Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Syst Biol ; 15(5): e8339, 2019 05 22.
Article in English | MEDLINE | ID: mdl-31118277

ABSTRACT

In chronic lymphocytic leukemia (CLL), a diverse set of genetic mutations is embedded in a deregulated epigenetic landscape that drives cancerogenesis. To elucidate the role of aberrant chromatin features, we mapped DNA methylation, seven histone modifications, nucleosome positions, chromatin accessibility, binding of EBF1 and CTCF, as well as the transcriptome of B cells from CLL patients and healthy donors. A globally increased histone deacetylase activity was detected and half of the genome comprised transcriptionally downregulated partially DNA methylated domains demarcated by CTCF CLL samples displayed a H3K4me3 redistribution and nucleosome gain at promoters as well as changes of enhancer activity and enhancer linkage to target genes. A DNA binding motif analysis identified transcription factors that gained or lost binding in CLL at sites with aberrant chromatin features. These findings were integrated into a gene regulatory enhancer containing network enriched for B-cell receptor signaling pathway components. Our study predicts novel molecular links to targets of CLL therapies and provides a valuable resource for further studies on the epigenetic contribution to the disease.


Subject(s)
Chromatin/chemistry , Gene Expression Regulation, Leukemic , Gene Regulatory Networks , Histones/chemistry , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Aged , Amino Acid Motifs , Binding Sites , CCCTC-Binding Factor/genetics , DNA/chemistry , DNA Methylation , Down-Regulation , Enhancer Elements, Genetic , Histone Deacetylases/genetics , Humans , Middle Aged , Promoter Regions, Genetic , Protein Binding , Trans-Activators/genetics
2.
PLoS One ; 10(4): e0124652, 2015.
Article in English | MEDLINE | ID: mdl-25894556

ABSTRACT

The Drosophila gene putzig (pzg) encodes a nuclear protein that is an integral component of the Trf2/Dref complex involved in the transcription of proliferation-related genes. Moreover, Pzg is found in a complex together with the nucleosome remodeling factor NURF, where it promotes Notch target gene activation. Here we show that downregulation of pzg activity in the developing wing imaginal discs induces an apoptotic response, accompanied by the induction of the pro-apoptotic gene reaper, repression of Drosophila inhibitor of apoptosis protein accumulation and the activation of the caspases Drice, Caspase3 and Dcp1. As a further consequence 'Apoptosis induced Proliferation' (AiP) and 'Apoptosis induced Apoptosis' (AiA) are triggered. As expected, the activity of the stress kinase Jun N-terminal kinase (JNK), proposed to mediate both processes, is ectopically induced in response to pzg loss. In addition, the expression of the mitogen wingless (wg) but not of decapentaplegic (dpp) is observed. We present evidence that downregulation of Notch activates Dcp1 caspase and JNK signaling, however, neither induces ectopic wg nor dpp expression. In contrast, the consequences of Dref-RNAi were largely indistinguishable from pzg-RNAi with regard to apoptosis induction. Moreover, overexpression of Dref ameliorated the downregulation of pzg compatible with the notion that the two are required together to maintain cell and tissue homeostasis in Drosophila.


Subject(s)
Apoptosis , Cell Cycle Proteins/deficiency , Drosophila Proteins/deficiency , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Imaginal Discs/growth & development , Wings, Animal/cytology , Animals , Apoptosis/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Cell Survival , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Epistasis, Genetic , Gene Silencing , Genes, Insect , Imaginal Discs/cytology , JNK Mitogen-Activated Protein Kinases/metabolism , Larva/genetics , Mutation/genetics , Receptors, Notch/metabolism , Signal Transduction/genetics , Stress, Physiological/genetics , Transcription Factors/metabolism , Wings, Animal/growth & development
4.
Genetics ; 188(1): 127-39, 2011 May.
Article in English | MEDLINE | ID: mdl-21385730

ABSTRACT

Putzig (Pzg) was originally identified as being an integral component of the TRF2/DREF complex in Drosophila melanogaster, thereby regulating the transcriptional activation of replication-related genes. In a DREF-independent manner, Pzg was shown to mediate Notch target gene activation. This function of Pzg entails an association with the nucleosome remodeling factor complex NURF, which directly binds the ecdysone receptor EcR and coregulates targets of the EcR via the NURF-specific subunit Nurf-301. In contrast, Nurf-301 acts as a negative regulator of JAK/STAT signaling. Here, we provide evidence to show that Pzg is fundamental for these functions of NURF, apart from the regulation of Notch signaling activity. A jump-out mutagenesis provided us with a pzg null mutant displaying early larval lethality, defects in growth, and molting accompanied by aberrant feeding behavior. We show that Pzg is associated with EcR in vivo and required for the transcriptional induction of EcR target genes, whereas reduced ecdysteroid levels imply a NURF-independent function of Pzg. Moreover, pzg interferes with JAK/STAT-signaling activity by acting as a corepressor of Ken. Lamellocyte differentiation was consistently affected in a JAK/STAT mutant background and the expression level of defense response genes was elevated in pzg mutants, leading to the formation of melanotic tumors. Our results suggest that Pzg acts as an important partner of NURF in the regulation of EcR and JAK/STAT signaling.


Subject(s)
Cell Cycle Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/immunology , Immunity, Innate/immunology , Multiprotein Complexes/metabolism , Nucleosomes/metabolism , Receptors, Steroid/metabolism , Signal Transduction , Animals , Cell Cycle Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Gene Silencing , Janus Kinases/metabolism , Metamorphosis, Biological , Mutation/genetics , Neoplasms/pathology , Protein Binding , Reproducibility of Results , STAT Transcription Factors/metabolism
5.
Mol Biol Cell ; 21(19): 3443-8, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20685964

ABSTRACT

Drosophila putzig was identified as a member of the TRF2-DREF complex that is involved in core promoter selection. Additionally, putzig regulates Notch signaling, however independently of DREF. Here, we show that Putzig associates with the NURF complex. Loss of any NURF component including the NURF-specific subunit Nurf 301 impedes binding of Putzig to Notch target genes, suggesting that NURF recruits Putzig to these sites. Accordingly, Putzig can be copurified with any NURF member. Moreover, Nurf 301 mutants show reduced Notch target gene activity and enhance Notch mutant phenotypes. These data suggest a novel Putzig-NURF chromatin complex required for epigenetic activation of Notch targets.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation , Receptors, Notch/genetics , Animals , Clone Cells , DNA/metabolism , Drosophila melanogaster/cytology , Mutation/genetics , Protein Binding , Receptors, Notch/metabolism
6.
Mol Biol Cell ; 18(10): 3733-40, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17634285

ABSTRACT

We have identified the gene putzig (pzg) as a key regulator of cell proliferation and of Notch signaling in Drosophila. pzg encodes a Zn-finger protein that was found earlier within a macromolecular complex, including TATA-binding protein-related factor 2 (TRF2)/DNA replication-related element factor (DREF). This complex is involved in core promoter selection, where DREF functions as a transcriptional activator of replication-related genes. Here, we provide the first in vivo evidence that pzg is required for the expression of cell cycle and replication-related genes, and hence for normal developmental growth. Independent of its role in the TRF2/DREF complex, pzg acts as a positive regulator of Notch signaling that may occur by chromatin activation. Down-regulation of pzg activity inhibits Notch target gene activation, whereas Hedgehog (Hh) signal transduction and growth regulation are unaffected. Our findings uncover different modes of operation of pzg during imaginal development of Drosophila, and they provide a novel mechanism of Notch regulation.


Subject(s)
Cell Cycle Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Receptors, Notch/metabolism , Animals , Cell Cycle , Cell Proliferation , Chromatin/metabolism , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcriptional Activation
7.
BMC Dev Biol ; 7: 74, 2007 Jun 25.
Article in English | MEDLINE | ID: mdl-17592635

ABSTRACT

BACKGROUND: Protein Kinase D (PKD) is an effector of diacylglycerol-regulated signaling pathways. Three isoforms are known in mammals that have been linked to diverse cellular functions including regulation of cell proliferation, differentiation, motility and secretory transport from the trans-Golgi network to the plasma membrane. In Drosophila, there is a single PKD orthologue, whose broad expression implicates a more general role in development. RESULTS: We have employed tissue specific overexpression of various PKD variants as well as tissue specific RNAi, in order to investigate the function of the PKD gene in Drosophila. Apart from a wild type (WT), a kinase dead (kd) and constitutively active (SE) Drosophila PKD variant, we also analyzed two human isoforms hPKD2 and hPKD3 for their capacity to substitute PKD activity in the fly. Overexpression of either WT or kd-PKD variants affected primarily wing vein development. However, overexpression of SE-PKD and PKD RNAi was deleterious. We observed tissue loss, wing defects and degeneration of the retina. The latter phenotype conforms to a role of PKD in the regulation of cytoskeletal dynamics. Strongest phenotypes were larval to pupal lethality. RNAi induced phenotypes could be rescued by a concurrent overexpression of Drosophila wild type PKD or either human isoform hPKD2 and hPKD3. CONCLUSION: Our data confirm the hypothesis that Drosophila PKD is a multifunctional kinase involved in diverse processes such as regulation of the cytoskeleton, cell proliferation and death as well as differentiation of various fly tissues.


Subject(s)
Drosophila melanogaster/enzymology , Drosophila melanogaster/growth & development , Isoenzymes/metabolism , Protein Kinase C/metabolism , Aging/physiology , Animals , Animals, Genetically Modified , Cell Line , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/anatomy & histology , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Isoenzymes/genetics , Larva/anatomy & histology , Larva/physiology , Light , Phenotype , Photoreceptor Cells, Invertebrate/metabolism , Photoreceptor Cells, Invertebrate/pathology , Protein Kinase C/genetics , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Wings, Animal/anatomy & histology , Wings, Animal/growth & development
8.
Gene Expr Patterns ; 6(8): 849-56, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16750940

ABSTRACT

Protein kinase D belongs to the subfamily of CaMK. In mammals, three isoforms are known. They have been linked to diverse cellular functions including regulation of cell proliferation, differentiation, apoptosis and motility as well as secretory transport from the trans-Golgi compartment to the plasma membrane. Accordingly, the mammalian PKDs show different intracellular locations, with reported dynamic redistribution, between cytosol, Golgi, plasma membranes and the nucleus, depending on the cell type and exogenous stimuli. The genome of Drosophila melanogaster harbours just one, highly conserved PKD homologue, which is expressed throughout development. PKD mRNA expression during late embryogenesis is restricted to ectodermal derivatives including those involved in cuticle secretion. In imaginal tissues, transcription appears more uniform. PKD protein is detected predominantly in the cytosol with an enrichment in lateral apodemes of late embryos as well as in larval fascicles. In secretory tissues like salivary glands, the protein is concentrated in dotted structures. A PKD-GFP transgene reveals a similar punctuate protein accumulation juxtaposed to a resident Golgi-marker. In cultured cells, transfected Drosophila PKD-GFP colocalizes with a marker of the trans-Golgi compartment like human PKD1-GFP. Similar to the mammalian homologues, Drosophila PKD may be multifunctional including a role in secretory transport in accordance with its subcellular distribution.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Golgi Apparatus/metabolism , Protein Kinase C/metabolism , Amino Acid Sequence , Animals , Biological Transport/physiology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Drosophila melanogaster/genetics , Embryo, Nonmammalian , Embryonic Development/physiology , Gene Dosage , Green Fluorescent Proteins/metabolism , Humans , Models, Biological , Molecular Sequence Data , Protein Kinase C/genetics , Protein Kinase C/physiology , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Tissue Distribution , Transcription, Genetic , Transfection
9.
Genetics ; 171(3): 1137-52, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16118195

ABSTRACT

Overexpression of Hairless (H) causes a remarkable degree of tissue loss and apoptosis during imaginal development. H functions as antagonist in the Notch-signaling pathway in Drosophila, and the link to growth and apoptosis is poorly understood. To further our insight into H-mediated apoptosis, we performed two large-scale screens for modifiers of a small rough eye phenotype caused by H overexpression. Both loss- and gain-of-function screens revealed known and new genetic interactors representing diverse cellular functions. Many of them did not cause eye phenotypes on their own, emphasizing a specific genetic interaction with H. As expected, we also identified components of different signaling pathways supposed to be involved in the regulation of cell growth and cell death. Accordingly, some of them also acted as modifiers of proapoptotic genes, suggesting a more general involvement in the regulation of apoptosis. Overall, these screens highlight the importance of H and the Notch pathway in mediating cell death in response to developmental and environmental cues and emphasize their role in maintaining developmental cellular homeostasis.


Subject(s)
Apoptosis/genetics , Cell Differentiation/genetics , Cell Enlargement , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Transcription Factors/genetics , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , ErbB Receptors/physiology , Eye Abnormalities/genetics , Female , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/physiology , Male , Protein Kinases/physiology , Receptors, Invertebrate Peptide/physiology , Signal Transduction/physiology , Transcription Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...