Histone Modification
Judith Erkmann1 (judith_erkmann at yahoo dot com), Nikolaos Parisis2 (nnparisis at gmail dot com)
1 University of Massachusetts Medical School, United States. 2 Université Paris Diderot, Institut Jacques Monod, Paris, France
DOI
//dx.doi.org/10.13070/mm.en.1.92
Date
last modified : 2022-11-05; original version : 2011-09-28
Cite as
MATER METHODS 2011;1:92
Abstract

A detailed review of methods in histone modification research and a summary of histone antibodies cited among the over 60,000 formal publications Labome has surveyed in its Validated Antibody Database.

Background

Histone proteins are subject to a variety of posttranslational modifications (PTMs), many of which (i.e., methylation, acetylation, ubiquitylation and SUMOylation) occur at lysines [4], or citrullination at arginine [5], or serotonylation at glutamine [6]. While only a handful of modification sites have been identified in the buried histone-fold domains, PTMs on the flexible N-terminal tail regions are quite common. Many histone modifications mediate functionally significant changes in chromatin structure and do so either by altering chromatin structure/dynamics directly or through the recruitment of histone modifiers and/or nucleosome remodeling complexes [4, 7]. Histones are the components of nucleosomes, which turn over several times within each cell cycle, suggesting that histone modifications are rapidly erased by nucleosome turnover. This turnover of modified nucleosomes may propagate epigenetic changes and determine the valency of methylation [8].

Histone PTMs have been found to affect a host of chromatin-based reactions, including transcription, heterochromatic gene silencing and genome stability [9-12]. For example, C Weber et al showed that Kdm6b, a histone demethylase, mediated the temperature-dependent sex determination in reptiles [13]. Modifications implicated in gene expression are especially significant as they have the potential to influence whole transcription programs. H3K4me3 initiates transcription, H3K27ac promotes gene transcription, while H3K27me3 represses the transcription of developmental genes [14, 15], although such a characterization remains debatable [16]. Defects in histone PTM metabolism have been linked to misregulated gene expression in various in vitro models and, in some cases, also correlate with human disease, as has been demonstrated for immunodeficiency disorders and a variety of human cancers [17-22]. Thus, how histone markers are regulated and influence the association of PTM-specific binding proteins will continue to be an area of significant investigation [7, 23-25].

Determining the function of histone PTMs often involves investigating the modification's abundance and interacting partners. The methods described here address these areas and include summaries and references for protocols detailing histone purification methods, the production of recombinant site-specifically modified histones, peptide-based systems to characterize PTM-binding proteins and histone modification-specific antibodies, in vivo histone modifications imaging, and different methods for analyzing chromatin immunoprecipitation samples. Histones and their modifications such as those with circulating cell-free nucleosomes have been proposed as diagnostic tools for cancers or kidney transplant rejection [26].

Methods for Histone Modification Research
Cell lysate

For detection of histone modifications by Western blotting, it often is possible to use whole-cell lysates made by extraction with SDS Laemmli sample buffer. In the case of animal cell lines, cells collected by centrifugation can be directly resuspended in sample buffer and boiled for loading [27, 28] ; note, however, that some protocols additionally recommend sonicating the samples after the extraction step. Protein extracts of fungi can be prepared the same way with the exception of an optional alkali pre-treatment step [29]. This step can be omitted, however, if it is experimentally necessary to minimize sample handling time, so long as the omission is noted and samples for follow-up experiments are treated the same way. Following extraction, the sample's insoluble fraction is removed by centrifugation, leaving behind the soluble whole cell extract in the supernatant.

Enriched histone fraction

For some applications, it is necessary to examine either histone-enriched fractions or purified histone proteins; examples of an enriched sample would be isolated nuclei or a crude chromatin preparation. The isolation of nuclei from cultured metazoan cells or yeast is very straightforward and requires essentially three steps: hypotonic swelling (after prior cell wall digestion in the case of yeast), cell membrane lysis by mechanical shearing (i.e., breakage with a Dounce homogenizer or mild agitation on a rotator), and isolation of nuclei by centrifugation [30, 31] (Figure 1A). The isolation of crude chromatin is also quite simple and includes only a detergent lysis step followed by the sedimentation of chromatin by centrifugation [32-34] (Figure 1B).

Histone purification

Several of the available histone purification protocols are excellent and easy to follow [31]. In the method described here, histones are extracted from nuclei using a dilute sulfuric acid solution and then purified by column chromatography (Figure 1C). The beauty of this protocol is that nucleic acids and many of the non-histone proteins can be readily removed by centrifugation because of their insolubility at acidic pH. The soluble histone fraction is then precipitated with trichloroacetic acid (TCA) and, if desired, purified over a reversed-phase HPLC column. The histones at this point can be used in a variety of applications, including Western blotting and mass spectrometry. Please note that another publication [31] also describes an alternate, high-salt histone extraction method.

Histone Modification figure 1
Figure 1. Protocols for isolating intact nuclei (A), preparing crude chromatin fractions (B), and purifying histones (C).

In addition, the technique of Chromatin Affinity Purification with Mass Spectrometry (ChAP-MS) can be used for affinity purification of ~1 kb regions of chromatin for analysis of histone PTMs [35].

Histone PTM detection by antibody recognition

Histone PTMs are generally detected through the antibodies. Phospho-histone H3 (ser10) is commonly used as a marker for mitosis [14, 36]. M Coolen et al identified mitotic killifish cells through phospho-histone H3 staining [36]. Rhodes JDP et al stained mouse ESCs with CST phospho-histone H3 (ser10) mouse mAb ( 9706) to detect mitotic cells [37] and Y Shwartz et al used CST rabbit monoclonal antibody 3377 [38]. Butler AA et al detected histone methylation through MilliporeSigma antibodies against H3K9me2 ( 07-441), H3K27me3 ( 07-449) and H3K4me3 ( 04-745) in Western blots [39] ; Batie M et al used Cell Signaling Technology antibodies against H3K4me3 (9751), H3K9me3 (9754 / 13969), H3K27me3 (9733), H3K36me2 (2901) and H3K36me3 (4909) in immunoblotting Hela and HFF cell lysates [40]. The quality and specificity of anti-PTM antibodies should be carefully evaluated prior to experimental application. Concerns to be addressed include cross-reactivity with alternate histone modification sites, recognition of the unmodified (recombinant) protein and cross-reactivity with other nuclear species. The procedures for this type of evaluation are very straightforward and involve either Western blotting of nuclear extract preparations against recombinant histones or immunoblotting an array of modified and unmodified peptides spotted on nitrocellulose membrane. Table 1 lists antibodies against histones and histone-related products cited among the formal articles Labome has surveyed for Validated Antibody Database.

SymProteinTop three suppliers
ENL MLLT1 super elongation complex subunit Abnova H00004298-M01 (2)
HAT1 histone acetyltransferase 1 Santa Cruz Biotechnology sc-376200 (2), Abcam ab194296 (1)
HDAC1 histone deacetylase 1 Cell Signaling Technology 5356 (41), Santa Cruz Biotechnology sc-81598 (11), Abcam ab109411 (4)
HDAC10 histone deacetylase 10 Santa Cruz Biotechnology sc-376121 (1)
HDAC11 histone deacetylase 11 Santa Cruz Biotechnology sc-390737 (1)
HDAC2 histone deacetylase 2 Cell Signaling Technology 5113 (20), Abcam ab32117 (13), Santa Cruz Biotechnology sc-9959 (10)
HDAC3 histone deacetylase 3 Cell Signaling Technology 3949 (11), Abcam ab32369 (8), Santa Cruz Biotechnology sc-17795 (4)
HDAC4 histone deacetylase 4 Cell Signaling Technology 5392 (8), Santa Cruz Biotechnology sc-46672 (3)
HDAC5 histone deacetylase 5 Santa Cruz Biotechnology sc-133225 (7)
HDAC6 histone deacetylase 6 Cell Signaling Technology 7612 (10), Santa Cruz Biotechnology sc-28386 (3), Abcam ab253033 (1)
HDAC7 histone deacetylase 7 Cell Signaling Technology 3443 (7), Santa Cruz Biotechnology sc-74563 (2), Abcam ab166911 (2)
HDAC8 histone deacetylase 8 Santa Cruz Biotechnology sc-17778 (2), Abcam ab187139 (2)
HDAC9 histone deacetylase 9 Abcam ab109446 (5), Santa Cruz Biotechnology sc-398003 (4)
HIRA histone cell cycle regulator Active Motif 39557 (4), Abcam ab129169 (1)
HIST1H1C histone cluster 1 H1 family member c Santa Cruz Biotechnology sc-8030 (28)
HIST1H2AA histone cluster 1 H2A family member a Cell Signaling Technology 9718 (203)
HIST1H2AC histone cluster 1 H2A family member c Abnova H00008334-M01 (1)
HIST1H2AD histone cluster 1 H2A family member d Santa Cruz Biotechnology sc-130356 (2)
HIST1H2AG histone cluster 1 H2A family member g Cell Signaling Technology 3636 (6)
HIST1H2AI histone cluster 1 H2A family member i Cell Signaling Technology 3636 (6)
HIST1H2AK histone cluster 1 H2A family member k Cell Signaling Technology 3636 (6)
HIST1H2AL histone cluster 1 H2A family member l Cell Signaling Technology 3636 (6)
HIST1H2AM histone cluster 1 H2A family member m Cell Signaling Technology 3636 (6)
HIST1H2BB histone cluster 1 H2B family member b Cell Signaling Technology 5546 (11)
HIST1H2BO histone cluster 1 H2B family member o Cell Signaling Technology 2934 (7), MédiMabs MM-0029 (5), Active Motif 39623 (2)
HIST1H3A histone cluster 1 H3 family member a Cell Signaling Technology 9733 (115), Abcam ab1012 (28), MilliporeSigma H9908 (15)
HIST1H3B histone cluster 1 H3 family member b Cell Signaling Technology 9733 (115), MilliporeSigma H9908 (15), Abcam ab12209 (10)
HIST1H3C histone cluster 1 H3 family member c Cell Signaling Technology 9733 (115), Abcam ab12209 (10), Active Motif 61251 (5)
HIST1H3D histone cluster 1 H3 family member d Abcam ab1220 (139), Cell Signaling Technology 9733 (115), MilliporeSigma H9908 (15)
HIST1H3E histone cluster 1 H3 family member e Cell Signaling Technology 9733 (115), MilliporeSigma H9908 (15), Abcam ab12209 (10)
HIST1H3F histone cluster 1 H3 family member f Cell Signaling Technology 9733 (115), Abcam ab10799 (18), MilliporeSigma H9908 (15)
HIST1H3G histone cluster 1 H3 family member g Cell Signaling Technology 9733 (115), Abcam ab12209 (10), Invitrogen AHO1432 (2)
HIST1H3H histone cluster 1 H3 family member h Cell Signaling Technology 9733 (115), MilliporeSigma H9908 (15), Abcam ab12209 (10)
HIST1H3I histone cluster 1 H3 family member i Cell Signaling Technology 9733 (115), MilliporeSigma H9908 (15), Abcam ab12209 (10)
HIST1H3J histone cluster 1 H3 family member j Cell Signaling Technology 9733 (115), Abcam ab12209 (10)
HIST1H4A histone cluster 1 H4 family member a Cell Signaling Technology 2935 (7), Invitrogen MA5-14816 (1), Active Motif 61529 (1)
HIST1H4B histone cluster 1 H4 family member b Cell Signaling Technology 2935 (7), Abcam ab51997 (6), Invitrogen MA5-14816 (1)
HIST1H4C histone cluster 1 H4 family member c Cell Signaling Technology 2935 (7), Invitrogen MA5-14816 (1)
HIST1H4D histone cluster 1 H4 family member d Cell Signaling Technology 2935 (7), Invitrogen MA5-15150 (1), Abcam ab197515 (1)
HIST1H4E histone cluster 1 H4 family member e Abcam ab109463 (8), Cell Signaling Technology 2935 (7), Invitrogen MA5-14816 (1)
HIST1H4F histone cluster 1 H4 family member f Cell Signaling Technology 2935 (7), Abcam ab197515 (1)
HIST1H4H histone cluster 1 H4 family member h Cell Signaling Technology 2935 (7)
HIST1H4I histone cluster 1 H4 family member i Cell Signaling Technology 2935 (7), Invitrogen MA5-14816 (1), Abcam ab197515 (1)
HIST1H4J histone cluster 1 H4 family member j Abcam ab31830 (9), Cell Signaling Technology 2935 (7)
HIST1H4K histone cluster 1 H4 family member k Cell Signaling Technology 2935 (7), Santa Cruz Biotechnology sc-25260 (3), Abcam ab197515 (1)
HIST1H4L histone cluster 1 H4 family member l Cell Signaling Technology 2935 (7), Invitrogen MA5-14816 (1)
HIST2H2AC histone cluster 2 H2A family member c Abcam ab45152 (3)
HIST2H2BF histone cluster 2 H2B family member f Abcam ab40886 (6)
HIST2H3A histone cluster 2 H3 family member a Invitrogen AHO1432 (2)
HIST2H3C histone cluster 2 H3 family member c Novus Biologicals NBP1-30141 (4), Invitrogen AHO1432 (2), Active Motif 61623 (1)
HIST2H3D histone cluster 2 H3 family member d MilliporeSigma H9908 (15)
HIST2H4A histone cluster 2 H4 family member a Cell Signaling Technology 2935 (7), Santa Cruz Biotechnology sc-134216 (2), Invitrogen MA5-14816 (1)
HIST2H4B histone cluster 2 H4 family member b Cell Signaling Technology 2935 (7), Invitrogen MA5-14816 (1), Abcam ab197515 (1)
HIST3H2A histone cluster 3 H2A Enzo Life Sciences ADI-KAM-CC255-D (1)
HIST3H3 histone cluster 3 H3 Abcam ab12209 (10), BioLegend 641005 (1), Santa Cruz Biotechnology sc-518011 (1)
HIST4H4 histone cluster 4 H4 Cell Signaling Technology 2935 (7), Active Motif 39671 (4), Abcam ab17036 (2)
KDM1A lysine demethylase 1A Cell Signaling Technology 2184 (14), Abcam ab129195 (4), Santa Cruz Biotechnology sc-271720 (2)
KDM1B lysine demethylase 1B Abcam ab193080 (1)
KDM3A lysine demethylase 3A Santa Cruz Biotechnology sc-376608 (2), ProMab 30134 (1)
KDM3B lysine demethylase 3B Cell Signaling Technology 5377 (1)
KDM4A lysine demethylase 4A Cell Signaling Technology 3393 (3), Neuromab 75-189 (2), Abcam ab191433 (1)
KDM4B lysine demethylase 4B Cell Signaling Technology 8639 (4)
KDM5A lysine demethylase 5A Cell Signaling Technology 3876 (4), Abcam ab194286 (1)
KDM5B lysine demethylase 5B Bio-Rad MCA4340Z (1)
KDM5C lysine demethylase 5C Cell Signaling Technology 5361 (1)
KDM6A lysine demethylase 6A Cell Signaling Technology 33510 (6), Santa Cruz Biotechnology sc-514859 (1)
Table 1. Histones and histone-related proteins and top cited antibodies against them among the over 60,000 formal publications in Validated Antibody Database. The most cited monoclonal antibody from each supplier is listed.
Anti-PTM histone antibody quality

Recent initiatives to address anti-PTM histone antibody quality issue have resulted in the characterization of more than 200 antibodies against 57 different histone modifications [41]. About 20% of antibodies were deemed not effective. The supplementary data to this report catalogues the performance of the different antibodies tested in dot blots, Western blots (across different species), and chromatin immunoprecipitation (ChIP) assays. Xia W et al used CST H3K27me3 antibody ( 9733) for CUT&RUN and immunocytochemistry and Active motif H3K27ac antibody ( 39133) on human oocytes or embryos [15].

A new peptide array application, the MODified™ Histone Peptide Array, has recently become available for the analysis of the binding specificity of histone modification-specific antibodies [42]. Additionally, a Histone Antibody Specificity Database has been created [43]. The database catalogs commercially available histone antibodies and their behavior in peptide microarray-based experiments.

Recombinant site-specifically modified histones

One method for producing site-specifically modified histones involves the ligation of protein fragment in vitro [44-48] (Figure 2A). The premise of this method is to chemically ligate a synthetic, modified peptide (corresponding to either the very N- or C-terminal end of the protein) to a recombinant fragment containing the complementary part of the protein, followed by purification of the full-length ligation product [49]. A related method, published more recently, describes the use of native chemical ligation to produce a fully synthetic modified histone by the sequential addition of synthetic peptides corresponding to consecutive parts of the protein [50].

Histone Modification figure 2
Figure 2. Production of site-specifically modified histones by expressed protein ligation (A) and direct incorporation in vivo (B).

Besides by chemical ligation, lysine-acetylated histones can also be produced by direct incorporation in vivo using E.coli that have been genetically engineered to incorporate acetyl-lysine at UAG codons [51] (Figure 2B). The premise of this system is to introduce into E. coli an M. barkeri pyrrolysyl tRNA synthetase variant that has been sequence-optimized to charge its cognate tRNACUA with acetyl-lysine. Thus, to produce a site-specifically acetylated histone, one would need only to add to the strain a construct containing a UAG codon at the desired modification site.

Characterization of histone PTM binding partners - co-immunoprecipitation (CoIP)/pulldown experiments

In cases where it is of interest to study the interaction of a protein with endogenous histones, it is necessary to first convert the chromatin to mononucleosome-sized pieces by digestion with micrococcal nuclease (MNase). The protein of interest can then be immunoprecipitated from the soluble chromatin fraction and evaluated for association with the core histones and different histone modifications by Western blotting [52, 53]. This experiment can also be done as a pulldown assay using a recombinant bait protein that has been incubated in the solubilized nucleosome fraction and then purified [54]. If it is obvious that a particular modification is enriched in complexes containing the target protein, it may then be of interest to characterize the interaction in binding assays using modified peptides (see below). In cases where no such preference is determined, yet it is still of interest to know whether an interaction is modification dependent, relative affinities for modified versus unmodified histones can be determined in binding assays using native and recombinant histones [55].

One common method of purifying native histones from tissue culture cells is to produce oligonucleosome-sized pieces of chromatin by either limited digestion with MNase or mechanical shearing, followed by chromatography of the fragments on a hydroxyapatite column and elution at high salt [56, 57] (Figure 3A). Recombinant histones are insoluble when overproduced as monomers; however, it is possible to recover the overexpressed proteins from inclusion bodies [58, 59] (Figure 3B). To start, preparations of inclusion bodies are made from bacterial cultures individually overexpressing each histone. The histones are then extracted from the inclusion bodies, purified over sequential ion-exchange resins, and eluted using a linear salt gradient. To generate histone octamers, equimolar amounts of H3, H4, H2A and H2B are first unfolded, combined, refolded by dialysis, and then purified over a sizing column.

Histone Modification figure 3
Figure 3. Purification schemes for isolating native (A) and recombinant histones (B).
Characterization of histone PTM binding partners - peptide pulldown assays

The modification preferences of a protein can be determined by comparing the relative binding affinities of the protein for differentially modified peptides. This can be accomplished in a few different ways. In one format, bead-immobilized peptides are used to pull down a test protein - using either recombinant protein or a nuclear extract – and the relative recoveries of the protein are determined by Western blotting [60-62] (Figure 4A). The principle of short peptides as bait molecules has also been applied in unbiased experiments to discover previously unknown histone PTM-binding proteins [53, 54, 63, 64]. In this assay, binding proteins from a nuclear extract are identified on the basis of enrichment on the modified peptide relative to an unmodified peptide or a peptide carrying the same modification at a different amino acid.

Characterization of histone PTM binding partners - peptide microarrays

Also described in the literature are array-based methods capable of screening the interaction of probe proteins with multiple peptides simultaneously (Figure 4B). For such experiments, the protein of interest is incubated over the surface of a peptide microarray, which is essentially a streptavidin-coated slide spotted with different biotinylated peptides, and the protein peptide complexes are visualized by detection with a fluorophore-conjugated antibody and an array scanner [65-67]. Studies using an inverse setup, i.e., protein arrays incubated with fluorescently-labeled peptide, have been described as well [68].

Histone Modification figure 4
Figure 4. Characterization of histone modifications using peptide-based pulldown assays (A) and peptide microarrays (B).
Determination of the genomic loci for specific PTM - chromatin immunoprecipitation

Genomic sites that are enriched for a particular PTM can be characterized by immunoprecipitating chromatin fragments containing the mark of interest and then quantifying the relative proportion of different loci that the PTM is associated with (Figure 5). Detailed ChIP protocols are available [69-71]. The first step of most ChIP protocols is to treat cells with formaldehyde to "freeze" the position of chromatin-associated proteins by crosslinking. Whereas cells of metazoan origin are then lysed directly, yeast cells must first undergo cell wall breakage, which can be accomplished by either mechanical shearing or enzymatic digestion. Depending on the nucleotide resolution desired at detection, chromatin is cut into short pieces in one of two ways: shearing by sonication to yield fragments of between 200 and 500 bp or digestion with MNase down to individual nucleosomes. Immunoprecipitation of the extracts is performed the same way as in a conventional IP experiment except that after the elution step, the eluate is incubated overnight at 65°C to reverse cross-links. On the following day, the samples are treated with Proteinase K and then phenol/chloroform extracted to recover the co-precipitating DNA fragments.

Histone Modification figure 5
Figure 5. Chromatin immunoprecipitation (A) results in the isolation of co-purifying DNA fragments, whose sequence and enrichment can be analyzed by PCR (B), ChIP-chip (C), and/or Chip-seq (D).

A variation of the ChIP procedure, ZipChip (Figure 6), was recently designed by Harmeyer et al [1]. The method facilitates the detection of histone modifications that are more difficult to identify, while reduces the procedure time. Moreover, it allows for the screening of multiple loci. Another variation of CHiP is CUT&RUN [15, 72].

Determination of the genomic loci for specific PTM - CHiP detection - PCR

If it is of interest to analyze PTM levels at only a handful of loci, this can be accomplished by either real-time PCR or quantification of PCR products on an ethidium-stained gel (Figure 5B). Briefly, the loci to be analyzed are amplified from dilutions of the IP and input fractions and compared, ideally, against these fractions from a parallel IP of the histone the PTM is attached to. To determine whether the PTM is enriched at a particular location, the normalized PTM/histone ratios should be compared with those at a region nearby that the PTM is not predicted to be associated with. For example, Butler AA et al immunoprecipitated formaldehyde-treated N2a cell chromatins with an H3K9me2 antibody and assayed the c-Fos gene promoter through RT-PCR [39].

Histone Modification figure 6
Figure 6. ZipChip procedure diagram as in [1].
Determination of the genomic loci for specific PTM - CHiP detection - ChIP-chip

Microarray-based methods enable the analysis of histone modification enrichment at large numbers of loci simultaneously. The microarray itself is a coated glass slide affixed with different oligonucleotides – ranging in number from the tens of thousands to the tens of millions. Sites where a protein is enriched are determined by co-hybridizing to the array fluorescently labeled DNAs derived from the IP and input fractions and comparing the normalized IP/input intensities across the array (Figure 5C).

The first steps of preparing DNAs for microarray analysis are to amplify the DNA from each test (IP) and reference (input) sample and differentially label the amplification products with either of two fluorophores. DNA amplification can be accomplished in several ways, and the methods that are PCR based begin with the addition of primer binding sites to the DNA ends. This part can be done by either ligating the DNAs to short linkers of a known sequence [73] or performing an initial two rounds of annealing and extension using primers that have degenerate 3' sequence and known 5' sequence [69, 74]. The end-tagged products are then PCR amplified for several rounds using primers that recognize the added linker sequence.

Another widely used method for DNA amplification involves the conversion of DNAs into transcription templates, followed by linear amplification of the products by transcription with T7 RNA polymerase [75]. To start, short polyT tails are added to the DNA 3' ends to create a priming site for the first-strand synthesis by Klenow. The oligo used for priming contains a stretch of A's at the 3' end of the minimal T7 RNA polymerase promoter sequence, which enables subsequent amplification of the first-strand products by in vitro transcription.

Samples that have been amplified by PCR can be fluorescently labeled in one of two ways: (1) by incorporating a fluorophore-modified dNTP during the final set of PCR cycles or (2) indirectly, using an amine-modified dNTP that can be dye coupled later [76]. The same labeling principles apply to products amplified by transcription, except that the modified nucleotides are incorporated during a final reverse transcription step [76]. When preparing samples for labeling, it is important also to label an appropriate reference sample that the test sample can be co-hybridized with. Thus, the labelings are set up such that one of the two samples is labeled with Cy3 and the other with Cy5. After the final amplification or post-labeling step, the samples are purified, and the labeled test and reference samples are mixed and prepared for hybridization.

Determination of the genomic loci for specific PTM - CHiP detection - ChIP-seq

Large-scale enrichment analysis can also be performed using a variety of massively parallel DNA sequencing methods. Such methods make possible the parallel sequencing of millions of DNA molecules in real time. The majority of ChIP-seq studies published so far were done using the "sequencing by synthesis" platform by Illumina. The premise of this platform is to perform parallel sequencing of millions of clonal DNA clusters on the surface of a flow-cell (Figure 5D).

ChIP-seq samples are prepared by ligating the IP'd DNAs to oligonucleotide adapter molecules, after which the ligation products (in some protocols) are amplified for several rounds by PCR and then purified [77, 78]. Samples are then injected into a flow cell whose surface is coated with oligonucleotides complementary to the ligation product adapter sequences. The density of the tethered oligonucleotides is such that, during the amplification steps, the DNAs remain spatially close to the parent template, as the newly synthesized molecules originate from primers that are attached to the flow-cell surface adjacently. The sequencing phase of the protocol is accomplished by single-base extension using nucleotides that are fluorescently labeled and can terminate elongation reversibly. The sequencing reaction therefore proceeds as follows: a labeled nucleotide is added to the free 3' end, after which elongation pauses to enable detection of the incorporated nucleotide. Subsequently, the terminator group is cleaved to permit the addition of the next nucleotide. Nucleotide extension is iterated for several more cycles, which typically results in read-lengths in the 40-bp range. For an in-depth review of next-generation sequencing methods, and comparison with microarray-based approaches, please see [79-82].

Bias in ChIP-seq can be introduced due to the chromatin state [83]. Open chromatin regions often lead to false positive, and algorithmic adjustment should be made.

ChIP-seq proved very useful for the detection of the presence or absence binding sites. Determining any changes in the binding intensity caused by different biological condition would be more informative. Chen et al have recently developed a statistical method, ChIPComp, to perform a quantitative comparison of multiple ChIP-seq datasets in order to detect differential protein binding or histone modification [84]. In this method, read counts from all datasets are compared, and peaks are selected to generate a set of candidate regions, which are further analyzed via an R software package freely available at: http://web1.sph.emory.edu/users/hwu30/software/ChIPComp.html. Computational workflows for differential binding analysis are also emerging [85].

Quantitative information on the level of histone modification at any particular chromosomal loci can now be obtained experimentally. Van Galen et al suggest a quantitative multiplexed ChIP-seq technology, Mint-Chip-seq, to profile chromatin quantitatively [86]. They map the relative levels of multiple histone modifications across multiple samples, while monitoring the dynamic changes of the chromatin following different cellular treatments, both at global and locus-specific levels. A detailed protocol as used in [86], as well as a further optimized version are available online from the Bernstein laboratory: http://bernstein.mgh.harvard.edu/resources/.

Determination of specific gene loci for a specific PTM - PLA assay

A method has been developed to identify specific histone PTM for a specific gene in a particular cell type in histological sections [87]. The cell type is marked through cell-specific marker with an antibody. The gene of interest is detected through in situ hybridization with a biotin-conjugated probe, and histone with specific PTM is labeled through a histone modification-specific antibody. Proximity ligation assay [88] is used to identify the co-location of the biotin label and histone antibody. The method enables detection at the single cell level.

Histone PTM detection by mass spectrometry (MS)

Mass spectrometry is a technique that proved effective in identifying and quantifying histone PTMs and their binding proteins, beyond the limitations of specific antibodies. The method has proved useful in the study of host-pathogen interactions [89], epigenetics-associated diseases [90], as well as in the analysis of clinical samples [91]. Histone PTM analysis can be done either by using intact histones (the top-down approach), intact histone tails (the middle-down approach) or shorter fragments (the bottom-up approach) with equal reliability [92, 93]. These approaches have been previously described [90, 94], while a new workflow for analysis by the bottom-up approach became available as of 2016 [95].

Apart from the identification of novel PTMs, the technique was shown to be useful in the analysis of the histone modification dynamics during the cell cycle [96]. Meharena et al used Global Chromatin Profiling (GCP) assay, an approach of quantitative targeted mass spectrometry with spiked-in internal control peptides, to quantify changes of histone H3 modifications [97]. Comprehensive reviews of the most influential PTMs, PTM purification methods, the fundamentals of PTMs-related proteomics experiments have recently been published [98, 99], and include suggestions for accurate interpretation of data in the context of the biological processes that are being studied.

Histone Modification figure 7
Figure 7. A. Schematic diagram of the split-reporter complementation system; protein-protein interactions between domains A and B lead to the formation of an integral, functional luciferase B. RLuc system for H3-methylation imaging; Chr – histone chromodomain; K9 – Lys9; Met-K9 – methylated Lys9.
Histone PTM imaging

Post-translational modifications of histone proteins regulate gene expression during development, as well as the pathogenesis of cellular diseases. Several techniques for in vitro and in vivo imaging of PT-modified histones are known. Most of them are based on the use of a split-reporter complementation system (Figure 7A). Most recently, the potential of the Renilla luciferase (RLuc) split reporter complementation system was extended to image histone 3 methylation [100] (Figure 7B) . In addition, Kanno et al [101], Lin et al [102] and others [103] used the fluorescence resonance energy transfer (FRET) method to image histone modifications in live cells. Histone modifications have also been tracked with the help of fluorescent modification-specific antibodies [104].

Histone Modification figure 8
Figure 8. Overview of the EpiTOF platform. Adapted from [2].
EpiTOF

EpiTOF (epigenetic landscape profiling using cytometry by time-of-flight) is a method that measures the overall cellular levels of chromatin marks at a single-cell level and in high-throughput format (Figure 8). The authors used it to measure the levels of 8 classes of histone PTMs and 4 histone variants in 22 subsets of immune cells [2].

The key steps of the procedure are the following: PBMCs (isolated from healthy volunteers and cryopreserved) were labelled with lanthanide-labelled antibodies against chromatin marks and in the presence of Fc receptor blocker. This was followed by extracellular marker staining, fixation in 1.6% paraformaldehyde (PFA), permeabilisation and mass-tag sample barcoding was performed. After an overnight incubation with intracellular antibodies in CyTOF buffer containing the Fc receptor blocker, they were stained with 250 nM 191/193Ir DNA intercalator. Cells were resuspended in ddH2O containing four element calibration beads and the final cell suspension was analysed on CyTOF2 (Fluidigm) in Stanford Shared FACS Facility while raw data were analysed with FlowJo software following a dedicated gating hierarchy. Single-cell signal from chromatin markers was normalised to basal levels of histone H3 and H4 proteins using a multivariate linear regression model [2] :

Mi,j = β0 + β1H3i + β2H4i

where H3 and H4 represent the raw values for the respective histone proteins in cell I; and Mi,j is the raw value for a given chromatin mark j in cell i across all cell types and subjects. The normalized chromatin score Si,j was defined as the residual of the regression, corresponding to:

Si,j = Mi,j–(Mi,j)^

where (Mi,j)^ indicates the best fit of the multivariate regression model [2].

The key findings of the study using 40 lanthanide-labelled antibodies against chromatin marks. showed that the chromatin profiles of immune cells is distinct in each cell type indicative of the hematopoietic lineages from which individual populations are derived. Interestingly, a signature of chromatin marks could predict the identity of immune cells.

In conclusion, EpiTOF is a versatile method with the ability to detect different immunophenotypic markers in immune single cells or any other sample with single cells in suspension derived from solid tissues and detect chromatin dysregulation in a variety of human pathological conditions.

Acetylated Histones

Protein acetylation is the addition of an acetyl group (CH3CO) on the ε-amino group of lysine residues by substituting an active hydrogen atom. Histones are heavily acetylated and the reaction is catalysed by histone acetyltransferases (HATs) using the metabolite acetyl-coenzyme A (acetyl CoA) as a substrate and it is reversed by deacetylases (HDAC) [105]. ENL protein, a chromatin reader, interacts with acetylated histoness and recruits/stabilizes the transcriptional machinery to increate gene expression [106].

Histone Modification figure 9
Figure 9. Lysine acetylation.

HATs comprise two groups: type A, HATs localised in the nucleus acetylating chromatin, and type B, HATs localised in the cytoplasm acetylating nascent histone proteins [107] ; and 5 families of which the p300/CBP is the most prevalent [108]. HDACs are divided into 4 classes based on their sequence similarity, Class I, II, III, IV [109].

While lysine is a positively charged amino acid, acetylation removes the charge thereby reducing the affinity to the negatively charged DNA molecule. This usually results in a more relaxed chromatin structure and is associated with chromatin loosening and increased gene transcription [110]. Besides, acetylated lysines can promote or inhibit the binding of transcriptional regulators [111]. These regulated acetylation and deacetylation cycles control chromatin reorganisation in response to various signals and regulate transcription, DNA repair and DNA replication [4].

Detection of acetylated lysines is achieved by mass spectrometry and acetylation-specific antibodies: The substitution of the hydrogen group by the acetyl group causes a +42.01056 Da mass shift which is easily detectable by mass spectrometers. As histones are lysine-rich, a couple of additional steps can improve the identification of acetylated lysines. This involves the chemical propionylation of unmodified (and monomethylated) lysines before protein digestion inhibiting cleavage by trypsin thereby allowing the generation of larger peptides [112]. Besides, trypsin does not cleave after acetylated lysines. Middle-down proteomics strategies can also be valuable [113].

Furthermore, anti-acetyl antibodies have been produced in animals immunizing with synthetic histone peptides bearing an acetylated lysine. Noteworthy, extra care needs to be taken to validate such antibodies [43]. Histones richness in lysines, adjacent amino acid modifications, and the repetition of certain motifs e.g., K9S10T11, K27S28T29, K56S57T58 on H3, render antibodies less specific. A terrific example is an anti-H3K56ac antibody which has caused significant irritation to the scientific community with its cross-reactivity towards the H3K9ac [114, 115].

Detection of increased histone acetylation is an established way for detection of euchromatic regions, promoters of active genes [116] and active enhancers (H3K27ac) [117]. Saito T et al used anti-H3K27ac antibody MABI0309 from MAB Institute for ChIP-qPCR assay to study the promoter regions of PPAR-alpha target genes [118]. Recently, hypoacetylated coding gene regions have been found to inhibit chromatin phase separation [119], promote transcriptional elongation [120]. Acetylation of specific amino acids on histones can have crucial roles in DNA replication and repair, either by serving as docking motifs for replication and repair proteins or as activators of the DNA damage and repair pathways [121-124].

For their detrimental effect on DNA metabolism and cell proliferation, the regulation of histone modification is associated with pathological conditions, including cancer [125], Rubinstein–Taybi syndrome (RTS) [126], inflammation [127], allergy [128], neurodegenerative diseases [129] and others [130]. The regulators of histone acetylation, namely HATs and HDACs are found mutated in multiple cancer types and are targets of several therapeutic strategies [131, 132] thus specific histone PTMs can serve as predictors for prognosis and classification of cancer subtypes [125].

Methylated Histones

Protein methylation is the transfer of one to three methyl groups to lysine or arginine residues. Histones are heavily methylated and its regulation is more complex than the other modifications because lysines can be mono-, di-, or tri-methylated while arginines can be mono- or di-methylated. Histone methyltransferases (HMTs) are the enzymes that use an S-adenosyl-L-methionine (SAM) to catalyse the methylation reaction on lysines while protein arginine methyltransferases (PRMTs) to arginines. HMTs are divided into the SET domain-containing and non-SET domain-containing. The methylation reactions are reversible through the action of histone demethylases, comprised of two families: lysine-specific demethylase 1 (LSD1) and Jumonji domain-containing histone demethylases (JmjC domain)

Histone Modification figure 10
Figure 10. Lysine and arginine methylation.

Histone arginines are the most frequent hydrogen bond donors in the association of histones with the DNA. While the methylation does not alter the charge of arginine or lysines significantly [133], methyl groups are hydrophobic and bulkier, therefore the main change they incur to histones may be to inhibit or promote the binding of proteins [134].

Methylation is detected by mass spectrometry and anti-methyl specific antibodies. Mono-methylation results in 14.0156 Da, di-methylation in 28.0312 Da and tri-methylation in 42.0468 Da increase on the mass of the modified peptide [135]. Pan anti-methyl antibodies can enrich methylated species prior to mass spectrometry and improve detection of methylation events of low stoichiometry [136]. As with most histone PTMs, histone propionylation improves their detection by mass spectrometry significantly. Likewise, the specificity of anti-methyl specific antibodies should be validated not only for identifying the amino acid of interest, but also to distinguish one methylation status from another [137].

H3K9me has been established as a marker of heterochromatic regions [92]. H3K9me serves as a docking motif to the inner nuclear membrane [92]. H3K4 methylation patterns can signal different chromatin events e.g., H3K4me3 is associated with transcriptional start sites and actively transcribed genes, especially during development [77, 138], H3K4me with enhancers but can also have roles in gene repression in a cell-type specific manner [139]. A handful of histone lysine methylations also play a role in RNA splicing [140, 141].

As with acetylation, histone methylation is associated with a variety of diseases, including poor survival in cancer [142], mental retardation [143], neurodevelopmental disorders [144], ageing [145] and others [146].

Phosphorylated Histones

The addition of a phosphoryl group (PO4) on serine, threonine, tyrosine and histidine residues is a reaction termed phosphorylation. Phosphorylation is the most abundant post-translational modification in most organisms mediated by kinases which transfer a phosphate from an ATP molecule to the hydroxyl group of the amino acid’s side chain. The reaction is reversed through hydrolysis by phosphatases. Phosphorylation regulates cellular signalling by switching on or off receptor and enzyme functions. Histone phosphorylation, albeit not as prevalent, it has a great impact on chromatin structure.

Histone Modification figure 11
Figure 11. Phosphorylated serine, threonine and tyrosine residues. From [3].

Serine, threonine and tyrosines have uncharged polar R groups thus the addition of the phosphate group gives a negative charge thereby reducing the affinity to the negatively charged DNA molecule. There is no rule that predicts the chromatin state from its phosphorylation status, however there are important histone phosphorylation events that are cell cycle-regulated and mark certain chromatin functions or cell cycle phases. Notable examples are the H3S10ph, H3S28ph and H3T3ph in mitosis or the H2AS139ph (γH2A.X) as one of the very first events in the formation of double strands breaks (DSBs) and in DNA damage signalling [147, 148]. γH2A.X Ser139 labeling is commonly used to indicate DNA double-stranded break [149, 150].

The main mechanisms through which histone phosphorylations affect chromatin are: i) the interplay with adjacent modifications, especially acetylation and methylation events [151] ; ii) the inhibition [152, 153] or promotion of binding of “reader” proteins which exert their function [154] ; iii) the potential change in nucleosome dynamics [155].

Detection of phosphorylation events on histones is achieved, as always, by mass spectrometry or phosphor-specific antibodies. In tandem mass spectrometry during the Collision Induced Dissociation (CID), the fragmentation of the ionised peptide that bears the phosphor group results in the loss of a H3PO4 molecule and the neutral loss is detected by mass spectrometers. As the phosphorylation is quite labile, enrichment is a usually prerequisite. This is achieved by metal oxide based methods, such as Titanium dioxide, Fe or Zn [156]. In addition, chromatographic fractionation methods, such as hydrophilic interaction chromatography (HILIC) or electrostatic repulsion HILIC (ERLIC) can further enrich the peptide sample for phosphopeptides before mass spectrometry [157]. With respect to phosphotyrosines, pan-phosphotyrosine antibodies work well in enriching tyrosine phosphopeptides [158]. Furthermore, anti-phospho-specific antibodies have been produced in and purified from animals injected with synthetic histone peptides bearing the phosphorylated amino acid. Noteworthy, as with any histone PTM antibody, extra care needs to be taken to validate such antibodies.

Most chemotherapeutic strategies target the stage of DNA replication resulting in double-strand breaks. Thus, γH2A.X can be a marker for patient responses and the efficiency of a therapy [159]. Microscopy examinations of biopsies [160] or peripheral blood mononuclear cells (PBMCs; [161] ) can be excellent tools for the detection and quantification of gH2A.X foci. H4S47ph has been associated with resistance to temozolomide (TMZ) treatment against glioblastoma [162].

Histone Citrullination

Protein citrullination, the conversion of the amino acid arginine (Arg) to the non-conventional amino acid citrulline, can also take place on histones. This epigenetic mechanism includes the deimination of arginine’s side chain and is mediated by a specific enzyme called peptidylarginine deiminases (PADIs), a family that includes PAD1, PAD2, PAD3, PAD4, and PAD6. Besides, no enzymes have been found yet that converts citrulline back to arginine.

Histone Modification figure 12
Figure 12. The chemical conversion (deimination) of arginine to citrulline. From Wikipedia.

PAD4 is the main PADI (PAD2 to a lesser extent [5] ) responsible for histone H3 and H4 citrullination (H3cit/H4cit) and have the capacity to citrullinate unmodified as well as monomethylated arginines. Citrullination of H3R2, H3R8, H3R17, H3R26, H2AR3 and H4R3 has been shown to inhibit further methylation of these arginines thus altering the transcriptional activity at target genes [163, 164]. While arginine is positively charged, citrulline has no net charge thus increasing the hydrophobicity. This change from basic to neutral isoelectric point may alter the tertiary structure of or inhibit binding of histone “readers” at the protein domain [165]. Knockdown of PAD2 results in the decrease of H3R26cit and reverses the chromatin state from closed to open locally at ER-regulated genes [166].

Detection of citrullinated arginines can be achieved by anti-citrullinated antibodies and by mass spectrometry. In the former case, commercial antibodies facilitate detection of citrullinated proteins [167]. Likewise, fluorescent chemical probes have been developed for use in western blots and immunoassays [168, 169]. However, the currently available antibodies are characterised by lack of sensitivity and specificity for broad citrullination events as they may recognise epitopes that are dependent on the surrounding amino acids of the citrulline [170].

In proteomics strategies, the conversion of arginine to citrulline causes a mass shift of +0.98 Da which, albeit small, can be detected by state-of-the-art mass spectrometers. Besides, trypsin (the classical enzyme used in bottom-up proteomics, which normally digests the peptide bonds of polypeptides right after the positively charged arginines and lysines) can offer a significant advantage as it doesn’t cleave after uncharged citrulline. This, together with the mass shift will change the time of elution in HPLC columns and facilitate detection of deimination events [171]. Other methods that biotinylate the citrullinated residue have been successful in enriching the detection of citrullinated peptides prior to mass spectrometry [172, 173]. However, as with most histone PTM identifications, the richness of histones in lysines and arginines is a bottleneck for most proteomics approaches [170]. A recent study has reported new histone citrullination events, H2AR88, H3R42, H3R116, H4R23, H4R55, H4R92, [174] which await validation and their functions to be uncovered.

Detection of increased protein citrullination by antibodies is an established way for detection of rheumatoid arthritis (RA). More recent studies have implicated citrullination of histones with other pathologies and is associated with the formation of neutrophil and macrophage extracellular traps (NETs and METs) [175, 176]. H3cit, for example, is increased upon stimulation with various agents, including LPS, TNF, etc and is found elevated in plasma of patients with advanced cancer [177] and acute kidney injury [178] while pan-PAD inhibitors suppressed miRNA expression affecting oncogene expression involved in prolactinoma and somatoprolactinoma tumors [179]. H4cit is increased in obese mice models of acute pancreatitis [180].

Common Questions
I want to obtain Hela whole cell lysate without destroying its histone structure. What kind of cell lysis method should be used?

Ultrasonic cell disrupter (200W, 4 min for twice) or mechanical microfluidics. Add some glycerin in the buffer, absolutely no SDS in the buffer.

Which kind of Western blot internal control should I use?

If it is the whole cell lysates, then the regular cytoplasmic controls such as beta-actin can be used. If it is the nuclear fraction, then the nuclear controls such as lamin B1 or PCNA can be used. If it is the purified histone fraction, then other members of the histone family, such as H2A can be used.

No positive histone bands are detected after Western blot?

One of the important considerations is that histone proteins have low molecular weights and can easily be transferred through the membrane during Western blotting. Therefore, it's important to reduce the transfer time, and also make sure the markers with similar molecular weights show up on the membrane.

Note

Dr. Nikolaos Parisis contributed the sections about acetylation, methylation, phosphorylation, citrullination of histones in 2018 and contributed the section EpiTOF in October 2020.

References
  1. Harmeyer K, South P, Bishop B, Ogas J, Briggs S. Immediate chromatin immunoprecipitation and on-bead quantitative PCR analysis: a versatile and rapid ChIP procedure. Nucleic Acids Res. 2015;43:e38 pubmed publisher
  2. Cheung P, Vallania F, Warsinske H, Donato M, Schaffert S, Chang S, et al. Single-Cell Chromatin Modification Profiling Reveals Increased Epigenetic Variations with Aging. Cell. 2018;173:1385-1397.e14 pubmed publisher
  3. Boersema P, Mohammed S, Heck A. Phosphopeptide fragmentation and analysis by mass spectrometry. J Mass Spectrom. 2009;44:861-78 pubmed publisher
  4. Kouzarides T. Chromatin modifications and their function. Cell. 2007;128:693-705 pubmed
  5. Cherrington B, Zhang X, McElwee J, Morency E, Anguish L, Coonrod S. Potential role for PAD2 in gene regulation in breast cancer cells. PLoS ONE. 2012;7:e41242 pubmed publisher
  6. Farrelly L, Thompson R, Zhao S, Lepack A, Lyu Y, Bhanu N, et al. Histone serotonylation is a permissive modification that enhances TFIID binding to H3K4me3. Nature. 2019;567:535-539 pubmed publisher
  7. Suganuma T, Workman J. Signals and combinatorial functions of histone modifications. Annu Rev Biochem. 2011;80:473-99 pubmed publisher
  8. Chory E, Calarco J, Hathaway N, Bell O, Neel D, Crabtree G. Nucleosome Turnover Regulates Histone Methylation Patterns over the Genome. Mol Cell. 2019;73:61-72.e3 pubmed publisher
  9. Lejeune E, Allshire R. Common ground: small RNA programming and chromatin modifications. Curr Opin Cell Biol. 2011;23:258-65 pubmed publisher
  10. Grewal S, Jia S. Heterochromatin revisited. Nat Rev Genet. 2007;8:35-46 pubmed
  11. Altaf M, Saksouk N, Cote J. Histone modifications in response to DNA damage. Mutat Res. 2007;618:81-90 pubmed
  12. Li B, Carey M, Workman J. The role of chromatin during transcription. Cell. 2007;128:707-19 pubmed
  13. Weber C, Zhou Y, Lee J, Looger L, Qian G, Ge C, et al. Temperature-dependent sex determination is mediated by pSTAT3 repression of Kdm6b. Science. 2020;368:303-306 pubmed publisher
  14. Wang W, Hu C, Zeng A, Alegre D, Hu D, Gotting K, et al. Changes in regeneration-responsive enhancers shape regenerative capacities in vertebrates. Science. 2020;369: pubmed publisher
  15. Xia W, Xu J, Yu G, Yao G, Xu K, Ma X, et al. Resetting histone modifications during human parental-to-zygotic transition. Science. 2019;365:353-360 pubmed publisher
  16. Wang Z, Chivu A, Choate L, Rice E, Miller D, Chu T, et al. Prediction of histone post-translational modification patterns based on nascent transcription data. Nat Genet. 2022;54:295-305 pubmed publisher
  17. Chi P, Allis C, Wang G. Covalent histone modifications--miswritten, misinterpreted and mis-erased in human cancers. Nat Rev Cancer. 2010;10:457-69 pubmed publisher
  18. Meissner A. Epigenetic modifications in pluripotent and differentiated cells. Nat Biotechnol. 2010;28:1079-88 pubmed publisher
  19. Moss T, Wallrath L. Connections between epigenetic gene silencing and human disease. Mutat Res. 2007;618:163-74 pubmed
  20. Bibikova M, Laurent L, Ren B, Loring J, Fan J. Unraveling epigenetic regulation in embryonic stem cells. Cell Stem Cell. 2008;2:123-34 pubmed publisher
  21. Liu C, Tangsombatvisit S, Rosenberg J, Mandelbaum G, Gillespie E, Gozani O, et al. Specific post-translational histone modifications of neutrophil extracellular traps as immunogens and potential targets of lupus autoantibodies. Arthritis Res Ther. 2012;14:R25 pubmed publisher
  22. Liu B, Cheng J, Zhang X, Wang R, Zhang W, Lin H, et al. Global histone modification patterns as prognostic markers to classify glioma patients. Cancer Epidemiol Biomarkers Prev. 2010;19:2888-96 pubmed publisher
  23. Lee J, Smith E, Shilatifard A. The language of histone crosstalk. Cell. 2010;142:682-5 pubmed publisher
  24. Gardner K, Allis C, Strahl B. Operating on chromatin, a colorful language where context matters. J Mol Biol. 2011;409:36-46 pubmed publisher
  25. Henikoff S, Shilatifard A. Histone modification: cause or cog?. Trends Genet. 2011;27:389-96 pubmed publisher
  26. Verhoeven J, Baan C, Peeters A, Clahsen van Groningen M, Nieboer D, Herzog M, et al. Circulating cell-free nucleosomes as biomarker for kidney transplant rejection: a pilot study. Clin Epigenetics. 2021;13:32 pubmed publisher
  27. Duncan E, Muratore Schroeder T, Cook R, Garcia B, Shabanowitz J, Hunt D, et al. Cathepsin L proteolytically processes histone H3 during mouse embryonic stem cell differentiation. Cell. 2008;135:284-94 pubmed publisher
  28. Hurd P, Bannister A, Halls K, Dawson M, Vermeulen M, Olsen J, et al. Phosphorylation of histone H3 Thr-45 is linked to apoptosis. J Biol Chem. 2009;284:16575-83 pubmed publisher
  29. Kushnirov V. Rapid and reliable protein extraction from yeast. Yeast. 2000;16:857-60 pubmed
  30. Kizer K, Xiao T, Strahl B. Accelerated nuclei preparation and methods for analysis of histone modifications in yeast. Methods. 2006;40:296-302 pubmed
  31. Shechter D, Dormann H, Allis C, Hake S. Extraction, purification and analysis of histones. Nat Protoc. 2007;2:1445-57 pubmed
  32. Frei C, Gasser S. The yeast Sgs1p helicase acts upstream of Rad53p in the DNA replication checkpoint and colocalizes with Rad53p in S-phase-specific foci. Genes Dev. 2000;14:81-96 pubmed
  33. Pallier C, Scaffidi P, Chopineau Proust S, Agresti A, Nordmann P, Bianchi M, et al. Association of chromatin proteins high mobility group box (HMGB) 1 and HMGB2 with mitotic chromosomes. Mol Biol Cell. 2003;14:3414-26 pubmed
  34. Das C, Lucia M, Hansen K, Tyler J. CBP/p300-mediated acetylation of histone H3 on lysine 56. Nature. 2009;459:113-7 pubmed publisher
  35. Byrum S, Taverna S, Tackett A. Purification of specific chromatin loci for proteomic analysis. Methods Mol Biol. 2015;1228:83-92 pubmed publisher
  36. Coolen M, Labusch M, Mannioui A, Bally Cuif L. Mosaic Heterochrony in Neural Progenitors Sustains Accelerated Brain Growth and Neurogenesis in the Juvenile Killifish N. furzeri. Curr Biol. 2020;30:736-745.e4 pubmed publisher
  37. Rhodes J, Feldmann A, Hernández Rodríguez B, Díaz N, Brown J, Fursova N, et al. Cohesin Disrupts Polycomb-Dependent Chromosome Interactions in Embryonic Stem Cells. Cell Rep. 2020;30:820-835.e10 pubmed publisher
  38. Shwartz Y, Gonzalez Celeiro M, Chen C, Pasolli H, Sheu S, Fan S, et al. Cell Types Promoting Goosebumps Form a Niche to Regulate Hair Follicle Stem Cells. Cell. 2020;: pubmed publisher
  39. Butler A, Johnston D, Kaur S, Lubin F. Long noncoding RNA NEAT1 mediates neuronal histone methylation and age-related memory impairment. Sci Signal. 2019;12: pubmed publisher
  40. Batie M, Frost J, Frost M, Wilson J, Schofield P, Rocha S. Hypoxia induces rapid changes to histone methylation and reprograms chromatin. Science. 2019;363:1222-1226 pubmed publisher
  41. Egelhofer T, Minoda A, Klugman S, Lee K, Kolasinska Zwierz P, Alekseyenko A, et al. An assessment of histone-modification antibody quality. Nat Struct Mol Biol. 2011;18:91-3 pubmed publisher
  42. Kungulovski G, Kycia I, Mauser R, Jeltsch A. Specificity Analysis of Histone Modification-Specific Antibodies or Reading Domains on Histone Peptide Arrays. Methods Mol Biol. 2015;1348:275-84 pubmed publisher
  43. Rothbart S, Dickson B, Raab J, Grzybowski A, Krajewski K, Guo A, et al. An Interactive Database for the Assessment of Histone Antibody Specificity. Mol Cell. 2015;59:502-11 pubmed publisher
  44. Allahverdi A, Yang R, Korolev N, Fan Y, Davey C, Liu C, et al. The effects of histone H4 tail acetylations on cation-induced chromatin folding and self-association. Nucleic Acids Res. 2011;39:1680-91 pubmed publisher
  45. He S, Bauman D, Davis J, Loyola A, Nishioka K, Gronlund J, et al. Facile synthesis of site-specifically acetylated and methylated histone proteins: reagents for evaluation of the histone code hypothesis. Proc Natl Acad Sci U S A. 2003;100:12033-8 pubmed
  46. Shogren Knaak M, Ishii H, Sun J, Pazin M, Davie J, Peterson C. Histone H4-K16 acetylation controls chromatin structure and protein interactions. Science. 2006;311:844-7 pubmed
  47. McGinty R, Kim J, Chatterjee C, Roeder R, Muir T. Chemically ubiquitylated histone H2B stimulates hDot1L-mediated intranucleosomal methylation. Nature. 2008;453:812-6 pubmed publisher
  48. Manohar M, Mooney A, North J, Nakkula R, Picking J, Edon A, et al. Acetylation of histone H3 at the nucleosome dyad alters DNA-histone binding. J Biol Chem. 2009;284:23312-21 pubmed publisher
  49. Shogren Knaak M, Peterson C. Creating designer histones by native chemical ligation. Methods Enzymol. 2004;375:62-76 pubmed
  50. Shimko J, North J, Bruns A, Poirier M, Ottesen J. Preparation of fully synthetic histone H3 reveals that acetyl-lysine 56 facilitates protein binding within nucleosomes. J Mol Biol. 2011;408:187-204 pubmed publisher
  51. Neumann H, Hancock S, Buning R, Routh A, Chapman L, Somers J, et al. A method for genetically installing site-specific acetylation in recombinant histones defines the effects of H3 K56 acetylation. Mol Cell. 2009;36:153-63 pubmed publisher
  52. Wysocka J, Reilly P, Herr W. Loss of HCF-1-chromatin association precedes temperature-induced growth arrest of tsBN67 cells. Mol Cell Biol. 2001;21:3820-9 pubmed
  53. Wysocka J, Swigut T, Xiao H, Milne T, Kwon S, Landry J, et al. A PHD finger of NURF couples histone H3 lysine 4 trimethylation with chromatin remodelling. Nature. 2006;442:86-90 pubmed
  54. Vermeulen M, Mulder K, Denissov S, Pijnappel W, van Schaik F, Varier R, et al. Selective anchoring of TFIID to nucleosomes by trimethylation of histone H3 lysine 4. Cell. 2007;131:58-69 pubmed
  55. Sims R, Trojer P, Li G, Reinberg D. Methods to identify and functionally analyze factors that specifically recognize histone lysine methylation. Methods. 2006;40:331-8 pubmed
  56. Simon R, Felsenfeld G. A new procedure for purifying histone pairs H2A + H2B and H3 + H4 from chromatin using hydroxylapatite. Nucleic Acids Res. 1979;6:689-96 pubmed
  57. Loyola A, Reinberg D. Histone deposition and chromatin assembly by RSF. Methods. 2003;31:96-103 pubmed
  58. Luger K, Rechsteiner T, Richmond T. Expression and purification of recombinant histones and nucleosome reconstitution. Methods Mol Biol. 1999;119:1-16 pubmed
  59. Gelbart M, Rechsteiner T, Richmond T, Tsukiyama T. Interactions of Isw2 chromatin remodeling complex with nucleosomal arrays: analyses using recombinant yeast histones and immobilized templates. Mol Cell Biol. 2001;21:2098-106 pubmed
  60. Lachner M, O Carroll D, Rea S, Mechtler K, Jenuwein T. Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins. Nature. 2001;410:116-20 pubmed
  61. Bannister A, Zegerman P, Partridge J, Miska E, Thomas J, Allshire R, et al. Selective recognition of methylated lysine 9 on histone H3 by the HP1 chromo domain. Nature. 2001;410:120-4 pubmed
  62. Shi X, Hong T, Walter K, Ewalt M, Michishita E, Hung T, et al. ING2 PHD domain links histone H3 lysine 4 methylation to active gene repression. Nature. 2006;442:96-9 pubmed
  63. Wysocka J, Swigut T, Milne T, Dou Y, Zhang X, Burlingame A, et al. WDR5 associates with histone H3 methylated at K4 and is essential for H3 K4 methylation and vertebrate development. Cell. 2005;121:859-72 pubmed
  64. Wysocka J. Identifying novel proteins recognizing histone modifications using peptide pull-down assay. Methods. 2006;40:339-43 pubmed
  65. Shi X, Kachirskaia I, Walter K, Kuo J, Lake A, Davrazou F, et al. Proteome-wide analysis in Saccharomyces cerevisiae identifies several PHD fingers as novel direct and selective binding modules of histone H3 methylated at either lysine 4 or lysine 36. J Biol Chem. 2007;282:2450-5 pubmed
  66. Matthews A, Kuo A, Ramon Maiques S, Han S, Champagne K, Ivanov D, et al. RAG2 PHD finger couples histone H3 lysine 4 trimethylation with V(D)J recombination. Nature. 2007;450:1106-10 pubmed
  67. Bua D, Kuo A, Cheung P, Liu C, Migliori V, Espejo A, et al. Epigenome microarray platform for proteome-wide dissection of chromatin-signaling networks. PLoS ONE. 2009;4:e6789 pubmed publisher
  68. Kim J, Daniel J, Espejo A, Lake A, Krishna M, Xia L, et al. Tudor, MBT and chromo domains gauge the degree of lysine methylation. EMBO Rep. 2006;7:397-403 pubmed
  69. Lieb J, Liu X, Botstein D, Brown P. Promoter-specific binding of Rap1 revealed by genome-wide maps of protein-DNA association. Nat Genet. 2001;28:327-34 pubmed
  70. Liu C, Kaplan T, Kim M, Buratowski S, Schreiber S, Friedman N, et al. Single-nucleosome mapping of histone modifications in S. cerevisiae. PLoS Biol. 2005;3:e328 pubmed
  71. Boyer L, Lee T, Cole M, Johnstone S, Levine S, Zucker J, et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell. 2005;122:947-56 pubmed
  72. Skene P, Henikoff S. An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites. elife. 2017;6: pubmed publisher
  73. Ren B, Robert F, Wyrick J, Aparicio O, Jennings E, Simon I, et al. Genome-wide location and function of DNA binding proteins. Science. 2000;290:2306-9 pubmed
  74. Gerton J, DeRisi J, Shroff R, Lichten M, Brown P, Petes T. Global mapping of meiotic recombination hotspots and coldspots in the yeast Saccharomyces cerevisiae. Proc Natl Acad Sci U S A. 2000;97:11383-90 pubmed
  75. Liu C, Schreiber S, Bernstein B. Development and validation of a T7 based linear amplification for genomic DNA. BMC Genomics. 2003;4:19 pubmed
  76. Pat Brown Lab Experimental Protocols. Available from: cmgm.stanford.edu/pbrown/protocols/index.html
  77. Barski A, Cuddapah S, Cui K, Roh T, Schones D, Wang Z, et al. High-resolution profiling of histone methylations in the human genome. Cell. 2007;129:823-37 pubmed
  78. Johnson D, Mortazavi A, Myers R, Wold B. Genome-wide mapping of in vivo protein-DNA interactions. Science. 2007;316:1497-502 pubmed
  79. Ho J, Bishop E, Karchenko P, Negre N, White K, Park P. ChIP-chip versus ChIP-seq: lessons for experimental design and data analysis. BMC Genomics. 2011;12:134 pubmed publisher
  80. Bentley D. Whole-genome re-sequencing. Curr Opin Genet Dev. 2006;16:545-52 pubmed
  81. Shendure J, Ji H. Next-generation DNA sequencing. Nat Biotechnol. 2008;26:1135-45 pubmed publisher
  82. Park P. ChIP-seq: advantages and challenges of a maturing technology. Nat Rev Genet. 2009;10:669-80 pubmed publisher
  83. Chen Y, Negre N, Li Q, Mieczkowska J, Slattery M, Liu T, et al. Systematic evaluation of factors influencing ChIP-seq fidelity. Nat Methods. 2012;9:609-14 pubmed publisher
  84. Chen L, Wang C, Qin Z, Wu H. A novel statistical method for quantitative comparison of multiple ChIP-seq datasets. Bioinformatics. 2015;31:1889-96 pubmed publisher
  85. Lun A, Smyth G. From reads to regions: a Bioconductor workflow to detect differential binding in ChIP-seq data. F1000Res. 2015;4:1080 pubmed publisher
  86. van Galen P, Viny A, Ram O, Ryan R, Cotton M, Donohue L, et al. A Multiplexed System for Quantitative Comparisons of Chromatin Landscapes. Mol Cell. 2016;61:170-80 pubmed publisher
  87. Gomez D, Shankman L, Nguyen A, Owens G. Detection of histone modifications at specific gene loci in single cells in histological sections. Nat Methods. 2013;10:171-7 pubmed publisher
  88. Söderberg O, Gullberg M, Jarvius M, Ridderstråle K, Leuchowius K, Jarvius J, et al. Direct observation of individual endogenous protein complexes in situ by proximity ligation. Nat Methods. 2006;3:995-1000 pubmed
  89. Kulej K, Avgousti D, Weitzman M, Garcia B. Characterization of histone post-translational modifications during virus infection using mass spectrometry-based proteomics. Methods. 2015;90:8-20 pubmed publisher
  90. Onder O, Sidoli S, Carroll M, Garcia B. Progress in epigenetic histone modification analysis by mass spectrometry for clinical investigations. Expert Rev Proteomics. 2015;12:499-517 pubmed publisher
  91. Lahiri S, Sun N, Buck A, Imhof A, Walch A. MALDI imaging mass spectrometry as a novel tool for detecting histone modifications in clinical tissue samples. Expert Rev Proteomics. 2016;13:275-84 pubmed publisher
  92. Cabianca D, Muñoz Jiménez C, Kalck V, Gaidatzis D, Padeken J, Seeber A, et al. Active chromatin marks drive spatial sequestration of heterochromatin in C. elegans nuclei. Nature. 2019;: pubmed publisher
  93. Israel A, del Rosario Garrido M, Barbella Y, Becemberg I. Effect of water deprivation and salt loading on atrial natriuretic peptide-stimulated guanylate cyclase activity in the rat subfornical organ. Neuroendocrinology. 1989;50:334-7 pubmed
  94. Moradian A, Kalli A, Sweredoski M, Hess S. The top-down, middle-down, and bottom-up mass spectrometry approaches for characterization of histone variants and their post-translational modifications. Proteomics. 2014;14:489-97 pubmed publisher
  95. Sidoli S, Bhanu N, Karch K, Wang X, Garcia B. Complete Workflow for Analysis of Histone Post-translational Modifications Using Bottom-up Mass Spectrometry: From Histone Extraction to Data Analysis. J Vis Exp. 2016;: pubmed publisher
  96. Yamamoto K, Chikaoka Y, Hayashi G, Sakamoto R, Yamamoto R, Sugiyama A, et al. Middle-Down and Chemical Proteomic Approaches to Reveal Histone H4 Modification Dynamics in Cell Cycle: Label-Free Semi-Quantification of Histone Tail Peptide Modifications Including Phosphorylation and Highly Sensitive Capture of Histone PTM Binding. Mass Spectrom (Tokyo). 2015;4:A0039 pubmed publisher
  97. Meharena H, Marco A, Dileep V, Lockshin E, Akatsu G, Mullahoo J, et al. Down-syndrome-induced senescence disrupts the nuclear architecture of neural progenitors. Cell Stem Cell. 2022;29:116-130.e7 pubmed publisher
  98. Swaney D, Villen J. Proteomic Analysis of Protein Posttranslational Modifications by Mass Spectrometry. Cold Spring Harb Protoc. 2016;2016:pdb.top077743 pubmed publisher
  99. Kim M, Zhong J, Pandey A. Common errors in mass spectrometry-based analysis of post-translational modifications. Proteomics. 2016;16:700-14 pubmed publisher
  100. Sekar T, Foygel K, Gelovani J, Paulmurugan R. Genetically encoded molecular biosensors to image histone methylation in living animals. Anal Chem. 2015;87:892-9 pubmed publisher
  101. Kanno T, Kanno Y, Siegel R, Jang M, Lenardo M, Ozato K. Selective recognition of acetylated histones by bromodomain proteins visualized in living cells. Mol Cell. 2004;13:33-43 pubmed
  102. Lin C, Jao C, Ting A. Genetically encoded fluorescent reporters of histone methylation in living cells. J Am Chem Soc. 2004;126:5982-3 pubmed
  103. Sasaki K, Ito A, Yoshida M. Development of live-cell imaging probes for monitoring histone modifications. Bioorg Med Chem. 2012;20:1887-92 pubmed publisher
  104. Sato Y, Mukai M, Ueda J, Muraki M, Stasevich T, Horikoshi N, et al. Genetically encoded system to track histone modification in vivo. Sci Rep. 2013;3:2436 pubmed publisher
  105. Drazic A, Myklebust L, Ree R, Arnesen T. The world of protein acetylation. Biochim Biophys Acta. 2016;1864:1372-401 pubmed publisher
  106. Wan L, Chong S, Xuan F, Liang A, Cui X, Gates L, et al. Impaired cell fate through gain-of-function mutations in a chromatin reader. Nature. 2020;577:121-126 pubmed publisher
  107. Richman R, Chicoine L, Collini M, Cook R, Allis C. Micronuclei and the cytoplasm of growing Tetrahymena contain a histone acetylase activity which is highly specific for free histone H4. J Cell Biol. 1988;106:1017-26 pubmed
  108. Dancy B, Cole P. Protein lysine acetylation by p300/CBP. Chem Rev. 2015;115:2419-52 pubmed publisher
  109. Seto E, Yoshida M. Erasers of histone acetylation: the histone deacetylase enzymes. Cold Spring Harb Perspect Biol. 2014;6:a018713 pubmed publisher
  110. Sadakierska Chudy A, Filip M. A comprehensive view of the epigenetic landscape. Part II: Histone post-translational modification, nucleosome level, and chromatin regulation by ncRNAs. Neurotox Res. 2015;27:172-97 pubmed publisher
  111. Benton C, Fiskus W, Bhalla K. Targeting Histone Acetylation: Readers and Writers in Leukemia and Cancer. Cancer J. 2017;23:286-291 pubmed publisher
  112. Poliantsev A. [Case of effective surgery of esophageal cancer, with subsequent Roux-Hertzen' plastic technic]. Vestn Khir Im I I Grek. 1950;70:47-8 pubmed
  113. Sidoli S, Garcia B. Middle-down proteomics: a still unexploited resource for chromatin biology. Expert Rev Proteomics. 2017;14:617-626 pubmed publisher
  114. Pal S, Graves H, Ohsawa R, Huang T, Wang P, Harmacek L, et al. The Commercial Antibodies Widely Used to Measure H3 K56 Acetylation Are Non-Specific in Human and Drosophila Cells. PLoS ONE. 2016;11:e0155409 pubmed publisher
  115. Drogaris P, Villeneuve V, Pomiès C, Lee E, Bourdeau V, Bonneil E, et al. Histone deacetylase inhibitors globally enhance h3/h4 tail acetylation without affecting h3 lysine 56 acetylation. Sci Rep. 2012;2:220 pubmed publisher
  116. Eberharter A, Becker P. Histone acetylation: a switch between repressive and permissive chromatin. Second in review series on chromatin dynamics. EMBO Rep. 2002;3:224-9 pubmed
  117. Creyghton M, Cheng A, Welstead G, Kooistra T, Carey B, Steine E, et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc Natl Acad Sci U S A. 2010;107:21931-6 pubmed publisher
  118. Saito T, Kuma A, Sugiura Y, Ichimura Y, Obata M, Kitamura H, et al. Autophagy regulates lipid metabolism through selective turnover of NCoR1. Nat Commun. 2019;10:1567 pubmed publisher
  119. Gibson B, Doolittle L, Schneider M, Jensen L, Gamarra N, Henry L, et al. Organization of Chromatin by Intrinsic and Regulated Phase Separation. Cell. 2019;179:470-484.e21 pubmed publisher
  120. Greer C, Tanaka Y, Kim Y, Xie P, Zhang M, Park I, et al. Histone Deacetylases Positively Regulate Transcription through the Elongation Machinery. Cell Rep. 2015;13:1444-1455 pubmed publisher
  121. Unnikrishnan A, Gafken P, Tsukiyama T. Dynamic changes in histone acetylation regulate origins of DNA replication. Nat Struct Mol Biol. 2010;17:430-7 pubmed publisher
  122. Vogelauer M, Rubbi L, Lucas I, Brewer B, Grunstein M. Histone acetylation regulates the time of replication origin firing. Mol Cell. 2002;10:1223-33 pubmed
  123. Williamson E, Wray J, Bansal P, Hromas R. Overview for the histone codes for DNA repair. Prog Mol Biol Transl Sci. 2012;110:207-27 pubmed publisher
  124. Dhar S, Gursoy Yuzugullu O, Parasuram R, Price B. The tale of a tail: histone H4 acetylation and the repair of DNA breaks. Philos Trans R Soc Lond B Biol Sci. 2017;372: pubmed publisher
  125. Chervona Y, Costa M. Histone modifications and cancer: biomarkers of prognosis?. Am J Cancer Res. 2012;2:589-97 pubmed
  126. Roelfsema J, Peters D. Rubinstein-Taybi syndrome: clinical and molecular overview. Expert Rev Mol Med. 2007;9:1-16 pubmed
  127. Glauben R, Siegmund B. Molecular basis of histone deacetylase inhibitors as new drugs for the treatment of inflammatory diseases and cancer. Methods Mol Biol. 2009;512:365-76 pubmed publisher
  128. Bergeron C, Fukakusa M, Olivenstein R, Lemiere C, Shannon J, Ernst P, et al. Increased glucocorticoid receptor-beta expression, but not decreased histone deacetylase 2, in severe asthma. J Allergy Clin Immunol. 2006;117:703-5 pubmed
  129. Nucifora F, Sasaki M, Peters M, Huang H, Cooper J, Yamada M, et al. Interference by huntingtin and atrophin-1 with cbp-mediated transcription leading to cellular toxicity. Science. 2001;291:2423-8 pubmed
  130. Khan S, Khan A. Role of histone acetylation in cell physiology and diseases: An update. Clin Chim Acta. 2010;411:1401-11 pubmed publisher
  131. Ropero S, Esteller M. The role of histone deacetylases (HDACs) in human cancer. Mol Oncol. 2007;1:19-25 pubmed publisher
  132. Chervona Y, Costa M, Dai W. Epigenomics: Pioneering a New Frontier in Cancer Research. J Pharmacogenomics Pharmacoproteomics. 2012;3: pubmed
  133. Evich M, Stroeva E, Zheng Y, Germann M. Effect of methylation on the side-chain pKa value of arginine. Protein Sci. 2016;25:479-86 pubmed publisher
  134. Bedford M, Clarke S. Protein arginine methylation in mammals: who, what, and why. Mol Cell. 2009;33:1-13 pubmed publisher
  135. Zhang K, Yau P, Chandrasekhar B, New R, Kondrat R, Imai B, et al. Differentiation between peptides containing acetylated or tri-methylated lysines by mass spectrometry: an application for determining lysine 9 acetylation and methylation of histone H3. Proteomics. 2004;4:1-10 pubmed
  136. Cao X, Garcia B. Global Proteomics Analysis of Protein Lysine Methylation. Curr Protoc Protein Sci. 2016;86:24.8.1-24.8.19 pubmed publisher
  137. Shah R, Grzybowski A, Cornett E, Johnstone A, Dickson B, Boone B, et al. Examining the Roles of H3K4 Methylation States with Systematically Characterized Antibodies. Mol Cell. 2018;72:162-177.e7 pubmed publisher
  138. Ruthenburg A, Allis C, Wysocka J. Methylation of lysine 4 on histone H3: intricacy of writing and reading a single epigenetic mark. Mol Cell. 2007;25:15-30 pubmed
  139. Cheng J, Blum R, Bowman C, Hu D, Shilatifard A, Shen S, et al. A role for H3K4 monomethylation in gene repression and partitioning of chromatin readers. Mol Cell. 2014;53:979-92 pubmed publisher
  140. Luco R, Allo M, Schor I, Kornblihtt A, Misteli T. Epigenetics in alternative pre-mRNA splicing. Cell. 2011;144:16-26 pubmed publisher
  141. Luco R, Pan Q, Tominaga K, Blencowe B, Pereira Smith O, Misteli T. Regulation of alternative splicing by histone modifications. Science. 2010;327:996-1000 pubmed publisher
  142. Greer E, Shi Y. Histone methylation: a dynamic mark in health, disease and inheritance. Nat Rev Genet. 2012;13:343-57 pubmed publisher
  143. Iwase S, Shi Y. Histone and DNA modifications in mental retardation. Prog Drug Res. 2011;67:147-73 pubmed
  144. Kim J, Lee J, Lee I, Lee S, Cho K. Histone Lysine Methylation and Neurodevelopmental Disorders. Int J Mol Sci. 2017;18: pubmed publisher
  145. Sarg B, Koutzamani E, Helliger W, Rundquist I, Lindner H. Postsynthetic trimethylation of histone H4 at lysine 20 in mammalian tissues is associated with aging. J Biol Chem. 2002;277:39195-201 pubmed
  146. Wen K, Milić J, El Khodor B, Dhana K, Nano J, Pulido T, et al. The Role of DNA Methylation and Histone Modifications in Neurodegenerative Diseases: A Systematic Review. PLoS ONE. 2016;11:e0167201 pubmed publisher
  147. Huang X, Halicka H, Darzynkiewicz Z. Detection of histone H2AX phosphorylation on Ser-139 as an indicator of DNA damage (DNA double-strand breaks). Curr Protoc Cytom. 2004;Chapter 7:Unit 7.27 pubmed publisher
  148. Rogakou E, Pilch D, Orr A, Ivanova V, Bonner W. DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J Biol Chem. 1998;273:5858-68 pubmed
  149. Adaikkan C, Middleton S, Marco A, Pao P, Mathys H, Kim D, et al. Gamma Entrainment Binds Higher-Order Brain Regions and Offers Neuroprotection. Neuron. 2019;102:929-943.e8 pubmed publisher
  150. Cole T, Zhao H, Collier T, Sandoval I, Sortwell C, Steece Collier K, et al. α-Synuclein antisense oligonucleotides as a disease-modifying therapy for Parkinson's disease. JCI Insight. 2021;6: pubmed publisher
  151. Josefowicz S, Shimada M, Armache A, Li C, Miller R, Lin S, et al. Chromatin Kinases Act on Transcription Factors and Histone Tails in Regulation of Inducible Transcription. Mol Cell. 2016;64:347-361 pubmed publisher
  152. Lau P, Cheung P. Histone code pathway involving H3 S28 phosphorylation and K27 acetylation activates transcription and antagonizes polycomb silencing. Proc Natl Acad Sci U S A. 2011;108:2801-6 pubmed publisher
  153. Dawson M, Bannister A, Göttgens B, Foster S, Bartke T, Green A, et al. JAK2 phosphorylates histone H3Y41 and excludes HP1alpha from chromatin. Nature. 2009;461:819-22 pubmed publisher
  154. Yun M, Wu J, Workman J, Li B. Readers of histone modifications. Cell Res. 2011;21:564-78 pubmed publisher
  155. Kang B, Pu M, Hu G, Wen W, Dong Z, Zhao K, et al. Phosphorylation of H4 Ser 47 promotes HIRA-mediated nucleosome assembly. Genes Dev. 2011;25:1359-64 pubmed publisher
  156. Thingholm T, Jensen O, Larsen M. Enrichment and separation of mono- and multiply phosphorylated peptides using sequential elution from IMAC prior to mass spectrometric analysis. Methods Mol Biol. 2009;527:67-78, xi pubmed publisher
  157. Leitner A. Enrichment Strategies in Phosphoproteomics. Methods Mol Biol. 2016;1355:105-21 pubmed publisher
  158. Schreiber T, Mäusbacher N, Breitkopf S, Grundner Culemann K, Daub H. Quantitative phosphoproteomics--an emerging key technology in signal-transduction research. Proteomics. 2008;8:4416-32 pubmed publisher
  159. Redon C, Weyemi U, Parekh P, Huang D, Burrell A, Bonner W. γ-H2AX and other histone post-translational modifications in the clinic. Biochim Biophys Acta. 2012;1819:743-56 pubmed publisher
  160. Bonner W, Redon C, Dickey J, Nakamura A, Sedelnikova O, Solier S, et al. GammaH2AX and cancer. Nat Rev Cancer. 2008;8:957-67 pubmed publisher
  161. Redon C, Nakamura A, Zhang Y, Ji J, Bonner W, Kinders R, et al. Histone gammaH2AX and poly(ADP-ribose) as clinical pharmacodynamic biomarkers. Clin Cancer Res. 2010;16:4532-42 pubmed publisher
  162. Lee S. Temozolomide resistance in glioblastoma multiforme. Genes Dis. 2016;3:198-210 pubmed publisher
  163. Cuthbert G, Daujat S, Snowden A, Erdjument Bromage H, Hagiwara T, Yamada M, et al. Histone deimination antagonizes arginine methylation. Cell. 2004;118:545-53 pubmed
  164. Wang Y, Wysocka J, Sayegh J, Lee Y, Perlin J, Leonelli L, et al. Human PAD4 regulates histone arginine methylation levels via demethylimination. Science. 2004;306:279-83 pubmed
  165. Fenley A, Anandakrishnan R, Kidane Y, Onufriev A. Modulation of nucleosomal DNA accessibility via charge-altering post-translational modifications in histone core. Epigenetics Chromatin. 2018;11:11 pubmed publisher
  166. Zhang X, Bolt M, Guertin M, Chen W, Zhang S, Cherrington B, et al. Peptidylarginine deiminase 2-catalyzed histone H3 arginine 26 citrullination facilitates estrogen receptor ? target gene activation. Proc Natl Acad Sci U S A. 2012;109:13331-6 pubmed publisher
  167. Moelants E, Van Damme J, Proost P. Detection and quantification of citrullinated chemokines. PLoS ONE. 2011;6:e28976 pubmed publisher
  168. Bicker K, Subramanian V, Chumanevich A, Hofseth L, Thompson P. Seeing citrulline: development of a phenylglyoxal-based probe to visualize protein citrullination. J Am Chem Soc. 2012;134:17015-8 pubmed publisher
  169. Kunieda K, Yamauchi H, Kawaguchi M, Ieda N, Nakagawa H. Development of a fluorescent probe for detection of citrulline based on photo-induced electron transfer. Bioorg Med Chem Lett. 2018;28:969-973 pubmed publisher
  170. Fert Bober J, Sokolove J. Proteomics of citrullination in cardiovascular disease. Proteomics Clin Appl. 2014;8:522-33 pubmed publisher
  171. Bennike et al, Optimizing the Identification of Citrullinated Peptides by Mass Spectrometry: Utilizing the Inability of Trypsin to Cleave after Citrullinated Amino Acids. 2013. J Proteomics Bioinform 6:288-295. Available from: dx.doi.org/10.4172/jpb.1000293
  172. Lewallen D, Bicker K, Subramanian V, Clancy K, Slade D, Martell J, et al. Chemical Proteomic Platform To Identify Citrullinated Proteins. ACS Chem Biol. 2015;10:2520-8 pubmed publisher
  173. Tutturen A, Holm A, Fleckenstein B. Specific biotinylation and sensitive enrichment of citrullinated peptides. Anal Bioanal Chem. 2013;405:9321-31 pubmed publisher
  174. Li Q, Hao J, Zhang Z, Krane L, Hammerich K, Sanford T, et al. Proteomic analysis of proteome and histone post-translational modifications in heat shock protein 90 inhibition-mediated bladder cancer therapeutics. Sci Rep. 2017;7:201 pubmed publisher
  175. Binet F, Cagnone G, Crespo Garcia S, Hata M, Neault M, Dejda A, et al. Neutrophil extracellular traps target senescent vasculature for tissue remodeling in retinopathy. Science. 2020;369: pubmed publisher
  176. Mohanan S, Horibata S, McElwee J, Dannenberg A, Coonrod S. Identification of macrophage extracellular trap-like structures in mammary gland adipose tissue: a preliminary study. Front Immunol. 2013;4:67 pubmed publisher
  177. Thålin C, Lundström S, Seignez C, Daleskog M, Lundström A, Henriksson P, et al. Citrullinated histone H3 as a novel prognostic blood marker in patients with advanced cancer. PLoS ONE. 2018;13:e0191231 pubmed publisher
  178. Okubo K, Kurosawa M, Kamiya M, Urano Y, Suzuki A, Yamamoto K, et al. Macrophage extracellular trap formation promoted by platelet activation is a key mediator of rhabdomyolysis-induced acute kidney injury. Nat Med. 2018;24:232-238 pubmed publisher
  179. DeVore S, Young C, Li G, Sundararajan A, Ramaraj T, Mudge J, et al. Histone Citrullination Represses MicroRNA Expression, Resulting in Increased Oncogene mRNAs in Somatolactotrope Cells. Mol Cell Biol. 2018;38: pubmed publisher
  180. Pérez S, Finamor I, Martí Andrés P, Pereda J, Campos A, Domingues R, et al. Role of obesity in the release of extracellular nucleosomes in acute pancreatitis: a clinical and experimental study. Int J Obes (Lond). 2019;43:158-168 pubmed publisher
ISSN : 2329-5139