Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
PLoS One ; 15(10): e0240829, 2020.
Article En | MEDLINE | ID: mdl-33104722

Histone post-translational modifications (PTMs) create a powerful regulatory mechanism for maintaining chromosomal integrity in cells. Histone acetylation and methylation, the most widely studied histone PTMs, act in concert with chromatin-associated proteins to control access to genetic information during transcription. Alterations in cellular histone PTMs have been linked to disease states and have crucial biomarker and therapeutic potential. Traditional bottom-up mass spectrometry of histones requires large numbers of cells, typically one million or more. However, for some cell subtype-specific studies, it is difficult or impossible to obtain such large numbers of cells and quantification of rare histone PTMs is often unachievable. An established targeted LC-MS/MS method was used to quantify the abundance of histone PTMs from cell lines and primary human specimens. Sample preparation was modified by omitting nuclear isolation and reducing the rounds of histone derivatization to improve detection of histone peptides down to 1,000 cells. In the current study, we developed and validated a quantitative LC-MS/MS approach tailored for a targeted histone assay of 75 histone peptides with as few as 10,000 cells. Furthermore, we were able to detect and quantify 61 histone peptides from just 1,000 primary human stem cells. Detection of 37 histone peptides was possible from 1,000 acute myeloid leukemia patient cells. We anticipate that this revised method can be used in many applications where achieving large cell numbers is challenging, including rare human cell populations.


Histones/genetics , Histones/metabolism , Proteomics/methods , Acetylation , Cell Line , Chromatography, Liquid/methods , Humans , Methylation , Peptides/chemistry , Protein Processing, Post-Translational/genetics , Tandem Mass Spectrometry/methods
2.
J Am Soc Mass Spectrom ; 30(12): 2526-2534, 2019 Dec.
Article En | MEDLINE | ID: mdl-31286445

Histone posttranslational modifications (PTMs) are essential for regulating chromatin and maintaining gene expression throughout cell differentiation. Despite the deep level of understanding of immunophenotypic differentiation pathways in hematopoietic cells, few studies have investigated global levels of histone PTMs required for differentiation and maintenance of these distinct cell types. Here, we describe an approach to couple fluorescence-activated cell sorting (FACS) with targeted mass spectrometry to define global "epi-proteomic" signatures for primary leukocytes. FACS was used to sort closely and distantly related leukocytes from normal human peripheral blood for quantitation of histone PTMs with a multiple reaction monitoring LC-MS/MS method measuring histone PTMs on histones H3 and H4. We validate cell sorting directly into H2SO4 for immediate histone extraction to decrease time and number of steps after FACS to analyze histone PTMs. Relative histone PTM levels vary in T cells across healthy donors, and the majority of PTMs remain stable up to 2 days following initial blood draw. Large differences in the levels of histone PTMs are observed across the mature lymphoid and myeloid lineages, as well as between different types within the same lineage, though no differences are observed in closely related T cell subtypes. The results show a streamlined approach for quantifying global changes in histone PTMs in cell types separated by FACS that is poised for clinical deployment.


Flow Cytometry/methods , Histone Code , Leukocytes/cytology , Tandem Mass Spectrometry/methods , Cells, Cultured , Chromatography, Liquid/methods , Histones/analysis , Humans , Leukocytes/chemistry
3.
Clin Cancer Res ; 25(1): 222-239, 2019 01 01.
Article En | MEDLINE | ID: mdl-30224337

PURPOSE: T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive disease, affecting children and adults. Chemotherapy treatments show high response rates but have debilitating effects and carry risk of relapse. Previous work implicated NOTCH1 and other oncogenes. However, direct inhibition of these pathways affects healthy tissues and cancer alike. Our goal in this work has been to identify enzymes active in T-ALL whose activity could be targeted for therapeutic purposes. EXPERIMENTAL DESIGN: To identify and characterize new NOTCH1 druggable partners in T-ALL, we coupled studies of the NOTCH1 interactome to expression analysis and a series of functional analyses in cell lines, patient samples, and xenograft models. RESULTS: We demonstrate that ubiquitin-specific protease 7 (USP7) interacts with NOTCH1 and controls leukemia growth by stabilizing the levels of NOTCH1 and JMJD3 histone demethylase. USP7 is highly expressed in T-ALL and is transcriptionally regulated by NOTCH1. In turn, USP7 controls NOTCH1 levels through deubiquitination. USP7 binds oncogenic targets and controls gene expression through stabilization of NOTCH1 and JMJD3 and ultimately H3K27me3 changes. We also show that USP7 and NOTCH1 bind T-ALL superenhancers, and inhibition of USP7 leads to a decrease of the transcriptional levels of NOTCH1 targets and significantly blocks T-ALL cell growth in vitro and in vivo. CONCLUSIONS: These results provide a new model for USP7 deubiquitinase activity through recruitment to oncogenic chromatin loci and regulation of both oncogenic transcription factors and chromatin marks to promote leukemia. Our studies also show that targeting USP7 inhibition could be a therapeutic strategy in aggressive leukemia.


Jumonji Domain-Containing Histone Demethylases/genetics , Leukemia, T-Cell/genetics , Receptor, Notch1/genetics , Ubiquitin-Specific Peptidase 7/genetics , Animals , Carcinogenesis/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Genetic Therapy , Humans , Jurkat Cells , Leukemia, T-Cell/pathology , Leukemia, T-Cell/therapy , Mice , Signal Transduction/genetics , Xenograft Model Antitumor Assays
4.
PLoS One ; 13(9): e0203351, 2018.
Article En | MEDLINE | ID: mdl-30192817

Chromatin structure, a key contributor to the regulation of gene expression, is modulated by a broad array of histone post-translational modifications (PTMs). Taken together, these "histone marks" comprise what is often referred to as the "histone code". The quantitative analysis of histone PTMs by mass spectrometry (MS) offers the ability to examine the response of the histone code to physiological signals. However, few studies have examined the stability of histone PTMs through the process of isolating and culturing primary cells. To address this, we used bottom-up, MS-based analysis of histone PTMs in liver, freshly isolated hepatocytes, and cultured hepatocytes from adult male Fisher F344 rats. Correlations between liver, freshly isolated cells, and primary cultures were generally high, with R2 values exceeding 0.9. However, a number of acetylation marks, including those on H2A K9, H2A1 K13, H3 K4, H3 K14, H4 K8, H4 K12 and H4 K16 differed significantly among the three sources. Inducing proliferation of primary adult hepatocytes in culture affected several marks on histones H3.1/3.2 and H4. We conclude that hepatocyte isolation, culturing and cell cycle status all contribute to steady-state changes in the levels of a number of histone PTMs, indicating changes in histone marks that are rapidly induced in response to alterations in the cellular milieu. This has implications for studies aimed at assigning biological significance to histone modifications in tumors versus cancer cells, the developmental behavior of stem cells, and the attribution of changes in histone PTMs to altered cell metabolism.


Histone Code , Liver/metabolism , Animals , Cell Culture Techniques , Cell Cycle , Cell Separation , Cells, Cultured , Hepatocytes/cytology , Hepatocytes/metabolism , Histones/chemistry , Histones/metabolism , Male , Mass Spectrometry , Protein Processing, Post-Translational , Protein Stability , Rats , Rats, Inbred F344
5.
Genes Dev ; 31(19): 2003-2014, 2017 10 01.
Article En | MEDLINE | ID: mdl-29089422

Histone H3 Lys4 (H3K4) methylation is a chromatin feature enriched at gene cis-regulatory sequences such as promoters and enhancers. Here we identify an evolutionarily conserved factor, BRWD2/PHIP, which colocalizes with histone H3K4 methylation genome-wide in human cells, mouse embryonic stem cells, and Drosophila Biochemical analysis of BRWD2 demonstrated an association with the Cullin-4-RING ubiquitin E3 ligase-4 (CRL4) complex, nucleosomes, and chromatin remodelers. BRWD2/PHIP binds directly to H3K4 methylation through a previously unidentified chromatin-binding module related to Royal Family Tudor domains, which we named the CryptoTudor domain. Using CRISPR-Cas9 genetic knockouts, we demonstrate that COMPASS H3K4 methyltransferase family members differentially regulate BRWD2/PHIP chromatin occupancy. Finally, we demonstrate that depletion of the single Drosophila homolog dBRWD3 results in altered gene expression and aberrant patterns of histone H3 Lys27 acetylation at enhancers and promoters, suggesting a cross-talk between these chromatin modifications and transcription through the BRWD protein family.


Drosophila melanogaster/genetics , Gene Expression Regulation , Histones/metabolism , Tudor Domain , Acetylation , Animals , CRISPR-Cas Systems , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Enhancer Elements, Genetic , Epigenesis, Genetic , Gene Knockout Techniques , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Humans , Methylation , Mice , Promoter Regions, Genetic , Protein Binding/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Nat Genet ; 49(11): 1647-1653, 2017 Nov.
Article En | MEDLINE | ID: mdl-28967912

Histone H3 lysine 4 monomethylation (H3K4me1) is an evolutionarily conserved feature of enhancer chromatin catalyzed by the COMPASS-like methyltransferase family, which includes Trr in Drosophila melanogaster and MLL3 (encoded by KMT2C) and MLL4 (encoded by KMT2D) in mammals. Here we demonstrate that Drosophila embryos expressing catalytically deficient Trr eclose and develop to productive adulthood. Parallel experiments with a trr allele that augments enzyme product specificity show that conversion of H3K4me1 at enhancers to H3K4me2 and H3K4me3 is also compatible with life and results in minimal changes in gene expression. Similarly, loss of the catalytic SET domains of MLL3 and MLL4 in mouse embryonic stem cells (mESCs) does not disrupt self-renewal. Drosophila embryos with trr alleles encoding catalytic mutants manifest subtle developmental abnormalities when subjected to temperature stress or altered cohesin levels. Collectively, our findings suggest that animal development can occur in the context of Trr or mammalian COMPASS-like proteins deficient in H3K4 monomethylation activity and point to a possible role for H3K4me1 on cis-regulatory elements in specific settings to fine-tune transcriptional regulation in response to environmental stress.


Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Amino Acid Sequence , Animals , Baculoviridae/genetics , CRISPR-Cas Systems , Chromatin/chemistry , Chromatin/metabolism , Drosophila Proteins/deficiency , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Larva/genetics , Larva/growth & development , Larva/metabolism , Methylation , Mice , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Sf9 Cells , Spodoptera
7.
Elife ; 62017 09 12.
Article En | MEDLINE | ID: mdl-28895528

The repeating subunit of chromatin, the nucleosome, includes two copies of each of the four core histones, and several recent studies have reported that asymmetrically-modified nucleosomes occur at regulatory elements in vivo. To probe the mechanisms by which histone modifications are read out, we designed an obligate pair of H3 heterodimers, termed H3X and H3Y, which we extensively validated genetically and biochemically. Comparing the effects of asymmetric histone tail point mutants with those of symmetric double mutants revealed that a single methylated H3K36 per nucleosome was sufficient to silence cryptic transcription in vivo. We also demonstrate the utility of this system for analysis of histone modification crosstalk, using mass spectrometry to separately identify modifications on each H3 molecule within asymmetric nucleosomes. The ability to generate asymmetric nucleosomes in vivo and in vitro provides a powerful and generalizable tool to probe the mechanisms by which H3 tails are read out by effector proteins in the cell.


Histones/analysis , Nucleosomes/chemistry , Saccharomyces cerevisiae Proteins/analysis , Saccharomyces cerevisiae/chemistry , Histones/genetics , Mass Spectrometry/methods , Protein Processing, Post-Translational , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Synthetic Biology/methods
8.
Nat Med ; 23(4): 493-500, 2017 Apr.
Article En | MEDLINE | ID: mdl-28263307

Diffuse intrinsic pontine glioma (DIPG) is a highly aggressive pediatric brainstem tumor characterized by rapid and uniform patient demise. A heterozygous point mutation of histone H3 occurs in more than 80% of these tumors and results in a lysine-to-methionine substitution (H3K27M). Expression of this histone mutant is accompanied by a reduction in the levels of polycomb repressive complex 2 (PRC2)-mediated H3K27 trimethylation (H3K27me3), and this is hypothesized to be a driving event of DIPG oncogenesis. Despite a major loss of H3K27me3, PRC2 activity is still detected in DIPG cells positive for H3K27M. To investigate the functional roles of H3K27M and PRC2 in DIPG pathogenesis, we profiled the epigenome of H3K27M-mutant DIPG cells and found that H3K27M associates with increased H3K27 acetylation (H3K27ac). In accordance with previous biochemical data, the majority of the heterotypic H3K27M-K27ac nucleosomes colocalize with bromodomain proteins at the loci of actively transcribed genes, whereas PRC2 is excluded from these regions; this suggests that H3K27M does not sequester PRC2 on chromatin. Residual PRC2 activity is required to maintain DIPG proliferative potential, by repressing neuronal differentiation and function. Finally, to examine the therapeutic potential of blocking the recruitment of bromodomain proteins by heterotypic H3K27M-K27ac nucleosomes in DIPG cells, we performed treatments in vivo with BET bromodomain inhibitors and demonstrate that they efficiently inhibit tumor progression, thus identifying this class of compounds as potential therapeutics in DIPG.


Brain Stem Neoplasms/genetics , Chromatin/metabolism , Gene Expression Regulation, Neoplastic/genetics , Glioma/genetics , Histone Code/genetics , Histones/genetics , Nucleosomes/metabolism , Polycomb Repressive Complex 2/metabolism , RNA-Binding Proteins/metabolism , Acetylation/drug effects , Animals , Azepines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chromatin/drug effects , Epigenomics , Gene Expression Regulation, Neoplastic/drug effects , Histone Code/drug effects , Histones/drug effects , Humans , Methylation/drug effects , Mice , Molecular Targeted Therapy , Mutation , Neurogenesis/drug effects , Neurogenesis/genetics , Nucleosomes/drug effects , Polycomb Repressive Complex 2/drug effects , Protein Transport , RNA-Binding Proteins/antagonists & inhibitors , Triazoles/pharmacology , Xenograft Model Antitumor Assays
...