression and hence contributes to barrier properties. The fact that ectopic Grh arrests dorsal closure also suggests that this dynamic process relies on epithelia having distinct adhesive properties conferred by differential deployment of Grh.


Subject(s)
DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Epithelium/embryology , Membrane Proteins/genetics , Transcription Factors/physiology , Animals , Binding Sites , Cell Adhesion , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Epidermal Cells , Epidermis/embryology , Epithelial Cells/cytology , Epithelium/metabolism , Intercellular Junctions/metabolism , Membrane Proteins/metabolism , Morphogenesis , Mutation , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL