Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Mol Cell ; 75(6): 1092-1101, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31539507

RESUMEN

Landmark discoveries made nearly two decades ago identified known transcriptional regulators as histone lysine methyltransferases. Since then, the field of lysine methylation signaling has been dominated by studies of how this small chemical posttranslational modification regulates gene expression and other chromatin-based processes. However, recent advances in mass-spectrometry-based proteomics have revealed that histones are just a subset of the thousands of eukaryotic proteins marked by lysine methylation. As the writers, erasers, and readers of histone lysine methylation are emerging as a promising therapeutic target class for cancer and other diseases, a key challenge for the field is to define the full spectrum of activities for these proteins. Here we summarize recent discoveries implicating non-histone lysine methylation as a major regulator of diverse cellular processes. We further discuss recent technological innovations that are enabling the expanded study of lysine methylation signaling. Collectively, these findings are shaping our understanding of the fundamental mechanisms of non-histone protein regulation through this dynamic and multi-functional posttranslational modification.


Asunto(s)
Epigenoma , Lisina/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Procesamiento Proteico-Postraduccional , Transducción de Señal , Animales , Humanos , Metilación
2.
Mol Cell ; 72(1): 162-177.e7, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30244833

RESUMEN

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.


Asunto(s)
Anticuerpos/genética , Inmunoprecipitación de Cromatina/métodos , Heterocromatina/genética , Histonas/genética , Anticuerpos/química , Anticuerpos/inmunología , Especificidad de Anticuerpos , Heterocromatina/química , Heterocromatina/inmunología , Código de Histonas/genética , Histonas/química , Histonas/inmunología , Humanos , Metilación , Nucleosomas/genética , Regiones Promotoras Genéticas/genética , Procesamiento Proteico-Postraduccional/genética
3.
J Biol Chem ; 299(5): 104651, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36972790

RESUMEN

Lysine methylation is a dynamic, posttranslational mark that regulates the function of histone and nonhistone proteins. Many of the enzymes that mediate lysine methylation, known as lysine methyltransferases (KMTs), were originally identified to modify histone proteins but have also been discovered to methylate nonhistone proteins. In this work, we investigate the substrate selectivity of the KMT PRDM9 to identify both potential histone and nonhistone substrates. Though normally expressed in germ cells, PRDM9 is significantly upregulated across many cancer types. The methyltransferase activity of PRDM9 is essential for double-strand break formation during meiotic recombination. PRDM9 has been reported to methylate histone H3 at lysine residues 4 and 36; however, PRDM9 KMT activity had not previously been evaluated on nonhistone proteins. Using lysine-oriented peptide libraries to screen potential substrates of PRDM9, we determined that PRDM9 preferentially methylates peptide sequences not found in any histone protein. We confirmed PRDM9 selectivity through in vitro KMT reactions using peptides with substitutions at critical positions. A multisite λ-dynamics computational analysis provided a structural rationale for the observed PRDM9 selectivity. The substrate selectivity profile was then used to identify putative nonhistone substrates, which were tested by peptide spot array, and a subset was further validated at the protein level by in vitro KMT assays on recombinant proteins. Finally, one of the nonhistone substrates, CTNNBL1, was found to be methylated by PRDM9 in cells.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Lisina , Metilación , Procesamiento Proteico-Postraduccional , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Especificidad por Sustrato , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo
4.
J Biol Chem ; 298(5): 101894, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35378129

RESUMEN

Extensive portions of the human genome have unknown function, including those derived from transposable elements. One such element, the DNA transposon Hsmar1, entered the primate lineage approximately 50 million years ago leaving behind terminal inverted repeat (TIR) sequences and a single intact copy of the Hsmar1 transposase, which retains its ancestral TIR-DNA-binding activity, and is fused with a lysine methyltransferase SET domain to constitute the chimeric SETMAR gene. Here, we provide a structural basis for recognition of TIRs by SETMAR and investigate the function of SETMAR through genome-wide approaches. As elucidated in our 2.37 Å crystal structure, SETMAR forms a dimeric complex with each DNA-binding domain bound specifically to TIR-DNA through the formation of 32 hydrogen bonds. We found that SETMAR recognizes primarily TIR sequences (∼5000 sites) within the human genome as assessed by chromatin immunoprecipitation sequencing analysis. In two SETMAR KO cell lines, we identified 163 shared differentially expressed genes and 233 shared alternative splicing events. Among these genes are several pre-mRNA-splicing factors, transcription factors, and genes associated with neuronal function, and one alternatively spliced primate-specific gene, TMEM14B, which has been identified as a marker for neocortex expansion associated with brain evolution. Taken together, our results suggest a model in which SETMAR impacts differential expression and alternative splicing of genes associated with transcription and neuronal function, potentially through both its TIR-specific DNA-binding and lysine methyltransferase activities, consistent with a role for SETMAR in simian primate development.


Asunto(s)
Genoma Humano , N-Metiltransferasa de Histona-Lisina/genética , Primates/genética , Animales , Evolución Biológica , Encéfalo/metabolismo , Elementos Transponibles de ADN/genética , Estudio de Asociación del Genoma Completo , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Secuencias Invertidas Repetidas , Lisina/genética , Primates/metabolismo , Transposasas/química
5.
J Biol Chem ; 295(47): 15826-15837, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-32994221

RESUMEN

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.


Asunto(s)
Secuenciación de Inmunoprecipitación de Cromatina , Análisis de Secuencia de ADN , Humanos
6.
Proc Natl Acad Sci U S A ; 115(35): 8775-8780, 2018 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-30104358

RESUMEN

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.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT , Cromatina , Metilación de ADN , Histonas , Modelos Biológicos , Ubiquitinación , Proteínas Potenciadoras de Unión a CCAAT/química , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Cromatina/química , Cromatina/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/química , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Histonas/química , Histonas/metabolismo , Humanos , Especificidad por Sustrato , Ubiquitina-Proteína Ligasas
7.
Nucleic Acids Res ; 46(9): 4405-4416, 2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29506131

RESUMEN

UHRF1 is a histone- and DNA-binding E3 ubiquitin ligase that functions with DNMT1 to maintain mammalian DNA methylation. UHRF1 facilitates DNMT1 recruitment to replicating chromatin through a coordinated mechanism involving histone and DNA recognition and histone ubiquitination. UHRF2 shares structural homology with UHRF1, but surprisingly lacks functional redundancy to facilitate DNA methylation maintenance. Molecular mechanisms uncoupling UHRF2 from DNA methylation maintenance are poorly defined. Through comprehensive and comparative biochemical analysis of recombinant human UHRF1 and UHRF2 reader and writer activities, we reveal conserved modes of histone PTM recognition but divergent DNA binding properties. While UHRF1 and UHRF2 diverge in their affinities toward hemi-methylated DNA, we surprisingly show that both hemi-methylated and hemi-hydroxymethylated DNA oligonucleotides stimulate UHRF2 ubiquitin ligase activity toward histone H3 peptide substrates. This is the first example of an E3 ligase allosterically regulated by DNA hydroxymethylation. However, UHRF2 is not a productive histone E3 ligase toward purified mononucleosomes, suggesting UHRF2 has an intra-domain architecture distinct from UHRF1 that is conformationally constrained when bound to chromatin. Collectively, our studies reveal that uncoupling of UHRF2 from the DNA methylation maintenance program is linked to differences in the molecular readout of chromatin signatures that connect UHRF1 to ubiquitination of histone H3.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Metilación de ADN , Ubiquitina-Proteína Ligasas/metabolismo , Regulación Alostérica , Proteínas Potenciadoras de Unión a CCAAT/química , Cromatina/metabolismo , ADN/metabolismo , Células HeLa , Histonas/metabolismo , Humanos , Dominios Proteicos , Ubiquitina-Proteína Ligasas/química
8.
Sci Adv ; 10(13): eadk4423, 2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38536911

RESUMEN

DNA methyltransferase inhibitor (DNMTi) efficacy in solid tumors is limited. Colon cancer cells exposed to DNMTi accumulate lysine-27 trimethylation on histone H3 (H3K27me3). We propose this Enhancer of Zeste Homolog 2 (EZH2)-dependent repressive modification limits DNMTi efficacy. Here, we show that low-dose DNMTi treatment sensitizes colon cancer cells to select EZH2 inhibitors (EZH2is). Integrative epigenomic analysis reveals that DNMTi-induced H3K27me3 accumulates at genomic regions poised with EZH2. Notably, combined EZH2i and DNMTi alters the epigenomic landscape to transcriptionally up-regulate the calcium-induced nuclear factor of activated T cells (NFAT):activating protein 1 (AP-1) signaling pathway. Blocking this pathway limits transcriptional activating effects of these drugs, including transposable element and innate immune response gene expression involved in viral defense. Analysis of primary human colon cancer specimens reveals positive correlations between DNMTi-, innate immune response-, and calcium signaling-associated transcription profiles. Collectively, we show that compensatory EZH2 activity limits DNMTi efficacy in colon cancer and link NFAT:AP-1 signaling to epigenetic therapy-induced viral mimicry.


Asunto(s)
Neoplasias del Colon , Proteína Potenciadora del Homólogo Zeste 2 , Histonas , Humanos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Histonas/metabolismo , Metilación , Transducción de Señal , Factor de Transcripción AP-1/metabolismo
9.
Chembiochem ; 14(16): 2087-90, 2013 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-24106198

RESUMEN

In the blink of the eye: a cascade of two deoxyribozymes was designed for rapid visual detection of bacterial 16S rRNA. The detection limit is 12.5 ng by the naked eye, with the ability to differentiate between closely related pathogenic and nonpathogenic species.


Asunto(s)
Bacterias/genética , Colorimetría , ADN Catalítico/metabolismo , ARN Bacteriano/análisis , 3,3'-Diaminobencidina/química , Secuencia de Bases , Humanos , Mycobacterium tuberculosis/genética , Oligonucleótidos/química , ARN Ribosómico 16S/análisis
10.
Bioorg Med Chem ; 21(7): 1988-91, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23411398

RESUMEN

Inhibitors that covalently damage proteins or nucleic acids offer great potency, but are difficult to rationally design and suffer from poor specificity. Here we outline a general concept for constructing covalent inhibitors, called the two-component covalent inhibitor (TCCI). The approach takes advantage of two ligand analogs equipped with pre-reactive groups. Binding of the analogs to the adjacent sites of a target biopolymer brings the pre-reactive groups in close proximity and causes their interaction followed by covalent damage of the target. In the present study we used light-activated pre-reactive groups to inactivate a DNA polymerase. It was found that the efficiency of a traditional single-component inhibitor was greatly reduced in the presence of a non-target protein, while the TCCI was not significantly affected. Our findings suggest that TCCI approach has advantages in inactivation of biopolymers in complex multi-component systems.


Asunto(s)
Bacteriófago T4/enzimología , Diseño de Fármacos , Inhibidores de la Síntesis del Ácido Nucleico , Nucleótidos de Timina/química , Nucleótidos de Timina/farmacología , Bacteriófago T4/química , Sitios de Unión , ADN Polimerasa Dirigida por ADN/química , ADN Polimerasa Dirigida por ADN/metabolismo , Luz , Unión Proteica
11.
Sci Rep ; 13(1): 377, 2023 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-36611042

RESUMEN

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.


Asunto(s)
Lisina , Proteoma , Humanos , Lisina/metabolismo , Proteoma/metabolismo , Epigenoma , Metilación , Péptidos/metabolismo , Anticuerpos/metabolismo
13.
ACS Omega ; 5(8): 4167-4171, 2020 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-32149246

RESUMEN

We synthesized two dTTP analogues for copper-free "click" chemistry-coupling in the active sites of DNA polymerases. We found that in the presence of both analogues, human immunodeficiency virus (HIV) reverse transcriptase (RT) activity was suppressed by up to 93%. This inhibitory effect was not recovered by an excess amount of primer-template unlike that for a conventional HIV RT inhibitor, azidothymidine. This finding may become the basis for the development of efficient in vivo inhibitors of HIV RT and other DNA polymerases.

14.
iScience ; 21: 773-788, 2019 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-31727574

RESUMEN

Acetylation of the histone variant H2A.Z (H2A.Zac) occurs at active regulatory regions associated with gene expression. Although the Tip60 complex is proposed to acetylate H2A.Z, functional studies suggest additional enzymes are involved. Here, we show that p300 acetylates H2A.Z at multiple lysines. In contrast, we found that although Tip60 does not efficiently acetylate H2A.Z in vitro, genetic inhibition of Tip60 reduces H2A.Zac in cells. Importantly, we found that interaction between the p300-bromodomain and H4 acetylation (H4ac) enhances p300-driven H2A.Zac. Indeed, H2A.Zac and H4ac show high genomic overlap, especially at active promoters. We also reveal unique chromatin features and transcriptional states at enhancers correlating with co-occurrence or exclusivity of H4ac and H2A.Zac. We propose that differential H4 and H2A.Z acetylation signatures can also define the enhancer state. In conclusion, we show both Tip60 and p300 contribute to H2A.Zac and reveal molecular mechanisms of writer/reader crosstalk between H2A.Z and H4 acetylation through p300.

15.
Nat Commun ; 10(1): 2314, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-31127101

RESUMEN

Histone methyltransferase MLL4 is centrally involved in transcriptional regulation and is often mutated in human diseases, including cancer and developmental disorders. MLL4 contains a catalytic SET domain that mono-methylates histone H3K4 and seven PHD fingers of unclear function. Here, we identify the PHD6 finger of MLL4 (MLL4-PHD6) as a selective reader of the epigenetic modification H4K16ac. The solution NMR structure of MLL4-PHD6 in complex with a H4K16ac peptide along with binding and mutational analyses reveal unique mechanistic features underlying recognition of H4K16ac. Genomic studies show that one third of MLL4 chromatin binding sites overlap with H4K16ac-enriched regions in vivo and that MLL4 occupancy in a set of genomic targets depends on the acetyltransferase activity of MOF, a H4K16ac-specific acetyltransferase. The recognition of H4K16ac is conserved in the PHD7 finger of paralogous MLL3. Together, our findings reveal a previously uncharacterized acetyllysine reader and suggest that selective targeting of H4K16ac by MLL4 provides a direct functional link between MLL4, MOF and H4K16 acetylation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Histona Acetiltransferasas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Dedos de Zinc PHD/fisiología , Acetilación , Animales , Sitios de Unión , Cromatina/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/aislamiento & purificación , Técnicas de Inactivación de Genes , Células HEK293 , Histona Acetiltransferasas/genética , N-Metiltransferasa de Histona-Lisina/química , Histonas/química , Humanos , Ratones Transgénicos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Resonancia Magnética Nuclear Biomolecular , Procesamiento Proteico-Postraduccional/fisiología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo
16.
Sci Adv ; 4(11): eaav2623, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30498785

RESUMEN

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.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Proteoma/análisis , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Metilación , Mutación Missense , Especificidad por Sustrato
17.
Science ; 362(6419): 1182-1186, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30523112

RESUMEN

DNA methylation generally functions as a repressive transcriptional signal, but it is also known to activate gene expression. In either case, the downstream factors remain largely unknown. By using comparative interactomics, we isolated proteins in Arabidopsis thaliana that associate with methylated DNA. Two SU(VAR)3-9 homologs, the transcriptional antisilencing factor SUVH1, and SUVH3, were among the methyl reader candidates. SUVH1 and SUVH3 bound methylated DNA in vitro, were associated with euchromatic methylation in vivo, and formed a complex with two DNAJ domain-containing homologs, DNAJ1 and DNAJ2. Ectopic recruitment of DNAJ1 enhanced gene transcription in plants, yeast, and mammals. Thus, the SUVH proteins bind to methylated DNA and recruit the DNAJ proteins to enhance proximal gene expression, thereby counteracting the repressive effects of transposon insertion near genes.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/genética , Metilación de ADN , Regulación de la Expresión Génica de las Plantas , Proteínas del Choque Térmico HSP40/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Metiltransferasas/metabolismo , Transcripción Genética , Arabidopsis/enzimología , Proteínas del Choque Térmico HSP40/química , Dominios Proteicos
18.
J Vis Exp ; (126)2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28809825

RESUMEN

Post-translational modifications (PTMs) on histone proteins are widely studied for their roles in regulating chromatin structure and gene expression. The mass production and distribution of antibodies specific to histone PTMs has greatly facilitated research on these marks. As histone PTM antibodies are key reagents for many chromatin biochemistry applications, rigorous analysis of antibody specificity is necessary for accurate data interpretation and continued progress in the field. This protocol describes an integrated pipeline for the design, fabrication and use of peptide microarrays for profiling the specificity of histone antibodies. The design and analysis aspects of this procedure are facilitated by ArrayNinja, an open-source and interactive software package we recently developed to streamline the customization of microarray print formats. This pipeline has been used to screen a large number of commercially available and widely used histone PTM antibodies, and data generated from these experiments are freely available through an online and expanding Histone Antibody Specificity Database. Beyond histones, the general methodology described herein can be applied broadly to the analysis of PTM-specific antibodies.


Asunto(s)
Especificidad de Anticuerpos , Histonas/inmunología , Análisis por Matrices de Proteínas/instrumentación , Análisis por Matrices de Proteínas/métodos , Anticuerpos/química , Anticuerpos/inmunología , Histonas/química , Histonas/metabolismo , Lisina/metabolismo , Péptidos/química , Péptidos/inmunología , Procesamiento Proteico-Postraduccional , Robótica/instrumentación , Programas Informáticos , Treonina/metabolismo
19.
Artículo en Inglés | MEDLINE | ID: mdl-27826357

RESUMEN

BACKGROUND: The robustness of ChIP-seq datasets is highly dependent upon the antibodies used. Currently, polyclonal antibodies are the standard despite several limitations: They are non-renewable, vary in performance between lots and need to be validated with each new lot. In contrast, monoclonal antibody lots are renewable and provide consistent performance. To increase ChIP-seq standardization, we investigated whether monoclonal antibodies could replace polyclonal antibodies. We compared monoclonal antibodies that target five key histone modifications (H3K4me1, H3K4me3, H3K9me3, H3K27ac and H3K27me3) to their polyclonal counterparts in both human and mouse cells. RESULTS: Overall performance was highly similar for four monoclonal/polyclonal pairs, including when we used two distinct lots of the same monoclonal antibody. In contrast, the binding patterns for H3K27ac differed substantially between polyclonal and monoclonal antibodies. However, this was most likely due to the distinct immunogen used rather than the clonality of the antibody. CONCLUSIONS: Altogether, we found that monoclonal antibodies as a class perform equivalently to polyclonal antibodies for the detection of histone post-translational modifications in both human and mouse. Accordingly, we recommend the use of monoclonal antibodies in ChIP-seq experiments.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos/inmunología , Histonas/metabolismo , Animales , Inmunoprecipitación de Cromatina , ADN/química , ADN/aislamiento & purificación , ADN/metabolismo , Histonas/química , Histonas/inmunología , Humanos , Células K562 , Ratones , Células Madre Embrionarias de Ratones , Mapeo Peptídico , Procesamiento Proteico-Postraduccional , Análisis de Secuencia de ADN
20.
Cell Rep ; 16(12): 3195-3207, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27653685

RESUMEN

MORC3 is linked to inflammatory myopathies and cancer; however, the precise role of MORC3 in normal cell physiology and disease remains poorly understood. Here, we present detailed genetic, biochemical, and structural analyses of MORC3. We demonstrate that MORC3 is significantly upregulated in Down syndrome and that genetic abnormalities in MORC3 are associated with cancer. The CW domain of MORC3 binds to the methylated histone H3K4 tail, and this interaction is essential for recruitment of MORC3 to chromatin and accumulation in nuclear bodies. We show that MORC3 possesses intrinsic ATPase activity that requires DNA, but it is negatively regulated by the CW domain, which interacts with the ATPase domain. Natively linked CW impedes binding of the ATPase domain to DNA, resulting in a decrease in the DNA-stimulated enzymatic activity. Collectively, our studies provide a molecular framework detailing MORC3 functions and suggest that its modulation may contribute to human disease.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Histidina Quinasa/metabolismo , Adenosina Trifosfatasas/química , Células Cultivadas , Cromatina/metabolismo , Proteínas de Unión al ADN/química , Síndrome de Down/genética , Síndrome de Down/metabolismo , Histidina Quinasa/química , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Conformación Proteica , Dominios Proteicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA