Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Mol Cell ; 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38955181

ABSTRACT

The essential Mediator (MED) coactivator complex plays a well-understood role in regulation of basal transcription in all eukaryotes, but the mechanism underlying its role in activator-dependent transcription remains unknown. We investigated modulation of metazoan MED interaction with RNA polymerase II (RNA Pol II) by antagonistic effects of the MED26 subunit and the CDK8 kinase module (CKM). Biochemical analysis of CKM-MED showed that the CKM blocks binding of the RNA Pol II carboxy-terminal domain (CTD), preventing RNA Pol II interaction. This restriction is eliminated by nuclear receptor (NR) binding to CKM-MED, which enables CTD binding in a MED26-dependent manner. Cryoelectron microscopy (cryo-EM) and crosslinking-mass spectrometry (XL-MS) revealed that the structural basis for modulation of CTD interaction with MED relates to a large intrinsically disordered region (IDR) in CKM subunit MED13 that blocks MED26 and CTD interaction with MED but is repositioned upon NR binding. Hence, NRs can control transcription initiation by priming CKM-MED for MED26-dependent RNA Pol II interaction.

2.
bioRxiv ; 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38915666

ABSTRACT

Viral invasion of the host cell causes some of the most dramatic changes in biology. Human cytomegalovirus (HCMV) extensively remodels host cells, altering nuclear shape and generating a cytoplasmic viral-induced assembly compartment (vIAC). How these striking morphology changes take place in the context of host gene regulation is still emerging. Here, we discovered that histone variant macroH2A1 is essential for producing infectious progeny. Because virion maturation and cellular remodeling are closely linked processes, we investigated structural changes in the host cell upon HCMV infection. We discovered that macroH2A1 is necessary for HCMV-induced reorganization of the host nucleus, cytoskeleton, and endoplasmic reticulum. Furthermore, using RNA-seq we found that while all viral genes were highly expressed in the absence of macroH2A1, many HCMV-induced host genes were not. Remarkably, hundreds of these HCMV-induced macroH2A1-dependent host genes are associated with neuronal synapse formation and vesicle trafficking. Knock-down of these HCMV-induced neuronal genes during infection resulted in malformed vIACs and smaller plaques, establishing their importance to HCMV infection. Together, our findings demonstrate that HCMV manipulates host gene expression by hijacking a dormant neuronal secretory pathway for efficient virion maturation.

3.
Cell Rep ; 43(4): 114090, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38607915

ABSTRACT

Gene repression by the Polycomb pathway is essential for metazoan development. Polycomb domains, characterized by trimethylation of histone H3 lysine 27 (H3K27me3), carry the memory of repression and hence need to be maintained to counter the dilution of parental H3K27me3 with unmodified H3 during replication. Yet, how locus-specific H3K27me3 is maintained through replication is unclear. To understand H3K27me3 recovery post-replication, we first define nucleation sites within each Polycomb domain in mouse embryonic stem cells. To map dynamics of H3K27me3 domains across the cell cycle, we develop CUT&Flow (coupling cleavage under target and tagmentation with flow cytometry). We show that post-replication recovery of Polycomb domains occurs by nucleation and spreading, using the same nucleation sites used during de novo domain formation. By using Polycomb repressive complex 2 (PRC2) subunit-specific inhibitors, we find that PRC2 targets nucleation sites post-replication independent of pre-existing H3K27me3. Thus, competition between H3K27me3 deposition and nucleosome turnover drives both de novo domain formation and maintenance during every cell cycle.


Subject(s)
Cell Cycle , Histones , Polycomb Repressive Complex 2 , Animals , Mice , Histones/metabolism , Polycomb Repressive Complex 2/metabolism , Methylation , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/cytology , Polycomb-Group Proteins/metabolism , Polycomb-Group Proteins/genetics , Protein Domains , Nucleosomes/metabolism
4.
bioRxiv ; 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38645261

ABSTRACT

The immunosuppressive function of regulatory T (Treg) cells is essential for maintaining immune homeostasis. Enhancer of zeste homolog 2 (EZH2), a histone H3 lysine 27 (H3K27) methyltransferase, plays a key role in maintaining Treg cell function upon CD28 co-stimulation, and Ezh2 deletion in Treg cells causes autoimmunity. Here we assessed whether increased EZH2 activity in Treg cells would improve Treg cell function. Using an Ezh2 gain-of-function mutation, Ezh2 Y641F , we found that Treg cells expressing Ezh2 Y641F displayed an increased effector Treg phenotype and were poised for improved homing to organ tissues. Expression of Ezh2 Y641F in Treg cells led to more rapid remission from autoimmunity. H3K27me3 profiling and transcriptomic analysis revealed a redistribution of H3K27me3, which prompted a gene expression profile in naïve Ezh2 Y641F Treg cells that recapitulated aspects of CD28-activated Ezh2 WT Treg cells. Altogether, increased EZH2 activity promotes the differentiation of effector Treg cells that can better suppress autoimmunity. Highlights: EZH2 function promotes effector differentiation of Treg cells.EZH2 function promotes Treg cell migration to organ tissues.EZH2 function in Treg cells improves remission from autoimmunity.EZH2 function poises naïve Treg cells to adopt a CD28-activated phenotype.

5.
bioRxiv ; 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38559248

ABSTRACT

Spn1 is a multifunctional histone chaperone essential for life in eukaryotes. While previous work has elucidated regions of the protein important for its many interactions, it is unknown how these domains contribute to the maintenance of chromatin structure. Here, we employ digestion by micrococcal nuclease followed by single-stranded library preparation and sequencing (MNase-SSP) to characterize chromatin structure in yeast expressing wild-type or mutants of Spn1. We mapped nucleosome and subnucleosomal protections genome-wide, and surprisingly, we observed a genome-wide loss of subnucleosomal protection over nucleosome-depleted regions (NDRs) in the Spn1-K192N-containing strain, indicating critical functions of Spn1 in maintaining normal chromatin architecture in promoter regions. Additionally, alterations in nucleosome and hexasome positioning were observed in markedly different mutant Spn1 strains, demonstrating that multiple functions of Spn1 are required to maintain proper chromatin structure in open reading frames, particularly at higher expressed and longer genes. Taken together, our results reveal a previously unknown role of Spn1 in the maintenance of NDR architecture and deepen our understanding of Spn1-dependent chromatin maintenance over transcribed regions.

6.
bioRxiv ; 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38370615

ABSTRACT

Facultative heterochromatinization of genomic regulators by Polycomb repressive complex (PRC) 1 and 2 is essential in development and differentiation; however, the underlying molecular mechanisms remain obscure. Using genetic engineering, molecular approaches, and live-cell single-molecule imaging, we quantify the number of proteins within condensates formed through liquid-liquid phase separation (LLPS) and find that in mouse embryonic stem cells (mESCs), approximately 3 CBX2 proteins nucleate many PRC1 and PRC2 subunits to form one non-stoichiometric condensate. We demonstrate that sparse CBX2 prevents Polycomb proteins from migrating to constitutive heterochromatin, demarcates the spatial boundaries of facultative heterochromatin, controls the deposition of H3K27me3, regulates transcription, and impacts cellular differentiation. Furthermore, we show that LLPS of CBX2 is required for the demarcation and deposition of H3K27me3 and is essential for cellular differentiation. Our findings uncover new functional roles of LLPS in the formation of facultative heterochromatin and unravel a new mechanism by which low-abundant proteins nucleate many other proteins to form compartments that enable them to execute their functions.

7.
J Cell Biol ; 222(9)2023 09 04.
Article in English | MEDLINE | ID: mdl-37516914

ABSTRACT

Herpes simplex virus (HSV-1) progeny form in the nucleus and exit to successfully infect other cells. Newly formed capsids navigate complex chromatin architecture to reach the inner nuclear membrane (INM) and egress. Here, we demonstrate by transmission electron microscopy (TEM) that HSV-1 capsids traverse heterochromatin associated with trimethylation on histone H3 lysine 27 (H3K27me3) and the histone variant macroH2A1. Through chromatin profiling during infection, we revealed global redistribution of these marks whereby massive host genomic regions bound by macroH2A1 and H3K27me3 correlate with decreased host transcription in active compartments. We found that the loss of these markers resulted in significantly lower viral titers but did not impact viral genome or protein accumulation. Strikingly, we discovered that loss of macroH2A1 or H3K27me3 resulted in nuclear trapping of capsids. Finally, by live-capsid tracking, we quantified this decreased capsid movement. Thus, our work demonstrates that HSV-1 takes advantage of the dynamic nature of host heterochromatin formation during infection for efficient nuclear egress.


Subject(s)
Herpesvirus 1, Human , Heterochromatin , Virus Release , Cell Nucleus/virology , Chromatin , Herpesvirus 1, Human/genetics , Heterochromatin/genetics , Histones/genetics , Capsid/ultrastructure
8.
Mol Cell ; 82(19): 3632-3645.e4, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36206739

ABSTRACT

The pause-release model of transcription proposes that 40-100 bases from the start site RNA Pol II pauses, followed by release into productive elongation. Pause release is facilitated by the PTEFb phosphorylation of the RNA Pol II elongation factor, Spt5. We mapped paused polymerases by eNET-seq and found frequent pausing in zones that extend ∼0.3-3 kb into genes even when PTEFb is inhibited. The fraction of paused polymerases or pausing propensity declines gradually over several kb and not abruptly as predicted for a discrete pause-release event. Spt5 depletion extends pausing zones, suggesting that it promotes the maturation of elongation complexes to a low-pausing state. The expression of mutants after Spt5 depletion showed that phosphomimetic substitutions in the CTR1 domain diminished pausing throughout genes. By contrast, mutants that prevent the phosphorylation of the Spt5 RNA-binding domain strengthened pausing. Thus, distinct Spt5 phospho-isoforms set the balance between pausing and elongation.


Subject(s)
RNA Polymerase II , Transcriptional Elongation Factors , Peptide Elongation Factors/metabolism , Phosphorylation , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Transcription, Genetic , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism
9.
Sci Adv ; 8(34): eabm4358, 2022 08 26.
Article in English | MEDLINE | ID: mdl-36001652

ABSTRACT

Genome-wide binding profiles of estrogen receptor (ER) and FOXA1 reflect cancer state in ER+ breast cancer. However, routine profiling of tumor transcription factor (TF) binding is impractical in the clinic. Here, we show that plasma cell-free DNA (cfDNA) contains high-resolution ER and FOXA1 tumor binding profiles for breast cancer. Enrichment of TF footprints in plasma reflects the binding strength of the TF in originating tissue. We defined pure in vivo tumor TF signatures in plasma using ER+ breast cancer xenografts, which can distinguish xenografts with distinct ER states. Furthermore, state-specific ER-binding signatures can partition human breast tumors into groups with significantly different ER expression and mortality. Last, TF footprints in human plasma samples can identify the presence of ER+ breast cancer. Thus, plasma TF footprints enable minimally invasive mapping of the regulatory landscape of breast cancer in humans and open vast possibilities for clinical applications across multiple tumor types.


Subject(s)
Breast Neoplasms , Cell-Free Nucleic Acids , Breast Neoplasms/pathology , Female , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Nucleosomes , Receptors, Estrogen/genetics
10.
STAR Protoc ; 3(2): 101299, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35463472

ABSTRACT

Here, we present a pipeline to map states of protein-binding DNA in vivo. Our pipeline infers as well as quantifies cooperative binding. Using dual-enzyme single-molecule footprinting (dSMF) data, we show how our workflow identifies binding states at an enhancer in Drosophila S2 cells. Data from cells lacking endogenous DNA methylation are a prerequisite for this pipeline. For complete details on the use and execution of this protocol, please refer to Rao et al. (2021) and Krebs et al. (2017).


Subject(s)
DNA Methylation , DNA , DNA/genetics , DNA Methylation/genetics , Protein Binding , Workflow
11.
Annu Rev Biochem ; 91: 183-195, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35303789

ABSTRACT

Gene regulation arises out of dynamic competition between nucleosomes, transcription factors, and other chromatin proteins for the opportunity to bind genomic DNA. The timescales of nucleosome assembly and binding of factors to DNA determine the outcomes of this competition at any given locus. Here, we review how these properties of chromatin proteins and the interplay between the dynamics of different factors are critical for gene regulation. We discuss how molecular structures of large chromatin-associated complexes, kinetic measurements, and high resolution mapping of protein-DNA complexes in vivo set the boundary conditions for chromatin dynamics, leading to models of how the steady state behaviors of regulatory elements arise.


Subject(s)
Chromatin , Nucleosomes , Chromatin/genetics , Chromatin Assembly and Disassembly , DNA/genetics , DNA/metabolism , Nucleosomes/genetics , Transcription Factors/genetics
12.
Mol Cell ; 81(8): 1651-1665.e4, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33705711

ABSTRACT

Enhancers harbor binding motifs that recruit transcription factors (TFs) for gene activation. While cooperative binding of TFs at enhancers is known to be critical for transcriptional activation of a handful of developmental enhancers, the extent of TF cooperativity genome-wide is unknown. Here, we couple high-resolution nuclease footprinting with single-molecule methylation profiling to characterize TF cooperativity at active enhancers in the Drosophila genome. Enrichment of short micrococcal nuclease (MNase)-protected DNA segments indicates that the majority of enhancers harbor two or more TF-binding sites, and we uncover protected fragments that correspond to co-bound sites in thousands of enhancers. From the analysis of co-binding, we find that cooperativity dominates TF binding in vivo at the majority of active enhancers. Cooperativity is highest between sites spaced 50 bp apart, indicating that cooperativity occurs without apparent protein-protein interactions. Our findings suggest nucleosomes promoting cooperativity because co-binding may effectively clear nucleosomes and promote enhancer function.


Subject(s)
Enhancer Elements, Genetic/genetics , Protein Binding/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Binding Sites/genetics , Cell Line , Drosophila/genetics , Drosophila/metabolism , Genome/genetics , Micrococcal Nuclease/genetics , Nucleosomes/genetics , Nucleosomes/metabolism , Protein Interaction Maps/genetics , Transcriptional Activation/genetics
13.
Open Biol ; 10(9): 200119, 2020 09.
Article in English | MEDLINE | ID: mdl-32873154

ABSTRACT

Cell-free DNA (cfDNA) has the potential to enable non-invasive detection of disease states and progression. Beyond its sequence, cfDNA also represents the nucleosomal landscape of cell(s)-of-origin and captures the dynamics of the epigenome. In this review, we highlight the emergence of cfDNA epigenomic methods that assess disease beyond the scope of mutant tumour genotyping. Detection of tumour mutations is the gold standard for sequencing methods in clinical oncology. However, limitations inherent to mutation targeting in cfDNA, and the possibilities of uncovering molecular mechanisms underlying disease, have made epigenomics of cfDNA an exciting alternative. We discuss the epigenomic information revealed by cfDNA, and how epigenomic methods exploit cfDNA to detect and characterize cancer. Future applications of cfDNA epigenomic methods to act complementarily and orthogonally to current clinical practices has the potential to transform cancer management and improve cancer patient outcomes.


Subject(s)
Biomarkers , Cell-Free Nucleic Acids , Phenotype , Chromatin/chemistry , Chromatin/genetics , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Clinical Decision-Making , DNA Methylation , Disease Management , Epigenesis, Genetic , Epigenomics/methods , Gene Expression , Histones/metabolism , Humans , Nucleosomes/genetics , Nucleosomes/metabolism , Outcome Assessment, Health Care , Prognosis
14.
Proc Natl Acad Sci U S A ; 117(33): 19888-19895, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32747552

ABSTRACT

More than 30% of genes in higher eukaryotes are regulated by RNA polymerase II (Pol II) promoter proximal pausing. Pausing is released by the positive transcription elongation factor complex (P-TEFb). However, the exact mechanism by which this occurs and whether phosphorylation of the carboxyl-terminal domain of Pol II is involved in the process remains unknown. We previously reported that JMJD5 could generate tailless nucleosomes at position +1 from transcription start sites (TSS), thus perhaps enable progression of Pol II. Here we find that knockout of JMJD5 leads to accumulation of nucleosomes at position +1. Absence of JMJD5 also results in loss of or lowered transcription of a large number of genes. Interestingly, we found that phosphorylation, by CDK9, of Ser2 within two neighboring heptad repeats in the carboxyl-terminal domain of Pol II, together with phosphorylation of Ser5 within the second repeat, HR-Ser2p (1, 2)-Ser5p (2) for short, allows Pol II to bind JMJD5 via engagement of the N-terminal domain of JMJD5. We suggest that these events bring JMJD5 near the nucleosome at position +1, thus allowing JMJD5 to clip histones on this nucleosome, a phenomenon that may contribute to release of Pol II pausing.


Subject(s)
Cyclin-Dependent Kinase 9/metabolism , Histone Demethylases/metabolism , RNA Polymerase II/metabolism , Transcription, Genetic , Cell Line, Tumor , Cyclin-Dependent Kinase 9/genetics , Histone Demethylases/chemistry , Histone Demethylases/genetics , Humans , Nucleosomes/genetics , Nucleosomes/metabolism , Phosphorylation , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Domains , RNA Polymerase II/genetics
15.
Mol Biol Cell ; 30(23): 2929-2942, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31553691

ABSTRACT

The Hippo signaling pathway regulates tissue growth and organ development in many animals, including humans. Pathway activity leads to inactivation of Yorkie (Yki), a transcriptional coactivator that drives expression of growth-promoting genes. In addition, Yki has been shown to recruit chromatin modifiers that enhance chromatin accessibility and thereby enhance Yki function. Here, we asked whether changes in chromatin accessibility that occur during DNA replication could also affect Yki function. We found that depletion of the chromatin assembly complex-1 (CAF-1) complex, a histone chaperone that is required for nucleosome assembly after DNA replication, in the wing imaginal epithelium leads to increased Hippo pathway target gene expression but does not affect expression of other genes. Yki shows greater association with target sites when CAF-1 is depleted and misregulation of target gene expression is Yki-dependent, suggesting that nucleosome assembly competes with Yki for pathway targets post-DNA replication. Consistent with this idea, increased target gene expression is DNA replication dependent and newly replicated chromatin at target sites shows marked nucleosome depletion when CAF-1 function is reduced. These observations suggest a connection between cell cycle progression and Hippo pathway target expression, providing insights into functions of the Hippo pathway in normal and abnormal tissue growth.


Subject(s)
Chromatin Assembly Factor-1/metabolism , DNA Replication/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Drosophila melanogaster/growth & development , Genetic Loci , Histone Code , Histones/metabolism , Models, Biological , Nuclear Proteins/metabolism , Protein Binding , Signal Transduction/genetics , Trans-Activators/metabolism , YAP-Signaling Proteins
16.
Cell ; 178(4): 774-776, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398334

ABSTRACT

In this issue of Cell, Cassidy et al. (2019) show that, in Drosophila melanogaster, developmental abnormalities resulting from loss of repressors such as microRNAs can be suppressed by slow metabolism. They additionally provide insight into the underlying mechanism that connects metabolic state with developmental outcomes.


Subject(s)
Drosophila melanogaster/genetics , MicroRNAs , Animals , Gene Expression Regulation , Transcription Factors
17.
Methods Mol Biol ; 1832: 159-168, 2018.
Article in English | MEDLINE | ID: mdl-30073526

ABSTRACT

The epigenome has been mapped in different cell types to understand the relationship between the chromatin landscape and the control of gene expression. Most mapping studies profile a large population of cells in various stages of the cell cycle, which results in an average snapshot of the chromatin landscape. However, chromatin is highly dynamic, undergoing rapid changes during active processes such as replication, transcription, repair, and remodeling. Hence, we need methods to map chromatin as a function of time. To address this problem in the context of replication, we developed the method MINCE-seq (Mapping In vivo Nascent Chromatin with EdU and sequencing). MINCE-seq is a genome-wide method that uses the passage of replication fork as a starting point to map the chromatin landscape as a function of time. MINCE-seq can measure chromatin dynamics in a time scale of minutes and at the resolution of individual nucleosome positions and transcription factor-binding sites genome-wide.


Subject(s)
Chromatin/metabolism , Deoxyuridine/analogs & derivatives , High-Throughput Nucleotide Sequencing/methods , Animals , Cell Line , Click Chemistry , Deoxyuridine/metabolism , Drosophila melanogaster/cytology
18.
Mol Cell ; 71(2): 193-194, 2018 07 19.
Article in English | MEDLINE | ID: mdl-30028998

ABSTRACT

Two papers in Molecular Cell (Kubik et al., 2018; Yan et al., 2018) explore the mechanisms by which transcription factors bind their sites in chromatin, providing fresh insights into the much-debated question of how transcription factors can be "pioneers."


Subject(s)
Saccharomycetales , Transcription Factors/genetics , Chromatin , Chromatin Assembly and Disassembly , Nucleosomes
19.
Genome Biol ; 19(1): 19, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29426353

ABSTRACT

We developed a chemical cleavage method that releases single nucleosome dyad-containing fragments, allowing us to precisely map both single nucleosomes and linkers with high accuracy genome-wide in yeast. Our single nucleosome positioning data reveal that nucleosomes occupy preferred positions that differ by integral multiples of the DNA helical repeat. By comparing nucleosome dyad positioning maps to existing genomic and transcriptomic data, we evaluated the contributions of sequence, transcription, and histones H1 and H2A.Z in defining the chromatin landscape. We present a biophysical model that neglects DNA sequence and shows that steric occlusion suffices to explain the salient features of nucleosome positioning.


Subject(s)
Genomics/methods , Nucleosomes , Genes , Histones , Models, Biological , Transcription, Genetic
20.
Mol Cell ; 68(6): 1038-1053.e4, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29225036

ABSTRACT

Nucleosomes are disrupted during transcription and other active processes, but the structural intermediates during nucleosome disruption in vivo are unknown. To identify intermediates, we mapped subnucleosomal protections in Drosophila cells using Micrococcal Nuclease followed by sequencing. At the first nucleosome position downstream of the transcription start site, we identified unwrapped intermediates, including hexasomes that lack either proximal or distal contacts. Inhibiting topoisomerases or depleting histone chaperones increased unwrapping, whereas inhibiting release of paused RNAPII or reducing RNAPII elongation decreased unwrapping. Our results indicate that positive torsion generated by elongating RNAPII causes transient loss of histone-DNA contacts. Using this mapping approach, we found that nucleosomes flanking human CTCF insulation sites are similarly disrupted. We also identified diagnostic subnucleosomal particle remnants in cell-free human DNA data as a relic of transcribed genes from apoptosing cells. Thus identification of subnucleosomal fragments from nuclease protection data represents a general strategy for structural epigenomics.


Subject(s)
Chromatin Assembly and Disassembly , Drosophila melanogaster/metabolism , Epigenomics , Gene Expression Regulation , Histones/chemistry , Nucleosomes/chemistry , Transcription, Genetic , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Histones/genetics , Histones/metabolism , Micrococcal Nuclease/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Transcription Initiation Site
SELECTION OF CITATIONS
SEARCH DETAIL
...