Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
iScience ; 26(11): 108193, 2023 Nov 17.
Article in English | MEDLINE | ID: mdl-37920666

ABSTRACT

DNMT3 in Hymenoptera has a unique duplication of the essential PWWP domain. Using GST-tagged PWWP fusion proteins and histone arrays we show that these domains have gained new properties and represent the first case of PWWP domains binding to H3K27 chromatin modifications, including H3K27me3, a key modification that is important during development. Phylogenetic analyses of 107 genomes indicate that the duplicated PWWP domains separated into two sister clades, and their distinct binding capacities are supported by 3D modeling. Other features of this unique DNA methylation system include variable copies, losses, and duplications of DNMT1 and DNMT3, and combinatorial generations of DNMT3 isoforms including variants missing the catalytic domain. Some of these losses and duplications of are found only in parasitic wasps. We discuss our findings in the context of the crosstalk between DNA methylation and histone methylation, and the expanded potential of epigenomic modifications in Hymenoptera to drive evolutionary novelties.

2.
Nucleic Acids Res ; 50(20): 11550-11562, 2022 11 11.
Article in English | MEDLINE | ID: mdl-36330958

ABSTRACT

The honey bee genome has the capacity to produce three phenotypically distinct organisms (two diploid female castes: queen and worker, and a haploid male drone). Previous studies have implicated metabolic flux acting via epigenetic regulation in directing nutrition-driven phenotypic plasticity in the honey bee. However, the cis-acting DNA regulatory elements that establish tissue and polyphenism -specific epigenomes and gene expression programmes, remain unclear. Using a high resolution multiomic approach including assay for transposase-accessible chromatin by sequencing (ATAC-seq), RNA-seq and ChIP-seq, we produce the first genome-wide maps of the regulatory landscape across all three adult honey bee phenotypes identifying > 5000 regulatory regions in queen, 7500 in worker and 6500 in drone, with the vast majority of these sites located within intronic regions. These regions are defined by positive enrichment of H3K27ac and depletion of H3K4me3 and show a positive correlation with gene expression. Using ATAC-seq footprinting we determine queen, worker and drone -specific transcription factor occupancy and uncover novel phenotype-specific regulatory networks identifying two key nuclear receptors that have previously been implicated in caste-determination and adult behavioural maturation in honey bees; ecdysone receptor and ultraspiracle. Collectively, this study provides novel insights into key gene regulatory networks that are associated with these distinct polyphenisms in the honey bee.


Subject(s)
Bees , Brain , Chromatin , Gene Regulatory Networks , Animals , Female , Male , Bees/genetics , Chromatin/genetics , Chromatin/metabolism , Epigenesis, Genetic , Larva/genetics
3.
Data Brief ; 36: 107019, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33937454

ABSTRACT

Forager Apis melliefera honeybees were collected from four localities located in Europe, i.e.: London, UK; Athens, Greece; Marchamalo, Spain and Lublin, Poland. Furthermore, from Asia we have collected A. mellifera as well as A. cerana foragers form Chiang Mai in Thailand We used next generation sequencing (NGS) to analyse the 16S rRNA bacterial gene amplicons based on the V3-V4 region and the ITS2 region from fungi and plants derived from honeybee samples. Amplicon libraries, were prepared using the 16S Metagenomic Sequencing Library Preparation, Preparing 16S Ribosomal RNA Gene Amplicons for the Illumina MiSeq System (Illumina®) protocol. NGS raw data are available at https://www.ncbi.nlm.nih.gov/bioproject/PRJNA686953. Furthermore, isolated DNA was used as the template for screening pathogens: Nosema apis, N. ceranae, N. bombi, tracheal mite (Acarapis woodi), any organism in the parasitic order Trypanosomatida, including Crithidia spp. (i.e., Crithidia mellificae), neogregarines including Mattesia and Apicystis spp. (i.e., Apicistis bombi). The presented data can be used to compare the metagenomic samples from different honeybee population all over the world. A higher load of fungi, and bacteria groups such as: Firmicutes (Lactobacillus); γ- proteobacteria, Neisseriaceae, and other unidentified bacteria was observed for Nosema cearana and neogregarines infected honeybees. Healthy honeybees had a higher load of plant pollens, and bacteria groups such as: Orbales, Gilliamella, Snodgrassella, and Enterobacteriaceae. More details can be found in research article [1] Ptaszynska et al. 2021.

4.
Pathogens ; 10(3)2021 Mar 22.
Article in English | MEDLINE | ID: mdl-33810160

ABSTRACT

European Apis mellifera and Asian Apis cerana honeybees are essential crop pollinators. Microbiome studies can provide complex information on health and fitness of these insects in relation to environmental changes, and plant availability. Amplicon sequencing of variable regions of the 16S rRNA from bacteria and the internally transcribed spacer (ITS) regions from fungi and plants allow identification of the metabiome. These methods provide a tool for monitoring otherwise uncultured microbes isolated from the gut of the honeybees. They also help monitor the composition of the gut fungi and, intriguingly, pollen collected by the insect. Here, we present data from amplicon sequencing of the 16S rRNA from bacteria and ITS2 regions from fungi and plants derived from honeybees collected at various time points from anthropogenic landscapes such as urban areas in Poland, UK, Spain, Greece, and Thailand. We have analysed microbial content of honeybee intestine as well as fungi and pollens. Furthermore, isolated DNA was used as the template for screening pathogens: Nosema apis, N. ceranae, N. bombi, tracheal mite (Acarapis woodi), any organism in the parasitic order Trypanosomatida, including Crithidia spp. (i.e., Crithidia mellificae), neogregarines including Mattesia and Apicystis spp. (i.e., Apicistis bombi). We conclude that differences between samples were mainly influenced by the bacteria, plant pollen and fungi, respectively. Moreover, honeybees feeding on a sugar based diet were more prone to fungal pathogens (Nosema ceranae) and neogregarines. In most samples Nosema sp. and neogregarines parasitized the host bee at the same time. A higher load of fungi, and bacteria groups such as Firmicutes (Lactobacillus); γ-proteobacteria, Neisseriaceae, and other unidentified bacteria was observed for Nosema ceranae and neogregarine infected honeybees. Healthy honeybees had a higher load of plant pollen, and bacteria groups such as: Orbales, Gilliamella, Snodgrassella, and Enterobacteriaceae. Finally, the period when honeybees switch to the winter generation (longer-lived forager honeybees) is the most sensitive to diet perturbations, and hence pathogen attack, for the whole beekeeping season. It is possible that evolutionary adaptation of bees fails to benefit them in the modern anthropomorphised environment.

5.
Sci Rep ; 11(1): 6852, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33767244

ABSTRACT

In the course of a screen designed to produce antibodies (ABs) with affinity to proteins in the honey bee brain we found an interesting AB that detects a highly specific epitope predominantly in the nuclei of Kenyon cells (KCs). The observed staining pattern is unique, and its unfamiliarity indicates a novel previously unseen nuclear structure that does not colocalize with the cytoskeletal protein f-actin. A single rod-like assembly, 3.7-4.1 µm long, is present in each nucleus of KCs in adult brains of worker bees and drones with the strongest immuno-labelling found in foraging bees. In brains of young queens, the labelling is more sporadic, and the rod-like structure appears to be shorter (~ 2.1 µm). No immunostaining is detectable in worker larvae. In pupal stage 5 during a peak of brain development only some occasional staining was identified. Although the cellular function of this unexpected structure has not been determined, the unusual distinctiveness of the revealed pattern suggests an unknown and potentially important protein assembly. One possibility is that this nuclear assembly is part of the KCs plasticity underlying the brain maturation in adult honey bees. Because no labelling with this AB is detectable in brains of the fly Drosophila melanogaster and the ant Camponotus floridanus, we tentatively named this antibody AmBNSab (Apis mellifera Brain Neurons Specific antibody). Here we report our results to make them accessible to a broader community and invite further research to unravel the biological role of this curious nuclear structure in the honey bee central brain.


Subject(s)
Bees/growth & development , Brain/cytology , Cell Nucleus/metabolism , Drosophila melanogaster/growth & development , Larva/cytology , Neurons/cytology , Pupa/cytology , Animals , Bees/immunology , Bees/metabolism , Brain/immunology , Brain/metabolism , Drosophila melanogaster/immunology , Drosophila melanogaster/metabolism , Immunohistochemistry , Larva/immunology , Larva/metabolism , Neurons/immunology , Neurons/metabolism , Pupa/immunology , Pupa/metabolism
6.
Ecol Evol ; 10(14): 7616-7626, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32760552

ABSTRACT

Understanding the effects of temperature on ecological and evolutionary processes is crucial for generating future climate adaptation scenarios. Using experimental evolution, we evolved the model ciliate Tetrahymena thermophila in an initially novel high temperature environment for more than 35 generations, closely monitoring population dynamics and morphological changes. We observed initially long lag phases in the high temperature environment that over about 26 generations reduced to no lag phase, a strong reduction in cell size and modifications in cell shape at high temperature. When exposing the adapted populations to their original temperature, most phenotypic traits returned to the observed levels in the ancestral populations, indicating phenotypic plasticity is an important component of this species thermal stress response. However, persistent changes in cell size were detected, indicating possible costs related to the adaptation process. Exploring the molecular basis of thermal adaptation will help clarify the mechanisms driving these phenotypic responses.

7.
Proc Natl Acad Sci U S A ; 117(38): 23311-23316, 2020 09 22.
Article in English | MEDLINE | ID: mdl-31332005

ABSTRACT

Prolonged social isolation has negative effects on brain and behavior in humans and other social organisms, but neural mechanisms leading to these effects are not understood. Here we tested the hypothesis that even brief periods of social isolation can alter gene expression and DNA methylation in higher cognitive centers of the brain, focusing on the auditory/associative forebrain of the highly social zebra finch. Using RNA sequencing, we first identified genes that individually increase or decrease expression after isolation and observed general repression of gene sets annotated for neurotrophin pathways and axonal guidance functions. We then pursued 4 genes of large effect size: EGR1 and BDNF (decreased by isolation) and FKBP5 and UTS2B (increased). By in situ hybridization, each gene responded in different cell subsets, arguing against a single cellular mechanism. To test whether effects were specific to the social component of the isolation experience, we compared gene expression in birds isolated either alone or with a single familiar partner. Partner inclusion ameliorated the effect of solo isolation on EGR1 and BDNF, but not on FKBP5 and UTS2B nor on circulating corticosterone. By bisulfite sequencing analysis of auditory forebrain DNA, isolation caused changes in methylation of a subset of differentially expressed genes, including BDNF. Thus, social isolation has rapid consequences on gene activity in a higher integrative center of the brain, triggering epigenetic mechanisms that may influence processing of ongoing experience.


Subject(s)
Finches/genetics , Prosencephalon/metabolism , Social Isolation , Animals , Behavior, Animal , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Corticosterone/blood , DNA Methylation , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Female , Finches/blood , Finches/physiology , Male , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism
8.
Genome Res ; 28(10): 1532-1542, 2018 10.
Article in English | MEDLINE | ID: mdl-30135090

ABSTRACT

The capacity of the honey bee to produce three phenotypically distinct organisms (two female castes; queens and sterile workers, and haploid male drones) from one genotype represents one of the most remarkable examples of developmental plasticity in any phylum. The queen-worker morphological and reproductive divide is environmentally controlled during post-embryonic development by differential feeding. Previous studies implicated metabolic flux acting via epigenetic regulation, in particular DNA methylation and microRNAs, in establishing distinct patterns of gene expression underlying caste-specific developmental trajectories. We produce the first genome-wide maps of chromatin structure in the honey bee at a key larval stage in which developmental canalization into queen or worker is virtually irreversible. We find extensive genome-wide differences in H3K4me3, H3K27ac, and H3K36me3, many of which correlate with caste-specific transcription. Furthermore, we identify H3K27ac as a key chromatin modification, with caste-specific regions of intronic H3K27ac directing the worker caste. These regions may harbor the first examples of caste-specific enhancer elements in the honey bee. Our results demonstrate a key role for chromatin modifications in the establishment and maintenance of caste-specific transcriptional programs in the honey bee. We show that at 96 h of larval growth, the queen-specific chromatin pattern is already established, whereas the worker determination is not, thus providing experimental support for the perceived timing of this critical point in developmental heterochrony in two types of honey bee females. In a broader context, our study provides novel data on environmentally regulated organismal plasticity and the molecular foundation of the evolutionary origins of eusociality.


Subject(s)
Bees/growth & development , Chromatin/genetics , Epigenesis, Genetic , Insect Proteins/genetics , Animals , Bees/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Female , Gene Expression Regulation, Developmental , Genotype , Histones/metabolism , Larva/genetics , Larva/growth & development , Male , Phenotype , Sequence Analysis, DNA , Sequence Analysis, RNA
9.
PLoS One ; 13(7): e0200410, 2018.
Article in English | MEDLINE | ID: mdl-30044811

ABSTRACT

Proper bioelement content is crucial for the health and wellness of all organisms, including honeybees. However, the situation is more complicated in these important pollinators due to the fact that they change their physiology during winter in order to survive the relatively harsh climatic conditions. Additionally, honeybees are susceptible to many diseases such as nosemosis, which during winter can depopulate an entire colony. Here we show that summer bees have a markedly higher content of important bioelements such as: Al, Cu, P, V, (physiologically essential); Ca, K, Mg, (electrolytic); Cr, Se, Zn, (enzymatic); As, Hg, (toxic). In contrast, a markedly higher content of: Fe (physiologically essential); Mn, Ni, (enzymatic); Cd (exclusively toxic) were present in winter bees. Importantly, N. ceranae infection resulted in an increased honeybee bioelement content of: S, Sr (physiologically essential) and Pb (exclusively toxic), whereas the Nosema-free worker-bees had higher amounts of B and Si (physiologically essential). We propose that the shortages of Fe, Mn, Ni, and Na observed in Nosema-infected bees, could be the reason for the higher mortality of Nosema-infected bees throughout overwintering. In addition, a shortage of bioelements such as B and Si may be a reason for accelerated aging in foragers that is observed following N. ceranae infection. Therefore, in winter, bioelement content was more strongly affected by N. ceranae infection than during summer. We found a strong correlation between the bioelement content of bees and seasons (summer or winter) and also with Nosema infection. We conclude that the balance of bioelements in the honeybee is altered by both seasonal affects and by Nosema infection.


Subject(s)
Bees/metabolism , Bees/microbiology , Honey/analysis , Microsporidiosis/veterinary , Nosema , Animals , Female , Microsporidiosis/metabolism , Seasons
10.
Mol Hum Reprod ; 21(11): 865-83, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26405173

ABSTRACT

Human parturition is associated with many pro-inflammatory mediators which are regulated by the nuclear factor-kappaB (NF-κB) family of transcription factors. In the present study, we employed a ChIP-on-chip approach to define genomic loci within chromatin of PHM1-31 myometrial cells that were occupied by RelA-containing NF-κB dimers in response to a TNF stimulation of 1 h. In TNF-stimulated PHM1-31 cells, anti-RelA serum enriched 13 300 chromatin regions; importantly, 11 110 regions were also enriched by anti-RelA antibodies in the absence of TNF. DNA sequences in these regions, from both unstimulated or TNF-stimulated PHM1-31 cultures, were associated with genic regions including IκBα, COX-2, IL6RN, Jun and KCNMB3. TNF-induced binding events at a consensus κB site numbered 1667; these were represented by 112 different instances of the consensus κB motif. Of the 1667 consensus κB motif occurrences, 770 (46.2%) were identified within intronic regions. In unstimulated PHM1-31 cells, anti-RelA-serum-enriched regions were associated with sequences corresponding to open reading frames of ion channel subunit genes including CACNB3 and KCNB1. Moreover, in unstimulated cells, the consensus κB site was identified 2116 times, being defined by 103 different sequence instances of this motif. Of these 2116 consensus κB motifs, 1089 (51.5%) were identified within intronic regions. Parallel expression array analyses in PHM1-31 cultures demonstrated that TNF stimulated a >2-fold induction in 51 genes and a fold repression of >1.5 in 18 others. We identified 14 anti-RelA-serum-enriched genomic regions that correlated with 17 TNF-inducible genes, such as COX2, Egr-1, Jun, IκBα and IL6, as well as five regions associated with TNF-mediated gene repression, including Col1A2.


Subject(s)
Myocytes, Smooth Muscle/metabolism , Myometrium/cytology , NF-kappa B/metabolism , Promoter Regions, Genetic/genetics , Transcription Factor RelA/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Female , Humans , Myocytes, Smooth Muscle/drug effects , NF-kappa B/genetics , Pregnancy , Protein Multimerization , Shab Potassium Channels/genetics , Shab Potassium Channels/metabolism , Transcription Factor RelA/genetics , Transcriptional Activation/drug effects , Tumor Necrosis Factor-alpha/pharmacology
11.
RNA ; 21(3): 347-59, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25605962

ABSTRACT

Splicing factor proline- and glutamine-rich (SFPQ) also commonly known as polypyrimidine tract-binding protein-associated-splicing factor (PSF) and its binding partner non-POU domain-containing octamer-binding protein (NONO/p54nrb), are highly abundant, multifunctional nuclear proteins. However, the exact role of this complex is yet to be determined. Following purification of the endogeneous SFPQ/NONO complex, mass spectrometry analysis identified a wide range of interacting proteins, including those involved in RNA processing, RNA splicing, and transcriptional regulation, consistent with a multifunctional role for SFPQ/NONO. In addition, we have identified several sites of arginine methylation in SFPQ/PSF using mass spectrometry and found that several arginines in the N-terminal domain of SFPQ/PSF are asymmetrically dimethylated. Furthermore, we find that the protein arginine N-methyltransferase, PRMT1, catalyzes this methylation in vitro and that this is antagonized by citrullination of SFPQ. Arginine methylation and citrullination of SFPQ/PSF does not affect complex formation with NONO. However, arginine methylation was shown to increase the association with mRNA in mRNP complexes in mammalian cells. Finally we show that the biochemical properties of the endogenous complex from cell lysates are significantly influenced by the ionic strength during purification. At low ionic strength, the SFPQ/NONO complex forms large heterogeneous protein assemblies or aggregates, preventing the purification of the SFPQ/NONO complex. The ability of the SFPQ/NONO complex to form varying protein assemblies, in conjunction with the effect of post-translational modifications of SFPQ modulating mRNA binding, suggests key roles affecting mRNP dynamics within the cell.


Subject(s)
Nuclear Matrix-Associated Proteins/genetics , Octamer Transcription Factors/genetics , RNA Splicing/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Transcription, Genetic , Animals , Arginine/genetics , Arginine/metabolism , DNA-Binding Proteins , Gene Expression Regulation , HeLa Cells , Humans , Methylation , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Nuclear Matrix-Associated Proteins/chemistry , Octamer Transcription Factors/chemistry , PTB-Associated Splicing Factor , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/genetics , RNA-Binding Proteins/chemistry , Repressor Proteins/genetics , Ribonucleoproteins/genetics
12.
J Cell Biochem ; 115(6): 1048-62, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23564320

ABSTRACT

The activity of S6 kinases (S6K) is highly induced in cancer cells highlighting an essential role in carcinogenesis. The S6K family has two members: S6K1 and S6K2 which bear common as well as distinct features. In an attempt to identify S6K2 unique sequence features compared to S6K1, we applied extensive bioinformatic analysis and motif search approaches. Interestingly, we identified 14 unique protein signatures which are present in proteins directly connected to chromatin and/or involved in transcription regulation. Using chromatin binding assay, we biochemically showed that S6K2 is bound to chromatin as well as nuclear matrix cellular fractions in HEK293 cells. The presence of S6K2 in chromatin fractions raised the possibility that it may be in close proximity to a number of chromatin substrates. For that, we then searched for S6K phosphorylation consensus sites RXRXXT/S in mammalian proteins using the SWISS-PROT database. Interestingly, we identified some potential phosphorylation sites in histone H3 (Thr45). Using in vitro kinase assays and siRNA-based knockdown strategy; we confirmed that S6K2 but not S6K1 or AKT is essential for histone H3-Thr45 phosphorylation in HEK293 cells. Furthermore, we show that the nuclear localisation sequence in the S6K2 C-terminus is essential for this modification. We have found that, H3-Thr45 phosphorylation correlates to S6K activation in response to mitogens and TPA-induced cell differentiation of leukaemic cell lines U937, HL60 and THP1. Overall, we demonstrate that S6K2 is a novel kinase that can phosphorylate histone H3 at position Thr45, which may play a role during cell proliferation and/or differentiation.


Subject(s)
Chromatin/metabolism , Histones/metabolism , Nuclear Matrix/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Threonine/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Cell Differentiation/drug effects , Cell Fractionation , Cell Line, Tumor , Chromatin/genetics , HEK293 Cells , HL-60 Cells , Humans , Mice , Middle Aged , Molecular Sequence Data , NIH 3T3 Cells , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Tetradecanoylphorbol Acetate/pharmacology , U937 Cells
13.
Biochem Soc Trans ; 41(3): 697-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23697928

ABSTRACT

Our advances in technology allow us to sequence DNA to uncover genetic differences not only between individuals, but also between normal and diseased cells within an individual. However, there is still a lot we have yet to understand regarding the epigenetic mechanisms that also contribute to our individuality and to disease. The 80th Biochemical Society Annual Symposium entitled Epigenetic Mechanisms in Development and Disease brought together some leading researchers in the field who discussed their latest insights into epigenetic mechanisms. Methylation of DNA has been the focus of much study from both a developmental perspective and imprinting of genes to its contribution to diseases such as cancer. Recently, the modification of methylcytosine to hydoxymethylcytosine within cells was uncovered, which opened a host of potential new mechanisms, and a flurry of new studies are underway to uncover its significance. Epigenetics is not confined to a study of DNA, and the post-translational modifications on the histone proteins have a significant role to play in regulating gene expression. There are many different modifications and, as shown at the Symposium, new variations used by cells are still being uncovered. We are some way to identifying how these modifications are added and removed and the protein complexes responsible for these changes. A focus on the function of the complexes and the interactions between individual modifications to regulate gene expression is advancing our knowledge, as discussed in the accompanying papers, although there are clearly plenty of opportunities for new breakthroughs to be made.


Subject(s)
Disease/genetics , Epigenesis, Genetic/physiology , Growth and Development/genetics , Animals , DNA Methylation , Epigenesis, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histone Deacetylases/physiology , Histones/metabolism , Humans , Protein Processing, Post-Translational
14.
Genome Biol ; 14(5): R43, 2013 May 25.
Article in English | MEDLINE | ID: mdl-23706135

ABSTRACT

BACKGROUND: Inter-individual epigenetic variation, due to genetic, environmental or random influences, is observed in many eukaryotic species. In mammals, however, the molecular nature of epiallelic variation has been poorly defined, partly due to the restricted focus on DNA methylation. Here we report the first genome-scale investigation of mammalian epialleles that integrates genomic, methylomic, transcriptomic and histone state information. RESULTS: First, in a small sample set, we demonstrate that non-genetically determined inter-individual differentially methylated regions (iiDMRs) can be temporally stable over at least 2 years. Then, we show that iiDMRs are associated with changes in chromatin state as measured by inter-individual differences in histone variant H2A.Z levels. However, the correlation of promoter iiDMRs with gene expression is negligible and not improved by integrating H2A.Z information. We find that most promoter epialleles, whether genetically or non-genetically determined, are associated with low levels of transcriptional activity, depleted for housekeeping genes, and either depleted for H3K4me3/enriched for H3K27me3 or lacking both these marks in human embryonic stem cells. The preferential enrichment of iiDMRs at regions of relative transcriptional inactivity validates in a larger independent cohort, and is reminiscent of observations previously made for promoters that undergo hypermethylation in various cancers, in vitro cell culture and ageing. CONCLUSIONS: Our work identifies potential key features of epiallelic variation in humans, including temporal stability of non-genetically determined epialleles, and concomitant perturbations of chromatin state. Furthermore, our work suggests a novel mechanistic link among inter-individual epialleles observed in the context of normal variation, cancer and ageing.


Subject(s)
Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Genomics/methods , Promoter Regions, Genetic , Twins, Monozygotic/genetics , Alleles , DNA Methylation , Female , Gene Expression Regulation , Genome, Human , Histones/metabolism , Humans , Molecular Sequence Data
15.
Insect Biochem Mol Biol ; 43(2): 125-37, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23174121

ABSTRACT

Histone post-translational modifications (PTMs) play a key role in regulating a variety of cellular processes including the establishment, maintenance and reversal of transcriptional programmes in eukaryotes. However, little is known about such modifications in the economically and ecologically important insect pollinator, the honey bee (Apis mellifera). Using mass spectrometry approaches, we show that histone H3.1, H3.3 and H4 of the honey bee are extensively modified by lysine acetylation and lysine methylation. We analysed histones isolated from queen ovaries and 96 hr-old larvae, in toto we quantified 23 specific modification states on 23 distinct peptides. In addition, we have identified and characterised patterns of histone PTMs that reside on the same peptide, generating detailed combinatorial information. Overall, we observed similar profiles of histone PTMs in both samples, with combinatorial patterns of lysine methylations on H3K27 and H3K36 more frequently identified in histones extracted from queen ovaries than from larvae. To our knowledge, this comprehensive dataset represents the first identification and quantitation of histone PTMs in this eusocial insect and emerging epigenetic model.


Subject(s)
Bees/metabolism , Histones/metabolism , Insect Proteins/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Animals , Bees/chemistry , Bees/genetics , Histones/chemistry , Histones/genetics , Insect Proteins/chemistry , Insect Proteins/genetics , Mass Spectrometry , Methylation , Molecular Sequence Data
16.
Brief Funct Genomics ; 9(5-6): 425-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21266345
17.
Brief Funct Genomic Proteomic ; 8(3): 174-83, 2009 May.
Article in English | MEDLINE | ID: mdl-19535508

ABSTRACT

Several recent studies from the field of epigenetics have combined chromatin-immunoprecipitation (ChIP) with next-generation high-throughput sequencing technologies to describe the locations of histone post-translational modifications (PTM) and DNA methylation genome-wide. While these reports begin to quench the chromatin biologists thirst for visualizing where in the genome epigenetic marks are placed, they also illustrate several advantages of sequencing based genomics compared to microarray analysis. Accordingly, next-generation sequencing (NGS) technologies are now challenging microarrays as the tool of choice for genome analysis. The increased affordability of comprehensive sequence-based genomic analysis will enable new questions to be addressed in many areas of biology. It is inevitable that massively-parallel sequencing platforms will supercede the microarray for many applications, however, there are niches for microarrays to fill and interestingly we may very well witness a symbiotic relationship between microarrays and high-throughput sequencing in the future.


Subject(s)
Epigenesis, Genetic , Genetic Research , Oligonucleotide Array Sequence Analysis/methods , Sequence Analysis, DNA/methods , Animals , Humans
18.
J Biol Chem ; 284(24): 16575-16583, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19363025

ABSTRACT

Numerous post-translational modifications have been identified in histones. Most of these occur within the histone tails, but a few have been identified within the histone core sequences. Histone core post-translational modifications have the potential to directly modulate nucleosome structure and consequently DNA accessibility. Here, we identify threonine 45 of histone H3 (H3T45) as a site of phosphorylation in vivo. We find that phosphorylation of H3T45 (H3T45ph) increases dramatically in apoptotic cells, around the time of DNA nicking. To further explore this connection, we analyzed human neutrophil cells because they are short-lived cells that undergo apoptosis in vivo. Freshly isolated neutrophils contain very little H3T45ph, whereas cells cultured for 20 h possess significant amounts; the kinetics of H3T45ph induction closely parallel those of caspase-3 activation. Cytokine inhibition of neutrophil apoptosis leads to reduced levels of H3T45ph. We identify protein kinase C-delta as the kinase responsible for H3T45ph in vitro and in vivo. Given the nucleosomal position of H3T45, we postulate that H3T45ph induces structural change within the nucleosome to facilitate DNA nicking and/or fragmentation.


Subject(s)
Apoptosis/physiology , Histones/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Cell Differentiation/physiology , HL-60 Cells , Histones/chemistry , Humans , Nucleosomes/metabolism , Phosphorylation/physiology , Protein Conformation , Protein Kinase C-delta/metabolism , Threonine/metabolism
19.
J Biol Chem ; 283(15): 9878-85, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18272522

ABSTRACT

The ability to exogenously impose targeted epigenetic changes in the genome represents an attractive route for the simulation of genomic de novo epigenetic events characteristic of some diseases and for the study of their downstream effects and also provides a potential therapeutic approach for the heritable repression of selected genes. Here we demonstrate for the first time the ability of zinc finger peptides to deliver DNA cytosine methylation in vivo to a genomic integrated target promoter when expressed as fusions with a mutant prokaryotic DNA cytosine methyltransferase enzyme, thus mimicking cellular genomic de novo methylation events and allowing a direct analysis of the mechanics of de novo DNA methylation-mediated gene silencing at a genomic locus. We show that targeted methylation leads to gene silencing via the initiation of a repressive chromatin signature at the targeted genomic locus. This repression is maintained after the loss of targeted methyltransferase enzyme from the cell, confirming epigenetic maintenance purely through the action of cellular enzymes. The inherited DNA methylation pattern is restricted only to targeted sites, suggesting that the establishment of repressive chromatin structure does not drive further de novo DNA methylation in this system. As well as demonstrating the potential of these enzymes as tools for the exogenous, heritable control of cellular gene expression, this work also provides the most definitive confirmation to date for a transcriptionally repressive role for de novo DNA methylation in the cell and lends some weight to the hypothesis that the aberrant methylation associated with certain diseases may well be a cause rather than a consequence of transcriptional gene repression.


Subject(s)
Bacterial Proteins/biosynthesis , Chromosomes/metabolism , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA Methylation , Gene Silencing , Quantitative Trait Loci , Animals , Bacterial Proteins/genetics , COS Cells , Chlorocebus aethiops , Chromosomes/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Genome , Genomic Imprinting , Mice , NIH 3T3 Cells , Quantitative Trait Loci/genetics
20.
Biochim Biophys Acta ; 1783(1): 131-43, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17931718

ABSTRACT

Hesx1 is a highly conserved homeobox gene present in vertebrates, but absent from invertebrates. Gene targeting experiments in mice have shown that this transcriptional repressor is required for normal forebrain and pituitary development. In humans, mutations in HESX1 impairing either its repressing activity or DNA binding properties lead to a comparable phenotype to that observed in Hesx1 deficient mice. In an attempt to gain insights into the molecular function of HESX1, we have performed a yeast two-hybrid screen and identified DNA methyltransferase 1 (DNMT1) as a HESX1 binding protein. We show that Dnmt1 is co-expressed with Hesx1 within the anterior forebrain and in the developing Rathke's pouch. Mapping of the interacting regions indicates that the entire HESX1 protein is required to establish binding to a portion of the N-terminus of DNMT1 and its catalytic domain in the C-terminus. The HESX1-DNMT1 complexes can be immunoprecipitated in cells and co-localise in the nucleus. These results establish a link between HESX1 and DNMT1 and suggest a novel mechanism for the repressing properties of HESX1.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Repressor Proteins/metabolism , Transcription, Genetic/genetics , Animals , Cell Line , Cricetinae , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Homeodomain Proteins/genetics , Humans , Mice , Protein Binding , Repressor Proteins/genetics , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...