Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
Add more filters










Publication year range
1.
Nat Genet ; 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816647

ABSTRACT

Catalytic activity of the imitation switch (ISWI) family of remodelers is critical for nucleosomal organization and DNA binding of certain transcription factors, including the insulator protein CTCF. Here we define the contribution of individual subcomplexes by deriving a panel of isogenic mouse stem cell lines, each lacking one of six ISWI accessory subunits. Individual deletions of subunits of either CERF, RSF, ACF, WICH or NoRC subcomplexes only moderately affect the chromatin landscape, while removal of the NURF-specific subunit BPTF leads to a strong reduction in chromatin accessibility and SNF2H ATPase localization around CTCF sites. This affects adjacent nucleosome occupancy and CTCF binding. At a group of sites with reduced chromatin accessibility, CTCF binding persists but cohesin occupancy is reduced, resulting in decreased insulation. These results suggest that CTCF binding can be separated from its function as an insulator in nuclear organization and identify a specific role for NURF in mediating SNF2H localization and chromatin opening at bound CTCF sites.

2.
FEBS Lett ; 598(7): 719-724, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38514456

ABSTRACT

The diverse range of organizations contributing to the global research ecosystem is believed to enhance the overall quality and resilience of its output. Mid-sized autonomous research institutes, distinct from universities, play a crucial role in this landscape. They often lead the way in new research fields and experimental methods, including those in social and organizational domains, which are vital for driving innovation. The EU-LIFE alliance was established with the goal of fostering excellence by developing and disseminating best practices among European biomedical research institutes. As directors of the 15 EU-LIFE institutes, we have spent a decade comparing and refining our processes. Now, we are eager to share the insights we've gained. To this end, we have crafted this Charter, outlining 10 principles we deem essential for research institutes to flourish and achieve ground-breaking discoveries. These principles, detailed in the Charter, encompass excellence, independence, training, internationality and inclusivity, mission focus, technological advancement, administrative innovation, cooperation, societal impact, and public engagement. Our aim is to inspire the establishment of new institutes that adhere to these principles and to raise awareness about their significance. We are convinced that they should be viewed a crucial component of any national and international innovation strategies.


Subject(s)
Biological Science Disciplines , Biomedical Research , Academies and Institutes
3.
Nature ; 619(7969): 385-393, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37407816

ABSTRACT

The basic helix-loop-helix (bHLH) family of transcription factors recognizes DNA motifs known as E-boxes (CANNTG) and includes 108 members1. Here we investigate how chromatinized E-boxes are engaged by two structurally diverse bHLH proteins: the proto-oncogene MYC-MAX and the circadian transcription factor CLOCK-BMAL1 (refs. 2,3). Both transcription factors bind to E-boxes preferentially near the nucleosomal entry-exit sites. Structural studies with engineered or native nucleosome sequences show that MYC-MAX or CLOCK-BMAL1 triggers the release of DNA from histones to gain access. Atop the H2A-H2B acidic patch4, the CLOCK-BMAL1 Per-Arnt-Sim (PAS) dimerization domains engage the histone octamer disc. Binding of tandem E-boxes5-7 at endogenous DNA sequences occurs through direct interactions between two CLOCK-BMAL1 protomers and histones and is important for circadian cycling. At internal E-boxes, the MYC-MAX leucine zipper can also interact with histones H2B and H3, and its binding is indirectly enhanced by OCT4 elsewhere on the nucleosome. The nucleosomal E-box position and the type of bHLH dimerization domain jointly determine the histone contact, the affinity and the degree of competition and cooperativity with other nucleosome-bound factors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , DNA , Histones , ARNTL Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA/genetics , DNA/metabolism , Helix-Loop-Helix Motifs/genetics , Histones/chemistry , Histones/metabolism , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/metabolism , Protein Binding , CLOCK Proteins/chemistry , CLOCK Proteins/metabolism , Proto-Oncogene Proteins c-myc/chemistry , Proto-Oncogene Proteins c-myc/metabolism , Allosteric Regulation , Leucine Zippers , Octamer Transcription Factor-3/metabolism , Protein Multimerization
4.
Nat Commun ; 14(1): 3907, 2023 07 03.
Article in English | MEDLINE | ID: mdl-37400441

ABSTRACT

YAP is a key transcriptional co-activator of TEADs, it regulates cell growth and is frequently activated in cancer. In Malignant Pleural Mesothelioma (MPM), YAP is activated by loss-of-function mutations in upstream components of the Hippo pathway, while, in Uveal Melanoma (UM), YAP is activated in a Hippo-independent manner. To date, it is unclear if and how the different oncogenic lesions activating YAP impact its oncogenic program, which is particularly relevant for designing selective anti-cancer therapies. Here we show that, despite YAP being essential in both MPM and UM, its interaction with TEAD is unexpectedly dispensable in UM, limiting the applicability of TEAD inhibitors in this cancer type. Systematic functional interrogation of YAP regulatory elements in both cancer types reveals convergent regulation of broad oncogenic drivers in both MPM and UM, but also strikingly selective programs. Our work reveals unanticipated lineage-specific features of the YAP regulatory network that provide important insights to guide the design of tailored therapeutic strategies to inhibit YAP signaling across different cancer types.


Subject(s)
Adaptor Proteins, Signal Transducing , Neoplasms , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , YAP-Signaling Proteins , Epigenomics , Transcription Factors/genetics , Transcription Factors/metabolism , Signal Transduction/genetics
5.
Nat Struct Mol Biol ; 30(7): 948-957, 2023 07.
Article in English | MEDLINE | ID: mdl-37386214

ABSTRACT

The genomic binding sites of the transcription factor (TF) and tumor suppressor p53 are unusually diverse with regard to their chromatin features, including histone modifications, raising the possibility that the local chromatin environment can contextualize p53 regulation. Here, we show that epigenetic characteristics of closed chromatin, such as DNA methylation, do not influence the binding of p53 across the genome. Instead, the ability of p53 to open chromatin and activate its target genes is locally restricted by its cofactor Trim24. Trim24 binds to both p53 and unmethylated histone 3 lysine 4 (H3K4), thereby preferentially localizing to those p53 sites that reside in closed chromatin, whereas it is deterred from accessible chromatin by H3K4 methylation. The presence of Trim24 increases cell viability upon stress and enables p53 to affect gene expression as a function of the local chromatin state. These findings link H3K4 methylation to p53 function and illustrate how specificity in chromatin can be achieved, not by TF-intrinsic sensitivity to histone modifications, but by employing chromatin-sensitive cofactors that locally modulate TF function.


Subject(s)
Chromatin , Histones , Histones/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , DNA Methylation , Protein Processing, Post-Translational , Transcription Factors/metabolism
6.
Nat Cell Biol ; 25(1): 42-55, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36604593

ABSTRACT

ZNF462 haploinsufficiency is linked to Weiss-Kruszka syndrome, a genetic disorder characterized by neurodevelopmental defects, including autism. Though conserved in vertebrates and essential for embryonic development, the molecular functions of ZNF462 remain unclear. We identified its murine homologue ZFP462 in a screen for mediators of epigenetic gene silencing. Here we show that ZFP462 safeguards neural lineage specification of mouse embryonic stem cells (ESCs) by targeting the H3K9-specific histone methyltransferase complex G9A/GLP to silence meso-endodermal genes. ZFP462 binds to transposable elements that are potential enhancers harbouring pluripotency and meso-endoderm transcription factor binding sites. Recruiting G9A/GLP, ZFP462 seeds heterochromatin, restricting transcription factor binding. Loss of ZFP462 in ESCs results in increased chromatin accessibility at target sites and ectopic expression of meso-endodermal genes. Taken together, ZFP462 confers lineage and locus specificity to the broadly expressed epigenetic regulator G9A/GLP. Our results suggest that aberrant activation of lineage non-specific genes in the neuronal lineage underlies ZNF462-associated neurodevelopmental pathology.


Subject(s)
Heterochromatin , Histone-Lysine N-Methyltransferase , Animals , Mice , Heterochromatin/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Chromatin , Embryonic Stem Cells , Transcription Factors/genetics , Transcription Factors/metabolism , DNA-Binding Proteins/genetics , Nerve Tissue Proteins/genetics
7.
Nat Genet ; 54(12): 1895-1906, 2022 12.
Article in English | MEDLINE | ID: mdl-36471082

ABSTRACT

Cytosine methylation efficiently silences CpG-rich regulatory regions of genes and repeats in mammalian genomes. To what extent this entails direct inhibition of transcription factor (TF) binding versus indirect inhibition via recruitment of methyl-CpG-binding domain (MBD) proteins is unclear. Here we show that combinatorial genetic deletions of all four proteins with functional MBDs in mouse embryonic stem cells, derived neurons or a human cell line do not reactivate genes or repeats with methylated promoters. These do, however, become activated by methylation-restricted TFs if DNA methylation is removed. We identify several causal TFs in neurons, including ONECUT1, which is methylation sensitive only at a motif variant. Rampantly upregulated retrotransposons in methylation-free neurons feature a CRE motif, which activates them in the absence of DNA methylation via methylation-sensitive binding of CREB1. Our study reveals methylation-sensitive TFs in vivo and argues that direct inhibition, rather than indirect repression by the tested MBD proteins, is the prevailing mechanism of methylation-mediated repression at regulatory regions and repeats.


Subject(s)
DNA Methylation , Transcription Factors , Animals , Humans , Mice , DNA Methylation/genetics , Hepatocyte Nuclear Factor 6 , Mammals , Transcription Factors/genetics
8.
Nat Genet ; 54(11): 1702-1710, 2022 11.
Article in English | MEDLINE | ID: mdl-36333500

ABSTRACT

Genomic imprinting is regulated by parental-specific DNA methylation of imprinting control regions (ICRs). Despite an identical DNA sequence, ICRs can exist in two distinct epigenetic states that are memorized throughout unlimited cell divisions and reset during germline formation. Here, we systematically study the genetic and epigenetic determinants of this epigenetic bistability. By iterative integration of ICRs and related DNA sequences to an ectopic location in the mouse genome, we first identify the DNA sequence features required for maintenance of epigenetic states in embryonic stem cells. The autonomous regulatory properties of ICRs further enabled us to create DNA-methylation-sensitive reporters and to screen for key components involved in regulating their epigenetic memory. Besides DNMT1, UHRF1 and ZFP57, we identify factors that prevent switching from methylated to unmethylated states and show that two of these candidates, ATF7IP and ZMYM2, are important for the stability of DNA and H3K9 methylation at ICRs in embryonic stem cells.


Subject(s)
DNA Methylation , Genomic Imprinting , Mice , Animals , Base Sequence , DNA Methylation/genetics , Epigenomics , Chromatin/genetics , Repressor Proteins/genetics
9.
Nat Rev Genet ; 23(12): 728-740, 2022 12.
Article in English | MEDLINE | ID: mdl-35831531

ABSTRACT

Cell type-specific gene expression relies on transcription factors (TFs) binding DNA sequence motifs embedded in chromatin. Understanding how motifs are accessed in chromatin is crucial to comprehend differential transcriptional responses and the phenotypic impact of sequence variation. Chromatin obstacles to TF binding range from DNA methylation to restriction of DNA access by nucleosomes depending on their position, composition and modification. In vivo and in vitro approaches now enable the study of TF binding in chromatin at unprecedented resolution. Emerging insights suggest that TFs vary in their ability to navigate chromatin states. However, it remains challenging to link binding and transcriptional outcomes to molecular characteristics of TFs or the local chromatin substrate. Here, we discuss our current understanding of how TFs access DNA in chromatin and novel techniques and directions towards a better understanding of this critical step in genome regulation.


Subject(s)
Chromatin , Transcription Factors , Chromatin/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Binding Sites/genetics , Nucleosomes/genetics , Protein Binding , DNA/genetics
10.
Bioinformatics ; 38(9): 2624-2625, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35199152

ABSTRACT

SUMMARY: Proteins binding to specific nucleotide sequences, such as transcription factors, play key roles in the regulation of gene expression. Their binding can be indirectly observed via associated changes in transcription, chromatin accessibility, DNA methylation and histone modifications. Identifying candidate factors that are responsible for these observed experimental changes is critical to understand the underlying biological processes. Here, we present monaLisa, an R/Bioconductor package that implements approaches to identify relevant transcription factors from experimental data. The package can be easily integrated with other Bioconductor packages and enables seamless motif analyses without any software dependencies outside of R. AVAILABILITY AND IMPLEMENTATION: monaLisa is implemented in R and available on Bioconductor at https://bioconductor.org/packages/monaLisa with the development version hosted on GitHub at https://github.com/fmicompbio/monaLisa. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Software , Transcription Factors
11.
Nature ; 596(7870): 133-137, 2021 08.
Article in English | MEDLINE | ID: mdl-34234345

ABSTRACT

The majority of gene transcripts generated by RNA polymerase II in mammalian genomes initiate at CpG island (CGI) promoters1,2, yet our understanding of their regulation remains limited. This is in part due to the incomplete information that we have on transcription factors, their DNA-binding motifs and which genomic binding sites are functional in any given cell type3-5. In addition, there are orphan motifs without known binders, such as the CGCG element, which is associated with highly expressed genes across human tissues and enriched near the transcription start site of a subset of CGI promoters6-8. Here we combine single-molecule footprinting with interaction proteomics to identify BTG3-associated nuclear protein (BANP) as the transcription factor that binds this element in the mouse and human genome. We show that BANP is a strong CGI activator that controls essential metabolic genes in pluripotent stem and terminally differentiated neuronal cells. BANP binding is repelled by DNA methylation of its motif in vitro and in vivo, which epigenetically restricts most binding to CGIs and accounts for differential binding at aberrantly methylated CGI promoters in cancer cells. Upon binding to an unmethylated motif, BANP opens chromatin and phases nucleosomes. These findings establish BANP as a critical activator of a set of essential genes and suggest a model in which the activity of CGI promoters relies on methylation-sensitive transcription factors that are capable of chromatin opening.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin Assembly and Disassembly , Chromatin/genetics , Chromatin/metabolism , CpG Islands/genetics , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Base Sequence , Cell Line, Tumor , Chromatin/chemistry , Chromatin Assembly and Disassembly/genetics , DNA Methylation , Gene Expression Regulation , Genes, Essential , Humans , Mice , Single Molecule Imaging
12.
Sci Adv ; 7(27)2021 07.
Article in English | MEDLINE | ID: mdl-34215580

ABSTRACT

Millions of putative transcriptional regulatory elements (TREs) have been cataloged in the human genome, yet their functional relevance in specific pathophysiological settings remains to be determined. This is critical to understand how oncogenic transcription factors (TFs) engage specific TREs to impose transcriptional programs underlying malignant phenotypes. Here, we combine cutting edge CRISPR screens and epigenomic profiling to functionally survey ≈15,000 TREs engaged by estrogen receptor (ER). We show that ER exerts its oncogenic role in breast cancer by engaging TREs enriched in GATA3, TFAP2C, and H3K27Ac signal. These TREs control critical downstream TFs, among which TFAP2C plays an essential role in ER-driven cell proliferation. Together, our work reveals novel insights into a critical oncogenic transcription program and provides a framework to map regulatory networks, enabling to dissect the function of the noncoding genome of cancer cells.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Gene Regulatory Networks , Carcinogenesis/genetics , Epigenomics , Genome, Human , Humans , Regulatory Elements, Transcriptional
13.
Nat Commun ; 12(1): 2442, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33903593

ABSTRACT

The transcription factor PAX8 is critical for the development of the thyroid and urogenital system. Comprehensive genomic screens furthermore indicate an additional oncogenic role for PAX8 in renal and ovarian cancers. While a plethora of PAX8-regulated genes in different contexts have been proposed, we still lack a mechanistic understanding of how PAX8 engages molecular complexes to drive disease-relevant oncogenic transcriptional programs. Here we show that protein isoforms originating from the MECOM locus form a complex with PAX8. These include MDS1-EVI1 (also called PRDM3) for which we map its interaction with PAX8 in vitro and in vivo. We show that PAX8 binds a large number of genomic sites and forms transcriptional hubs. At a subset of these, PAX8 together with PRDM3 regulates a specific gene expression module involved in adhesion and extracellular matrix. This gene module correlates with PAX8 and MECOM expression in large scale profiling of cell lines, patient-derived xenografts (PDXs) and clinical cases and stratifies gynecological cancer cases with worse prognosis. PRDM3 is amplified in ovarian cancers and we show that the MECOM locus and PAX8 sustain in vivo tumor growth, further supporting that the identified function of the MECOM locus underlies PAX8-driven oncogenic functions in ovarian cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , MDS1 and EVI1 Complex Locus Protein/genetics , Ovarian Neoplasms/genetics , PAX8 Transcription Factor/genetics , Animals , Cell Line, Tumor , Female , HEK293 Cells , Humans , MDS1 and EVI1 Complex Locus Protein/metabolism , Mice, Nude , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , PAX8 Transcription Factor/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tumor Burden/genetics , Xenograft Model Antitumor Assays/methods
14.
Nat Genet ; 53(3): 279-287, 2021 03.
Article in English | MEDLINE | ID: mdl-33558757

ABSTRACT

Chromatin accessibility is a hallmark of regulatory regions, entails transcription factor (TF) binding and requires nucleosomal reorganization. However, it remains unclear how dynamic this process is. In the present study, we use small-molecule inhibition of the catalytic subunit of the mouse SWI/SNF remodeler complex to show that accessibility and reduced nucleosome presence at TF-binding sites rely on persistent activity of nucleosome remodelers. Within minutes of remodeler inhibition, accessibility and TF binding decrease. Although this is irrespective of TF function, we show that the activating TF OCT4 (POU5F1) exhibits a faster response than the repressive TF REST. Accessibility, nucleosome depletion and gene expression are rapidly restored on inhibitor removal, suggesting that accessible chromatin is regenerated continuously and in a largely cell-autonomous fashion. We postulate that TF binding to chromatin and remodeler-mediated nucleosomal removal do not represent a stable situation, but instead accessible chromatin reflects an average of a dynamic process under continued renewal.


Subject(s)
Chromatin/metabolism , Multiprotein Complexes/metabolism , Transcription Factors/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Binding Sites , Cell Line/drug effects , Chromatin/genetics , Chromatin Assembly and Disassembly/drug effects , Chromatin Assembly and Disassembly/physiology , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/antagonists & inhibitors , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Histones/genetics , Histones/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Multiprotein Complexes/drug effects , Multiprotein Complexes/genetics , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Small Molecule Libraries/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
15.
Mol Cell ; 81(2): 255-267.e6, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33290745

ABSTRACT

Gene activation requires the cooperative activity of multiple transcription factors at cis-regulatory elements (CREs). Yet, most transcription factors have short residence time, questioning the requirement of their physical co-occupancy on DNA to achieve cooperativity. Here, we present a DNA footprinting method that detects individual molecular interactions of transcription factors and nucleosomes with DNA in vivo. We apply this strategy to quantify the simultaneous binding of multiple transcription factors on single DNA molecules at mouse CREs. Analysis of the binary occupancy patterns at thousands of motif combinations reveals that high DNA co-occupancy occurs for most types of transcription factors, in the absence of direct physical interaction, at sites of competition with nucleosomes. Perturbation of pairwise interactions demonstrates the function of molecular co-occupancy in binding cooperativity. Our results reveal the interactions regulating CREs at molecular resolution and identify DNA co-occupancy as a widespread cooperativity mechanism used by transcription factors to remodel chromatin.


Subject(s)
DNA Footprinting/methods , DNA/genetics , Nucleosomes/chemistry , Regulatory Elements, Transcriptional , Transcription Factors/genetics , Animals , Binding Sites , DNA/chemistry , DNA/metabolism , Male , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Nucleosomes/metabolism , Protein Binding , Transcription Factors/chemistry , Transcription Factors/metabolism , Transcription, Genetic
16.
Nat Commun ; 11(1): 2680, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32471981

ABSTRACT

DNA methylation is considered a stable epigenetic mark, yet methylation patterns can vary during differentiation and in diseases such as cancer. Local levels of DNA methylation result from opposing enzymatic activities, the rates of which remain largely unknown. Here we developed a theoretical and experimental framework enabling us to infer methylation and demethylation rates at 860,404 CpGs in mouse embryonic stem cells. We find that enzymatic rates can vary as much as two orders of magnitude between CpGs with identical steady-state DNA methylation. Unexpectedly, de novo and maintenance methylation activity is reduced at transcription factor binding sites, while methylation turnover is elevated in transcribed gene bodies. Furthermore, we show that TET activity contributes substantially more than passive demethylation to establishing low methylation levels at distal enhancers. Taken together, our work unveils a genome-scale map of methylation kinetics, revealing highly variable and context-specific activity for the DNA methylation machinery.


Subject(s)
CpG Islands/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Demethylation , DNA Methylation/genetics , DNA-Binding Proteins/metabolism , Mouse Embryonic Stem Cells/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Binding Sites/genetics , Cell Line , Chromosome Mapping , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Dioxygenases/metabolism , Epigenesis, Genetic/genetics , Genome/genetics , Histones/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/genetics , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics , DNA Methyltransferase 3B
17.
Science ; 368(6498): 1460-1465, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32327602

ABSTRACT

Transcription factors (TFs) regulate gene expression through chromatin where nucleosomes restrict DNA access. To study how TFs bind nucleosome-occupied motifs, we focused on the reprogramming factors OCT4 and SOX2 in mouse embryonic stem cells. We determined TF engagement throughout a nucleosome at base-pair resolution in vitro, enabling structure determination by cryo-electron microscopy at two preferred positions. Depending on motif location, OCT4 and SOX2 differentially distort nucleosomal DNA. At one position, OCT4-SOX2 removes DNA from histone H2A and histone H3; however, at an inverted motif, the TFs only induce local DNA distortions. OCT4 uses one of its two DNA-binding domains to engage DNA in both structures, reading out a partial motif. These findings explain site-specific nucleosome engagement by the pluripotency factors OCT4 and SOX2, and they reveal how TFs distort nucleosomes to access chromatinized motifs.


Subject(s)
Gene Expression Regulation , Nucleosomes/chemistry , Octamer Transcription Factor-3/chemistry , SOXB1 Transcription Factors/chemistry , Animals , Cryoelectron Microscopy , DNA/chemistry , Histones/chemistry , Mice , Mouse Embryonic Stem Cells/metabolism
18.
Nat Neurosci ; 22(8): 1345-1356, 2019 08.
Article in English | MEDLINE | ID: mdl-31285614

ABSTRACT

Targeting genes to specific neuronal or glial cell types is valuable for both understanding and repairing brain circuits. Adeno-associated viruses (AAVs) are frequently used for gene delivery, but targeting expression to specific cell types is an unsolved problem. We created a library of 230 AAVs, each with a different synthetic promoter designed using four independent strategies. We show that a number of these AAVs specifically target expression to neuronal and glial cell types in the mouse and non-human primate retina in vivo and in the human retina in vitro. We demonstrate applications for recording and stimulation, as well as the intersectional and combinatorial labeling of cell types. These resources and approaches allow economic, fast and efficient cell-type targeting in a variety of species, both for fundamental science and for gene therapy.


Subject(s)
Dependovirus/genetics , Gene Targeting/methods , Neuroglia/virology , Neurons/virology , Animals , Gene Transfer Techniques , Humans , Macaca fascicularis , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Retina/virology
19.
Nature ; 571(7764): E6, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31239520

ABSTRACT

In this Article, in Fig. 1a, the 5' and 3' labels were reversed in the DNA sequence, and Fig. 4 was missing panel labels a-e. These errors have been corrected online.

20.
Nature ; 571(7763): 79-84, 2019 07.
Article in English | MEDLINE | ID: mdl-31142837

ABSTRACT

Access to DNA packaged in nucleosomes is critical for gene regulation, DNA replication and DNA repair. In humans, the UV-damaged DNA-binding protein (UV-DDB) complex detects UV-light-induced pyrimidine dimers throughout the genome; however, it remains unknown how these lesions are recognized in chromatin, in which nucleosomes restrict access to DNA. Here we report cryo-electron microscopy structures of UV-DDB bound to nucleosomes bearing a 6-4 pyrimidine-pyrimidone dimer or a DNA-damage mimic in various positions. We find that UV-DDB binds UV-damaged nucleosomes at lesions located in the solvent-facing minor groove without affecting the overall nucleosome architecture. In the case of buried lesions that face the histone core, UV-DDB changes the predominant translational register of the nucleosome and selectively binds the lesion in an accessible, exposed position. Our findings explain how UV-DDB detects occluded lesions in strongly positioned nucleosomes, and identify slide-assisted site exposure as a mechanism by which high-affinity DNA-binding proteins can access otherwise occluded sites in nucleosomal DNA.


Subject(s)
DNA Damage , DNA-Binding Proteins/metabolism , DNA/metabolism , DNA/ultrastructure , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Pyrimidine Dimers/analysis , Cryoelectron Microscopy , DNA/chemistry , DNA/radiation effects , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/ultrastructure , Histones/chemistry , Histones/metabolism , Histones/ultrastructure , Humans , Models, Molecular , Nucleosomes/genetics , Nucleosomes/radiation effects , Pyrimidine Dimers/chemistry , Pyrimidine Dimers/genetics , Thermodynamics , Ultraviolet Rays/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...