Fact-checked by Grok 2 weeks ago

Histone-modifying enzymes

Histone-modifying enzymes are a diverse class of proteins that catalyze the addition or removal of covalent chemical groups, such as acetyl, methyl, , or moieties, on the amino-terminal tails of proteins, thereby regulating structure, DNA accessibility, and in eukaryotic cells. These enzymes play a central role in epigenetic regulation by establishing and maintaining heritable patterns of histone modifications that influence transcriptional activation or repression without altering the underlying DNA sequence. By dynamically altering the charge and interactions of histones with DNA and other proteins, they control fundamental cellular processes including , , and response to environmental cues. Histone-modifying enzymes are broadly classified into writers, which deposit modifications, and erasers, which remove them, with a third category of readers that recognize these marks to propagate or interpret epigenetic signals. Writers include histone acetyltransferases (HATs), such as p300/CBP and Rtt109, which transfer acetyl groups from to residues, typically promoting open and activation; and histone methyltransferases (HMTs), like in the Polycomb repressive complex 2 (PRC2) or SET domain-containing enzymes such as MLL1-4, which add methyl groups to or residues using S-adenosylmethionine as a cofactor. Erasers encompass histone deacetylases (HDACs), which hydrolyze acetyl groups to condense and repress transcription, and histone demethylases (HDMs), including flavin-dependent enzymes like LSD1 that oxidize mono- and dimethylated s, or Jumonji C (JmjC) domain-containing enzymes like JMJD2A and UTX that employ Fe(II) and α-ketoglutarate-dependent to remove methyl groups. Readers, such as proteins with bromodomains (for acetylated s) or chromodomains (for methylated s), often contain enzymatic domains themselves, enabling "writers that read" to reinforce modification patterns through feedback loops. The most prevalent histone modifications targeted by these enzymes include (e.g., H3K27ac associated with active enhancers), in various states (e.g., activating or repressive and ), and ubiquitylation (e.g., H2AK119ub1 linked to ). These modifications exhibit ; for instance, H2BK120 ubiquitylation stimulates H3K4 and H3K79 by writers like MLL and DOT1L, while H3K36me3 recruits erasers like HDACs to deacetylate nearby histones, preventing aberrant transcription. Positive and mechanisms ensure stable domains: activating marks like recruit additional writers via readers such as CFP1, whereas repressive marks like and H2AK119ub1 mutually reinforce each other through PRC1 and PRC2 complexes. Beyond basic regulation, histone-modifying enzymes are essential for developmental decisions and cellular identity, where they coordinate modification patterns to activate pluripotency genes (e.g., via MLL-mediated on OCT4 and NANOG) or repress differentiation programs (e.g., via EZH2-directed ). Dysregulation of these enzymes contributes to human diseases, including cancers where mutations in or UTX alter , and neurodevelopmental disorders linked to impaired enzymes like CREBBP or JARID1C. Their therapeutic targeting, such as HDAC inhibitors in , underscores their in modulating epigenetic landscapes.

Overview

Definition and classification

Histone-modifying enzymes are a diverse group of proteins that catalyze the addition (writers) or removal (erasers) of covalent post-translational modifications (PTMs) on tails, thereby regulating structure and . These enzymes target specific residues, primarily lysines, arginines, serines, and threonines, within the N-terminal tails of core s (H2A, H2B, H3, and H4) or the linker histone H1. Unlike reader proteins that recognize and bind to these modifications without altering them, histone-modifying enzymes actively install or erase PTMs such as , , , and ubiquitination, influencing the accessibility of DNA to transcriptional machinery. These enzymes are classified primarily by the type of chemical modification they catalyze, encompassing both writers that add functional groups and erasers that remove them. For , writers include acetyltransferases (HATs), divided into families such as (e.g., GCN5) and (e.g., MOF), while erasers comprise deacetylases (HDACs), categorized into four classes: class I (e.g., , HDAC2, HDAC3), class II (subdivided into IIA and IIB), class III (sirtuins), and class IV (HDAC11). involves methyltransferases (HMTs), which add methyl groups to lysines or arginines and include SET domain-containing families like EZ (e.g., in the Polycomb repressive complex) and SUV39, with erasers being demethylases (HDMs) such as the KDM family (e.g., /LSD1) and JmjC-domain proteins. is mediated by kinases (writers, e.g., kinases targeting serine/ residues) and phosphatases (erasers, e.g., PP2A), while ubiquitination relies on E3 ubiquitin ligases (writers, e.g., RING1B) and deubiquitinases (DUBs, erasers, e.g., BAP1). Less common modifications, such as sumoylation or , involve specialized enzymes but follow a similar writer-eraser . Histone-modifying enzymes exhibit strong evolutionary conservation across eukaryotes, from simple organisms like (e.g., conserved HDAC homologs such as Rpd3) to complex mammals including humans, underscoring their fundamental role in . In humans, the encodes hundreds of such enzymes, with approximately 130 identified writers and erasers for and alone, with recent genomic analyses identifying 32 HATs, 20 HDACs, 55 HMTs, and 23 HDMs. Key examples include the transcriptional co-activators p300 and CBP as versatile HATs that acetylate multiple histone lysines to promote open , as a repressive HMT that trimethylates H3K27, and class I HDACs (HDAC1-3) that deacetylate histones to facilitate compaction.
Modification TypeWritersErasersKey Families/Examples
AcetylationHATsHDACsGNAT (GCN5), MYST (MOF); Class I (HDAC1-3), Sirtuins
MethylationHMTsHDMsEZ (EZH2), SUV39; KDM (LSD1), JmjC
PhosphorylationKinasesPhosphatases kinases; PP2A
UbiquitinationE3 ligasesDUBsRING1B; BAP1

Biological significance

Histone-modifying enzymes play a pivotal role in epigenetic by catalyzing post-translational modifications (PTMs) on tails, which collectively form the " code" that dictates structure and function. This code enables the transition between compact , which represses , and open , which facilitates access for transcription factors and the transcriptional machinery. The code hypothesis, proposed by Jenuwein and Allis in 2001, posits that specific combinations of PTMs serve as binding platforms for effector proteins, thereby extending the informational content beyond the DNA sequence itself. Subsequent studies have validated this concept through the identification of combinatorial PTM patterns that recruit distinct regulators, influencing genome-wide transcriptional outputs. The dynamic balance between histone-modifying "writers" (e.g., acetyltransferases and methyltransferases) and "erasers" (e.g., deacetylases and demethylases) allows cells to rapidly adjust states in response to environmental cues such as stress or developmental signals. For instance, histone acetylation typically promotes activation by loosening structure, while can either activate or repress depending on the modified residue, enabling context-specific regulation. This reversibility ensures precise control over during processes like or adaptation to stressors, where PTM levels fluctuate to maintain epigenetic . Histone modifications also exhibit crosstalk with other epigenetic mechanisms, particularly and non-coding RNAs, to fine-tune dynamics with a focus on histone-specific effects. For example, certain histone PTMs can recruit DNA methyltransferases to reinforce , while non-coding RNAs may guide histone-modifying complexes to target loci, amplifying regulatory outcomes. These interactions highlight the integrated nature of epigenetic control, where histone PTMs serve as central hubs for coordinating broader genomic responses.

Histone context

Histone structure

Histones are small, basic proteins that package DNA into nucleosomes, the fundamental units of chromatin. The core histones—H2A, H2B, H3, and H4—each exist as two copies that assemble into a histone octamer around which approximately 147 base pairs of DNA are wrapped in about 1.65 left-handed superhelical turns. This octameric structure positions the histones in a disk-like configuration, with the DNA groove facing the histone surface to facilitate tight binding through electrostatic interactions and hydrogen bonds. In addition to these core histones, the linker histone H1 binds to the entry and exit points of the DNA on the nucleosome, stabilizing higher-order chromatin folding by bridging adjacent nucleosomes and promoting compaction into 30-nm fibers. The N-terminal tail domains of core histones protrude from the nucleosome core and are unstructured, allowing flexibility for interactions with DNA and other proteins. These tails, typically 20–40 residues long in H3 and H4, are rich in positively charged lysine (K) and arginine (R) residues, as well as serine (S) residues, making them prime targets for post-translational modifications (PTMs) such as acetylation, methylation, and phosphorylation. For instance, the N-terminal tail of histone H3 contains key modifiable sites including lysines at positions 4 (K4), 9 (K9), and 27 (K27), which are frequently methylated to influence chromatin accessibility. The tails' dynamic nature enables them to extend outward or associate weakly with adjacent DNA, contributing to the nucleosome's overall accessibility for regulatory factors. In contrast, the globular domains form the central, structured core of the histone octamer, mediating the primary contacts with DNA through a series of alpha-helices and loops that insert into the DNA minor groove at 14 distinct sites. These domains are more rigid and less prone to modifications compared to the tails, but certain residues within them, such as lysine 120 on H2B (K120), can undergo ubiquitination, which subtly alters nucleosome stability and histone-DNA interactions without disrupting the core architecture. Histone variants introduce sequence diversity that modulates properties and patterns relative to histones. For example, H2A.Z, which differs from H2A primarily in its C-terminal domain and acidic patch, incorporates into at promoter and enhancer regions, influencing the recruitment of modification enzymes and altering local dynamics. Such variants expand the epigenetic repertoire by fine-tuning the nucleosome's structural and functional landscape.

Chromatin dynamics

Chromatin dynamics refer to the structural transitions of from compact to open states, primarily driven by histone modifications that alter interactions and accessibility. , the basic units of , assemble into arrays where binds to between nucleosomes, promoting the folding of these arrays into a 30 nm solenoid-like fiber through electrostatic interactions between tails and adjacent nucleosomes. Post-translational modifications (PTMs) on tails modulate these interactions; for instance, neutralizes positive charges on residues, weakening internucleosomal contacts and loosening the 30 nm fiber structure, while certain methylations can enhance compaction by facilitating protein recruitment that stabilizes the fiber. This dynamic regulation allows to transition between condensed and decondensed states, influencing DNA accessibility for cellular processes. At higher-order levels, histone modifications regulate the formation of chromatin loops and topologically associating domains (TADs), which organize the into functional compartments. Modifications such as , deposited by Polycomb repressive complexes, promote chromatin compaction by inducing phase separation and loop extrusion independent of boundaries in some cases, thereby silencing gene clusters within TADs. In contrast, active marks like and H3K27ac enrich open chromatin regions, facilitating enhancer-promoter interactions within TADs and active compartments, with regulation-associated modules defined by these marks insulating specific genomic neighborhoods. These modifications thus dictate the of , linking local dynamics to genome-wide architecture. Histone-modifying enzymes collaborate with ATP-dependent remodeling complexes, such as , to reposition and expose or conceal DNA. Acetylation on histone tails, for example, enhances binding via bromodomains, promoting nucleosome sliding and eviction at promoters to increase accessibility. This synergy allows precise control over , where PTMs serve as signals that guide the mechanical actions of remodelers. Quantitatively, histone modifications exhibit site-specific densities that correlate with functional regions; for instance, is highly enriched at transcription start sites of active promoters, often spanning broad domains that amplify regulatory signals. Such targeted enrichment ensures that only a subset of nucleosomes bears specific marks, enabling efficient reconfiguration without global disruption.

Common modifications and enzymes

Acetylation and deacetylation

Histone acetyltransferases (HATs), also known as lysine acetyltransferases (KATs), are enzymes that catalyze the transfer of an acetyl group from acetyl-coenzyme A (acetyl-CoA) to the ε-amino group of lysine residues on histone tails, primarily on histones H3 and H4. This modification neutralizes the positive charge of lysine, influencing chromatin structure. HATs are classified into several families based on structural and sequence homology, including the GCN5-related N-acetyltransferase (GNAT) family, which encompasses enzymes like GCN5 and p300/CBP-associated factor (PCAF), and the MYST family, named after its founding members MOZ, Ybf2/Sas3, Sas2, and Tip60. The GNAT family members, such as GCN5 and PCAF, preferentially acetylate lysines like H3K9 and H3K14, while MYST family enzymes, including MOF and TIP60, target sites such as H4K16 and H3K56. The catalytic mechanism of HATs involves a conserved core domain where an glutamate residue acts as a general base to deprotonate the ε-amino group of the substrate , facilitating nucleophilic attack on the carbonyl carbon of and subsequent transfer of the acetyl group. This process is highly specific, with HATs often recruited to promoters and enhancers through interactions with transcription factors, enabling targeted that correlates with active states. In humans, there are approximately 30 known HATs distributed across these families, with the and families being prominent for histone modifications. Notable examples include p300 and CREB-binding protein (CBP), which belong to a distinct HAT family and function as transcriptional co-activators by acetylating multiple histone lysines, such as H3K27, in addition to non-histone proteins. These enzymes integrate signals from various activators and are essential for enhancer activation. Histone deacetylases (HDACs) counteract acetylation by hydrolytically removing acetyl groups from lysine residues, restoring the positive charge and promoting chromatin compaction. In humans, there are 18 HDACs divided into four classes: class I (HDAC1, HDAC2, HDAC3, HDAC8), which are zinc-dependent enzymes primarily localized in the nucleus; class II, further subdivided into IIa (HDAC4, HDAC5, HDAC7, HDAC9) and IIb (HDAC6, HDAC10), which shuttle between nucleus and cytoplasm; class III, the sirtuins (SIRT1–7), which are NAD+-dependent; and class IV (HDAC11). Class I HDACs, such as HDAC1, exhibit broad substrate specificity and are recruited to target sites via co-repressor complexes like Sin3, where they deacetylate marks like H3K9ac to facilitate gene repression. The mechanism for classical HDACs (classes I, II, and IV) involves a in the that coordinates a molecule to generate a , which performs nucleophilic attack on the acetyl carbonyl, leading to deacetylation and release; for example, employs this Zn2+-mediated to remove acetyl groups from H3K9ac. In contrast, class III sirtuins utilize NAD+ as a cofactor, coupling deacetylation to and O-acetyl-ADP-ribose production. HDACs often display specificity through complex formation, with class I enzymes associating with Sin3 for promoter targeting and class II enzymes responding to signaling cues for dynamic localization.

Methylation and demethylation

Histone methylation involves the covalent addition of one or more methyl groups to the ε-amino group of residues or the guanidino group of residues on tails, primarily and H4, resulting in mono-, di-, or tri-methylated states that influence structure and . This modification is dynamically regulated by histone methyltransferases (HMTs), which add methyl groups, and histone demethylases (HDMs), which remove them, with the specific state determining whether the mark is activating or repressive. In humans, there are approximately 50 such enzymes, including both HMTs and HDMs, enabling precise control over epigenetic landscapes. Histone methyltransferases for residues, known as lysine methyltransferases (KMTs), typically contain a conserved SET domain and utilize S-adenosylmethionine () as the methyl donor in an SN2-like nucleophilic attack , where the substrate attacks the electrophilic carbon of 's , displacing the adenosyl . A prominent example is SUV39H1, a SET domain-containing KMT that catalyzes di- and trimethylation of at 9 (H3K9me2/3), establishing repressive domains. For residues, protein arginine methyltransferases (PRMTs) perform similar -dependent ; PRMT1, for instance, monomethylates H4 at 3 (H4R3me1), promoting active transcription. Histone demethylation is mediated by two main classes of HDMs. The (FAD)-dependent (LSD1, also ) oxidatively removes methyl groups from mono- and dimethylated lysines, such as H3K4me1/2, via an oxidation mechanism that generates an intermediate, ultimately producing and as byproducts. The jumonji C (JmjC) domain-containing demethylases (KDM2-8), which constitute the majority of HDMs, function as (II)- and 2-oxoglutarate (2OG)-dependent dioxygenases; for example, KDM6A/B (UTX/JMJD3) demethylate through oxidative of 2OG, yielding succinate, CO2, and . The functional outcomes of methylation are highly context-dependent, dictated by the residue, methylation state, and genomic location. Trimethylation of H3K4 (H3K4me3), catalyzed by the MLL (KMT2) complex, marks active promoters and enhancers, facilitating transcription initiation. In contrast, H3K9me3, deposited by G9a (EHMT2), and H3K27me3, mediated by EZH2 within the Polycomb Repressive Complex 2 (PRC2), are repressive marks associated with gene silencing and chromatin compaction. The combinatorial nature of methylation states allows for recruitment of specific reader proteins that interpret these marks to propagate epigenetic memory. For instance, (HP1) isoforms bind H3K9me2/3 via their chromodomains, promoting further spreading and transcriptional repression. This reader-mediated recognition underscores the reversible and tunable regulation of by dynamics.

Phosphorylation and dephosphorylation

Histone involves the covalent addition of phosphate groups to specific residues, primarily , , and , on tails or cores, which introduces a negative charge that can alter structure and accessibility. This modification is dynamically regulated by kinases that catalyze the transfer of the γ-phosphate from ATP to the hydroxyl group of the target residue, facilitating rapid responses to cellular signals such as or . Unlike more stable modifications like , is highly reversible, enabling quick adjustments in conformation. Key histone kinases include Aurora B, which phosphorylates at serine 10 (H3S10ph) during to promote chromatin condensation by displacing (HP1). (CDK1) targets serine 10 on (H1S10ph), contributing to linker histone dissociation and mitotic . (JAK2) phosphorylates tyrosine 41 on H3 (H3Y41ph) in hematopoietic cells, leading to HP1α eviction and enhanced transcription upon signaling. These kinases represent a subset of the approximately 10 major enzymes dedicated to histone , underscoring the specificity of this modification despite the abundance of general kinases in the cell. Dephosphorylation is mediated by protein phosphatases that hydrolyze the phosphate ester bond, restoring the neutral residue and reversing the structural effects. Protein phosphatase 2A (PP2A) specifically removes the phosphate from H3S10 post-mitosis, facilitating decondensation and progression through the . PPM1D (also known as WIP1) dephosphorylates serine 139 on H2AX (γ-H2AX), terminating DNA damage signaling after repair and preventing prolonged checkpoint activation. This reversibility allows phosphorylation to serve as a transient switch in response to extracellular cues. Phosphorylation exhibits site-specific functions, with H3S10ph and H3S28ph marking mitotic entry to drive condensation and segregation. In contrast, γ-H2AX at S139ph forms foci at DNA double-strand breaks, recruiting repair factors like 53BP1 and to facilitate or . These modifications often interplay with others; for instance, H3S10 adjacent to methylated residues triggers a "phospho-methyl switch," ejecting methyl-binding readers like HP1 to expose for remodeling.

Ubiquitination and deubiquitination

Ubiquitination of histones involves the covalent attachment of , a 76-amino-acid protein, primarily as a monoubiquitin modification on residues of and H2B, which serves as a signaling platform rather than a signal. This process is mediated by a hierarchical enzymatic cascade consisting of E1 ubiquitin-activating enzymes, which use ATP to form a bond with ; E2 ubiquitin-conjugating enzymes, which receive the activated ; and E3 ubiquitin ligases, which provide substrate specificity and catalyze the transfer to target lysines on histones. In the context of histones, monoubiquitination predominates and distinguishes regulatory functions from polyubiquitination, which typically targets proteins for proteasomal . Key ligases include the RNF20/RNF40 heterodimer, which monoubiquitinates at 120 (H2Bub1) in a process requiring the E2 enzyme UBE2A/B (also known as UbcH5a/b) or UbcH6. RNF20/RNF40 is recruited to transcribing via the PAF1 complex (hPAF1 in humans), linking H2Bub1 to transcription elongation by facilitating processivity. Another major ligase is the Polycomb Repressive Complex 1 (PRC1), which monoubiquitinates at 119 (H2AK119ub1), promoting repression through compaction and recruitment of PRC2 for H3K27 . PRC1's domain subunits, such as RING1A/B, confer specificity in this repressive modification. Deubiquitination reverses these marks via deubiquitinases (DUBs), a family of over 90 enzymes in humans, with approximately 20 implicated in regulation through hydrolytic cleavage of the between ubiquitin's C-terminal 76 and the (Gly76-Gly77 linkage). Prominent DUBs include USP16, which specifically removes from H2AK119 to counteract PRC1-mediated repression and facilitate during processes like X-chromosome reactivation. BAP1, often in complex with ASXL1, deubiquitinates H2AK119ub1 and other H2A sites, influencing and tumor suppression by modulating Polycomb silencing. These DUBs exhibit context-dependent specificity, with USP16 associating with the MLL complex for balanced regulation. H2B K120 ubiquitination by RNF20/40 exemplifies functional specificity, as it acts as a prerequisite for downstream events, such as H3K4 and H3K79 trimethylation by SET1/ and DOT1L complexes, respectively, thereby enhancing transcriptional output without directly altering structure. In contrast, H2AK119ub1 by PRC1 reinforces stability. Monoubiquitination thus integrates signaling cascades, with polyubiquitin chains on histones being rare and typically linked to DNA damage responses rather than routine .

Less common and emerging modifications and enzymes

O-GlcNAcylation is a dynamic post-translational modification involving the attachment of a single β-N-acetylglucosamine (GlcNAc) moiety to serine or threonine residues of nuclear and cytoplasmic proteins, including core histones H2A, H2B, H3, and H4. This modification was first identified on histones in 2010, where it was shown to increase under heat stress conditions, promoting chromatin condensation and protecting genomic DNA. Unlike extracellular glycosylation, O-GlcNAcylation occurs entirely within the cell and serves as a nutrient sensor, reflecting the availability of glucose and other metabolites through the hexosamine biosynthetic pathway (HBP), which generates the donor substrate UDP-GlcNAc. Histone O-GlcNAcylation influences chromatin architecture, gene transcription, and DNA repair by competing with or modulating other modifications, such as phosphorylation at overlapping sites. The addition of O-GlcNAc to s is catalyzed exclusively by O-GlcNAc transferase (OGT), a complex that transfers GlcNAc from UDP-GlcNAc to target residues. OGT exists in multiple isoforms generated by , with the nucleocytoplasmic form featuring an N-terminal tetratricopeptide repeat (TPR) that mediates protein-protein interactions and substrate specificity, alongside catalytic domains for glycosyl transfer. For histone targeting, OGT interacts with regulators like TET2, which helps direct modification to specific sites such as H2BS112, thereby facilitating transcriptional activation through enhanced H2B monoubiquitination at K120. A seminal example is H2BS112-GlcNAc, which recruits the factor nibrin (NBN/NBS1) to promote and during DNA damage response. OGT's activity is tightly regulated by nutrient levels, as elevated glucose flux through the HBP increases UDP-GlcNAc pools, amplifying O-GlcNAcylation and linking metabolic status to epigenetic outcomes. The removal of O-GlcNAc from histones is performed by O-GlcNAcase (OGA), the sole enzyme hydrolyzing the N-acetylglucosamine-β linkage via its catalytic domain, which exhibits β-N-acetylglucosaminidase activity. OGA has two main isoforms: the longer form (lOGA) includes a C-terminal (HAT)-like domain that may influence interactions, while the shorter form (sOGA) lacks this region and predominates in the . Substrate specificity of OGA favors sites with nearby hydrophobic residues, allowing efficient deglycosylation of histones to restore Ser/Thr availability for other modifications. This cycling between OGT and OGA enables rapid responses to cellular signals, such as stress or metabolic shifts. O-GlcNAcylation on s often competes directly with at shared Ser/Thr sites, creating reciprocal regulation that fine-tunes dynamics; for instance, O-GlcNAc at H3S10 or H3S28 inhibits mitotic , reducing condensation. This is evident in the reciprocal modification of H2BS112, where GlcNAc and forms mutually exclude each other, with the GlcNAc variant supporting transcription and repair. Overall, histone O-GlcNAcylation occurs at low abundance, underscoring its role as a fine modulator rather than a dominant mark. Through these mechanisms, O-GlcNAcylation integrates sensing with epigenetic control, influencing and genomic stability.

Sumoylation and desumoylation

Sumoylation involves the covalent attachment of small ubiquitin-like modifier () proteins to residues on target proteins, including s, through a multi-step enzymatic cascade analogous to ubiquitination but utilizing SUMO paralogs SUMO1, SUMO2, and SUMO3. The process begins with the activation of SUMO by the E1 enzyme (SAE1/SAE2), followed by conjugation via the E2 enzyme UBC9, and enhancement of specificity by E3 ligases such as the PIAS family. Members of the PIAS family, including PIAS1, act as SUMO E3 ligases to promote histone sumoylation, with PIAS1 facilitating the modification of histones H3 and H2B in a manner dependent on its SAP domain and ligase activity. Desumoylation is mediated by SUMO-specific proteases known as SENPs, which cleave the isopeptide bond between SUMO and the target lysine, thereby reversing the modification. SENP1, for instance, deconjugates SUMO from histone H2B, influencing transcriptional regulation by altering chromatin accessibility. Other SENPs, such as SENP2 and SENP3, contribute to the dynamic control of sumoylation on histones, ensuring transient modifications that respond to cellular signals. Histone sumoylation exhibits specificity, particularly on H2A and H2B during DNA damage responses, where it facilitates repair processes by modulating structure. This modification also promotes formation and maintenance, often affecting less than 5% of total in a transient manner to fine-tune . For example, sumoylation enhances the recruitment of histone deacetylases (HDACs), such as , to , thereby reinforcing transcriptional repression through deacetylation and compaction.

ADP-ribosylation and de-ADP-ribosylation

involves the covalent attachment of ADP-ribose units, derived from NAD+, to specific residues on target proteins, including histones, thereby modulating structure and function in rapid cellular responses. This modification can occur as mono-ADP-ribosylation (MARylation) or poly-ADP-ribosylation (PARylation), with the latter forming linear or branched chains that amplify signaling. On histones, primarily targets glutamate (Glu), aspartate (Asp), and serine (Ser) residues, particularly on linker and core histone H2B, facilitating relaxation and access during responses. The primary writers of histone ADP-ribosylation are poly(ADP-ribose) polymerases (PARPs), a family of 17 enzymes in humans, with PARP1 being the most abundant and central to nuclear functions. PARP1, often in complex with histone PARylation factor 1 (HPF1), catalyzes serine-specific ADP-ribosylation on histones, such as Ser residues on H2B and H1, while also modifying Glu and Asp sites like Glu2 on H1.5 and Glu35 on H2B. These enzymes are activated by binding to DNA breaks, such as single-strand breaks, triggering auto-PARylation on PARP1 itself, which generates branched PAR chains to recruit repair factors like XRCC1 and amplify the damage signal at repair foci. For instance, PARP1 auto-PARylation at DNA damage sites promotes the assembly of nucleosomes and enhances histone H1 binding, thereby coordinating chromatin remodeling for efficient repair. Erasure of ADP-ribosylation is mediated by hydrolases, including poly(ADP-ribose) glycohydrolase (PARG) and ADP-ribosylhydrolase 3 (ARH3). PARG acts as an endo- and exo-glycohydrolase, efficiently degrading poly(ADP-ribose) chains and, in some cases, converting them to mono units, particularly on PARP1. In contrast, ARH3 is a mono-specific hydrolase that preferentially removes serine-linked mono-ADP-ribose from histones, such as H3 and H2B tails, achieving up to 80% demodification efficiency compared to PARG's lower activity on mono forms. This specificity ensures timely reversal of serine-ADP-ribosylation marks at repair sites, preventing persistent chromatin scars as seen in ARH3-deficient cells. Histone ADP-ribosylation, particularly on H1.5 at Glu2, marks repair foci to facilitate DNA damage response, though broader roles in transcription and replication are also noted.

Citrullination

Citrullination, also known as deimination, is a post-translational modification in which peptidylarginine deiminase (PAD) enzymes convert positively charged arginine residues in proteins, including histones, to neutral citrulline by hydrolyzing the guanidino group and releasing ammonia (NH3). This calcium-dependent process alters histone charge and reduces potential sites for arginine methylation, thereby influencing chromatin structure and gene expression. Among the PAD family, PAD2 and PAD4 are the primary enzymes responsible for histone citrullination in mammalian cells, with PAD4 showing nuclear localization and prominent activity on core histones. Specifically, PAD4 citrullinates histone H3 at arginine residues R2, R8, and R17, while PAD2 contributes to similar modifications, particularly in non-neutrophil contexts. Unlike many histone modifications, citrullination lacks a dedicated to directly reverse it back to , making it effectively irreversible at the covalent level. Reversal occurs indirectly through histone or degradation, allowing replacement with unmodified s. This irreversibility underscores citrullination's role in stable epigenetic changes, often competing with reversible modifications like . Citrullination exhibits site-specific effects on dynamics; for instance, citrullination at H3R17 prevents subsequent by PRMT4/CARM1, which normally promotes p300-mediated at adjacent H3K18, thereby inhibiting H3K18 and repressing receptor-regulated . This modification is prominently linked to (NETs), where PAD4-mediated citrullination promotes decondensation and extracellular release during immune responses. In , PAD4 drives citrullination at multiple sites, facilitating defense but also contributing to pathological processes when dysregulated. Overall, by neutralizing arginine's positive charge, citrullination disrupts electrostatic interactions in nucleosomes, often leading to transcriptional repression or .

Proline isomerization

Proline isomerization represents a non-covalent modification of tails, distinct from the covalent alterations like or , as it involves enzymatic flipping of the between a preceding residue and from to trans conformation or vice versa. This dynamic structural change modulates the flexibility and accessibility of tails, thereby influencing interactions with other proteins and the of downstream effectors without adding or removing chemical groups. Peptidyl-prolyl isomerases (PPIases) catalyze this process, accelerating the inherently slow interconversion that occurs spontaneously at rates too low for biological . The enzymes responsible belong to several PPIase families, including cyclophilins and parvulins, with specificity often dictated by adjacent modifications such as . In , the Fpr4 binds tails via its nucleophilin-like domain and isomerizes the bonds at H3P30 and H3P38 using its PPIase domain, thereby inhibiting Set2-mediated at H3K36 and when the proline is in the cis state. This specificity arises because the trans conformation of H3P38 positions the H3 tail optimally for Set2 binding, while the cis form disrupts it, demonstrating how enforces a conformational gate for subsequent modifications. In mammals, the parvulin Pin1 exemplifies a phosphorylation-dependent PPIase that targets Ser/Thr-Pro motifs following their phosphorylation, catalyzing to alter protein conformation and function. Although primarily studied in signaling proteins, Pin1 also acts on histones, such as binding phosphorylated sites in the C-terminal domain of (e.g., pS173 and pS187) to promote its dephosphorylation by protein phosphatase 2A (PP2A), which stabilizes H1 association with transcriptionally active . Pin1's preference for post-phosphorylation substrates links to core histone modifications in H2A and H3 tails, where it facilitates exposure of sites for further regulatory changes. Proline isomerization is a rare event, confined to select X-Pro bonds in histone tails—typically those adjacent to modifiable residues like serine or threonine—rather than the majority of prolines in the proteome. This selectivity ensures precise control over tail dynamics, often coupling with phosphorylation to trigger conformational shifts essential for processes like mitosis. For instance, Pin1 isomerization of phosphorylated motifs in mitotic regulators promotes cell cycle progression, indirectly supporting histone tail rearrangements that enable modifications such as H3S10 phosphorylation during chromosome condensation. Unlike covalent histone modifiers that directly alter charge or add bulk, PPIases like Pin1 and Fpr4 emphasize transient structural toggles, providing a rapid, reversible layer of epigenetic regulation. Proline isomerization often synergizes with phosphorylation, as seen in Pin1's reliance on prior Ser/Thr phosphorylation to engage histone substrates, thereby coordinating sequential tail modifications.

Novel acylation modifications (e.g., lactylation, crotonylation)

Novel modifications represent an expanding class of histone posttranslational modifications that extend beyond traditional , incorporating acyl groups derived from diverse metabolic intermediates such as , crotonyl-CoA, and succinyl-CoA. These modifications, primarily on residues, influence structure and by altering histone charge and recruiting specific reader proteins, often linking cellular directly to epigenetic . Since their initial discoveries in the early , advances in have revealed over a dozen new types of histone acylations, including butyrylation, propionylation, malonylation, and glutarylation, highlighting their prevalence in active genomic regions. Histone lactylation involves the attachment of a lactyl group to residues, notably H3K18la, catalyzed by the acetyltransferase p300 using as the acyl donor. This modification was first identified in in macrophages undergoing , where it accumulates at enhancer regions to promote the transcription of inflammation-related genes, such as those involved in signaling. Unlike , lactylation directly couples glycolytic flux to epigenetic activation, with elevated levels under driving its deposition. Crotonylation, marked by a four-carbon crotonyl group on lysines like H3K27cr, is mediated by histone acetyltransferases such as p300 and PCAF, utilizing crotonyl-CoA derived from or . Discovered in 2011, this modification enhances instability and accessibility more effectively than , facilitating robust activation at promoters and enhancers. Decrotonylation is primarily handled by family deacylases, including SIRT3, which uses NAD+ to remove the crotonyl group in a manner distinct from deacetylation due to the longer acyl chain. Succinylation attaches a five-carbon succinyl group to histones, such as H4K succinylation, catalyzed by enzymes such as HAT1 and p300, as well as non-enzymatic mechanisms driven by high levels from the tricarboxylic acid cycle. This modification neutralizes charge similarly to but is reversed by the mitochondrial SIRT5 through NAD+-dependent desuccinylation, regulating metabolic and stability. Butyrylation and propionylation, involving C4 and C3 acyl chains respectively, exhibit functional parallels to crotonylation, enriching at transcription start sites and promoting open , as revealed by quantitative profiling. These novel acylations underscore the metabolic sensitivity of the epigenome, with p300-like HATs moonlighting as versatile acyltransferases across multiple substrates.

Functional roles

Gene expression and epigenetic memory

Histone-modifying enzymes play a central in regulating by depositing specific marks on histone tails that either promote or inhibit transcriptional activity. Activating modifications, such as trimethylation of at lysine 4 () catalyzed by the MLL family of methyltransferases, facilitate the recruitment of (Pol II) to promoter regions, enabling efficient transcription initiation and elongation. Similarly, acetylation of H3 at lysine 27 (), primarily mediated by the p300/CBP acetyltransferases, enhances accessibility at enhancers and promoters, further supporting Pol II engagement and gene activation. In contrast, repressive marks like trimethylation of H3 at lysine 27 (), deposited by the subunit of the Polycomb Repressive Complex 2 (PRC2), compact and recruit additional silencing factors, thereby inhibiting Pol II progression and maintaining transcriptional repression at target loci. These modifications also contribute to epigenetic memory, ensuring the stable inheritance of states across cell divisions. In embryonic stem cells, bivalent domains—characterized by the coexistence of activating and repressive —poise developmental genes for rapid activation or repression during , allowing cells to retain while preventing premature commitment. This stability arises from self-propagating mechanisms involving "writer" enzymes: for instance, recruits PRC2 to propagate the mark on newly synthesized histones during replication, while is reinforced by MLL complexes in a that recognizes and methylates adjacent nucleosomes. Such read-write dynamics enable the transmission of states without altering the underlying DNA sequence, forming the basis of heritable epigenetic . Notable examples illustrate these principles in developmental processes. During X-chromosome inactivation in female mammals, PRC2-mediated spreads along the inactive , enforcing stable silencing of non-essential genes and maintaining dosage compensation across generations. In genomic imprinting, particularly in placental tissues, the G9a (EHMT2) methyltransferase deposits H3K9 dimethylation (H3K9me2) at imprinting control regions, cooperating with to silence the maternal or paternal selectively and ensuring parent-of-origin-specific expression. These histone modifications exhibit partial persistence through DNA replication, remaining enriched at target loci post-S phase, bolstered by ongoing enzymatic reinforcement to counteract dilution on new histones. This temporal window allows for the re-establishment of full modification levels, underscoring the dynamic yet faithful nature of epigenetic memory in steady-state gene regulation.

DNA repair and cell cycle regulation

Histone-modifying enzymes play crucial roles in DNA repair by establishing and interpreting specific modifications that facilitate the recruitment of repair machinery to sites of damage. Upon detection of double-strand breaks (DSBs), the ATM kinase phosphorylates histone H2AX at serine 139 to form γH2AX, which serves as a platform for recruiting poly(ADP-ribose) polymerase 1 (PARP1) and histone deacetylases (HDACs) to chromatin flanks, promoting efficient DSB signaling and repair initiation. In homologous recombination (HR), the methyltransferase DOT1L deposits H3K79 methylation (H3K79me), which is essential for recruiting HR factors and completing repair, as DOT1L deficiency impairs HR-mediated DSB resolution. Additionally, ubiquitination by E3 ligases RNF8 and RNF168 targets histones H2A and H2AX at lysine 13/15, amplifying damage signals and enabling downstream effector recruitment to orchestrate repair pathway choice. In cell cycle regulation, these enzymes coordinate dynamics to ensure proper progression through phases, particularly during transitions vulnerable to replication stress. The Aurora B phosphorylates at serine 10 (H3S10ph) during , which is required for condensation and segregation, as Aurora B inhibition disrupts H3S10ph and leads to condensation defects. For the , cyclin-dependent s (CDKs), such as CDK2, promote acetylation indirectly by phosphorylating transcriptional repressors like pRB, disrupting their association with HDACs and thereby increasing acetylation levels to facilitate S-phase entry and replication origin licensing. Specific histone marks guide repair pathway selection, exemplified by the reader protein 53BP1, which binds dimethylated H4K20 (H4K20me2) and ubiquitinated H2AK15 (H2AK15ub) to favor (NHEJ) over , ensuring accurate re-ligation of DSBs in . Following repair completion, enzymes actively reset these modifications; for instance, 4 (PP4) dephosphorylates γH2AX, while other phosphatases reverse ubiquitination and methylation to restore integrity and terminate signaling. DNA damage checkpoints integrate histone-modifying enzymes to maintain fidelity, where phosphatases such as PP1, PP2A, and PP4 reverse phosphorylation marks like γH2AX and H3S10ph upon repair, allowing checkpoint recovery and preventing premature re-entry that could propagate errors. This reversal ensures coordinated progression, as persistent marks would sustain arrest, while timely by these enzymes signals successful repair and resumption of the .

Disease associations

Cancer

Histone-modifying enzymes play critical roles in oncogenesis through mutations and aberrant expression that disrupt epigenetic landscapes, leading to uncontrolled and tumor progression. Dysregulation of these enzymes, including histone methyltransferases (HMTs) and deacetylases (HDACs), is frequently observed across various cancer types, contributing to the silencing of tumor suppressor genes and activation of oncogenes. Overexpression of the HMT , a key component of the Polycomb repressive complex 2 (PRC2), is a hallmark of certain lymphomas, where it drives hypertrimethylation of at lysine 27 (), resulting in repressive states that silence tumor suppressor genes and promote lymphomagenesis. Gain-of-function mutations in , such as Y641F, further enhance this hyper-repressive activity, distorting global profiles and facilitating development. Similarly, HDACs exhibit oncogenic roles in solid tumors by deacetylating histones, which compacts and represses pro-apoptotic and anti-proliferative genes, thereby enhancing survival and resistance to therapy. Aberrant HDAC expression is linked to tumor progression in cancers such as breast and colorectal, where it sustains oncogenic signaling pathways. In contrast, certain histone-modifying enzymes function as tumor suppressors when disrupted. Fusions involving the HMT MLL (also known as KMT2A), which normally catalyzes to activate transcription, are prevalent in acute leukemias and lead to aberrant deposition at ectopic loci, driving leukemogenic gene expression programs and disrupting normal hematopoietic differentiation. The INHAT complex, comprising SET oncoprotein and /2, silences the tumor suppressor by inhibiting its acetylation and transcriptional activity, thereby impairing DNA damage responses and promoting genomic instability in cancers. Mutations in HDACs and HMTs contribute to tumorigenesis across diverse malignancies, as shown by pan-cancer genomic analyses. For instance, in , the BRAF mutation is associated with upregulation of HDACs, particularly HDAC8, which sustains a resistant transcriptional state and enhances tumor cell invasiveness. Mechanistically, global hypoacetylation, often resulting from HDAC overexpression, correlates with poor clinical prognosis in multiple cancers, including and pancreatic, by promoting compact structures that favor oncogenic and inhibit . Studies from the 2010s further established links between dysregulated modifications, such as altered H3K27me3 and patterns, and metastatic potential, demonstrating how these changes facilitate epithelial-mesenchymal transition and distant colonization in and cancers. In cancer, emerging evidence (as of 2025) links HDAC6 dysregulation to both tumor progression and shared pathways with neurodegeneration.

Neurodevelopmental and neurodegenerative disorders

Histone-modifying enzymes play critical roles in neurodevelopment by regulating accessibility and essential for neuronal , , and . Mutations in these enzymes disrupt epigenetic landscapes, leading to neurodevelopmental disorders such as (ASD). For instance, disruptive mutations in the chromodomain helicase DNA-binding protein 8 (CHD8), a remodeler that interacts with mixed-lineage leukemia (MLL) complexes to promote 4 trimethylation (), are strongly associated with . These mutations impair the establishment of active marks at neurodevelopmental genes, resulting in altered integrity and synaptic dysfunction observed in affected individuals. Similarly, the enhancer of zeste homolog 2 (), part of the polycomb repressive complex 2 (PRC2), maintains the balance between self-renewal and in neural stem cells by catalyzing to repress differentiation-promoting genes. Dysregulation of , such as loss-of-function variants, leads to premature neuronal and defects in cortical , contributing to and ASD-like phenotypes. In , the leading inherited cause of , increased H3K9 dimethylation (H3K9me2) at the promoter silences the gene, and the methyltransferase EHMT2 (G9a) drives this repressive mark, exacerbating synaptic gene misexpression and abnormalities. In neurodegenerative disorders, imbalances in histone-modifying enzymes contribute to , neuronal loss, and cognitive decline by altering the epigenetic control of stress response and genes. In (AD), a characterized by neurofibrillary tangles, histone deacetylase 6 (HDAC6) is hyperactive, deacetylating and to promote tau aggregation and impair . This hyperactivity correlates with disease progression, as HDAC6 upregulation in AD brains exacerbates tau pathology and synaptic loss. Inhibition of HDAC6 has been shown to reduce tau hyperphosphorylation and improve memory in AD models, highlighting its pathological role. Recent studies (as of 2025) have implicated novel acylation modifications like lactylation in AD tau pathology. In (PD), deficits in sirtuins SIRT1 and SIRT2 diminish deacetylation of histone H4 lysine 16 (H4K16ac), leading to aberrant chromatin opening at pro-apoptotic genes and α-synuclein aggregation in neurons. Reduced SIRT1 activity in PD patients impairs neuroprotective pathways, while SIRT2's preference for H4K16ac deacetylation, when deficient, contributes to and mitochondrial dysfunction. Amyotrophic lateral sclerosis (ALS), a often overlapping with , involves dysregulation of serine 10 (H3S10ph), mediated by kinases like Aurora B. In ALS models with fused in sarcoma (FUS) mutations, decreased H3S10ph levels disrupt condensation and transcriptional fidelity, promoting toxic aggregation and death. This modification's imbalance leads to misexpression of synaptic and survival genes, accelerating neurodegeneration. Overall, epigenetic dysregulation by histone-modifying enzymes underlies synaptic dysfunction and neuronal vulnerability in neurodevelopmental and neurodegenerative disorders, emphasizing their convergence on -mediated gene regulation.

Research and applications

Therapeutic targeting

Therapeutic targeting of histone-modifying enzymes has emerged as a promising strategy in and other diseases, with several s and activators advancing to clinical use or trials. (HDAC) s represent one of the most established classes, primarily targeting cancers where aberrant contributes to oncogenesis. , a broad-spectrum HDAC , received FDA approval in 2006 for the treatment of (CTCL) in patients with progressive, persistent, or recurrent disease on or following two systemic therapies. , another pan-HDAC , was approved in 2015 for with and dexamethasone in patients with who have received at least two prior standard regimens, though its U.S. approval was withdrawn in 2022 following a sponsor request due to insufficient confirmatory data. , a selective class I HDAC , gained FDA approval in 2009 for relapsed or refractory CTCL after at least one prior systemic therapy, demonstrating response rates of around 38% in clinical studies. Histone methyltransferase (HMT) inhibitors have also shown clinical promise, particularly for epigenetically driven malignancies. Tazemetostat, an inhibitor targeting the polycomb repressive complex 2 (PRC2), was granted accelerated FDA approval in 2020 for adults and pediatric patients aged 16 years and older with locally advanced or metastatic not eligible for complete resection, based on an overall response rate of 15% and durable responses. For mixed-lineage leukemia (MLL)-rearranged acute s, DOT1L inhibitors such as pinometostat (EPZ-5676) have been evaluated in phase 1/2 trials, showing target engagement through reduced H3K79 methylation and modest clinical activity, including complete remissions in a subset of relapsed/refractory pediatric and adult patients, though no approvals have been achieved as of 2025. Beyond HDACs and HMTs, other agents modulating histone-related modifications are in development. PARP inhibitors like olaparib, which interfere with poly(ADP-ribosyl)ation of histones and other proteins to impair DNA repair, have been approved since 2014 for homologous recombination (HR)-deficient cancers such as ovarian and breast cancers with BRCA mutations, exploiting synthetic lethality in epigenetically dysregulated tumors. For sirtuins (SIRTs), activators mimicking resveratrol, such as SRT2104—a selective SIRT1 agonist—were investigated in phase 1/2 clinical trials for conditions including metabolic disorders and neurodegeneration, demonstrating tolerability and exercise-mimetic effects, but development has been discontinued as of the early 2020s without achieving regulatory approval. Despite these advances, therapeutic targeting of histone-modifying enzymes faces significant challenges, including achieving isoform-specific inhibition to minimize off-target effects on non-histone proteins and normal cells, which can lead to toxicities like and gastrointestinal issues observed with HDAC inhibitors. Numerous epigenetic modulators, including histone-modifying inhibitors, remain in clinical development as of 2025, underscoring the field's momentum but highlighting the need for improved selectivity and strategies to enhance response rates and overcome .

Recent advances (post-2020)

Recent advances in the study of histone-modifying enzymes have illuminated intricate interplay between these enzymes and their substrates, particularly through high-resolution structural analyses. In 2023, cryo-electron microscopy (cryo-EM) structures revealed how G-quadruplex RNA inactivates polycomb repressive complex 2 (PRC2) by binding to its core subunits, thereby inhibiting H3K27 trimethylation (H3K27me3) deposition and disrupting gene silencing dynamics. Building on this, a 2024 cryo-EM study demonstrated that automethylation of the EZH2 subunit in PRC2 promotes dimerization on chromatin, enhancing catalytic activity for H3K27 methylation and providing mechanistic insights into allosteric regulation. Complementing these structural findings, mass spectrometry approaches in 2024 enabled the mapping of multi-post-translational modification (PTM) codes on histones during embryonic development, identifying combinatorial patterns of acetylation, methylation, and phosphorylation that coordinate lineage specification in mammalian cells. Novel roles for histone modifications have emerged in linking metabolic states to physiological rhythms and disease progression. In glioblastoma, a 2025 epigenomic atlas underscored HDAC-PARP crosstalk, where histone deacetylases (HDACs) modulate poly(ADP-ribose) polymerase (PARP) activity to alter H3K27ac levels, promoting tumor heterogeneity and therapeutic resistance. These findings highlight how enzyme-mediated PTMs integrate environmental cues with oncogenic signaling. Technological innovations have accelerated the precise manipulation and prediction of histone PTMs. CRISPR-based epigenome editors, advanced since 2020, now enable site-specific deposition of PTMs such as H3K9me3 or H3K27ac by fusing deactivated Cas9 (dCas9) to histone-modifying domains, allowing reversible control of gene expression in vivo without DNA cleavage. In parallel, 2024 AI models have improved histone code prediction by integrating convolutional neural networks with chromatin accessibility data, achieving high accuracy in forecasting gene expression from histone mark profiles across cell types. Key discoveries have expanded the known repertoire of histone modifications and their enzymatic regulators. A 2022 study established histone succinylation as a critical link between mitochondrial metabolism and disease, showing how elevated levels drive non-enzymatic succinylation of H3K122, enhancing accessibility in metabolic disorders like . Furthermore, proteomic screening post-2020 has identified approximately 113 novel histone marks, including previously unrecognized acylations, along with new enzyme-substrate pairs such as HDAC11-H3K14succ and novel methyltransferases for H4 variants, broadening the epigenetic landscape.

References

  1. [1]
    Histone Modifying Enzymes: Structures, Mechanisms, and Specificities
    In this review, we summarize recent advances in understanding these enzymes, highlighting studies of the histone acetyltransferases (HATs) p300 (also now ...
  2. [2]
    Histone-modifying enzymes: regulators of developmental decisions ...
    This review will focus on our current understanding of how histone-modifying enzymes coordinately regulate different histone modification patterns, and thus ...
  3. [3]
    The interplay of histone modifications – writers that read - EMBO Press
    The most abundant histone modifications are acetylation, phosphorylation, methylation, and ubiquitylation, although many other modifications have been reported ...
  4. [4]
    WERAM: a database of writers, erasers and readers of histone ...
    Oct 26, 2016 · From the scientific literature, we first collected 584 experimentally identified histone regulators, including 72 HATs, 97 HDACs, 116 acetyl- ...
  5. [5]
    Translating the Histone Code - Science
    We propose that a “histone code” exists that may considerably extend the information potential of the genetic (DNA) code. ... Under the assumption that a histone ...
  6. [6]
    Chromatin proteomics reveals novel combinatorial histone ...
    Sep 13, 2017 · By applying N-ChroP to dissect enhancers, we have provided further support to the histone code hypothesis, showing that distinct combinations of ...
  7. [7]
    Writing, erasing and reading histone lysine methylations - Nature
    Apr 28, 2017 · In this review, we summarize how histone lysine methylations are regulated by histone methyltransferases ('writers') and demethylases ('erasers') ...
  8. [8]
    Regulation of chromatin by histone modifications | Cell Research
    Feb 15, 2011 · Here, we describe the known histone modifications, define where they are found genomically and discuss some of their functional consequences.
  9. [9]
    Crosstalk between Noncoding RNAs and the Epigenetics Machinery ...
    These ncRNAs regulate various epigenetic processes, including DNA methylation and histone modification, by controlling chromatin-modifying enzymes such as ...
  10. [10]
    Crystal structure of the nucleosome core particle at 2.8 Å resolution
    Sep 18, 1997 · Luger, K., Mäder, A., Richmond, R. et al. Crystal structure of the nucleosome core particle at 2.8 Å resolution. Nature 389, 251–260 (1997).
  11. [11]
    Emerging roles of linker histones in regulating chromatin structure ...
    Oct 11, 2017 · H1 binds to the nucleosome to form the next structural unit of metazoan chromatin, the chromatosome, which may help chromatin to fold into ...
  12. [12]
    SETDB1: a novel KAP-1-associated histone H3, lysine 9-specific ...
    (C) Amino acid sequence of the N-terminal tail of histone H3 (1–30) is shown at the top with the K4, K9, and K27 residues highlighted. The various lysine to ...
  13. [13]
    Histone structure and nucleosome stability - PMC - NIH
    This review focuses on the histones including structural information, sequence variants, PTMs, interactions and contributions to chromatin organization.
  14. [14]
    Post-Translational Modifications of Histones That Influence ...
    Nov 26, 2014 · The incorporation of this modified histone into nucleosomal arrays inhibits the formation of compact 30-nm-like fibers and impedes the ...2.1. Histone Tails Influence... · 3. Epigenetic Guidance Of... · Figure 11<|separator|>
  15. [15]
    Modulation of the high-order chromatin structure by Polycomb ...
    In this review, we review current advances as for how Polycomb complexes function to modulate and help define the high-order chromatin structure and topology.
  16. [16]
    Regulation associated modules reflect 3D genome modularity ...
    Sep 8, 2022 · Accumulating evidence has revealed the association of histone modifications with the topologically associating domains (TADs) and compartments ...
  17. [17]
  18. [18]
    Whole-genome landscape of histone H3K4me3 modification during ...
    Jun 27, 2024 · We show that H3K4me3 peaks were mainly enriched in the promoter regions, and intergenic H3K4me3 peaks expanded as sperm cell lineage ...
  19. [19]
    The role of histone modifications: from neurodevelopment to ...
    Jul 6, 2022 · Aberrant histone modifications contribute to the development of neurodegenerative and neuropsychiatric diseases. Herein, recent progress in ...
  20. [20]
    Camello, a novel family of Histone Acetyltransferases that acetylate ...
    Aug 15, 2014 · Phylogenetic analysis of these 33 HATs revealed that there are groups of proteins from MYST family, GNAT family (GCN5/PCAF, ATAC2, ARD) ...
  21. [21]
    Crystal Structure of Yeast Esa1 Suggests a Unified Mechanism for ...
    The structural similarity in the core domains of the HAT proteins correspond to motifs D and A of the extended Gcn5-related N-acetyltransferases (GNAT). ( ...
  22. [22]
    Genome-wide Mapping of HATs and HDACs Reveals Distinct ...
    Sep 4, 2009 · We describe here genome-wide mapping of HATs and HDACs binding on chromatin and find that both are found at active genes with acetylated histones.
  23. [23]
    Histone acetylation and the role of histone deacetylases in normal ...
    Aug 13, 2020 · So far, about 30 HATs have been identified in humans. HATs are primarily classified into two classes based on their subcellular localization: ...
  24. [24]
    The Transcriptional Coactivators p300 and CBP Are Histone ...
    Nov 29, 1996 · Mutations in the. N-terminal region lead to loss of the capacity for p300/. CBP binding without affecting RB binding (Moran, 1993;. Wang et al., ...
  25. [25]
    The p300/CBP acetyltransferases function as transcriptional ...
    CBP and p300 can form larger protein complexes that include other acetylases and serve as coactivators of numerous transcription factors. They can accomplish ...
  26. [26]
    HDACs, histone deacetylation and gene transcription - Nature
    Feb 27, 2007 · HDACs catalyze the inverse reaction by removing the acetyl group from the acetyl lysine residue. Higher organisms have evolved a considerable ...
  27. [27]
    A short guide to histone deacetylases including recent progress on ...
    Feb 19, 2020 · Histones can be modified by the acetylation or deacetylation of lysine residues by HATs or HDACs to regulate the interaction between histones ...
  28. [28]
    the many faces of the Sin3 histone deacetylase (HDAC) complex
    Sin3 appears to have properties of a co-repressor, co-activator and general transcription factor, and has thus been termed a co-regulator complex.
  29. [29]
    HATs and HDACs: from structure, function and regulation to novel ...
    Aug 13, 2007 · Members of the GNAT family include HAT1 (histone acetyltransferase 1), yeast Gcn5 and its metazoan orthologs GCN5 and PCAF (p300/CBP-associated ...
  30. [30]
    In Vitro Targeting Reveals Intrinsic Histone Tail Specificity of the ...
    A large number of in vivo studies have shown that HATs and HDACs can be recruited to target genes by activators and repressors. ... The Sin3/HDAC complex ...
  31. [31]
    Review - Histone Lysine Methylation Dynamics - Cell Press
    Aug 8, 2012 · The SET domain is a 130 amino acid catalytic domain initially found to be conserved in Su(var)3-9, E(z). (enhancer of zeste) and trithorax ( ...
  32. [32]
    Targeting histone methyltransferases and demethylases in clinical ...
    May 24, 2016 · Up to date more than 50 lysine human methyltransferases (KMTs) have been reported. These enzymes possess high selectivity concerning the histone ...
  33. [33]
    Catalytic Mechanism and Product Specificity of the Histone Lysine ...
    anism: it is a typical in-line SN2 nucleophilic substitution reaction with a ... formation of near-attack conformations for the di-methylation reaction.
  34. [34]
    Structure, Activity and Function of the Suv39h1 and Suv39h2 Protein ...
    Jul 16, 2021 · Both enzymes introduce di- and trimethylation at histone H3 lysine 9 (H3K9) and have important roles in the maintenance of heterochromatin and gene repression.
  35. [35]
    Kinetic mechanism of Protein Arginine Methyltransferase 1 - PMC
    Individual PRMT isozymes utilize S-adenosyl-L-methionine (SAM) as the methyl donor and catalyze methyl group transfer to the ω-nitrogen of a peptidyl arginine ...Missing: H4R3me1 | Show results with:H4R3me1
  36. [36]
    LSD1 and The Chemistry of Histone Demethylation - PMC - NIH
    The recent discovery that histone demethylation can be catalyzed by the flavin-dependent amine oxidase LSD1 has ushered in a new chapter in the chromatin ...
  37. [37]
    JMJD family proteins in cancer and inflammation - Nature
    Sep 1, 2022 · The JMJD family of KDM enzymes function as Fe2+ and 2-oxoglutarate-dependent dioxygenases and are able to demethylate histone lysines at ...
  38. [38]
    Global Analysis of H3K4 Methylation Defines MLL Family Member ...
    Abstract. A common landmark of activated genes is the presence of trimethylation on lysine 4 of histone H3 (H3K4) at promoter regions.
  39. [39]
    The Histone H3 Lysine 9 Methyltransferases G9a and GLP Regulate ...
    Jan 23, 2014 · G9a/GLP and Polycomb Repressive Complex 2 (PRC2) are two major epigenetic silencing machineries, which in particular methylate histone H3 on lysines 9 and 27.
  40. [40]
    A central role for G9a and EZH2 in the epigenetic silencing of ...
    This study explored the role of G9a- and enhancer of zeste homolog 2 (EZH2)-mediated methylation of histone H3 lysine 9 (H3K9me3) and histone H3 lysine 27 (H3K ...
  41. [41]
    HP1 oligomerization compensates for low-affinity H3K9me ... - Science
    Jul 8, 2022 · HP1 proteins traverse a complex and crowded chromatin landscape to bind with low affinity but high specificity to histone H3K9 methylation ...
  42. [42]
    Histone Phosphorylation - an overview | ScienceDirect Topics
    3.2.​​ Histone phosphorylation involves the transfer of phosphate groups from adenosine triphosphate (ATP) to hydroxyl groups of serine, threonine, or tyrosine ...
  43. [43]
    Histone H3 phosphorylation – A versatile chromatin modification for ...
    In this review we describe the importance of different histone H3 phosphorylation marks for chromatin condensation during mitosis.
  44. [44]
    Histone H3 serine 10 phosphorylation by Aurora B causes HP1 ...
    Dec 22, 2005 · H3 phosphorylation by Aurora B is therefore part of a 'methyl/phos switch' mechanism that displaces HP1 and perhaps other proteins from mitotic heterochromatin.Missing: paper | Show results with:paper
  45. [45]
    Phosphorylation of the linker histone H1 by CDK regulates its ...
    We propose that phosphorylation of H1 provides a signal for the disassembly of higher order chromatin structures during interphase.Missing: S10 | Show results with:S10
  46. [46]
    Histone Phosphorylation in DNA Damage Response - MDPI
    Aurora kinases, which regulate mitosis through histone H3 phosphorylation, have emerged as key therapeutic targets.Histone Phosphorylation In... · 2. Histone Phosphorylation... · 2.1. 1. γ H2ax
  47. [47]
    Phosphorylation of histone H3 at Ser10 facilitates RNA polymerase ...
    It has been proposed previously that phosphatase 2A (PP2A) plays a major role in transcription-dependent H3S10 phosphorylation (Nowak et al. 2003). Therefore, ...
  48. [48]
    Dephosphorylation of γH2AX by WIP1 - NIH
    A key early event in the DNA damage response (DDR) is ATM phosphorylation of the histone variant H2AX at serine 139 at the site of the DNA break. Phosphorylated ...
  49. [49]
    H2AX Phosphorylation: Its Role in DNA Damage Response ... - NIH
    Phosphorylation of H2AX plays a key role in DDR and is required for the assembly of DNA repair proteins at the sites containing damaged chromatin.Missing: S139 | Show results with:S139
  50. [50]
    A phospho/methyl switch at histone H3 regulates TFIID ... - PubMed
    Dec 1, 2010 · We propose that a histone H3 phospho-methyl switch regulates TFIID-mediated transcription during mitotic progression of the cell cycle.
  51. [51]
    Histone Ubiquitination: An Integrative Signaling Platform in Genome ...
    Jun 1, 2020 · Here we review the current knowledge on specific, low-abundant histone ubiquitination events that are highly regulated within the cellular DNA damage response ...
  52. [52]
    Protein ubiquitination involving an E1-E2-E3 enzyme ... - PubMed
    This cascade of ubiquitin thioester complexes suggests that E3s have a defined enzymatic activity and do not function simply as docking proteins. The cysteine ...
  53. [53]
    Monoubiquitination of Human Histone H2B: The Factors Involved ...
    We report that in humans, the 600 kDa RNF20/40 complex is the E3 ligase and UbcH6 is the ubiquitin E2-conjugating enzyme for H2B-Lys120 monoubiquitination.Missing: seminal papers<|separator|>
  54. [54]
    Histone Mono-Ubiquitination in Transcriptional Regulation and Its ...
    RNF20/40 and H2BK120ub facilitates transcriptional elongation. (A) To initiate H2B ubiquitination, RNF20/40, an E2 enzyme (UBE2A/B), and PAF1 create a ...<|control11|><|separator|>
  55. [55]
    Activity of PRC1 and histone H2AK119 monoubiquitination
    Mar 20, 2020 · PRC1-catalytic activity towards monoubiquitination of histone H2AK119, a well-established hallmark of PRC1 complexes whose importance has been long debated.
  56. [56]
    Polycomb group-mediated histone H2A monoubiquitination ... - Nature
    Nov 23, 2020 · Structural insights into the mechanism of H2A ubiquitination by PRC1. While the PRC1 complex was originally purified from Drosophila cells and ...
  57. [57]
    Histone Ubiquitination and Deubiquitination in Transcription, DNA ...
    USP16 catalyzes H2A deubiquitination in vitro and in vivo, and plays important roles in H2Aub mediated HOX gene silencing, X chromosome inactivation, cell cycle ...
  58. [58]
    USP16-mediated histone H2A lysine-119 deubiquitination during ...
    May 30, 2022 · Genetic evidences indicated that ubiquitin-specific peptidase 16 (USP16) is the major deubiquitinase for H2AK119ub1 in mouse oocytes.
  59. [59]
    The Pleiotropic Ubiquitin-Specific Peptidase 16 and Its Many ...
    Mar 13, 2023 · Ubiquitin-specific peptidase 16 (USP16) is a deubiquitinase that plays a role in the regulation of gene expression, cell cycle progression, and various other ...
  60. [60]
    Article Mechanism of Cross-talk between H2B Ubiquitination and H3 ...
    Mar 7, 2019 · Our study provides a comprehensive mechanism of cross-talk between histone ubiquitination and methylation and reveals structural plasticity in histones.
  61. [61]
  62. [62]
    O-GlcNAcylation: the sweet side of epigenetics
    Dec 14, 2023 · This review provides a current overview of our knowledge of how O-GlcNAcylation impacts the histone code both directly and by regulating other chromatin ...Missing: seminal | Show results with:seminal
  63. [63]
    The Beginner's Guide to O-GlcNAc: From Nutrient ... - Frontiers
    O-GlcNAcylation plays a role in a broad range of biological processes, such as transcription, translation, enzyme activity, cell division, protein localization ...O-GlcNAc Cycling Enzymes · O-GlcNAc's Involvement in the... · DiscussionMissing: seminal papers
  64. [64]
    OGT and OGA: Sweet guardians of the genome - PMC - NIH
    Here, we first review the recent structural insights of OGT and OGA, as scientists strive to answer why there is only one enzyme for both writing and erasing O- ...Missing: seminal papers
  65. [65]
  66. [66]
  67. [67]
    O-GlcNAc transferase and O-GlcNAcase: achieving target substrate ...
    This review will discuss factors known and hypothesized to regulate OGT and OGA substrate bias. These mechanisms appear to be built into the cellular ...Missing: seminal papers
  68. [68]
    Histone O-GlcNAcylation and Potential Biological Functions - lidsen
    In this review, we provide an overview of the current understanding of histone O-GlcNAcylation and discuss its potential roles in biological functions.Missing: seminal | Show results with:seminal
  69. [69]
    Histone sumoylation and chromatin dynamics - PMC - NIH
    Histone sumoylation is integral to complex signaling codes that prime additional histone PTMs as well as modifications of the RNA polymerase II carboxy-terminal ...Missing: H4K16 | Show results with:H4K16
  70. [70]
    Histone sumoylation is associated with transcriptional repression
    There appear to be a number of different SUMO ligases (E3s) in higher eukaryotes such as PIAS family proteins (9–11), RanBP2 (12), and the polycomb protein Pc2 ...
  71. [71]
    Linking nuclear matrix–localized PIAS1 to chromatin SUMOylation ...
    Here, we present evidence that multiple SUMOylation machinery components, including SUMO E1 proteins SAE1 and SAE2 and the PIAS (protein inhibitor of activated ...Missing: H4K16 | Show results with:H4K16
  72. [72]
    SENP1 Enhances Androgen Receptor-Dependent Transcription ...
    SENP1's ability to enhance AR-dependent transcription is not mediated through desumoylation of AR, but rather through its ability to deconjugate histone ...
  73. [73]
    Mitotic Phosphorylation of SENP3 Regulates DeSUMOylation of ...
    DeSUMOylation by SUMO isopeptidases (SENP) is essential to ensure the reversible nature of SUMO conjugation. There are six different isopeptidases (SENP1 ...
  74. [74]
    Histone sumoylation is a negative regulator in Saccharomyces ... - NIH
    We provide evidence that histone sumoylation negatively regulates transcription in S. cerevisiae. We show that all four core histones are sumoylated.
  75. [75]
  76. [76]
    Proteomic analyses identify ARH3 as a serine mono-ADP ... - Nature
    Dec 12, 2017 · PARG has both endo- and exo-glycosidase activities, whereas ARH3 seems to exert only exoglycosidase activity. Besides PAR, ARH3 hydrolyzes O- ...
  77. [77]
  78. [78]
    The regulatory landscape of the human HPF1- and ARH3 ... - Nature
    Oct 8, 2021 · We find that HPF1 and ARH3 inversely and homogenously regulate the serine ADP-ribosylome on a proteome-wide scale with consistent adherence to lysine-serine- ...
  79. [79]
    Role of mono‑ADP‑ribosylation histone modification (Review)
    Mar 31, 2021 · A number of studies have indicated that poly-ADP-ribosylation of histones have important roles in DNA repair, replication, transcription (9,10), ...
  80. [80]
    The nucleosomal surface is the main target of histone ADP ... - PMC
    Although ADP-ribosylation was discovered in the 1950s, these challenges precluded comprehensive analysis of histone ADP-ribosylation sites for decades. ... H1.5 ...
  81. [81]
    Protein Arginine Deiminases and Associated Citrullination
    In this mechanism, PAD4 is recruited to gene promoters where it mediates the citrullination of histone H4 at R3 and histone H3 at R2, R8, and R17 and the ...
  82. [82]
    Protein Arginine Methylation and Citrullination in Epigenetic ...
    Dec 19, 2015 · We show that peptidyl arginine deiminase 4 (PADI4) specifically deiminates, arginine residues R2, R8, R17, and R26 in the H3 tail. Deimination ...Figure 3 · Prmt Inhibitors · Pad Inhibitors
  83. [83]
    Protein Arginine Methylation and Citrullination in Epigenetic ...
    Specifically, the citrullination at H3R17 represses the expression of estrogen receptor regulated genes. Moreover, PAD4 seems to act as a p53 corepressor ...
  84. [84]
    Citrullination and the protein code: crosstalk between post ... - Journals
    Oct 2, 2023 · Below we outline some examples of cross-talk between citrullination and other modifications on both histone and non-histone targets. (e) ...
  85. [85]
    Protein arginine deiminase 4 antagonizes methylglyoxal-induced ...
    Jun 26, 2020 · We report that PAD4 antagonizes histone MGO-glycation by protecting the reactive arginine sites, as well as by converting already-glycated arginine residues ...
  86. [86]
    Citrullination/methylation crosstalk on histone H3 regulates ER ...
    As PAD2 and PAD4 citrullinate multiple sites on Histone H3 in vitro, we synthesized a series of Histone H3 peptide tail analogs, centered around H3K27, that ...
  87. [87]
    Histone citrullination: a new target for tumors - Molecular Cancer
    Jun 11, 2021 · Histone citrullination, a posttranscriptional modification catalyzed by peptidyl arginine deiminase (PAD) enzymes, is involved in human carcinogenesis.Citrullination · Nets And Tumors · Background<|control11|><|separator|>
  88. [88]
    Peptidylarginine deiminases in citrullination, gene regulation, health ...
    In PAD4 null neutrophils, the basal level of histone citrullination is undetectable. After stimulation with LPS, H2O2, PMA and bacteria, histone citrullination ...2. Pad4 And Pad2 In Gene... · 2.1. Pad4 In Gene Regulation · 3. Pad4 And Nets
  89. [89]
  90. [90]
    Histone acylations and chromatin dynamics: concepts, challenges ...
    Jun 23, 2021 · Histone acylations can link the metabolic state of a cell with chromatin architecture and contribute to cellular adaptation through changes in gene expression.
  91. [91]
    A glimpse into novel acylations and their emerging role in regulating ...
    Feb 5, 2024 · Here, we comprehensively review the identification and characterization of 11 novel acylations, and the corresponding modifying enzymes, highlighting their ...
  92. [92]
    Protein lysine crotonylation: past, present, perspective - Nature
    Jul 14, 2021 · Alignment of all sirtuins demonstrated that the residue Phe180 of SIRT3 is conserved in SIRT1-2, but not in other sirtuins, which may explain ...
  93. [93]
    Crossing epigenetic frontiers: the intersection of novel histone ...
    Sep 16, 2024 · Predominantly positively charged due to rich lysine and arginine residues, with about two-thirds of these charges located in the tails, histones ...Missing: modifiable | Show results with:modifiable
  94. [94]
    Histone γH2AX and Poly(ADP ribose) as Clinical Pharmacodynamic ...
    In response to DNA damage, PARP1 itself is the main PARylation acceptor (automodification) and over 90% of PAR is found on PARP1 (13). PARylation is a ...
  95. [95]
    The multifaceted influence of histone deacetylases on DNA damage ...
    The recruitment of the MRN complex to DSBs requires PARP1 (98). ATM in complex with the histone acetyltransferase (HAT) TIP60 is then recruited to the DSB ...
  96. [96]
    The histone methyltransferase DOT1L is required for proper DNA ...
    Jan 7, 2019 · DOT1L is required for homologous recombination-mediated DSB repair. Based on our results indicating that DOT1L is required for the early DNA ...
  97. [97]
    RNF8-dependent histone ubiquitination during DNA damage ... - PMC
    RNF8 catalyzes ubiquitination of the histones H2A and H2AX during the DDR, thus facilitating downstream recruitment of DDR factors.
  98. [98]
    RNF168 and the DNA damage response pathway - PMC - NIH
    Apr 16, 2021 · The ubiquitin E3 ligase RNF168 is essential in assembling a cohort of DNA repair proteins at the damaged chromatin via its enzymatic activity.
  99. [99]
    histone H3S10 phosphorylation and H3S10 kinases in cancer ...
    Oct 14, 2020 · H3S10ph is emerging as an important player in the initiation and propagation of cancer, as it facilitates cellular malignant transformation.
  100. [100]
    Dual Regulation by Pairs of Cyclin-Dependent Protein Kinases and ...
    In this study, we provide detailed mechanistic insights into Whi5-dependent regulation of G1-specific transcription and cell cycle progression. Specifically, we ...
  101. [101]
    Histone acetylation and the cell-cycle in cancer - PubMed - NIH
    Apr 1, 2001 · Transition through the cell cycle is regulated by a family of protein kinase holoenzymes, the cyclin-dependent kinases (Cdks), and their ...
  102. [102]
    Histone methylation in DNA repair and clinical practice: new findings ...
    It might contribute to DOT1L-mediated H3K79 methylation ... Transcriptionally active chromatin recruits homologous recombination at DNA double-strand breaks.
  103. [103]
    The histone DNA repair code: H4K20me2 makes its mark - PMC
    Loss of RNF168 results in abrogation of 53BP1 foci formation. Recognition of both H2AK15ub and H4K20me2 by 53BP1 is necessary for efficient NHEJ.
  104. [104]
    PP4 is a γH2AX phosphatase required for recovery from the DNA ...
    PP4 is a γH2AX phosphatase that contributes to the dephosphorylation of γH2AX and is required for recovery from the DNA damage checkpoint.
  105. [105]
    Role of protein phosphatases PP1, PP2A, PP4 and Cdc14 in the ...
    Phosphatases are involved in the control of DDR activation after a DNA lesion is generated, as well as to its inactivation when the DNA adduct has been repaired ...Missing: reset | Show results with:reset<|separator|>
  106. [106]
    phosphatases gate-crash the DNA damage response - PMC - NIH
    This finding suggests that the phosphatases have a redundant role in DNA repair, but a synergistic impact on checkpoint control [52, 53].
  107. [107]
    Histone phosphorylation: A chromatin modification involved in ...
    The corresponding phosphorylation on S10 of yeast H2B was confirmed ... Phosphorylation of the linker histone H1 by CDK regulates its binding to HP1alpha.
  108. [108]
    Targeting epigenetic regulators for cancer therapy - Nature
    Dec 17, 2019 · In this review, we summarize the aberrant functions of enzymes in DNA methylation, histone acetylation and histone methylation during tumor progression.
  109. [109]
    EZH2 mutations in follicular lymphoma distort H3K27me3 profiles ...
    Apr 24, 2024 · We show that Ezh2 Y641F induces aberrant H3K27 methylation patterns even without wild-type Ezh2, which are alleviated by partial PRC2 inhibition.
  110. [110]
    The hydroxamate based HDAC inhibitor WMJ-J-09 induces ... - Nature
    Jun 4, 2025 · Aberrant expression of histone deacetylases (HDACs) is associated with cancer drug resistance and tumor progression.
  111. [111]
    MLL-fusion-driven leukemia requires SETD2 to safeguard genomic ...
    May 18, 2018 · MLL-fusions represent a large group of leukemia drivers ... MLL complex-mediated H3K4me3 and transcription of leukemia-associated genes.
  112. [112]
    Targeted next-generation sequencing reveals high frequency of ...
    Jun 9, 2015 · Our study identified a high prevalence of somatic mutations in genes encoding epigenetic regulators, including those involved in DNA demethylation, histone ...
  113. [113]
    HDAC8-mediated inhibition of EP300 drives a transcriptional state ...
    Nov 29, 2023 · We previously identified the class I histone deacetylase HDAC8 as a driver of BRAF and BRAF-MEK inhibitor resistance in BRAF-mutant melanoma.
  114. [114]
    Prognostic significance of global histone modifications in resected ...
    Oct 24, 2008 · Patterns of global histone modifications have been recently suggested as outcome predictors in cancer patients. To date, there has been no ...Missing: hypoacetylation | Show results with:hypoacetylation
  115. [115]
    The Mechanisms of CHD8 in Neurodevelopment and Autism ...
    Jul 26, 2021 · CHD8 stimulates histone H3 lysine 4 (H3K4) methylation through interaction with mixed-lineage leukemia (MLL), and influences oligodendrocyte ...
  116. [116]
    CHD8 regulates neurodevelopmental pathways associated ... - PNAS
    Truncating mutation of chromodomain helicase DNA-binding protein 8 (CHD8) represents one of the strongest known risk factors for autism spectrum disorder (ASD).
  117. [117]
    Ezh2, the histone methyltransferase of PRC2, regulates the balance ...
    Ezh2 orchestrates gene expression for the stepwise differentiation of tissue-specific stem cells. Cell. 2009;136:1122–1135. doi: 10.1016/j.cell.2008.12.043.
  118. [118]
    Small Molecules Targeting H3K9 Methylation Prevent Silencing of ...
    Fragile X syndrome (FXS) is the leading cause of inherited intellectual disability, which affects approximately 1 in 5000 males and 1 in 4000 to 8000 females [1] ...Missing: H3K9me | Show results with:H3K9me
  119. [119]
    An HDAC6-dependent surveillance mechanism suppresses tau ...
    Nov 2, 2020 · Tauopathies including Alzheimer's disease (AD) are marked by the accumulation of aberrantly modified tau proteins.
  120. [120]
    Histone deacetylase 6 inhibition improves memory and reduces total ...
    These data support the use of HDAC6 inhibitors as potential therapeutic agents against tau pathology. Introduction. Tauopathies are neurodegenerative disorders ...
  121. [121]
    SirT2 is a histone deacetylase with preference for histone H4 Lys 16 ...
    Apr 28, 2006 · We found that SirT2 and its yeast counterpart Hst2 have a strong preference for histone H4K16Ac in their deacetylation activity in vitro and in ...
  122. [122]
    SIRT1 and SIRT2 Activity Control in Neurodegenerative Diseases
    Both SIRT1 and SIRT2 act independently on cell death in Parkinson's diseases. SIRT1 downregulates c-PARP and NF-κβ and reduces the protein aggregation in cells ...
  123. [123]
    The impact of histone post-translational modifications in ...
    Here, we review recent advances on the association of histone post-translational modifications with ALS, FTD, PD and several ataxias.
  124. [124]
    Epigenetics in Amyotrophic Lateral Sclerosis: A Role for Histone ...
    Here we review recent advances linking ALS and epigenetics, with a strong focus on histone modifications.
  125. [125]
    vorinostat for treatment of advanced primary cutaneous T ... - PubMed
    On October 6, 2006, the U.S. Food and Drug Administration granted regular approval to vorinostat (Zolinza(R); Merck & Co., Inc., Whitehouse Station, NJ), ...
  126. [126]
    FDA Approves New Agent for Multiple Myeloma - Oxford Academic
    Jun 1, 2015 · In February, the U.S. Food and Drug Administration (FDA) approved a new drug, panobinostat, for multiple myeloma. A histone deacetylase (HDAC) ...
  127. [127]
    A Natural Product Recently Approved for Cutaneous T-cell Lymphoma
    Romidepsin (Istodax®), a selective inhibitor of histone deacetylases (HDACs), was approved for the treatment of cutaneous T-cell lymphoma in November 2009 ...
  128. [128]
    FDA approves tazemetostat for advanced epithelioid sarcoma
    Jan 24, 2020 · On January 23, 2020, the Food and Drug Administration granted accelerated approval to tazemetostat (TAZVERIK, Epizyme, Inc.)Missing: EZH2 inhibitor
  129. [129]
    The DOT1L inhibitor pinometostat reduces H3K79 methylation and ...
    Jun 14, 2018 · Pinometostat demonstrates first evidence of DOT1L target inhibition and clinical responses in a subset of MLL-r advanced leukemia patients.
  130. [130]
    New perspectives on epigenetic modifications and PARP inhibitor ...
    Jan 4, 2023 · The clinical treatment of DNA-repair defective tumours has been revolutionised by the use of poly(ADP) ribose polymerase (PARP) inhibitors.
  131. [131]
    Emerging roles of SIRT1 activator, SRT2104, in disease treatment
    Mar 6, 2024 · SRT2104, as a novel, efficient, and specific SIRT1 activator, has shown promising tolerability in animal models and clinical trials.
  132. [132]
    Epigenetics-targeted drugs: current paradigms and future challenges
    Nov 26, 2024 · Epigenetic modifications and the enzymes involved can either activate or suppress the expression of specific genes at different levels (Table 1) ...
  133. [133]
    Drugging the epigenome in the age of precision medicine
    Jan 11, 2023 · Modulating the epigenome has long been considered a potential opportunity for therapeutic intervention in numerous disease areas with ...
  134. [134]
    Structural basis for inactivation of PRC2 by G-quadruplex RNA
    Sep 21, 2023 · In this work, we present a 3.3-Å-resolution cryo–electron microscopy structure of PRC2 bound to a G-quadruplex RNA.
  135. [135]
    Activation of automethylated PRC2 by dimerization on chromatin
    Oct 17, 2024 · Here, we set out to investigate how automethylation of EZH2 may activate human PRC2 using single-particle cryo-electron microscopy (cryo-EM) and ...
  136. [136]
    Mass Spectrometry-based Profiling of Single-cell Histone Post ...
    Jul 6, 2024 · We present a robust mass spectrometry-based method for the unbiased analysis of histone PTMs (hPTMs) at single-cell level.
  137. [137]
    Interplay Between the Circadian Clock and Sirtuins - PMC
    The circadian clock regulates the activity of sirtuins through multiple mechanisms, including indirectly modulating sirtuin activity by controlling the rhythmic ...Missing: OGT 2025
  138. [138]
    Epigenetic regulation of histone modifications in glioblastoma
    May 31, 2025 · This review provides a comprehensive overview of epigenetic regulation mechanisms involving histone modifications in GBM, covering their roles in tumor ...
  139. [139]
    Systematic epigenome editing captures the context-dependent ...
    May 9, 2024 · Here we develop a modular epigenome editing platform that programs nine key chromatin modifications, or combinations thereof, to precise loci in living cells.Missing: PTMs | Show results with:PTMs
  140. [140]
    Predicting gene expression from histone marks using chromatin ...
    We used convolutional and attention-based models to predict transcription from histone mark activity at promoters and distal regulatory elements. Our work shows ...
  141. [141]
    Succinylation and redox status in cancer cells - Frontiers
    Dec 19, 2022 · Succinylation is a post-translational modification (PTM) event that associates metabolic reprogramming with various pathological disorders including cancers.
  142. [142]
    Identification of 113 New Histone Marks by CHiMA
    Apr 5, 2023 · This tool not only offers a valuable approach for identifying histone modifications but also greatly expands the repertoire of histone marks.