Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Clin Epigenetics ; 15(1): 102, 2023 06 12.
Article in English | MEDLINE | ID: mdl-37309009

ABSTRACT

BACKGROUND: Epigenetic alterations are a near-universal feature of human malignancy and have been detected in malignant cells as well as in easily accessible specimens such as blood and urine. These findings offer promising applications in cancer detection, subtyping, and treatment monitoring. However, much of the current evidence is based on findings in retrospective studies and may reflect epigenetic patterns that have already been influenced by the onset of the disease. METHODS: Studying breast cancer, we established genome-scale DNA methylation profiles of prospectively collected buffy coat samples (n = 702) from a case-control study nested within the EPIC-Heidelberg cohort using reduced representation bisulphite sequencing (RRBS). RESULTS: We observed cancer-specific DNA methylation events in buffy coat samples. Increased DNA methylation in genomic regions associated with SURF6 and REXO1/CTB31O20.3 was linked to the length of time to diagnosis in the prospectively collected buffy coat DNA from individuals who subsequently developed breast cancer. Using machine learning methods, we piloted a DNA methylation-based classifier that predicted case-control status in a held-out validation set with 76.5% accuracy, in some cases up to 15 years before clinical diagnosis of the disease. CONCLUSIONS: Taken together, our findings suggest a model of gradual accumulation of cancer-associated DNA methylation patterns in peripheral blood, which may be detected long before clinical manifestation of cancer. Such changes may provide useful markers for risk stratification and, ultimately, personalized cancer prevention.


Subject(s)
Breast Neoplasms , Humans , Female , Case-Control Studies , Prospective Studies , Retrospective Studies , DNA Methylation , Nuclear Proteins
2.
Nat Commun ; 14(1): 232, 2023 01 16.
Article in English | MEDLINE | ID: mdl-36646694

ABSTRACT

Methylation of cytosines is a prototypic epigenetic modification of the DNA. It has been implicated in various regulatory mechanisms across the animal kingdom and particularly in vertebrates. We mapped DNA methylation in 580 animal species (535 vertebrates, 45 invertebrates), resulting in 2443 genome-scale DNA methylation profiles of multiple organs. Bioinformatic analysis of this large dataset quantified the association of DNA methylation with the underlying genomic DNA sequence throughout vertebrate evolution. We observed a broadly conserved link with two major transitions-once in the first vertebrates and again with the emergence of reptiles. Cross-species comparisons focusing on individual organs supported a deeply conserved association of DNA methylation with tissue type, and cross-mapping analysis of DNA methylation at gene promoters revealed evolutionary changes for orthologous genes. In summary, this study establishes a large resource of vertebrate and invertebrate DNA methylomes, it showcases the power of reference-free epigenome analysis in species for which no reference genomes are available, and it contributes an epigenetic perspective to the study of vertebrate evolution.


Subject(s)
DNA Methylation , Genome , Animals , DNA Methylation/genetics , Genome/genetics , Invertebrates/genetics , Vertebrates/genetics , Vertebrates/metabolism , Epigenesis, Genetic , DNA/metabolism
3.
Article in English | MEDLINE | ID: mdl-37214176

ABSTRACT

CRISPR screens are a powerful source of biological discovery, enabling the unbiased interrogation of gene function in a wide range of applications and species. In pooled CRISPR screens, various genetically encoded perturbations are introduced into pools of cells. The targeted cells proliferate under a biological challenge such as cell competition, drug treatment or viral infection. Subsequently, the perturbation-induced effects are evaluated by sequencing-based counting of the guide RNAs that specify each perturbation. The typical results of such screens are ranked lists of genes that confer sensitivity or resistance to the biological challenge of interest. Contributing to the broad utility of CRISPR screens, adaptations of the core CRISPR technology make it possible to activate, silence or otherwise manipulate the target genes. Moreover, high-content read-outs such as single-cell RNA sequencing and spatial imaging help characterize screened cells with unprecedented detail. Dedicated software tools facilitate bioinformatic analysis and enhance reproducibility. CRISPR screening has unravelled various molecular mechanisms in basic biology, medical genetics, cancer research, immunology, infectious diseases, microbiology and other fields. This Primer describes the basic and advanced concepts of CRISPR screening and its application as a flexible and reliable method for biological discovery, biomedical research and drug development - with a special emphasis on high-content methods that make it possible to obtain detailed biological insights directly as part of the screen.

4.
Nat Methods ; 18(6): 635-642, 2021 06.
Article in English | MEDLINE | ID: mdl-34059827

ABSTRACT

Cell atlas projects and high-throughput perturbation screens require single-cell sequencing at a scale that is challenging with current technology. To enable cost-effective single-cell sequencing for millions of individual cells, we developed 'single-cell combinatorial fluidic indexing' (scifi). The scifi-RNA-seq assay combines one-step combinatorial preindexing of entire transcriptomes inside permeabilized cells with subsequent single-cell RNA-seq using microfluidics. Preindexing allows us to load several cells per droplet and computationally demultiplex their individual expression profiles. Thereby, scifi-RNA-seq massively increases the throughput of droplet-based single-cell RNA-seq, and provides a straightforward way of multiplexing thousands of samples in a single experiment. Compared with multiround combinatorial indexing, scifi-RNA-seq provides an easy and efficient workflow. Compared to cell hashing methods, which flag and discard droplets containing more than one cell, scifi-RNA-seq resolves and retains individual transcriptomes from overloaded droplets. We benchmarked scifi-RNA-seq on various human and mouse cell lines, validated it for primary human T cells and applied it in a highly multiplexed CRISPR screen with single-cell transcriptome readout of T cell receptor activation.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Animals , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , Cost-Benefit Analysis , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/economics , Humans , Mice , Microfluidics/methods , Receptors, Antigen, T-Cell/genetics , Single-Cell Analysis/economics , Single-Cell Analysis/methods , Transcriptome
5.
Hum Mol Genet ; 29(7): 1154-1167, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32160291

ABSTRACT

Human longevity is a complex trait influenced by both genetic and environmental factors, whose interaction is mediated by epigenetic mechanisms like DNA methylation. Here, we generated genome-wide whole-blood methylome data from 267 individuals, of which 71 were long-lived (90-104 years), by applying reduced representation bisulfite sequencing. We followed a stringent two-stage analysis procedure using discovery and replication samples to detect differentially methylated sites (DMSs) between young and long-lived study participants. Additionally, we performed a DNA methylation quantitative trait loci analysis to identify DMSs that underlie the longevity phenotype. We combined the DMSs results with gene expression data as an indicator of functional relevance. This approach yielded 21 new candidate genes, the majority of which are involved in neurophysiological processes or cancer. Notably, two candidates (PVRL2, ERCC1) are located on chromosome 19q, in close proximity to the well-known longevity- and Alzheimer's disease-associated loci APOE and TOMM40. We propose this region as a longevity hub, operating on both a genetic (APOE, TOMM40) and an epigenetic (PVRL2, ERCC1) level. We hypothesize that the heritable methylation and associated gene expression changes reported here are overall advantageous for the LLI and may prevent/postpone age-related diseases and facilitate survival into very old age.


Subject(s)
Apolipoproteins E/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , Longevity/genetics , Membrane Transport Proteins/genetics , Nectins/genetics , Aged, 80 and over , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Epigenome/genetics , Female , Gene Expression Regulation/genetics , Genome, Human/genetics , Humans , Male , Mitochondrial Precursor Protein Import Complex Proteins
6.
Sci Rep ; 9(1): 15751, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31673055

ABSTRACT

The mutagenic repair of Cas9 generated breaks is thought to predominantly rely on non-homologous end-joining (NHEJ), leading to insertions and deletions within DNA that culminate in gene knock-out (KO). In this study, by taking focused as well as genome-wide approaches, we show that this pathway is dispensable for the repair of such lesions. Genetic ablation of NHEJ is fully compensated for by alternative end joining (alt-EJ), in a POLQ-dependent manner, resulting in a distinct repair signature with larger deletions that may be exploited for large-scale genome editing. Moreover, we show that cells deficient for both NHEJ and alt-EJ were still able to repair CRISPR-mediated DNA double-strand breaks, highlighting how little is yet known about the mechanisms of CRISPR-based genome editing.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/methods , CRISPR-Associated Protein 9/metabolism , Cell Line , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Gene Knockout Techniques , HSP90 Heat-Shock Proteins/genetics , Humans , RNA, Guide, Kinetoplastida/metabolism , Ubiquitin-Protein Ligases/genetics
7.
Nat Med ; 24(10): 1611-1624, 2018 10.
Article in English | MEDLINE | ID: mdl-30150718

ABSTRACT

Glioblastoma is characterized by widespread genetic and transcriptional heterogeneity, yet little is known about the role of the epigenome in glioblastoma disease progression. Here, we present genome-scale maps of DNA methylation in matched primary and recurring glioblastoma tumors, using data from a highly annotated clinical cohort that was selected through a national patient registry. We demonstrate the feasibility of DNA methylation mapping in a large set of routinely collected FFPE samples, and we validate bisulfite sequencing as a multipurpose assay that allowed us to infer a range of different genetic, epigenetic, and transcriptional characteristics of the profiled tumor samples. On the basis of these data, we identified subtle differences between primary and recurring tumors, links between DNA methylation and the tumor microenvironment, and an association of epigenetic tumor heterogeneity with patient survival. In summary, this study establishes an open resource for dissecting DNA methylation heterogeneity in a genetically diverse and heterogeneous cancer, and it demonstrates the feasibility of integrating epigenomics, radiology, and digital pathology for a national cohort, thereby leveraging existing samples and data collected as part of routine clinical practice.


Subject(s)
DNA Methylation/genetics , Genome, Human/genetics , Glioblastoma/genetics , Neoplasm Recurrence, Local/genetics , Chromosome Mapping , Disease Progression , Epigenesis, Genetic , Female , Genetic Heterogeneity , Glioblastoma/pathology , High-Throughput Nucleotide Sequencing , Humans , Male , Neoplasm Recurrence, Local/pathology
8.
Nat Med ; 23(3): 386-395, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28134926

ABSTRACT

Developmental tumors in children and young adults carry few genetic alterations, yet they have diverse clinical presentation. Focusing on Ewing sarcoma, we sought to establish the prevalence and characteristics of epigenetic heterogeneity in genetically homogeneous cancers. We performed genome-scale DNA methylation sequencing for a large cohort of Ewing sarcoma tumors and analyzed epigenetic heterogeneity on three levels: between cancers, between tumors, and within tumors. We observed consistent DNA hypomethylation at enhancers regulated by the disease-defining EWS-FLI1 fusion protein, thus establishing epigenomic enhancer reprogramming as a ubiquitous and characteristic feature of Ewing sarcoma. DNA methylation differences between tumors identified a continuous disease spectrum underlying Ewing sarcoma, which reflected the strength of an EWS-FLI1 regulatory signature and a continuum between mesenchymal and stem cell signatures. There was substantial epigenetic heterogeneity within tumors, particularly in patients with metastatic disease. In summary, our study provides a comprehensive assessment of epigenetic heterogeneity in Ewing sarcoma and thereby highlights the importance of considering nongenetic aspects of tumor heterogeneity in the context of cancer biology and personalized medicine.


Subject(s)
Bone Neoplasms/genetics , DNA Methylation/genetics , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/genetics , Adolescent , Adult , Cell Line, Tumor , Child , Child, Preschool , Epigenesis, Genetic , Female , Genetic Heterogeneity , Humans , Male , Middle Aged , Promoter Regions, Genetic/genetics , Young Adult
9.
Nat Methods ; 14(3): 297-301, 2017 03.
Article in English | MEDLINE | ID: mdl-28099430

ABSTRACT

CRISPR-based genetic screens are accelerating biological discovery, but current methods have inherent limitations. Widely used pooled screens are restricted to simple readouts including cell proliferation and sortable marker proteins. Arrayed screens allow for comprehensive molecular readouts such as transcriptome profiling, but at much lower throughput. Here we combine pooled CRISPR screening with single-cell RNA sequencing into a broadly applicable workflow, directly linking guide RNA expression to transcriptome responses in thousands of individual cells. Our method for CRISPR droplet sequencing (CROP-seq) enables pooled CRISPR screens with single-cell transcriptome resolution, which will facilitate high-throughput functional dissection of complex regulatory mechanisms and heterogeneous cell populations.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, RNA/methods , Transcriptome/genetics , Cell Line , Cell Proliferation , HEK293 Cells , Humans , RNA, Guide, Kinetoplastida/genetics , Single-Cell Analysis/methods
10.
Cell Rep ; 13(11): 2621-2633, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26673328

ABSTRACT

Genome-wide DNA methylation mapping uncovers epigenetic changes associated with animal development, environmental adaptation, and species evolution. To address the lack of high-throughput methods for DNA methylation analysis in non-model organisms, we developed an integrated approach for studying DNA methylation differences independent of a reference genome. Experimentally, our method relies on an optimized 96-well protocol for reduced representation bisulfite sequencing (RRBS), which we have validated in nine species (human, mouse, rat, cow, dog, chicken, carp, sea bass, and zebrafish). Bioinformatically, we developed the RefFreeDMA software to deduce ad hoc genomes directly from RRBS reads and to pinpoint differentially methylated regions between samples or groups of individuals (http://RefFreeDMA.computational-epigenetics.org). The identified regions are interpreted using motif enrichment analysis and/or cross-mapping to annotated genomes. We validated our method by reference-free analysis of cell-type-specific DNA methylation in the blood of human, cow, and carp. In summary, we present a cost-effective method for epigenome analysis in ecology and evolution, which enables epigenome-wide association studies in natural populations and species without a reference genome.


Subject(s)
DNA Methylation , DNA/metabolism , Genome , Genomics/methods , Animals , Blood Cells/metabolism , Carps , Cattle , Chromosome Mapping , CpG Islands , DNA/chemistry , High-Throughput Nucleotide Sequencing , Humans , Leukocytes/metabolism , Sequence Analysis, DNA , Software
11.
Dev Biol ; 402(2): 291-305, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25912690

ABSTRACT

A large subset of mammalian imprinted genes show extra-embryonic lineage (EXEL) specific imprinted expression that is restricted to placental trophectoderm lineages and to visceral yolk sac endoderm (ysE). Isolated ysE provides a homogenous in vivo model of a mid-gestation extra-embryonic tissue to examine the mechanism of EXEL-specific imprinted gene silencing, but an in vitro model of ysE to facilitate more rapid and cost-effective experiments is not available. Reports indicate that ES cells differentiated into cystic embryoid bodies (EBs) contain ysE, so here we investigate if cystic EBs model ysE imprinted expression. The imprinted expression pattern of cystic EBs is shown to resemble fetal liver and not ysE. To investigate the reason for this we characterized the methylome and transcriptome of cystic EBs in comparison to fetal liver and ysE, by whole genome bisulphite sequencing and RNA-seq. Cystic EBs show a fetal liver pattern of global hypermethylation and low expression of repeats, while ysE shows global hypomethylation and high expression of IAPEz retroviral repeats, as reported for placenta. Transcriptome analysis confirmed that cystic EBs are more similar to fetal liver than ysE and express markers of early embryonic endoderm. Genome-wide analysis shows that ysE shares epigenetic and repeat expression features with placenta. Contrary to previous reports, we show that cystic EBs do not contain ysE, but are more similar to the embryonic endoderm of fetal liver. This explains why cystic EBs reproduce the imprinted expression seen in the embryo but not that seen in the ysE.


Subject(s)
DNA Methylation , Embryoid Bodies/metabolism , Gene Expression Regulation, Developmental/genetics , Genomic Imprinting/genetics , Models, Biological , Animals , Base Sequence , Cell Differentiation/physiology , Chromatin Immunoprecipitation , Computational Biology , Endoderm/metabolism , Gene Expression Profiling , Histological Techniques , In Situ Hybridization , Mice , Molecular Sequence Data , Sequence Analysis, RNA , Yolk Sac/cytology , Yolk Sac/metabolism
12.
Cell Rep ; 10(8): 1386-97, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25732828

ABSTRACT

Methods for single-cell genome and transcriptome sequencing have contributed to our understanding of cellular heterogeneity, whereas methods for single-cell epigenomics are much less established. Here, we describe a whole-genome bisulfite sequencing (WGBS) assay that enables DNA methylation mapping in very small cell populations (µWGBS) and single cells (scWGBS). Our assay is optimized for profiling many samples at low coverage, and we describe a bioinformatic method that analyzes collections of single-cell methylomes to infer cell-state dynamics. Using these technological advances, we studied epigenomic cell-state dynamics in three in vitro models of cellular differentiation and pluripotency, where we observed characteristic patterns of epigenome remodeling and cell-to-cell heterogeneity. The described method enables single-cell analysis of DNA methylation in a broad range of biological systems, including embryonic development, stem cell differentiation, and cancer. It can also be used to establish composite methylomes that account for cell-to-cell heterogeneity in complex tissue samples.


Subject(s)
Computational Biology/methods , DNA Methylation , Epigenomics , High-Throughput Nucleotide Sequencing , Sequence Analysis, DNA , Single-Cell Analysis/methods , Animals , Cell Differentiation , Cell Line , DNA/chemistry , DNA/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , HL-60 Cells , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , K562 Cells , Mice , Sulfites/chemistry
13.
Cell Rep ; 10(7): 1082-95, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25704812

ABSTRACT

Transcription factor fusion proteins can transform cells by inducing global changes of the transcriptome, often creating a state of oncogene addiction. Here, we investigate the role of epigenetic mechanisms in this process, focusing on Ewing sarcoma cells that are dependent on the EWS-FLI1 fusion protein. We established reference epigenome maps comprising DNA methylation, seven histone marks, open chromatin states, and RNA levels, and we analyzed the epigenome dynamics upon downregulation of the driving oncogene. Reduced EWS-FLI1 expression led to widespread epigenetic changes in promoters, enhancers, and super-enhancers, and we identified histone H3K27 acetylation as the most strongly affected mark. Clustering of epigenetic promoter signatures defined classes of EWS-FLI1-regulated genes that responded differently to low-dose treatment with histone deacetylase inhibitors. Furthermore, we observed strong and opposing enrichment patterns for E2F and AP-1 among EWS-FLI1-correlated and anticorrelated genes. Our data describe extensive genome-wide rewiring of epigenetic cell states driven by an oncogenic fusion protein.


Subject(s)
Cellular Reprogramming , Epigenesis, Genetic , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Acetylation , Cell Line, Tumor , Chromatin/metabolism , Chromosome Mapping , Cluster Analysis , DNA Methylation , Down-Regulation/drug effects , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Oncogene Proteins, Fusion/genetics , Promoter Regions, Genetic , Proto-Oncogene Protein c-fli-1/genetics , RNA, Small Interfering/metabolism , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcriptome/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...