Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38405904

ABSTRACT

The RING E3 ubiquitin ligase UHRF1 is an established cofactor for DNA methylation inheritance. Nucleosomal engagement through histone and DNA interactions directs UHRF1 ubiquitin ligase activity toward lysines on histone H3 tails, creating binding sites for DNMT1 through ubiquitin interacting motifs (UIM1 and UIM2). Here, we profile contributions of UHRF1 and DNMT1 to genome-wide DNA methylation inheritance and dissect specific roles for ubiquitin signaling in this process. We reveal DNA methylation maintenance at low-density CpGs is vulnerable to disruption of UHRF1 ubiquitin ligase activity and DNMT1 ubiquitin reading activity through UIM1. Hypomethylation of low-density CpGs in this manner induces formation of partially methylated domains (PMD), a methylation signature observed across human cancers. Furthermore, disrupting DNMT1 UIM2 function abolishes DNA methylation maintenance. Collectively, we show DNMT1-dependent DNA methylation inheritance is a ubiquitin-regulated process and suggest a disrupted UHRF1-DNMT1 ubiquitin signaling axis contributes to the development of PMDs in human cancers.

2.
bioRxiv ; 2023 Dec 03.
Article in English | MEDLINE | ID: mdl-38077086

ABSTRACT

Kinase domains are highly conserved within protein kinases in both sequence and structure. Many factors, including phosphorylation, amino acid substitutions or mutations, and small molecule inhibitor binding, influence conformations of the kinase domain and enzymatic activity. The serine/threonine kinases ULK1 and ULK2 are highly conserved with N- and C-terminal domains, phosphate-binding P-loops, αC-helix, regulatory and catalytic spines, and activation loop DFG and APE motifs. Here, we performed molecular dynamics (MD) simulations to understand better the potency and selectivity of the ULK1/2 small molecule inhibitor, ULK-101. We observed stable bound states for ULK-101 to the adenosine triphosphate (ATP)-binding site of ULK2, coordinated by hydrogen bonding with the hinge backbone and the catalytic lysine sidechain. Notably, ULK-101 occupies a hydrophobic pocket associated with the N-terminus of the αC-helix. Large movements in the P-loop are also associated with ULK-101 inhibitor binding and exit from ULK2. Our data further suggests that ULK-101 could induce a folded P-loop conformation and hydrophobic pocket reflected in its nanomolar potency and kinome selectivity.

3.
Immunity ; 56(9): 2021-2035.e8, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37516105

ABSTRACT

Environmental nutrient availability influences T cell metabolism, impacting T cell function and shaping immune outcomes. Here, we identified ketone bodies (KBs)-including ß-hydroxybutyrate (ßOHB) and acetoacetate (AcAc)-as essential fuels supporting CD8+ T cell metabolism and effector function. ßOHB directly increased CD8+ T effector (Teff) cell cytokine production and cytolytic activity, and KB oxidation (ketolysis) was required for Teff cell responses to bacterial infection and tumor challenge. CD8+ Teff cells preferentially used KBs over glucose to fuel the tricarboxylic acid (TCA) cycle in vitro and in vivo. KBs directly boosted the respiratory capacity and TCA cycle-dependent metabolic pathways that fuel CD8+ T cell function. Mechanistically, ßOHB was a major substrate for acetyl-CoA production in CD8+ T cells and regulated effector responses through effects on histone acetylation. Together, our results identify cell-intrinsic ketolysis as a metabolic and epigenetic driver of optimal CD8+ T cell effector responses.


Subject(s)
CD8-Positive T-Lymphocytes , Histones , 3-Hydroxybutyric Acid/metabolism , 3-Hydroxybutyric Acid/pharmacology , Acetylation , Histones/metabolism , Ketone Bodies , Animals , Mice
4.
Sci Rep ; 13(1): 7508, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37160995

ABSTRACT

We recently introduced an absolute and physical quantitative scale for chromatin immunoprecipitation followed by sequencing (ChIP-seq). The scale itself was determined directly from measurements routinely made on sequencing samples without additional reagents or spike-ins. We called this approach sans spike-in quantitative ChIP, or siQ-ChIP. Herein, we extend those results in several ways. First, we simplified the calculations defining the quantitative scale, reducing practitioner burden. Second, we reveal a normalization constraint implied by the quantitative scale and introduce a new scheme for generating 'tracks'. The constraint requires that tracks are probability distributions so that quantified ChIP-seq is analogous to a mass distribution. Third, we introduce some whole-genome analyses that allow us, for example, to project the IP mass (immunoprecipitated mass) onto the genome to evaluate how much of any genomic interval was captured in the IP. We applied siQ-ChIP to p300/CBP inhibition and compare our results to those of others. We detail how the same data-level observations are misinterpreted in the literature when tracks are not understood as probability densities and are compared without correct quantitative scaling, and we offer new interpretations of p300/CBP inhibition outcomes.


Subject(s)
Histone Code , Nucleosomes , Nucleosomes/genetics , Chromatin Immunoprecipitation , Chromatin Immunoprecipitation Sequencing , Genomics
5.
Nucleic Acids Res ; 51(12): 5997-6005, 2023 07 07.
Article in English | MEDLINE | ID: mdl-37094063

ABSTRACT

CpG methylation generally occurs on both DNA strands and is essential for mammalian development and differentiation. Until recently, hemimethylation, in which only one strand is methylated, was considered to be simply a transitory state generated during DNA synthesis. The discovery that a subset of CCCTC-binding factor (CTCF) binding sites is heritably hemimethylated suggests that hemimethylation might have an unknown biological function. Here we show that the binding of CTCF is profoundly altered by which DNA strand is methylated and by the specific CTCF binding motif. CpG methylation on the motif strand can inhibit CTCF binding by up to 7-fold, whereas methylation on the opposite strand can stimulate binding by up to 4-fold. Thus, hemimethylation can alter binding by up to 28-fold in a strand-specific manner. The mechanism for sensing methylation on the opposite strand requires two critical residues, V454 and S364, within CTCF zinc fingers 7 and 4. Similar to methylation, CpG hydroxymethylation on the motif strand can inhibit CTCF binding by up to 4-fold. However, hydroxymethylation on the opposite strand removes the stimulatory effect. Strand-specific methylation states may therefore provide a mechanism to explain the transient and dynamic nature of CTCF-mediated chromatin interactions.


Subject(s)
CCCTC-Binding Factor , DNA Methylation , Repressor Proteins , Animals , Binding Sites , CCCTC-Binding Factor/metabolism , Chromatin , CpG Islands , DNA/metabolism , Mammals/genetics , Repressor Proteins/metabolism
6.
bioRxiv ; 2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36945621

ABSTRACT

We previously developed sans spike-in quantitative chromatin immunoprecipitation sequencing (siQ-ChIP), a technique that introduces an absolute quantitative scale to ChIP-seq data without reliance on spike-in normalization approaches. The physical model of siQ-ChIP predicted that the IP step of ChIP would produce a classical binding isotherm when antibody or epitope was titrated. Here, we define experimental conditions in which this titration is observable for antibodies that recognize modified states of histone proteins. We show that minimally sequenced points along an isotherm can reveal differential binding specificities that are associated with on- and off-target epitope interactions. This work demonstrates that the interpretation of histone post-translational modification distribution from ChIP-seq data has a dependence on antibody concentration. Collectively, these studies introduce a simplified and reproducible experimental method to generate quantitative ChIP-seq data without spike-in normalization and demonstrate that histone antibody specificity can be analyzed directly in ChIP-seq experiments.

7.
Sci Rep ; 13(1): 377, 2023 01 07.
Article in English | MEDLINE | ID: mdl-36611042

ABSTRACT

Lysine methylation modulates the function of histone and non-histone proteins, and the enzymes that add or remove lysine methylation-lysine methyltransferases (KMTs) and lysine demethylases (KDMs), respectively-are frequently mutated and dysregulated in human diseases. Identification of lysine methylation sites proteome-wide has been a critical barrier to identifying the non-histone substrates of KMTs and KDMs and for studying functions of non-histone lysine methylation. Detection of lysine methylation by mass spectrometry (MS) typically relies on the enrichment of methylated peptides by pan-methyllysine antibodies. In this study, we use peptide microarrays to show that pan-methyllysine antibodies have sequence bias, and we evaluate how the differential selectivity of these reagents impacts the detection of methylated peptides in MS-based workflows. We discovered that most commercially available pan-Kme antibodies have an in vitro sequence bias, and multiple enrichment approaches provide the most comprehensive coverage of the lysine methylome. Overall, global lysine methylation proteomics with multiple characterized pan-methyllysine antibodies resulted in the detection of 5089 lysine methylation sites on 2751 proteins from two human cell lines, nearly doubling the number of reported lysine methylation sites in the human proteome.


Subject(s)
Lysine , Proteome , Humans , Lysine/metabolism , Proteome/metabolism , Epigenome , Methylation , Peptides/metabolism , Antibodies/metabolism
8.
Epigenetics Chromatin ; 13(1): 44, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33097091

ABSTRACT

The chromatin-binding E3 ubiquitin ligase ubiquitin-like with PHD and RING finger domains 1 (UHRF1) contributes to the maintenance of aberrant DNA methylation patterning in cancer cells through multivalent histone and DNA recognition. The tandem Tudor domain (TTD) of UHRF1 is well-characterized as a reader of lysine 9 di- and tri-methylation on histone H3 (H3K9me2/me3) and, more recently, lysine 126 di- and tri-methylation on DNA ligase 1 (LIG1K126me2/me3). However, the functional significance and selectivity of these interactions remain unclear. In this study, we used protein domain microarrays to search for additional readers of LIG1K126me2, the preferred methyl state bound by the UHRF1 TTD. We show that the UHRF1 TTD binds LIG1K126me2 with high affinity and selectivity compared to other known methyllysine readers. Notably, and unlike H3K9me2/me3, the UHRF1 plant homeodomain (PHD) and its N-terminal linker (L2) do not contribute to multivalent LIG1K126me2 recognition along with the TTD. To test the functional significance of this interaction, we designed a LIG1K126me2 cell-penetrating peptide (CPP). Consistent with LIG1 knockdown, uptake of the CPP had no significant effect on the propagation of DNA methylation patterning across the genomes of bulk populations from high-resolution analysis of several cancer cell lines. Further, we did not detect significant changes in DNA methylation patterning from bulk cell populations after chemical or genetic disruption of lysine methyltransferase activity associated with LIG1K126me2 and H3K9me2. Collectively, these studies identify UHRF1 as a selective reader of LIG1K126me2 in vitro and further implicate the histone and non-histone methyllysine reader activity of the UHRF1 TTD as a dispensable domain function for cancer cell DNA methylation maintenance.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , DNA Methylation , Gene Expression Regulation, Neoplastic , Histone Code , Ubiquitin-Protein Ligases/metabolism , CCAAT-Enhancer-Binding Proteins/chemistry , Epigenesis, Genetic , HCT116 Cells , HeLa Cells , Histones/chemistry , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Protein Processing, Post-Translational , Tudor Domain , Ubiquitin-Protein Ligases/chemistry
9.
J Biol Chem ; 295(47): 15826-15837, 2020 11 20.
Article in English | MEDLINE | ID: mdl-32994221

ABSTRACT

ChIP followed by next-generation sequencing (ChIP-Seq) is a key technique for mapping the distribution of histone posttranslational modifications (PTMs) and chromatin-associated factors across genomes. There is a perceived challenge to define a quantitative scale for ChIP-Seq data, and as such, several approaches making use of exogenous additives, or "spike-ins," have recently been developed. Herein, we report on the development of a quantitative, physical model defining ChIP-Seq. The quantitative scale on which ChIP-Seq results should be compared emerges from the model. To test the model and demonstrate the quantitative scale, we examine the impacts of an EZH2 inhibitor through the lens of ChIP-Seq. We report a significant increase in immunoprecipitation of presumed off-target histone PTMs after inhibitor treatment, a trend predicted by the model but contrary to spike-in-based indications. Our work also identifies a sensitivity issue in spike-in normalization that has not been considered in the literature, placing limitations on its utility and trustworthiness. We call our new approach the sans-spike-in method for quantitative ChIP-sequencing (siQ-ChIP). A number of changes in community practice of ChIP-Seq, data reporting, and analysis are motivated by this work.


Subject(s)
Chromatin Immunoprecipitation Sequencing , Sequence Analysis, DNA , Humans
10.
J Chem Phys ; 153(5): 054125, 2020 Aug 07.
Article in English | MEDLINE | ID: mdl-32770912

ABSTRACT

In an infrequent metadynamics (iMetaD) simulation, a well-tempered metadynamics bias accumulates in the reactant basin, accelerating escapes to the product state. Like the earlier hyperdynamics strategy, iMetaD enables estimates of the unbiased escape rates. However, iMetaD applies the bias to visited locations in a collective variable (CV) space, not to the more specific visited locations in a full configuration space as done in hyperdynamics. This difference makes rate estimates from iMetaD sensitive to the choice of CVs, to parameters that control the bias deposition rate, and to the preparation of the initial state within the reactant basin. This paper uses an extremely simple discrete state model to illustrate complications that can arise in systems that exhibit fluxional transitions between sub-basins of the reactant state. Specifically, we show how the reactant-to-product escape time and relaxation times within the reactant basin(s) impose bounds on the admissible parameter choices for an iMetaD calculation. Predictions from the discrete state model are validated by iMetaD simulations on a corresponding two-dimensional potential energy surface.

11.
Front Cell Dev Biol ; 8: 241, 2020.
Article in English | MEDLINE | ID: mdl-32328492

ABSTRACT

Lysine methylation facilitates protein-protein interactions through the activity of methyllysine (Kme) "reader" proteins. Functions of Kme readers have historically been studied in the context of histone interactions, where readers aid in chromatin-templated processes such as transcription, DNA replication and repair. However, there is growing evidence that Kme readers also function through interactions with non-histone proteins. To facilitate expanded study of Kme reader activities, we developed a high-throughput binding assay to reveal the sequence determinants of Kme-driven protein interactions. The assay queries a degenerate methylated lysine-oriented peptide library (Kme-OPL) to identify the key residues that modulate reader binding. The assay recapitulated methyl order and amino acid sequence preferences associated with histone Kme readers. The assay also revealed methylated sequences that bound Kme readers with higher affinity than histones. Proteome-wide scoring was applied to assay results to help prioritize future study of Kme reader interactions. The platform was also used to design sequences that directed specificity among closely related reader domains, an application which may have utility in the development of peptidomimetic inhibitors. Furthermore, we used the platform to identify binding determinants of site-specific histone Kme antibodies and surprisingly revealed that only a few amino acids drove epitope recognition. Collectively, these studies introduce and validate a rapid, unbiased, and high-throughput binding assay for Kme readers, and we envision its use as a resource for expanding the study of Kme-driven protein interactions.

12.
PLoS Comput Biol ; 16(4): e1007394, 2020 04.
Article in English | MEDLINE | ID: mdl-32275713

ABSTRACT

The development of novel analgesics with improved safety profiles to combat the opioid epidemic represents a central question to G protein coupled receptor structural biology and pharmacology: What chemical features dictate G protein or ß-arrestin signaling? Here we use adaptively biased molecular dynamics simulations to determine how fentanyl, a potent ß-arrestin biased agonist, binds the µ-opioid receptor (µOR). The resulting fentanyl-bound pose provides rational insight into a wealth of historical structure-activity-relationship on its chemical scaffold. Following an in-silico derived hypothesis we found that fentanyl and the synthetic opioid peptide DAMGO require M153 to induce ß-arrestin coupling, while M153 was dispensable for G protein coupling. We propose and validate an activation mechanism where the n-aniline ring of fentanyl mediates µOR ß-arrestin through a novel M153 "microswitch" by synthesizing fentanyl-based derivatives that exhibit complete, clinically desirable, G protein biased coupling. Together, these results provide molecular insight into fentanyl mediated ß-arrestin biased signaling and a rational framework for further optimization of fentanyl-based analgesics with improved safety profiles.


Subject(s)
Fentanyl/pharmacology , beta-Arrestins/metabolism , beta-Arrestins/ultrastructure , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Fentanyl/metabolism , GTP-Binding Proteins/metabolism , Humans , Molecular Dynamics Simulation , Receptors, G-Protein-Coupled/metabolism , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , beta-Arrestins/agonists
13.
Cureus ; 12(2): e6910, 2020 Feb 07.
Article in English | MEDLINE | ID: mdl-32190465

ABSTRACT

Introduction Most medical residency training programs maintain websites to provide content for audiences including current residents, prospective residents, and medical students. This study seeks to characterize when and how a residency program website is being accessed in order to identify the primary audiences to provide appropriate and timely content. Methods The authors examined website analytics at a large urban Accreditation Council for Graduate Medical Education (ACGME)-accredited post-graduate year (PGY)1-PGY4 emergency medicine residency training program website. Analytics were performed from July 1, 2016 to June 30, 2018 with daily traffic cataloged along with referral sources, page views, and device type used to access the website. The top five dates by daily traffic were further analyzed with daily traffic trends during the residency interview season. Results There was an average of 45.8 unique visitors daily with 261.5 daily page views. Computer (67.2%), mobile device (29.6%), and tablet (3.1%) were the most common devices used for viewing. The most popular content areas by page-view were "people" (68,987 visits), "home" (38,569), "clinical curriculum" (35,556), and "medical students" (14,461). The five most-visited dates were all related to application processes including the opening of the Visiting Student Application Service (VSAS), the Electronic Residency Application Service (ERAS), and Match Day. During the interview season, peak visits occurred the dates immediately preceding interview dates. Conclusion Residency program websites appear to be accessed most commonly by medical students and prospective residency applicants. Website managers should take the needs of these audiences into account and provide appropriate content to maximally inform prospective residency program candidates.

14.
J Biol Chem ; 294(43): 15724-15732, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31481468

ABSTRACT

The Su(var)3-9, enhancer of zeste, and trithorax (SET) and really interesting new gene (RING) finger-associated (SRA) protein domain is conserved across bacteria and eukaryota and coordinates extrahelical or "flipped" DNA bases. A functional SRA domain is required for ubiquitin-like with PHD and RING finger domains 1 (UHRF1) E3 ubiquitin ligase activity toward histone H3, a mechanism for recruiting the DNA methylation maintenance enzyme DNA methyltransferase 1 (DNMT1). The SRA domain supports UHRF1 oncogenic activity in colon cancer cells, highlighting that UHRF1 SRA antagonism could be a cancer therapeutic strategy. Here we used molecular dynamics simulations, DNA binding assays, in vitro ubiquitination reactions, and DNA methylation analysis to identify the SRA finger loop as a regulator of UHRF1 ubiquitin targeting and DNA methylation maintenance. A chimeric UHRF1 (finger swap) with diminished E3 ligase activity toward nucleosomal histones, despite tighter binding to unmodified or asymmetric or symmetrically methylated DNA, uncouples DNA affinity from regulation of E3 ligase activity. Our model suggests that SRA domains sample DNA bases through flipping in the presence or absence of a cytosine modification and that specific interactions of the SRA finger loop with DNA are required for downstream host protein function. Our findings provide insight into allosteric regulation of UHRF1 E3 ligase activity, suggesting that UHRF1's SRA finger loop regulates its conformation and function.


Subject(s)
CCAAT-Enhancer-Binding Proteins/chemistry , CCAAT-Enhancer-Binding Proteins/metabolism , DNA Methylation/genetics , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Amino Acid Sequence , DNA/chemistry , HCT116 Cells , HEK293 Cells , Humans , Phosphates/metabolism , Protein Domains , Structure-Activity Relationship
16.
J Chem Theory Comput ; 15(1): 78-83, 2019 Jan 08.
Article in English | MEDLINE | ID: mdl-30468579

ABSTRACT

Recent combinations of hyperdynamics and adaptive biasing potentials or free-energy-based bias potentials introduce an implicit assumption: the bias potential needs only be zero on the prominent free-energy barriers of interest. Herein we demonstrate that this implicit assumption is flawed. Thus, hyperdynamics in collective variables is likely to fail unless there are no hidden barriers within the initial state. Moreoever, the one-sample Kolmogorov-Smirnov test used to declare "trust" in hyperdynamics is shown to be unable to classify this failure. In fact, nonzero bias potential on hidden barriers emerges as a contradiction of the hypothesis that a Poisson distributed set of hyperdynamics escape times is a correctly distributed set of escape times. We demonstrate failure in the alanine dipeptide benchmark, and we present an apparent failure for a protein conformation change. The standards for "trustworthy" hyperdynamics in collective variables must be raised.


Subject(s)
Thermodynamics , Molecular Dynamics Simulation
17.
Sci Adv ; 4(11): eaav2623, 2018 11.
Article in English | MEDLINE | ID: mdl-30498785

ABSTRACT

Lysine methylation is a key regulator of histone protein function. Beyond histones, few connections have been made to the enzymes responsible for the deposition of these posttranslational modifications. Here, we debut a high-throughput functional proteomics platform that maps the sequence determinants of lysine methyltransferase (KMT) substrate selectivity without a priori knowledge of a substrate or target proteome. We demonstrate the predictive power of this approach for identifying KMT substrates, generating scaffolds for inhibitor design, and predicting the impact of missense mutations on lysine methylation signaling. By comparing KMT selectivity profiles to available lysine methylome datasets, we reveal a disconnect between preferred KMT substrates and the ability to detect these motifs using standard mass spectrometry pipelines. Collectively, our studies validate the use of this platform for guiding the study of lysine methylation signaling and suggest that substantial gaps exist in proteome-wide curation of lysine methylomes.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lysine/metabolism , Proteome/analysis , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Humans , Methylation , Mutation, Missense , Substrate Specificity
19.
Mol Cell ; 72(1): 162-177.e7, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30244833

ABSTRACT

Histone post-translational modifications (PTMs) are important genomic regulators often studied by chromatin immunoprecipitation (ChIP), whereby their locations and relative abundance are inferred by antibody capture of nucleosomes and associated DNA. However, the specificity of antibodies within these experiments has not been systematically studied. Here, we use histone peptide arrays and internally calibrated ChIP (ICeChIP) to characterize 52 commercial antibodies purported to distinguish the H3K4 methylforms (me1, me2, and me3, with each ascribed distinct biological functions). We find that many widely used antibodies poorly distinguish the methylforms and that high- and low-specificity reagents can yield dramatically different biological interpretations, resulting in substantial divergence from the literature for numerous H3K4 methylform paradigms. Using ICeChIP, we also discern quantitative relationships between enhancer H3K4 methylation and promoter transcriptional output and can measure global PTM abundance changes. Our results illustrate how poor antibody specificity contributes to the "reproducibility crisis," demonstrating the need for rigorous, platform-appropriate validation.


Subject(s)
Antibodies/genetics , Chromatin Immunoprecipitation/methods , Heterochromatin/genetics , Histones/genetics , Antibodies/chemistry , Antibodies/immunology , Antibody Specificity , Heterochromatin/chemistry , Heterochromatin/immunology , Histone Code/genetics , Histones/chemistry , Histones/immunology , Humans , Methylation , Nucleosomes/genetics , Promoter Regions, Genetic/genetics , Protein Processing, Post-Translational/genetics
20.
Proc Natl Acad Sci U S A ; 115(35): 8775-8780, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30104358

ABSTRACT

Mitotic inheritance of DNA methylation patterns is facilitated by UHRF1, a DNA- and histone-binding E3 ubiquitin ligase that helps recruit the maintenance DNA methyltransferase DNMT1 to replicating chromatin. The DNA methylation maintenance function of UHRF1 is dependent on its ability to bind chromatin, where it facilitates monoubiquitination of histone H3 at lysines 18 and 23, a docking site for DNMT1. Because of technical limitations, this model of UHRF1-dependent DNA methylation inheritance has been constructed largely based on genetics and biochemical observations querying methylated DNA oligonucleotides, synthetic histone peptides, and heterogeneous chromatin extracted from cells. Here, we construct semisynthetic mononucleosomes harboring defined histone and DNA modifications and perform rigorous analysis of UHRF1 binding and enzymatic activity with these reagents. We show that multivalent engagement of nucleosomal linker DNA and dimethylated lysine 9 on histone H3 directs UHRF1 ubiquitin ligase activity toward histone substrates. Notably, we reveal a molecular switch, stimulated by recognition of hemimethylated DNA, which redirects UHRF1 ubiquitin ligase activity away from histones in favor of robust autoubiquitination. Our studies support a noncompetitive model for UHRF1 and DNMT1 chromatin recruitment to replicating chromatin and define a role for hemimethylated linker DNA as a regulator of UHRF1 ubiquitin ligase substrate selectivity.


Subject(s)
CCAAT-Enhancer-Binding Proteins , Chromatin , DNA Methylation , Histones , Models, Biological , Ubiquitination , CCAAT-Enhancer-Binding Proteins/chemistry , CCAAT-Enhancer-Binding Proteins/metabolism , Chromatin/chemistry , Chromatin/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/chemistry , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Histones/chemistry , Histones/metabolism , Humans , Substrate Specificity , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL
...