Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
EMBO J ; 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39020149

ABSTRACT

Tumor necrosis factor receptors (TNFRs) control pleiotropic pro-inflammatory functions that range from apoptosis to cell survival. The ability to trigger a particular function will depend on the upstream cues, association with regulatory complexes, and downstream pathways. In Drosophila melanogaster, two TNFRs have been identified, Wengen (Wgn) and Grindelwald (Grnd). Although several reports associate these receptors with JNK-dependent apoptosis, it has recently been found that Wgn activates a variety of other functions. We demonstrate that Wgn is required for survival by protecting cells from apoptosis. This is mediated by dTRAF1 and results in the activation of p38 MAP kinase. Remarkably, Wgn is required for apoptosis-induced regeneration and is activated by the reactive oxygen species (ROS) produced following apoptosis. This ROS activation is exclusive for Wgn, but not for Grnd, and can occur after knocking down Eiger/TNFα. The extracellular cysteine-rich domain of Grnd is much more divergent than that of Wgn, which is more similar to TNFRs from other animals, including humans. Our results show a novel TNFR function that responds to stressors by ensuring p38-dependent regeneration.

2.
Development ; 148(8)2021 04 15.
Article in English | MEDLINE | ID: mdl-33913483

ABSTRACT

Regeneration after damage requires early signals to trigger the tissue repair machinery. Reactive oxygen species (ROS) act as early signals that are sensed by the MAP3 kinase Ask1, which in turn activates by phosphorylation the MAP kinases p38 and JNK. The sustained or high activation of these kinases can result in apoptosis, whereas short or low activation can promote regeneration. Using the Ask1-dependent regeneration program, we demonstrate in Drosophila wing that PI3K/Akt signaling is necessary for Ask1 to activate p38, but not JNK. In addition, nutrient restriction or mutations that target Ser83 of the Drosophila Ask1 protein, a PI3K/Akt-sensitive residue, block regeneration. However, these effects can be reversed by the ectopic activation of p38, but not of JNK. Our results demonstrate that Ask1 controls the activation of p38 through Ser83, and that the phosphorylation of p38 during regeneration is nutrient sensitive. This mechanism is important for discriminating between p38 and JNK in the cells involved in tissue repair and regenerative growth.


Subject(s)
MAP Kinase Signaling System , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Regeneration , Wings, Animal/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , p38 Mitogen-Activated Protein Kinases/genetics
3.
Semin Cell Dev Biol ; 97: 55-62, 2020 01.
Article in English | MEDLINE | ID: mdl-31063813

ABSTRACT

During the process of regeneration, a switch in the transcription program occurs in cells that contribute to the reconstruction of the missing tissue. Early signals released upon damage are integrated into the chromatin of responding cells to change its activity and function. Changes in chromatin dynamics result in transcriptional reprogramming, this is the coordinated regulation of expression of a specific subset of genes required for the regeneration process. Here we summarize changes in gene expression and chromatin dynamics that occurs during the process of regeneration of Drosophila imaginal discs.


Subject(s)
Chromatin/metabolism , Drosophila/genetics , Imaginal Discs/drug effects , Regeneration/genetics , Animals
4.
Mamm Genome ; 33(2): 328-342, 2022 06.
Article in English | MEDLINE | ID: mdl-35098341

ABSTRACT

Over the last decade, the increasing interest in long non-coding RNAs (lncRNAs) has led to the discovery of these transcripts in multiple organisms. LncRNAs tend to be specifically, and often lowly, expressed in certain tissues, cell types and biological contexts. Although lncRNAs participate in the regulation of a wide variety of biological processes, including development and disease, most of their functions and mechanisms of action remain unknown. Poor conservation of the DNA sequences encoding for these transcripts makes the identification of lncRNAs orthologues among different species very challenging, especially between evolutionarily distant species such as flies and humans or mice. However, the functions of lncRNAs are unexpectedly preserved among different species supporting the idea that conservation occurs beyond DNA sequences and reinforcing the potential of characterising lncRNAs in animal models. In this review, we describe the features and roles of lncRNAs in the fruit fly Drosophila melanogaster, focusing on genomic and functional comparisons with human and mouse lncRNAs. We also discuss the current state of advances and limitations in the study of lncRNA conservation and future perspectives.


Subject(s)
RNA, Long Noncoding , Animals , Base Sequence , Drosophila melanogaster/genetics , Genome , Genomics , Humans , Mice , RNA, Long Noncoding/genetics
5.
PLoS Genet ; 15(1): e1007926, 2019 01.
Article in English | MEDLINE | ID: mdl-30677014

ABSTRACT

How cells communicate to initiate a regenerative response after damage has captivated scientists during the last few decades. It is known that one of the main signals emanating from injured cells is the Reactive Oxygen Species (ROS), which propagate to the surrounding tissue to trigger the replacement of the missing cells. However, the link between ROS production and the activation of regenerative signaling pathways is not yet fully understood. We describe here the non-autonomous ROS sensing mechanism by which living cells launch their regenerative program. To this aim, we used Drosophila imaginal discs as a model system due to its well-characterized regenerative ability after injury or cell death. We genetically-induced cell death and found that the Apoptosis signal-regulating kinase 1 (Ask1) is essential for regenerative growth. Ask1 senses ROS both in dying and living cells, but its activation is selectively attenuated in living cells by Akt1, the core kinase component of the insulin/insulin-like growth factor pathway. Akt1 phosphorylates Ask1 in a secondary site outside the kinase domain, which attenuates its activity. This modulation of Ask1 activity results in moderate levels of JNK signaling in the living tissue, as well as in activation of p38 signaling, both pathways required to turn on the regenerative response. Our findings demonstrate a non-autonomous activation of a ROS sensing mechanism by Ask1 and Akt1 to replace the missing tissue after damage. Collectively, these results provide the basis for understanding the molecular mechanism of communication between dying and living cells that triggers regeneration.


Subject(s)
Drosophila Proteins/genetics , Imaginal Discs/growth & development , MAP Kinase Kinase Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Regeneration/genetics , Animals , Apoptosis/genetics , Cell Communication/genetics , Cell Proliferation/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Humans , Imaginal Discs/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/genetics
6.
Genome Res ; 28(12): 1852-1866, 2018 12.
Article in English | MEDLINE | ID: mdl-30459214

ABSTRACT

One of the most important questions in regenerative biology is to unveil how and when genes change expression and trigger regeneration programs. The resetting of gene expression patterns during response to injury is governed by coordinated actions of genomic regions that control the activity of multiple sequence-specific DNA binding proteins. Using genome-wide approaches to interrogate chromatin function, we here identify the elements that regulate tissue recovery in Drosophila imaginal discs, which show a high regenerative capacity after genetically induced cell death. Our findings indicate there is global coregulation of gene expression as well as a regeneration program driven by different types of regulatory elements. Novel enhancers acting exclusively within damaged tissue cooperate with enhancers co-opted from other tissues and other developmental stages, as well as with endogenous enhancers that show increased activity after injury. Together, these enhancers host binding sites for regulatory proteins that include a core set of conserved transcription factors that control regeneration across metazoans.


Subject(s)
Drosophila/physiology , Gene Expression Regulation , Regeneration/genetics , Response Elements , Animals , Chromatin/genetics , Conserved Sequence , Gene Expression Profiling , Signal Transduction , Transcription, Genetic , Transcriptional Activation , Transcriptome
8.
Genome Res ; 25(9): 1256-67, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26194102

ABSTRACT

Selenoproteins are proteins that incorporate selenocysteine (Sec), a nonstandard amino acid encoded by UGA, normally a stop codon. Sec synthesis requires the enzyme Selenophosphate synthetase (SPS or SelD), conserved in all prokaryotic and eukaryotic genomes encoding selenoproteins. Here, we study the evolutionary history of SPS genes, providing a map of selenoprotein function spanning the whole tree of life. SPS is itself a selenoprotein in many species, although functionally equivalent homologs that replace the Sec site with cysteine (Cys) are common. Many metazoans, however, possess SPS genes with substitutions other than Sec or Cys (collectively referred to as SPS1). Using complementation assays in fly mutants, we show that these genes share a common function, which appears to be distinct from the synthesis of selenophosphate carried out by the Sec- and Cys- SPS genes (termed SPS2), and unrelated to Sec synthesis. We show here that SPS1 genes originated through a number of independent gene duplications from an ancestral metazoan selenoprotein SPS2 gene that most likely already carried the SPS1 function. Thus, in SPS genes, parallel duplications and subsequent convergent subfunctionalization have resulted in the segregation to different loci of functions initially carried by a single gene. This evolutionary history constitutes a remarkable example of emergence and evolution of gene function, which we have been able to trace thanks to the singular features of SPS genes, wherein the amino acid at a single site determines unequivocally protein function and is intertwined to the evolutionary fate of the entire selenoproteome.


Subject(s)
Biological Evolution , Phosphotransferases/genetics , Phosphotransferases/metabolism , Animals , Biomarkers , Eukaryota/genetics , Eukaryota/metabolism , Gene Duplication , Humans , Insecta , Phylogeny , Prokaryotic Cells/metabolism , Selection, Genetic , Selenium/metabolism , Selenoproteins/genetics , Selenoproteins/metabolism , Urochordata , Vertebrates
9.
PLoS Genet ; 11(10): e1005595, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26496642

ABSTRACT

Upon apoptotic stimuli, epithelial cells compensate the gaps left by dead cells by activating proliferation. This has led to the proposal that dying cells signal to surrounding living cells to maintain homeostasis. Although the nature of these signals is not clear, reactive oxygen species (ROS) could act as a signaling mechanism as they can trigger pro-inflammatory responses to protect epithelia from environmental insults. Whether ROS emerge from dead cells and what is the genetic response triggered by ROS is pivotal to understand regeneration of Drosophila imaginal discs. We genetically induced cell death in wing imaginal discs, monitored the production of ROS and analyzed the signals required for repair. We found that cell death generates a burst of ROS that propagate to the nearby surviving cells. Propagated ROS activate p38 and induce tolerable levels of JNK. The activation of JNK and p38 results in the expression of the cytokines Unpaired (Upd), which triggers the JAK/STAT signaling pathway required for regeneration. Our findings demonstrate that this ROS/JNK/p38/Upd stress responsive module restores tissue homeostasis. This module is not only activated after cell death induction but also after physical damage and reveals one of the earliest responses for imaginal disc regeneration.


Subject(s)
Drosophila Proteins/genetics , JNK Mitogen-Activated Protein Kinases/genetics , Regeneration/genetics , Transcription Factors/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Apoptosis/genetics , Cell Proliferation/genetics , Drosophila Proteins/biosynthesis , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Imaginal Discs/growth & development , JNK Mitogen-Activated Protein Kinases/biosynthesis , Reactive Oxygen Species/metabolism , Signal Transduction , Stress, Physiological/genetics , Transcription Factors/biosynthesis , Wings, Animal/growth & development , p38 Mitogen-Activated Protein Kinases/biosynthesis
11.
EMBO Rep ; 16(3): 362-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25572844

ABSTRACT

The Drosophila transcription factor Cabut/dTIEG (Cbt) is a growth regulator, whose expression is modulated by different stimuli. Here, we determine Cbt association with chromatin and identify Yorkie (Yki), the transcriptional co-activator of the Hippo (Hpo) pathway as its partner. Cbt and Yki co-localize on common gene promoters, and the expression of target genes varies according to changes in Cbt levels. Down-regulation of Cbt suppresses the overgrowth phenotypes caused by mutations in expanded (ex) and yki overexpression, whereas its up-regulation promotes cell proliferation. Our results imply that Cbt is a novel partner of Yki that is required as a transcriptional co-activator in growth control.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/growth & development , Intracellular Signaling Peptides and Proteins/metabolism , Juvenile Hormones/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Chromatin Immunoprecipitation , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Models, Biological , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Transcription Factors/genetics , YAP-Signaling Proteins
12.
PLoS Genet ; 9(7): e1003627, 2013.
Article in English | MEDLINE | ID: mdl-23874226

ABSTRACT

The capacity of tumour cells to maintain continual overgrowth potential has been linked to the commandeering of normal self-renewal pathways. Using an epithelial cancer model in Drosophila melanogaster, we carried out an overexpression screen for oncogenes capable of cooperating with the loss of the epithelial apico-basal cell polarity regulator, scribbled (scrib), and identified the cell fate regulator, Abrupt, a BTB-zinc finger protein. Abrupt overexpression alone is insufficient to transform cells, but in cooperation with scrib loss of function, Abrupt promotes the formation of massive tumours in the eye/antennal disc. The steroid hormone receptor coactivator, Taiman (a homologue of SRC3/AIB1), is known to associate with Abrupt, and Taiman overexpression also drives tumour formation in cooperation with the loss of Scrib. Expression arrays and ChIP-Seq indicates that Abrupt overexpression represses a large number of genes, including steroid hormone-response genes and multiple cell fate regulators, thereby maintaining cells within an epithelial progenitor-like state. The progenitor-like state is characterised by the failure to express the conserved Eyes absent/Dachshund regulatory complex in the eye disc, and in the antennal disc by the failure to express cell fate regulators that define the temporal elaboration of the appendage along the proximo-distal axis downstream of Distalless. Loss of scrib promotes cooperation with Abrupt through impaired Hippo signalling, which is required and sufficient for cooperative overgrowth with Abrupt, and JNK (Jun kinase) signalling, which is required for tumour cell migration/invasion but not overgrowth. These results thus identify a novel cooperating oncogene, identify mammalian family members of which are also known oncogenes, and demonstrate that epithelial tumours in Drosophila can be characterised by the maintenance of a progenitor-like state.


Subject(s)
Carcinogenesis , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , MAP Kinase Signaling System/genetics , Neoplasms, Glandular and Epithelial/genetics , Nuclear Proteins/genetics , Animals , Cell Proliferation , Disease Models, Animal , Drosophila Proteins/metabolism , Eye Neoplasms/genetics , Eye Neoplasms/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Glandular and Epithelial/pathology , Nuclear Proteins/metabolism , Oncogene Protein p65(gag-jun)/genetics , Oncogene Protein p65(gag-jun)/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
13.
J Cell Sci ; 126(Pt 1): 53-9, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23178937

ABSTRACT

In a genome-wide expression profile search for genes required for Drosophila R7 photoreceptor development we found ß amyloid protein precursor-like (Appl), the ortholog of human APP, which is a key factor in the pathogenesis of Alzheimer's disease. We analyzed Appl expression in the eye imaginal disc and found that is highly accumulated in R7 photoreceptor cells. The R7 photoreceptor is responsible for UV light detection. To explore the link between high expression of Appl and R7 function, we have analyzed Appl null mutants and found reduced preference for UV light, probably because of mistargeted R7 axons. Moreover, axon mistargeting and inappropriate light discrimination are enhanced in combination with neurotactin mutants. R7 differentiation is triggered by the inductive interaction between R8 and R7 precursors, which results in a burst of Ras1/MAPK, activated by the tyrosine kinase receptor Sevenless. Therefore, we examined whether Ras1/MAPK is responsible for the high Appl expression. Inhibition of Ras1 signaling leads to reduced Appl expression, whereas constitutive activation drives ectopic Appl expression. We show that Appl is directly regulated by the Ras/MAPK pathway through a mechanism mediated by PntP2, an ETS transcription factor that specifically binds ETS sites in the Appl regulatory region. We also found that zebrafish appb expression increased after ectopic fgfr activation in the neural tube of zebrafish embryos, suggesting a conserved regulatory mechanism.


Subject(s)
Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nerve Tissue Proteins/metabolism , Photoreceptor Cells/cytology , Photoreceptor Cells/metabolism , ras Proteins/metabolism , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila , Drosophila Proteins/genetics , Membrane Proteins/genetics , Mitogen-Activated Protein Kinases/genetics , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , ras Proteins/genetics
14.
EMBO Rep ; 14(3): 261-8, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23337628

ABSTRACT

The molecular mechanisms regulating tissue size represent an unsolved puzzle in developmental biology. One signalling pathway controlling growth of the Drosophila wing is Dpp. Dpp promotes growth by repression of the transcription factor Brk. The transcriptional targets of Brk that control cell growth and proliferation, however, are not yet fully elucidated. We report here a genome-wide ChIP-Seq of endogenous Brk from wing imaginal discs. We identify the growth regulator Myc as a target of Brk and show that repression of Myc and of the miRNA bantam explains a significant fraction of the growth inhibition caused by Brk. This work sheds light on the effector mechanisms by which Dpp signalling controls tissue growth.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Wings, Animal/growth & development , Animals , DNA-Binding Proteins/genetics , Down-Regulation , Drosophila/genetics , Drosophila/growth & development , Drosophila/metabolism , Drosophila Proteins/genetics , Genome, Insect/genetics , Imaginal Discs/growth & development , Imaginal Discs/metabolism , MicroRNAs/metabolism , Repressor Proteins/genetics , Transcription Factors/genetics , Wings, Animal/metabolism
15.
Mol Cancer ; 13: 74, 2014 Mar 31.
Article in English | MEDLINE | ID: mdl-24684754

ABSTRACT

BACKGROUND: PTOV1 is an adaptor protein with functions in diverse processes, including gene transcription and protein translation, whose overexpression is associated with a higher proliferation index and tumor grade in prostate cancer (PC) and other neoplasms. Here we report its interaction with the Notch pathway and its involvement in PC progression. METHODS: Stable PTOV1 knockdown or overexpression were performed by lentiviral transduction. Protein interactions were analyzed by co-immunoprecipitation, pull-down and/or immunofluorescence. Endogenous gene expression was analyzed by real time RT-PCR and/or Western blotting. Exogenous promoter activities were studied by luciferase assays. Gene promoter interactions were analyzed by chromatin immunoprecipitation assays (ChIP). In vivo studies were performed in the Drosophila melanogaster wing, the SCID-Beige mouse model, and human prostate cancer tissues and metastasis. The Excel package was used for statistical analysis. RESULTS: Knockdown of PTOV1 in prostate epithelial cells and HaCaT skin keratinocytes caused the upregulation, and overexpression of PTOV1 the downregulation, of the Notch target genes HEY1 and HES1, suggesting that PTOV1 counteracts Notch signaling. Under conditions of inactive Notch signaling, endogenous PTOV1 associated with the HEY1 and HES1 promoters, together with components of the Notch repressor complex. Conversely, expression of active Notch1 provoked the dismissal of PTOV1 from these promoters. The antagonist role of PTOV1 on Notch activity was corroborated in the Drosophila melanogaster wing, where human PTOV1 exacerbated Notch deletion mutant phenotypes and suppressed the effects of constitutively active Notch. PTOV1 was required for optimal in vitro invasiveness and anchorage-independent growth of PC-3 cells, activities counteracted by Notch, and for their efficient growth and metastatic spread in vivo. In prostate tumors, the overexpression of PTOV1 was associated with decreased expression of HEY1 and HES1, and this correlation was significant in metastatic lesions. CONCLUSIONS: High levels of the adaptor protein PTOV1 counteract the transcriptional activity of Notch. Our evidences link the pro-oncogenic and pro-metastatic effects of PTOV1 in prostate cancer to its inhibitory activity on Notch signaling and are supportive of a tumor suppressor role of Notch in prostate cancer progression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Biomarkers, Tumor/genetics , Cell Cycle Proteins/biosynthesis , Homeodomain Proteins/biosynthesis , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers, Tumor/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Drosophila melanogaster , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Humans , Male , Mice , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Prostatic Neoplasms/pathology , Receptors, Notch/biosynthesis , Signal Transduction/genetics , Transcription Factor HES-1 , Transcriptional Activation/genetics
16.
Nucleic Acids Res ; 40(19): 9493-505, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22904080

ABSTRACT

H3K4me3 is a histone modification that accumulates at the transcription-start site (TSS) of active genes and is known to be important for transcription activation. The way in which H3K4me3 is regulated at TSS and the actual molecular basis of its contribution to transcription remain largely unanswered. To address these questions, we have analyzed the contribution of dKDM5/LID, the main H3K4me3 demethylase in Drosophila, to the regulation of the pattern of H3K4me3. ChIP-seq results show that, at developmental genes, dKDM5/LID localizes at TSS and regulates H3K4me3. dKDM5/LID target genes are highly transcribed and enriched in active RNApol II and H3K36me3, suggesting a positive contribution to transcription. Expression-profiling show that, though weakly, dKDM5/LID target genes are significantly downregulated upon dKDM5/LID depletion. Furthermore, dKDM5/LID depletion results in decreased RNApol II occupancy, particularly by the promoter-proximal Pol llo(ser5) form. Our results also show that ASH2, an evolutionarily conserved factor that locates at TSS and is required for H3K4me3, binds and positively regulates dKDM5/LID target genes. However, dKDM5/LID and ASH2 do not bind simultaneously and recognize different chromatin states, enriched in H3K4me3 and not, respectively. These results indicate that, at developmental genes, dKDM5/LID and ASH2 coordinately regulate H3K4me3 at TSS and that this dynamic regulation contributes to transcription.


Subject(s)
Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Transcription Initiation Site , Transcription, Genetic , Animals , Cell Line , Drosophila/enzymology , Drosophila/genetics , Drosophila/metabolism , Histone Demethylases , Nuclear Proteins/metabolism , Transcription Factors/metabolism
17.
NAR Genom Bioinform ; 6(3): lqae091, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39157585

ABSTRACT

The discovery of functional long non-coding RNAs (lncRNAs) changed their initial concept as transcriptional noise. LncRNAs have been identified as regulators of multiple biological processes, including chromatin structure, gene expression, splicing, mRNA degradation, and translation. However, functional studies of lncRNAs are hindered by the usual lack of phenotypes upon deletion or inhibition. Here, we used Drosophila imaginal discs as a model system to identify lncRNAs involved in development and regeneration. We examined a subset of lncRNAs expressed in the wing, leg, and eye disc development. Additionally, we analyzed transcriptomic data from regenerating wing discs to profile the expression pattern of lncRNAs during tissue repair. We focused on the lncRNA CR40469, which is upregulated during regeneration. We generated CR40469 mutant flies that developed normally but showed impaired wing regeneration upon cell death induction. The ability of these mutants to regenerate was restored by the ectopic expression of CR40469. Furthermore, we found that the lncRNA CR34335 has a high degree of sequence similarity with CR40469 and can partially compensate for its function during regeneration in the absence of CR40469. Our findings point to a potential role of the lncRNA CR40469 in trans during the response to damage in the wing imaginal disc.

18.
NAR Genom Bioinform ; 6(3): lqae075, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39022326

ABSTRACT

The Catalan Initiative for the Earth BioGenome Project (CBP) is an EBP-affiliated project network aimed at sequencing the genome of the >40 000 eukaryotic species estimated to live in the Catalan-speaking territories (Catalan Linguistic Area, CLA). These territories represent a biodiversity hotspot. While covering less than 1% of Europe, they are home to about one fourth of all known European eukaryotic species. These include a high proportion of endemisms, many of which are threatened. This trend is likely to get worse as the effects of global change are expected to be particularly severe across the Mediterranean Basin, particularly in freshwater ecosystems and mountain areas. Following the EBP model, the CBP is a networked organization that has been able to engage many scientific and non-scientific partners. In the pilot phase, the genomes of 52 species are being sequenced. As a case study in biodiversity conservation, we highlight the genome of the Balearic shearwater Puffinus mauretanicus, sequenced under the CBP umbrella.

19.
Development ; 137(7): 1169-79, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20215351

ABSTRACT

Regeneration and tissue repair allow damaged or lost body parts to be replaced. After injury or fragmentation of Drosophila imaginal discs, regeneration leads to the development of normal adult structures. This process is likely to involve a combination of cell rearrangement and compensatory proliferation. However, the detailed mechanisms underlying these processes are poorly understood. We have established a system to allow temporally restricted induction of cell death in situ. Using Gal4/Gal80 and UAS-rpr constructs, targeted ablation of a region of the disc could be performed and regeneration monitored without the requirement for microsurgical manipulation. Using a ptc-Gal4 construct to drive rpr expression in the wing disc resulted in a stripe of dead cells in the anterior compartment flanking the anteroposterior boundary, whereas a sal-Gal4 driver generated a dead domain that includes both anterior and posterior cells. Under these conditions, regenerated tissues were derived from the damaged compartment, suggesting that compartment restrictions are preserved during regeneration. Our studies reveal that during regeneration the live cells bordering the domain in which cell death was induced first display cytoskeletal reorganisation and apical-to-basal closure of the epithelium. Then, proliferation begins locally in the vicinity of the wound and later more extensively in the affected compartment. Finally, we show that regeneration of genetically ablated tissue requires JNK activity. During cell death-induced regeneration, the JNK pathway is activated at the leading edges of healing tissue and not in the apoptotic cells, and is required for the regulation of healing and regenerative growth.


Subject(s)
Cell Death/physiology , Drosophila melanogaster , Embryo, Nonmammalian , JNK Mitogen-Activated Protein Kinases/metabolism , Regeneration/physiology , Signal Transduction/physiology , Animals , Cell Lineage , Cell Proliferation , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/embryology , Drosophila melanogaster/physiology , Embryo, Nonmammalian/anatomy & histology , Embryo, Nonmammalian/physiology , JNK Mitogen-Activated Protein Kinases/genetics , Mitosis/physiology , Wings, Animal/anatomy & histology , Wings, Animal/embryology , Wings, Animal/physiology
20.
Nucleic Acids Res ; 39(11): 4628-39, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21310711

ABSTRACT

An important mechanism for gene regulation involves chromatin changes via histone modification. One such modification is histone H3 lysine 4 trimethylation (H3K4me3), which requires histone methyltranferase complexes (HMT) containing the trithorax-group (trxG) protein ASH2. Mutations in ash2 cause a variety of pattern formation defects in the Drosophila wing. We have identified genome-wide binding of ASH2 in wing imaginal discs using chromatin immunoprecipitation combined with sequencing (ChIP-Seq). Our results show that genes with functions in development and transcriptional regulation are activated by ASH2 via H3K4 trimethylation in nearby nucleosomes. We have characterized the occupancy of phosphorylated forms of RNA Polymerase II and histone marks associated with activation and repression of transcription. ASH2 occupancy correlates with phosphorylated forms of RNA Polymerase II and histone activating marks in expressed genes. Additionally, RNA Polymerase II phosphorylation on serine 5 and H3K4me3 are reduced in ash2 mutants in comparison to wild-type flies. Finally, we have identified specific motifs associated with ASH2 binding in genes that are differentially expressed in ash2 mutants. Our data suggest that recruitment of the ASH2-containing HMT complexes is context specific and points to a function of ASH2 and H3K4me3 in transcriptional pausing control.


Subject(s)
Chromatin/metabolism , Drosophila Proteins/physiology , Drosophila/genetics , Nuclear Proteins/physiology , Transcription Factors/physiology , Transcriptional Activation , Animals , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Genome, Insect , Histones/chemistry , Histones/metabolism , Lysine/metabolism , Methylation , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL