Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 139
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 185(20): 3689-3704.e21, 2022 09 29.
Article in English | MEDLINE | ID: mdl-36179666

ABSTRACT

Regulatory landscapes drive complex developmental gene expression, but it remains unclear how their integrity is maintained when incorporating novel genes and functions during evolution. Here, we investigated how a placental mammal-specific gene, Zfp42, emerged in an ancient vertebrate topologically associated domain (TAD) without adopting or disrupting the conserved expression of its gene, Fat1. In ESCs, physical TAD partitioning separates Zfp42 and Fat1 with distinct local enhancers that drive their independent expression. This separation is driven by chromatin activity and not CTCF/cohesin. In contrast, in embryonic limbs, inactive Zfp42 shares Fat1's intact TAD without responding to active Fat1 enhancers. However, neither Fat1 enhancer-incompatibility nor nuclear envelope-attachment account for Zfp42's unresponsiveness. Rather, Zfp42's promoter is rendered inert to enhancers by context-dependent DNA methylation. Thus, diverse mechanisms enabled the integration of independent Zfp42 regulation in the Fat1 locus. Critically, such regulatory complexity appears common in evolution as, genome wide, most TADs contain multiple independently expressed genes.


Subject(s)
Chromatin , Placenta , Animals , CCCTC-Binding Factor/metabolism , Chromatin Assembly and Disassembly , Enhancer Elements, Genetic , Evolution, Molecular , Female , Genome , Mammals/metabolism , Placenta/metabolism , Pregnancy , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Nat Rev Mol Cell Biol ; 22(8): 511-528, 2021 08.
Article in English | MEDLINE | ID: mdl-33953379

ABSTRACT

Understanding how chromatin is folded in the nucleus is fundamental to understanding its function. Although 3D genome organization has been historically difficult to study owing to a lack of relevant methodologies, major technological breakthroughs in genome-wide mapping of chromatin contacts and advances in imaging technologies in the twenty-first century considerably improved our understanding of chromosome conformation and nuclear architecture. In this Review, we discuss methods of 3D genome organization analysis, including sequencing-based techniques, such as Hi-C and its derivatives, Micro-C, DamID and others; microscopy-based techniques, such as super-resolution imaging coupled with fluorescence in situ hybridization (FISH), multiplex FISH, in situ genome sequencing and live microscopy methods; and computational and modelling approaches. We describe the most commonly used techniques and their contribution to our current knowledge of nuclear architecture and, finally, we provide a perspective on up-and-coming methods that open possibilities for future major discoveries.


Subject(s)
Chromatin/chemistry , Genome , Chromatin/genetics , Chromatin/metabolism , Chromosome Mapping , Chromosomes/chemistry , Chromosomes/genetics , Chromosomes/metabolism , Computational Biology , High-Throughput Nucleotide Sequencing , Humans , Microscopy , Models, Molecular , Sequence Analysis, DNA
3.
Cell ; 171(1): 34-57, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28938122

ABSTRACT

Polycomb (PcG) and Trithorax (TrxG) group proteins are evolutionarily conserved chromatin-modifying factors originally identified as part of an epigenetic cellular memory system that maintains repressed or active gene expression states. Recently, they have been shown to globally control a plethora of cellular processes. This functional diversity is achieved by their ability to regulate chromatin at multiple levels, ranging from modifying local chromatin structure to orchestrating the three-dimensional organization of the genome. Understanding this system is a fascinating challenge of critical relevance for biology and medicine, since misexpression or mutation of multiple PcG components, as well as of TrxG members of the COMPASS family and of the SWI/SNF complex, is implicated in cancer and other diseases.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation , Polycomb-Group Proteins/metabolism , Animals , Chromosomal Proteins, Non-Histone/history , Embryonic Stem Cells/metabolism , Genome , History, 20th Century , History, 21st Century , Humans , Neoplasms/metabolism , Polycomb-Group Proteins/history
4.
Cell ; 171(3): 557-572.e24, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-29053968

ABSTRACT

Chromosome conformation capture technologies have revealed important insights into genome folding. Yet, how spatial genome architecture is related to gene expression and cell fate remains unclear. We comprehensively mapped 3D chromatin organization during mouse neural differentiation in vitro and in vivo, generating the highest-resolution Hi-C maps available to date. We found that transcription is correlated with chromatin insulation and long-range interactions, but dCas9-mediated activation is insufficient for creating TAD boundaries de novo. Additionally, we discovered long-range contacts between gene bodies of exon-rich, active genes in all cell types. During neural differentiation, contacts between active TADs become less pronounced while inactive TADs interact more strongly. An extensive Polycomb network in stem cells is disrupted, while dynamic interactions between neural transcription factors appear in vivo. Finally, cell type-specific enhancer-promoter contacts are established concomitant to gene expression. This work shows that multiple factors influence the dynamics of chromatin interactions in development.


Subject(s)
Chromatin/metabolism , Genome , Neurogenesis , Animals , CCCTC-Binding Factor , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Exons , Gene Expression , Gene Regulatory Networks , Mice , Promoter Regions, Genetic , Repressor Proteins/metabolism , Transcription Factors/metabolism
5.
Cell ; 160(6): 1049-59, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25768903

ABSTRACT

The genome must be highly compacted to fit within eukaryotic nuclei but must be accessible to the transcriptional machinery to allow appropriate expression of genes in different cell types and throughout developmental pathways. A growing body of work has shown that the genome, analogously to proteins, forms an ordered, hierarchical structure that closely correlates and may even be causally linked with regulation of functions such as transcription. This review describes our current understanding of how these functional genomic "secondary and tertiary structures" form a blueprint for global nuclear architecture and the potential they hold for understanding and manipulating genomic regulation.


Subject(s)
Chromosomes/chemistry , Animals , Cell Differentiation , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromosomes/genetics , Chromosomes/metabolism , Enhancer Elements, Genetic , Genome , Humans , Promoter Regions, Genetic , Stem Cells/cytology , Stem Cells/metabolism
6.
Mol Cell ; 82(4): 816-832.e12, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35081363

ABSTRACT

Gene silencing by heterochromatin plays a crucial role in cell identity. Here, we characterize the localization, the biogenesis, and the function of an atypical heterochromatin, which is simultaneously enriched in the typical H3K9me3 mark and in H3K36me3, a histone mark usually associated with gene expression. We identified thousands of dual regions in mouse embryonic stem (ES) cells that rely on the histone methyltransferases SET domain bifurcated 1 (SETDB1) and nuclear set domain (NSD)-containing proteins to generate H3K9me3 and H3K36me3, respectively. Upon SETDB1 removal, dual domains lose both marks, gain signatures of active enhancers, and come into contact with upregulated genes, suggesting that it might be an important pathway by which genes are controlled by heterochromatin. In differentiated tissues, a subset of these dual domains is destabilized and becomes enriched in active enhancer marks, providing a mechanistic insight into the involvement of heterochromatin in the maintenance of cell identity.


Subject(s)
Chromatin Assembly and Disassembly , DNA Methylation , Enhancer Elements, Genetic , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Mouse Embryonic Stem Cells/enzymology , Protein Processing, Post-Translational , Animals , Cell Line , Chromatin Immunoprecipitation Sequencing , Gene Expression Profiling , Gene Expression Regulation, Developmental , Heterochromatin/genetics , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Methylation , Mice , RNA-Seq , Transcriptome
7.
Nat Rev Genet ; 23(6): 325-341, 2022 06.
Article in English | MEDLINE | ID: mdl-34983971

ABSTRACT

Increasing evidence indicates that non-DNA sequence-based epigenetic information can be inherited across several generations in organisms ranging from yeast to plants to humans. This raises the possibility of heritable 'epimutations' contributing to heritable phenotypic variation and thus to evolution. Recent work has shed light on both the signals that underpin these epimutations, including DNA methylation, histone modifications and non-coding RNAs, and the mechanisms by which they are transmitted across generations at the molecular level. These mechanisms can vary greatly among species and have a more limited effect in mammals than in plants and other animal species. Nevertheless, common principles are emerging, with transmission occurring either via direct replicative mechanisms or indirect reconstruction of the signal in subsequent generations. As these processes become clearer we continue to improve our understanding of the distinctive features and relative contribution of DNA sequence and epigenetic variation to heritable differences in phenotype.


Subject(s)
Epigenesis, Genetic , Inheritance Patterns , Animals , DNA Methylation , Databases, Genetic , Epigenomics , Mammals
8.
Mol Cell ; 78(3): 522-538.e9, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32220303

ABSTRACT

To understand the role of the extensive senescence-associated 3D genome reorganization, we generated genome-wide chromatin interaction maps, epigenome, replication-timing, whole-genome bisulfite sequencing, and gene expression profiles from cells entering replicative senescence (RS) or upon oncogene-induced senescence (OIS). We identify senescence-associated heterochromatin domains (SAHDs). Differential intra- versus inter-SAHD interactions lead to the formation of senescence-associated heterochromatin foci (SAHFs) in OIS but not in RS. This OIS-specific configuration brings active genes located in genomic regions adjacent to SAHDs in close spatial proximity and favors their expression. We also identify DNMT1 as a factor that induces SAHFs by promoting HMGA2 expression. Upon DNMT1 depletion, OIS cells transition to a 3D genome conformation akin to that of cells in replicative senescence. These data show how multi-omics and imaging can identify critical features of RS and OIS and discover determinants of acute senescence and SAHF formation.


Subject(s)
Cellular Senescence/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Genome, Human , Oncogenes , Cells, Cultured , Chromatin Assembly and Disassembly/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , Fibroblasts , Heterochromatin/genetics , Humans , In Situ Hybridization, Fluorescence
9.
PLoS Biol ; 22(5): e3002629, 2024 May.
Article in English | MEDLINE | ID: mdl-38805504

ABSTRACT

Despite significant progress in understanding epigenetic reprogramming of cells, the mechanistic basis of "organ reprogramming" by (epi-)gene-environment interactions remained largely obscure. Here, we use the ether-induced haltere-to-wing transformations in Drosophila as a model for epigenetic "reprogramming" at the whole organism level. Our findings support a mechanistic chain of events explaining why and how brief embryonic exposure to ether leads to haltere-to-wing transformations manifested at the larval stage and on. We show that ether interferes with protein integrity in the egg, leading to altered deployment of Hsp90 and widespread repression of Trithorax-mediated establishment of active H3K4me3 chromatin marks throughout the genome. Despite this global reduction, Ubx targets and wing development genes preferentially retain higher levels of H3K4me3 that predispose these genes for later up-regulation in the larval haltere disc, hence the wing-like outcome. Consistent with compromised protein integrity during the exposure, the penetrance of bithorax transformations increases by genetic or chemical reduction of Hsp90 function. Moreover, joint reduction in Hsp90 and trx gene dosage can cause bithorax transformations without exposure to ether, supporting an underlying epistasis between Hsp90 and trx loss-of-functions. These findings implicate environmental disruption of protein integrity at the onset of histone methylation with altered epigenetic regulation of developmental patterning genes. The emerging picture provides a unique example wherein the alleviation of the Hsp90 "capacitor function" by the environment drives a morphogenetic shift towards an ancestral-like body plan. The morphogenetic impact of chaperone response during a major setup of epigenetic patterns may be a general scheme for organ transformation by environmental cues.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Epigenesis, Genetic , HSP90 Heat-Shock Proteins , Histones , Wings, Animal , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Histones/metabolism , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/genetics , Wings, Animal/metabolism , Wings, Animal/growth & development , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Larva/metabolism , Larva/genetics , Larva/growth & development , Gene Expression Regulation, Developmental , Gene-Environment Interaction , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromatin/metabolism , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Epigenetic Memory , Transcription Factors
10.
Cell ; 148(3): 458-72, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22265598

ABSTRACT

Chromosomes are the physical realization of genetic information and thus form the basis for its readout and propagation. Here we present a high-resolution chromosomal contact map derived from a modified genome-wide chromosome conformation capture approach applied to Drosophila embryonic nuclei. The data show that the entire genome is linearly partitioned into well-demarcated physical domains that overlap extensively with active and repressive epigenetic marks. Chromosomal contacts are hierarchically organized between domains. Global modeling of contact density and clustering of domains show that inactive domains are condensed and confined to their chromosomal territories, whereas active domains reach out of the territory to form remote intra- and interchromosomal contacts. Moreover, we systematically identify specific long-range intrachromosomal contacts between Polycomb-repressed domains. Together, these observations allow for quantitative prediction of the Drosophila chromosomal contact map, laying the foundation for detailed studies of chromosome structure and function in a genetically tractable system.


Subject(s)
Drosophila melanogaster/genetics , Genome, Insect , Animals , Cell Nucleus/genetics , Chromosomes, Insect , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Models, Statistical , Polycomb Repressive Complex 1
11.
Mol Cell ; 74(1): 212-222.e5, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30795893

ABSTRACT

Eukaryotic chromosomes are organized in multiple scales, from nucleosomes to chromosome territories. Recently, genome-wide methods identified an intermediate level of chromosome organization, topologically associating domains (TADs), that play key roles in transcriptional regulation. However, these methods cannot directly examine the interplay between transcriptional activation and chromosome architecture while maintaining spatial information. Here we present a multiplexed, sequential imaging approach (Hi-M) that permits simultaneous detection of chromosome organization and transcription in single nuclei. This allowed us to unveil the changes in 3D chromatin organization occurring upon transcriptional activation and homologous chromosome unpairing during awakening of the zygotic genome in intact Drosophila embryos. Excitingly, the ability of Hi-M to explore the multi-scale chromosome architecture with spatial resolution at different stages of development or during the cell cycle will be key to understanding the mechanisms and consequences of the 4D organization of the genome.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/genetics , Chromosomes, Insect/genetics , Drosophila melanogaster/genetics , Genome , High-Throughput Nucleotide Sequencing/methods , Microscopy, Fluorescence/methods , RNA/genetics , Single-Cell Analysis/methods , Transcription, Genetic , Transcriptional Activation , Animals , Cell Cycle/genetics , Chromatin/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , In Situ Hybridization, Fluorescence , RNA/biosynthesis
12.
Blood ; 144(5): 496-509, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38643512

ABSTRACT

ABSTRACT: Plasma cells (PCs) are highly specialized cells representing the end stage of B-cell differentiation. We have shown that PC differentiation can be reproduced in vitro using elaborate culture systems. The molecular changes occurring during PC differentiation are recapitulated in this in vitro differentiation model. However, a major challenge exists to decipher the spatiotemporal epigenetic and transcriptional programs that drive the early stages of PC differentiation. We combined single cell (sc) RNA sequencing (RNA-seq) and assay for transposase-accessible chromatin with high throughput sequencing (scATAC-seq) to decipher the trajectories involved in PC differentiation. ScRNA-seq experiments revealed a strong heterogeneity of the preplasmablastic and plasmablastic stages. Among genes that were commonly identified using scATAC-seq and scRNA-seq, we identified several transcription factors with significant stage specific potential importance in PC differentiation. Interestingly, differentially accessible peaks characterizing the preplasmablastic stage were enriched in motifs of BATF3, FOS and BATF, belonging to activating protein 1 (AP-1) transcription factor family that may represent key transcriptional nodes involved in PC differentiation. Integration of transcriptomic and epigenetic data at the single cell level revealed that a population of preplasmablasts had already undergone epigenetic remodeling related to PC profile together with unfolded protein response activation and are committed to differentiate in PC. These results and the supporting data generated with our in vitro PC differentiation model provide a unique resource for the identification of molecular circuits that are crucial for early and mature PC maturation and biological functions. These data thus provide critical insights into epigenetic- and transcription-mediated reprogramming events that sustain PC differentiation.


Subject(s)
Cell Differentiation , Chromatin , Gene Expression Profiling , Plasma Cells , Single-Cell Analysis , Humans , Cell Differentiation/genetics , Plasma Cells/metabolism , Plasma Cells/cytology , Single-Cell Analysis/methods , Chromatin/metabolism , Chromatin/genetics , Transcriptome , Epigenesis, Genetic , Cells, Cultured
13.
Cell ; 145(6): 815-7, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21663785

ABSTRACT

Embryonic stem cell (ESC) pluripotency is maintained by core transcriptional circuits whereby critical factors sustain their own expression while preventing the expression of genes required for differentiation. Thomson et al. (2011) now show that two core components of the pluripotency circuit, Oct4 and Sox2, are also critical for germ layer fate choice.

14.
Cell ; 144(2): 214-26, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21241892

ABSTRACT

In Drosophila melanogaster, Hox genes are organized in an anterior and a posterior cluster, called Antennapedia complex and bithorax complex, located on the same chromosome arm and separated by 10 Mb of DNA. Both clusters are repressed by Polycomb group (PcG) proteins. Here, we show that genes of the two Hox complexes can interact within nuclear PcG bodies in tissues where they are corepressed. This colocalization increases during development and depends on PcG proteins. Hox gene contacts are conserved in the distantly related Drosophila virilis species and they are part of a large gene interaction network that includes other PcG target genes. Importantly, mutations on one of the loci weaken silencing of genes in the other locus, resulting in the exacerbation of homeotic phenotypes in sensitized genetic backgrounds. Thus, the three-dimensional organization of Polycomb target genes in the cell nucleus stabilizes the maintenance of epigenetic gene silencing.


Subject(s)
Drosophila/genetics , Drosophila/metabolism , Genes, Homeobox , Repressor Proteins/metabolism , Animals , Antennapedia Homeodomain Protein/genetics , Cell Nucleus/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Silencing , Polycomb-Group Proteins , Regulatory Elements, Transcriptional
15.
Nature ; 587(7834): 377-386, 2020 11.
Article in English | MEDLINE | ID: mdl-32894860

ABSTRACT

Here we describe the LifeTime Initiative, which aims to track, understand and target human cells during the onset and progression of complex diseases, and to analyse their response to therapy at single-cell resolution. This mission will be implemented through the development, integration and application of single-cell multi-omics and imaging, artificial intelligence and patient-derived experimental disease models during the progression from health to disease. The analysis of large molecular and clinical datasets will identify molecular mechanisms, create predictive computational models of disease progression, and reveal new drug targets and therapies. The timely detection and interception of disease embedded in an ethical and patient-centred vision will be achieved through interactions across academia, hospitals, patient associations, health data management systems and industry. The application of this strategy to key medical challenges in cancer, neurological and neuropsychiatric disorders, and infectious, chronic inflammatory and cardiovascular diseases at the single-cell level will usher in cell-based interceptive medicine in Europe over the next decade.


Subject(s)
Cell- and Tissue-Based Therapy , Delivery of Health Care/methods , Delivery of Health Care/trends , Medicine/methods , Medicine/trends , Pathology , Single-Cell Analysis , Artificial Intelligence , Delivery of Health Care/ethics , Delivery of Health Care/standards , Early Diagnosis , Education, Medical , Europe , Female , Health , Humans , Legislation, Medical , Male , Medicine/standards
16.
Mol Cell ; 71(1): 73-88.e5, 2018 07 05.
Article in English | MEDLINE | ID: mdl-30008320

ABSTRACT

Interphase chromatin is organized into topologically associating domains (TADs). Within TADs, chromatin looping interactions are formed between DNA regulatory elements, but their functional importance for the establishment of the 3D genome organization and gene regulation during development is unclear. Using high-resolution Hi-C experiments, we analyze higher order 3D chromatin organization during Drosophila embryogenesis and identify active and repressive chromatin loops that are established with different kinetics and depend on distinct factors: Zelda-dependent active loops are formed before the midblastula transition between transcribed genes over long distances. Repressive loops within polycomb domains are formed after the midblastula transition between polycomb response elements by the action of GAGA factor and polycomb proteins. Perturbation of PRE function by CRISPR/Cas9 genome engineering affects polycomb domain formation and destabilizes polycomb-mediated silencing. Preventing loop formation without removal of polycomb components also decreases silencing efficiency, suggesting that chromatin architecture can play instructive roles in gene regulation during development. VIDEO ABSTRACT.


Subject(s)
Chromatin/metabolism , Drosophila Proteins/metabolism , Gene Silencing , Polycomb-Group Proteins/metabolism , Animals , CRISPR-Cas Systems , Chromatin/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Polycomb-Group Proteins/genetics
17.
Mol Biol Evol ; 41(2)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38364113

ABSTRACT

Evolutionary analyses have estimated that ∼60% of nucleotides in intergenic regions of the Drosophila melanogaster genome are functionally relevant, suggesting that regulatory information may be encoded more densely in intergenic regions than has been revealed by most functional dissections of regulatory DNA. Here, we approached this issue through a functional dissection of the regulatory region of the gene shavenbaby (svb). Most of the ∼90 kb of this large regulatory region is highly conserved in the genus Drosophila, though characterized enhancers occupy a small fraction of this region. By analyzing the regulation of svb in different contexts of Drosophila development, we found that the regulatory information that drives svb expression in the abdominal pupal epidermis is organized in a different way than the elements that drive svb expression in the embryonic epidermis. While in the embryonic epidermis svb is activated by compact enhancers separated by large inactive DNA regions, svb expression in the pupal epidermis is driven by regulatory information distributed over broader regions of svb cis-regulatory DNA. In the same vein, we observed that other developmental genes also display a dense distribution of putative regulatory elements in their regulatory regions. Furthermore, we found that a large percentage of conserved noncoding DNA of the Drosophila genome is contained within regions of open chromatin. These results suggest that part of the evolutionary constraint on noncoding DNA of Drosophila is explained by the density of regulatory information, which may be greater than previously appreciated.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila/metabolism , Transcription Factors/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental , DNA , DNA, Intergenic/genetics , DNA, Intergenic/metabolism , Enhancer Elements, Genetic
18.
Nature ; 571(7766): 489-499, 2019 07.
Article in English | MEDLINE | ID: mdl-31341302

ABSTRACT

Epigenetic research has accelerated rapidly in the twenty-first century, generating justified excitement and hope, but also a degree of hype. Here we review how the field has evolved over the last few decades and reflect on some of the recent advances that are changing our understanding of biology. We discuss the interplay between epigenetics and DNA sequence variation as well as the implications of epigenetics for cellular memory and plasticity. We consider the effects of the environment and both intergenerational and transgenerational epigenetic inheritance on biology, disease and evolution. Finally, we present some new frontiers in epigenetics with implications for human health.


Subject(s)
Disease/genetics , Epigenesis, Genetic/genetics , Epigenomics/trends , Gene-Environment Interaction , Aging/genetics , Animals , Chromatin/genetics , Chromatin/metabolism , DNA Methylation/genetics , Genetic Variation/genetics , Humans , Neoplasms/genetics
19.
PLoS Genet ; 18(3): e1010083, 2022 03.
Article in English | MEDLINE | ID: mdl-35294439

ABSTRACT

Gene duplications and transcriptional enhancer emergence/modifications are thought having greatly contributed to phenotypic innovations during animal evolution. Nevertheless, little is known about how enhancers evolve after gene duplication and how regulatory information is rewired between duplicated genes. The Drosophila melanogaster bric-a-brac (bab) complex, comprising the tandem paralogous genes bab1 and bab2, provides a paradigm to address these issues. We previously characterized an intergenic enhancer (named LAE) regulating bab2 expression in the developing legs. We show here that bab2 regulators binding directly the LAE also govern bab1 expression in tarsal cells. LAE excision by CRISPR/Cas9-mediated genome editing reveals that this enhancer appears involved but not strictly required for bab1 and bab2 co-expression in leg tissues. Instead, the LAE enhancer is critical for paralog-specific bab2 expression along the proximo-distal leg axis. Chromatin features and phenotypic rescue experiments indicate that LAE functions partly redundantly with leg-specific regulatory information overlapping the bab1 transcription unit. Phylogenomics analyses indicate that (i) the bab complex originates from duplication of an ancestral singleton gene early on within the Cyclorrhapha dipteran sublineage, and (ii) LAE sequences have been evolutionarily-fixed early on within the Brachycera suborder thus predating the gene duplication event. This work provides new insights on enhancers, particularly about their emergence, maintenance and functional diversification during evolution.


Subject(s)
Drosophila Proteins , Drosophila , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Enhancer Elements, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Histochem Cell Biol ; 162(1-2): 133-147, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38888809

ABSTRACT

Cancer initiation and progression are typically associated with the accumulation of driver mutations and genomic instability. However, recent studies demonstrated that cancer can also be driven purely by epigenetic alterations, without driver mutations. Specifically, a 24-h transient downregulation of polyhomeotic (ph-KD), a core component of the Polycomb complex PRC1, is sufficient to induce epigenetically initiated cancers (EICs) in Drosophila, which are proficient in DNA repair and characterized by a stable genome. Whether genomic instability eventually occurs when PRC1 downregulation is performed for extended periods of time remains unclear. Here, we show that prolonged depletion of PH, which mimics cancer initiating events, results in broad dysregulation of DNA replication and repair genes, along with the accumulation of DNA breaks, defective repair, and widespread genomic instability in the cancer tissue. A broad misregulation of H2AK118 ubiquitylation and to a lesser extent of H3K27 trimethylation also occurs and might contribute to these phenotypes. Together, this study supports a model where DNA repair and replication defects accumulate during the tumorigenic transformation epigenetically induced by PRC1 loss, resulting in genomic instability and cancer progression.


Subject(s)
DNA Repair , Epigenesis, Genetic , Genomic Instability , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 1/genetics , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , Drosophila/metabolism , Drosophila melanogaster/metabolism , Polycomb-Group Proteins/metabolism , Polycomb-Group Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL