ABSTRACT
Histone variants regulate chromatin accessibility and gene transcription. Given their distinct properties and functions, histone varint substitutions allow for profound alteration of nucleosomal architecture and local chromatin landscape. Skeletal myogenesis driven by the key transcription factor MyoD is characterized by precise temporal regulation of myogenic genes. Timed substitution of variants within the nucleosomes provides a powerful means to ensure sequential expression of myogenic genes. Indeed, growing evidence has shown H3.3, H2A.Z, macroH2A, and H1b to be critical for skeletal myogenesis. However, the relative importance of various histone variants and their associated chaperones in myogenesis is not fully appreciated. In this review, we summarize the role that histone variants play in altering chromatin landscape to ensure proper muscle differentiation. The temporal regulation and cross talk between histones variants and their chaperones in conjunction with other forms of epigenetic regulation could be critical to understanding myogenesis and their involvement in myopathies.
KEYWORDS: Skeletal myogenesis, epigenetics, histone variants, myopathies
Introduction
Chromatin in the nucleus is wrapped around basic, octameric protein structures known as nucleosomes. Around 147bp of genetic material is wrapped around one nucleosome, which allows for both compactions as well as regulation of gene transcription by controlling access to transcriptional machinery. Core nucleosomes consist of two copies of each of the histone subunits H2A, H2B, H3, and H4. There is also a linker arm, H1, which modulates interactions between the nucleosome and the associated DNA. Each of these subunits, with the sole exception of H4, have multiple variants in humans, many of which are also evolutionarily conserved in other species. Variants of a histone subunit are predominantly non-allelic, that is, produced by different genes, rather than through the production of splice isoforms. However, some variants of H2A are produced through alternative splicing. Histone variants are classified into one of two categories: replication-dependent or replication-independent. The synthesis of replication-dependent variants, as the name suggests, is linked to DNA replication. These variants form the bulk of nucleosomes and their synthesis is consequently tied to synthesis of DNA. In line with this role, these variants are often synthesized by several genes that are present in multiple copies within clusters, thus allowing for the production of a vast quantity in time for DNA replication and compaction. On the other hand, replication-independent variants can be synthesized at any time during the cell cycle, depending on context and requirement. These are often encoded by one or two genes, normally present outside the replication-dependent histone clusters. Some variants are limited to germ cells only. Replication-dependent histone genes lack introns and produce mRNA that contain a consensus stem-loop structure downstream of the stop codon, required for processing the 3’ ends. Conversely, replication-independent variants lack this structure. Some also possess introns that allows for alternative splicing and increased isoform diversity. The properties of histone variants, and the features of those that have been discovered thus far, have been summarized in several excellent reviews [2–8].
Histone variants can be exchanged within nucleosomes. This exchange can lead to changes in the structure, stability, and properties of the nucleosome. Therefore, the exchange of one variant for another, or the marking of a specific genetic locus with a particular variant or combination of variants, can alter the transcription of the target genes. This process is dependent on histone chaperone complexes, as each variant interacts preferentially with a few chaperones and chromatin remodellers. The complexes that have thus far been shown to associate with specific variants are shown in (Table 1). Both variants and their chaperones play important roles in modification of chromatin landscape and gene transcription. This process of modification of the chromatin landscape is one that is important to the regulation of skeletal myogenesis.
Table 1.
Variants of histone subunits
| Variant | Associated chaperones & remodelling complexes | Known functions |
|---|---|---|
| H1.1 | TAF1[163–165], NAP1[165,166], NPM1[167],ProTα[168–170], Nucleolin[171] | Regulator of gene expression[6,172] |
| H1.2 | TAF1[163–165], NAP1[165,166], NPM1[167],ProTα[168–170], Nucleolin[171] | Transcriptional repression173, Transcriptional activation[174,175], Cell cycle progression[176] |
| H1.3 | TAF1[163–165], NAP1[165,166], NPM1[167],ProTα[168–170], Nucleolin[171] | Repression of ncRNA expression[145] |
| H1.4 | TAF1[163–165], NAP1[165,166], NPM1[167],ProTα[168–170], Nucleolin[171] | Heterochromatin formation[177], Transcriptional activation[174,177,178], Cell cycle progression[176] |
| H1.5 | TAF1[163–165], NAP1[165,166], NPM1[167],ProTα[168–170], Nucleolin[171] | Transcriptional repression[123] |
| H1.0 | TAF1[163,164] | Replacement of replication-dependent variants in differentiated tissue[179] |
| H1.X | TAF1[163] | Mitotic progression[180] |
| H1t | - | Testes specific form[181] |
| H1T2 | - | Cell restructuring and polarity in spermiogenesis[182,183] |
| H1oo | - | Oocyte-specific form[184] |
| HILS | - | Spermatid-specific form[185,186] |
| H2A | FACT[187], NAP1[188–192], nucleolin[193] | Regulation of gene expression[194] |
| H2A.X | FACT[195] | Genome stability[196], proliferation[197], DNA damage response[198] |
| macroH2A.1.1 | Nucleolin[193], ATRX[199] | Development[103,106,109,200], X-chromosome inactivation[105], myogenesis[116] |
| macroH2A.1.2 | Nucleolin[193], ATRX[199] | Development[103,106,109,200], X-chromosome inactivation[105], myogenesis[116] |
| macroH2A2 | - | Developmental genes[103] |
| H2A.Z.2.1 | - | - |
| H2A.Z.2.2 | NuA4/TIP60/P400[201], SRCAP[201] | - |
| H2A.Z.1 | ANP32E[202,203], SRCAP[204,205], NuA4/TIP60/P400[75], INO80[203] | Transcriptional activation[205], Myogenesis[97], ES cell renewal and embryonic development [78,206] |
| H2A.Bbd | NAP1[207] | Stability of newly synthesized DNA[208], regulation of gene transcription[209] |
| H2B | FACT[187], NAP1[188–192], nucleolin[193] | Regulation of gene expression |
| TSH2B | - | Testis-specific form[210], reprogramming[211] |
| H2BFWT | SWI/SNF[212] | - |
| H3.1 | FACT[187], NAP1[189,213-215], ASF1[216–218], NASP1[219,220], CAF1[221–223] | Repression of myogenesis[65], Regulation of gene expression[224] |
| H3.2 | FACT[187], NAP1[189,213-215], ASF1[216–218], NASP1[219,220], CAF1[221–223] | Regulation of gene expression[224] |
| H3.3 | FACT[187], ASF1[218,225], ATRX/DAXX [30–53–54–226–230], HIRA[30–42–50–230–234], CHD1[56,235,236], CHD2 [60,235], DEK[237], EP400[238], RPA[234] | Myogenesis [58,60], genome integrity[228], embryonic development[44], transcriptional activation[30,31], lineage determination and epigenetic memory [41,42] |
| H3.1 t | - | Testis-specific form[224] |
| H3.5 | - | Transcriptional regulation in spermatogenesis[239,240] |
| H3.Y | - | Transcriptional response to stimuli[241,242] |
| H3.X | - | - |
| CENP-A | HJURP[243,244] | Centromere organization and chromosome segregation[244,245] |
| H4 | FACT[187], NAP1[189,213-215], CAF1[221–223] | Regulation of gene expression |
With the exception of H4, all core histone subunits have multiple variants in humans, which are deposited by specific chaperone complexes and associated chromatin remodellers. The known variants of all subunits, along with known physiological roles and associated chaperones/remodelling complexes are shown. For each subunit, dark colours indicate replication-dependent variants, whereas light colour indicate replication-independent variants. Variants within a single box are splice isoforms of each other.
Histone variants in skeletal myogenesis
Skeletal myogenesis is the process of formation of mature skeletal muscle tissue from precursor cells. While this occurs primarily during embryonic and foetal development, it is also be recapitulated in adult tissue, when resident muscle stem cells, known as satellite cells, are activated in response to injury or damage. Myogenic differentiation (Figure 1) is a tightly regulated process marked by the sequential expression of transcription factors and epigenetic complexes that regulate the expression of cell cycle and myogenic genes. Both foetal and adult stem cells express paired-box (Pax) transcription factors Pax7 and Pax3. Myoblast determination protein 1 (MyoD), the master regulator of myogenesis, and myogenic factor 5 (Myf5), are among the earliest expressed transcription factors involved in cell lineage specification [9–11]. Skeletal myoblasts irreversibly exit the cell cycle and fuse to form multinucleated myotubes. As differentiation proceeds, other myogenic regulatory factor (MRF) including myogenin (Myog) and Mrf4 as well as myocyte enhancing factor (MEF) drive the expression of subsequent genes, such as troponin, myosin heavy chain (MHC), muscle creatine kinase (MCK) and skeletal alpha (α)-actin. Terminal differentiation is marked by the presence of MHC (encoded by the Myh genes), skeletal muscle (α)- actin (encoded by Acta1) and MCK (encoded by the Ckm gene) [12–14].
Figure 1.

Histone variants and chaperones/remodelling complexes involved in myogenic differentiation. Skeletal myogenesis involves a series of well-defined steps the result in skeletal myofibres being produced from either foetal or adult stem cells. Each step is driven by the expression of transcription factors, and marked by the presence of certain components that can be used to determine the stage of differentiation. The histone variants and associated chaperones/remodelling complexes that are currently known to be involved at each stage are also shown. These are discussed later in this review
Gene expression is also controlled by epigenetic complexes, both activating and repressing. Many of these are histone-modifying enzymes. Histone modifications include methylation, demethylation, acetylation, deacetylation, phosphorylation, sumoylation, and ubiquitination among others [15]. Depending on the type and residue of modification, these marks can affect histone–chromatin interactions, or act as docking sites for enzymes and modifiers. Therefore, histone modifications are crucial to regulating gene expression, and many complexes play critical roles in regulating myogenesis [16–20].
ATP-dependent chromatin remodelling is also crucial to skeletal myogenesis [21]. SWItch/Sucrose Non-Fermentable (SWI/SNF)-linked chromatin remodelling is essential to MyoD-dependent activation of myogenic genes [22]. The activity of SWI/SNF complexes is regulated by external differentiation cues [23]. The SWI/SNF ATPase Brg1 interacts with both MyoD and Myog to induce expression of skeletal muscle genes [24]. During the onset of differentiation, there is extensive chromatin remodelling and altered expression of the HIST1 gene cluster on chromosome 13[1], which consists of a subset of 16 replication-independent histones. Downregulation of a majority of the genes in this locus (with the exceptions of HisIh2bc, Hist1h1a, and HistIh1c) corresponds with the idea that the expression of replication-dependent histones is progressively suppressed during myoblast differentiation [25].
Therefore, temporal regulation of chromatin landscape through a myriad of ways is critical to the process of myogenesis [26–28]. Our understanding of the role of histone variants in myogenesis is summarized below.
H3.3
Of all the replication-independent variants, the H3 variant H3.3 has been best-studied for its role in myogenesis. H3.3 varies from the two canonical subunits, H3.1 and H3.2, by only five and four amino acids, respectively, [29]. Nevertheless, in spite of the high degree of similarity between the canonical variants and H3.3, they diverge significantly in the roles they play when incorporated into a nucleosome. H3.3 is preferentially incorporated into promoters, although it has also been found in enhancers and in gene bodies of actively transcribed genes [30,31]. Unlike its canonical counterparts, it is enriched in activating histone marks and is thought to destabilize nucleosomes into which it is incorporated. Chromatin Immunoprecipitation sequencing (ChIP-seq) has shown that H3.3 enrichment correlates with markers of active transcription, such as active (Ser-5 phosphorylated) RNA polymerase II and H3K4me3, H3K36me3, and H3K4me1. In addition, H3.3 antagonizes binding of the repressive histone H1 [32,33]. Therefore, the role of H3.3 is predominantly activating in nature [30,34–40]. It is required for cellular ‘memory’ of lineage identity. This requires the accumulation of both activating (H3K4me3) and repressive (H3K27me3) marks [41]. Interestingly, H3.3 is required to maintain parental lineage early on during cellular reprogramming, but its role is later reversed to establish and maintain the newly established lineage [42]. Surprisingly, there is also some evidence to show that H3.3 accumulates at, and is required for, the maintenance of inactive regions of the chromatin, such as telomeres and pericentric heterochromatin [43].
Like all replication-independent variants, H3.3 is incorporated throughout the cell cycle, as opposed to the canonical form. The protein is encoded by two separate genes, H3F3A on chromosome 1 and H3F3B on chromosome 17. Double knockout of both genes leads to embryonic lethality, indicating the importance of H3.3 in embryonic development. While H3F3A-null mice are viable to adulthood, male fertility is impaired and there is some growth deficiency. Likewise, H3F3B-heterozygous mice show growth deficiency and male infertility. H3F3B-null mice show severe deficiencies in growth (both foetal and post-natal) and survival, resulting in embryonic or post-natal death [44]. Where mice do survive, knockout cells show karyotype defects, as well as defects in cell cycle. The defects are a result of ectopic localization of another variant of H3, CENP-A, in the absence of H3.3 [45]. H3.3 and its chaperone HIRA are also required for gastrulation in Xenopus [46,47]. GFP-tagged H3.3 knock-in mice are a useful tool to study genome-wide distribution of H3.3 and show correlation between H3.3 deposition and transcripts produced [48]. In addition, mouse models with SNAP-tagged H3.1/H3.3 have been used to study symmetric and asymmetric cell divisions in muscle stem cells. These have shown that muscle stem cell division patterns change when taken from in vivo to ex vivo, and that there is symmetric distribution of H3.1 and H3.3-containing nucleosomes among daughter cells [49].
Each variant has been shown to interact preferentially with different histone chaperones. H3.1 deposition is linked to the histone chaperone Asf1b and the CAF complex. H3.3 primarily interacts with the chaperone HIRA, responsible for its deposition at regions of active transcription, or those induced by stress or infection [30,50–52], as well as with the ATRX/DAXX complex, which is required for its incorporation into inactive regions of the genome [53–55]. H3.3 has also been previously shown to interact with the chromatin remodelling factor Chd1 [56].
Expression and function in myogenesis
Early studies in chicken myotubes showed that there are changes in the variants of H3 produced over the course of myogenic differentiation. H3.2 (the canonical subunit) is predominantly synthesized in proliferating myoblasts and a switch to H3.3 occurs differentiation proceeds. This is expected given that the switch to differentiation results in a permanent exit from the cell cycle, accompanied by a loss of synthesis of replication-dependent variants [57]. This pattern was also seen in mouse myotubes, where it was also seen that H2A.X, H2A.Z, macroH2A, and CENP-A were all downregulated upon cell cycle exit [58].
In C2C12 cells, similarly, the expression of H3.3 increased during the onset of differentiation, while the expression of the canonical variant of H3 decreased after cell cycle exit. In addition, the expression of HIRA and Asf1a were also maintained during myogenesis [58,59], as opposed to that of Chd1 and Asf1b.
An early study showed that H3.3 was deposited at the MyoD promoter, and promoted its expression, as well as that of Myf5 [41]. Later, the activation of MyoD was shown to be through the accumulation of H3.3 around the core enhancer region (CER), distal regulatory region (DRR) and proximal regulatory region (PRR) of the MyoD gene (Figure 2a). These have been shown to be important regulatory regions for MyoD expression. High levels of H3.3 were found around the CER and PRR prior to the onset of differentiation, during which process these levels increased even further. Combined loss of Chd1 and HIRA led to significant loss of H3.3 around these sites. Moreover, the role of H3.3 in regulating MyoD expression is dependent not on the changes in associated histone marks that usually accompany variant substitution, but through H3.3-dependent removal of repressive linker histone H1 and other structural changes to the chromatin around the PRR [58].
H3.3 deposition is also important for a later step in myogenic differentiation. In skeletal myoblasts, the SNF2-family chromatin remodelling enzyme chromodomain helicase DNA-binding domain 2 (Chd2) interacts with MyoD to deposit H3.3 at the loci of myogenic regulatory sequences. Binding of Chd2 to these myogenic loci is dependent on MyoD, and loss of either Chd2 or H3.3 reduced expression of the myogenic genes Myog, Ckm, Myh4 and Acta1, and impaired formation of myotubes. H3.3 was deposited around the transcription start site (TSS) of these myogenic loci prior to onset of differentiation, and levels of incorporation increased even further during differentiation. MyoD-dependent deposition of H3.3 at myogenic genes in myoblasts is a mechanism by which genes required for differentiation are marked prior to the induction of the process [60]. Thus, H3.3 deposition is important not only for MyoD expression (Figure 2(a)), but also for its subsequent activating function (Figure 3(a)).
Figure 2.

Regulation of MyoD expression through histone variant substitution. The effect of individual variants are shown separately (a) Replacement of H3.1 with H3.3 at the MyoD PRR and CER by a complex consisting of HIRA and Asf1a results in removal of the repressive linker histone H1. Removal of H1 also leads to removal of repressor Msx1.The resulting permissive chromatin structure causes activation of MyoD expression. (b) Around the transcriptional start site, canonical H2A is replaced with H2A.Z, which accumulates activating acetyl marks (Lys4, 7, 11, 13, and 15). This activates MyoD expression. It is important to note that the sequence of figures does not necessarily indicate a temporal sequence of events. The order in which these changes occurs is unclear as of now. The key for both parts (a) and (b) is shown on the right
Figure 3.

Regulation of Myog expression through histone variant substitution. Multiple histone variant substitutions are involved in the transcriptional activation of Myog. (a) Replacement of H3.1 with H3.3 by the HIRA-Chd2 complex, along with MyoD, results in the acquisition of activating H3K4me3 marks around the Myog promoter. Myog expression is activated. (b) Around the transcriptional start site, canonical H2A is replaced with H2A.Z, which results in accumulation of activating acetyl marks. This activates Myog expression. (c)
At the enhancer, replacement with macroH2A leads to an accumulation of H3K27Ac marks. This leads to recruitment of MyoD and co-activator Pbx1. Myog expression is activated. It is important to note that the sequence of figures does not necessarily indicate a temporal sequence of events. The order in which these changes occurs is unclear as of now. The key for parts (a), (b) and (c) is shown on the right
The forced expression of the canonical variant H3.1, which replaces H3.3 in the TSSs of myogenic genes when ectopically expressed, has a repressive effect on myogenesis. Overexpression of H3.1 in C2C12 myoblasts led to a reduction in the expression of myogenic genes Myog, Myh, Acta1 and Ckm, whereas overexpression of H3.3 led to increased expression of these genes. This was shown to be a result of ‘bivariant’ nucleosomes. There was an increase in H3K27me3 (repressive) and a decrease in H4K4me3 (activating) marks. The increase in the H3K27me3 mark was due to increased interaction between H3.1 and Ezh2, the catalytic subunit of the Polycomb Repressive Complex (PRC2). Conversely, knockdown of H3.3 led to loss of both H3K27me3 and H3K4me3, indicating that H3.3 is required for the maintenance of the bivalent state. Bivalent modifications have been shown to mark or prime the promoters of lineage-specific genes prior to induction [61–64]. In contrast, H3.1 leads to predominantly repressive marking. The recruitment of H3.3 to skeletal muscle genes is dependent on HIRA, the knockdown of which leads to reduction in the expression of genes such as Myog and Acta1 [65].
H3.3 is also involved in the reorganization of constitutive heterochromatin that takes place during myogenic differentiation. The ATRX/DAXX/H3.3 complex interacts with the muscle-specific long non-coding RNA ChRO1, which is induced during differentiation. This interaction and incorporation of H3.3 in satellite repeats of chromocenters is critical for chromocenter clustering and cell differentiation [66].
Satellite cells are capable of developing into mature muscle tissue in response to damage or injury [67–69]. One specific sub-variant, H3mm7, is expressed in satellite cells and is required for muscle regeneration. Although not much is known about the sub-variants of histone variants, due in part to the high degree of similarity between sub-variants, which makes detection difficult, one study has shown the presence of multiple sub-variants with tissue-specific distribution. The forms most highly expressed in mouse skeletal muscle are subvariants H3mm15, H3mm13, H3mm14, H3mm7, H3mm8, and H3F3A, whereas H3F3B is expressed at much lower levels [70]. H3mm7 is highly expressed in satellite cells, with reduced expression in muscle fibres. As a result of the alanine residue at position 57 (serine in other variants), H3mm7 forms weaker bonds with the arginine residue at position 40 on H4. The resulting nucleosome is less stable than the ones formed by H3.3-H4 subunits. Therefore, there is a more open chromatin structure around the promoter regions of many genes, including several myogenic genes. The difference between H3.3 and H3mm7 is the rate at which target genes are expressed upon incorporation, that is, how much nucleosomal instability and transcriptional accessibility each of these forms provides. Knockout of this sub-variant leads to defective adult skeletal muscle regeneration and differentiation, while there is no other dramatic phenotype. Thus, the incorporation of H3mm7 at myogenic genes is crucial for satellite cell differentiation [36].
H3.3 chaperones
In light of its role in depositing H3.3 in regions of active transcription at myogenic loci at the onset of differentiation, the HIRA chaperone complex has been recognized to be important for myogenesis. Loss of HIRA led to a reduction in the expression of MyoD, Myog, MCK, and MHC. In particular, loss of HIRA led to a loss of MyoD in differentiating cells, but did not significantly affect proliferating myoblasts [58,59]. Given that the HIRA complex localizes to myogenic genes before the onset of differentiation, after which there is not much change in the levels of protein, one possibility is that the activation of complex, rather than its levels, that are important for induction of differentiation. To this end, the regulation of HIRA has also been studied to a certain degree. Deposition of H3.3 during myogenesis is dependent on the activation of HIRA, which is phosphorylated at serine (S) 650 (in human) or 648 (in mouse) by Akt1. Phosphorylated HIRA is inactive and cannot undertake H3.3 deposition at myogenic genes and needs to be dephosphorylated before it can do so. Therefore, this provides a mechanism by which the deposition of H3.3, and the subsequent induction of myogenic genes can be regulated by differentiation cues such as the phosphorylation and activation of Akt [71].
Skeletal myocyte-specific HIRA depletion in mice resulted in myofibers hypertrophy, constant regeneration, sarcolemmal perforation and oxidative damage by 6 months of age. Fast-twitch muscle fibres of these knockout mice exhibited more profound effects. Hypertrophy and regeneration were accompanied by increased relative abundance of reactive oxygen species, which likely promoted muscle fibre degeneration and subsequent regeneration. Indeed, HIRA loss affects a large number of genes involved in cellular metabolism and DNA damage response [72]. HIRA also contributes to the activation of Mef2c-target genes during differentiation, and Asf1 was indispensable for transcriptional activation by this complex [73].
Interestingly, the H3.3 and macroH2A-linked chaperone ATRX also appears to be critical for muscle development; conditional skeletal muscle specific-knockout of ATRX in mice led to offspring that were viable at birth, but showed kyphosis and significant defects in muscle development within 3 weeks of birth due to profound genomic instability in myoblast cells, leading to poor expansion and regeneration [74].
H2A.Z
H2A.Z, one of the several replication-independent variants of the H2A subunit is, much like H3.3, linked to transcriptional activation [75–77]. H2A.Z constitutes about 10% of the total pool of H2A at any given time [78]. H3.3 and H2A.Z have been previously been shown to co-occupy nucleosomes that mark regions of transcriptional activity [37]. Like H3.3, H2A.Z is thought also to destabilize nucleosomes and confer a more open, accessible chromatin structure [78–81]. It is also thought to confer epigenetic memory of previous transcriptional states [82]. H2A.Z is required to maintain genomic stability and chromosome segregation [83]. It is enriched, and interacts with HP1α, at pericentric heterochromatic foci and is required to maintain heterochromatin through this association [83,84]. At the same time, it is present at boundary elements, where it plays a role in prevention of spread of silent heterochromatin [85]. Like H3.3, it can accumulate histone marks linked to ‘poised’ promoters, such as those involved in lineage specification and development [86].
H2A.Z deficiency results in embryonic lethality in mice, as in drosophila, indicating its importance in embryonic development [78,87]. Brain-specific knockouts of H2A.Z have been created, which exhibit altered proliferation and differentiation of neural progenitors [88]. ZNHIT1 knockouts in intestinal epithelial tissue have shown the importance of this SNF2‐related CBP activator protein (SRCAP) subunit in H2A.Z deposition and intestinal stem cell homoeostasis [89,90].
Recently, it has been shown that there are two isoforms of H2A.Z, H2A.Z.1 and H2A.Z.2, in vertebrates. Although these isoforms are deposited to the same extent by SRCAP, the two forms have some differences in the ways they affect nucleosomal stability and chromatin structure [91]. Nevertheless, most studies either take both to be one form, or focus on the form encoded by H2A.Z.1.
Expression and function in myogenesis
H2A.Z levels are low in proliferating myoblasts and accumulate rapidly in myotubes [57], as well as in dystrophic muscles [92]. However, downregulation of H3.1, H3.2, macroH2A, H2A.X, H2A.Z, and CENP-A has also been reported in C2C12 myotubes upon onset of differentiation [58].
H2A.Z is important for the expression of MyoD (Figure 2(b)). Ectopic expression of a non-acetylable form of H2A.Z in place of the wild-type form results in a less permissive chromatin structure around the MyoD transcriptional start site, reduced MyoD expression and a block in myogenic differentiation in C2C12 cells. There are five lysine residues that can be acetylated at the N-terminus of H2A.Z (Lysine-4, 7, 11, 13, and 15) and acetylation has been linked to its role in transcriptional activation, whereas ubiquitination (which usually occurs at Lysine-120, 121, and 125 at the C-terminus) results in transcriptional repression and polycomb signalling [93–95]. However, knockdown of H2A.Z did not result in the same phenotype, which could be due to the presence of isoforms that were not depleted by the chosen shRNA, resulting in residual H2A.Z [96].
The p38 MAPK substrate ZNHIT1 or p18Hamlet, a subunit of the SRCAP chromatin remodelling complex, is required for the deposition of H2A.Z at the Myog promoter. This accumulation of H2A.Z during the early stages of differentiation is required for the activation of Myog expression (Figure 3(b)). This is similar to the HIRA/Chd2/Asf1a-dependent deposition of H3.3 at the same locus. Furthermore, similar to the Akt-dependent phosphorylation of HIRA, which acts as a signal for the induction of differentiation through H3.3 deposition, p38 MAPK‐mediated p18Hamlet phosphorylation is also required for the deposition of H2A.Z [97]. Some work will be required to understand how the processes of H3.3 deposition and H2A.Z deposition interact at the Myog promoter and affect myogenic differentiation.
In post-mitotic cells, such as mature muscle, H2A.Z is not required either to activate or maintain transcription; it is a marker rather than a driver of transcription. RNA-sequencing and ATAC-sequencing showed that neither gene expression nor chromatin accessibility was significantly altered by loss of H2A.Z. Thus, while it is necessary for the transition from myoblasts to early differentiation, it is not required in differentiated tissue [98].
MacroH2A
MacroH2A is nearly thrice the size of canonical H2A due to the presence of an evolutionarily conserved non-histone macro domain of ∼25 kDa. This domain can interact with metabolites such as NAD, thus playing a role in regulation of energy and metabolism [99–102]. macroH2A is an important epigenetic regulator of developmentally regulated genes [103]. While histone variants tend to be non-allelic for the most part, macroH2A variants are produced by alternative splicing. In mammals, three macroH2A proteins-macroH2A1.1, macroH2A1.2 and macroH2A2- are produced through two genes and one mutually exclusive splicing event [101].
Knockout of both isoforms in mice leads to pre-natal and post-natal growth deficiencies and loss of reproductive capacity [104]. macroH2A plays an important role in X-chromosome inactivation in females [105]. Double knockout models show that these variants, along with H3K27me3, are found on important pluripotency genes, where they act as a barrier to induced pluripotency [106,107]. Cells harbouring the double knockout can differentiate, but retain the ability to return to a stem-cell like state [106]. While macroH2A1 has been mainly implicated in limiting chromatin accessibility [108] and promoting gene silencing, it has been suggested that it may have broader roles, including finetuning gene expression [109]. macroH2A is required to maintain nuclear organization and heterochromatin structure [110].
Expression and function in myogenesis
Both forms are found in proliferating myoblasts, but mRNA encoding macroH2A1.1 rapidly becomes predominant during differentiation. Indeed, while protein levels of macroH2A1.1 increase over differentiation, macroH2A1.2 levels rapidly decrease [111].
The macroH2A1.1 isoform binds to ADP-ribose, and in differentiating myotubes, where there is a switch towards increased carbon use for ATP production, macroH2A1.1 binds to and inhibits poly-ADP ribose polymerase (PARP-1) activity. Myotubes lacking macroH2A1.1 show defective mitochondrial respiratory capacity because the buffer of NAD+ precursors (NMN in particular) is disrupted [111]. Therefore, macroH2A plays an important role in protecting differentiated myotubes from oxidative stress [111–113].
Similar to H3.3 and H2A.Z, macroH2A1.2 is also recruited to the genes involved in the myogenic regulatory network and is required for the activation of the myogenic programme . Unlike H3.3 and H2A.Z, however, macroH2A is recruited to the prospective enhancers of genes such as Myog, where its presence is required for the activating H3K27 acetylation mark and the recruitment of transcription factor Pbx1 (Figure 3(c)), which cooperates with MyoD [114,115]. macroH2A1.2 ‘marks’ prospective enhancers, and although it is not critical for the maintenance of active enhancers, it is important for the activation of the myogenic gene network [116].
H1b
While the contribution of replication-independent histone variants in myogenic differentiation, for the most part, is activating in nature, there is also a repressive role played by the linker histone H1. H1 histones are ∼200 amino acid residues in length. H1 stabilizes the nucleosome through interactions with DNA and facilitates higher-order folding of nucleosomes [117]. There are 11 known variants histone H1 in humans. H1.1-H1.5 are replication-dependent, whereas H1.X and H1.0 are replication-independent. The remaining forms (H1oo, HILS, H1T, and H1T2) are specific to germ cells. Individual knockouts of the somatic variants are tolerated in mice [118], although combined knockout of three variants- H1c, H1d, and H1e- was embryonic lethal [119]. The loss of H10 in mice is also tolerated, and mice develop normally [120]. In Drosophila, however, H1 is important for embryonic development [121]. In Xenopus, H1 has been shown to be important for regulation of mesodermal differentiation [122].
Function in myogenesis
In muscle, H1b (H1.5), a variant of mouse histone H1, cooperates with the transcriptional repressor Msx1 to bind to the CER of MyoD, resulting in a closed chromatin conformation and preventing MyoD activation. There is consequently impaired myotube formation. The ability of Msx1 to repress myogenic differentiation depends on its interaction of H1b found at the CER of MyoD [123].
An extra-nuclear role for H1 in regulating skeletal muscle regeneration has also been shown. H1 is found at the plasma membrane and extracellular matrix in a pool of regenerating cells, where it interacts with the heparan sulphate chain of the proteoglycan perlecan. Perlecan is present on the plasma membrane of myoblasts, with reduced expression during differentiation [124]. H1-perlecan interaction results in a strong proliferative signal for myoblasts, indicating a role for H1 in skeletal muscle regeneration. The source of H1 in the extracellular matrix is hypothesized to be cell necrosis [125].
Conclusions
Hitherto, H3.3, H2A.Z, macroH2A, and H1b have been shown to be important for skeletal differentiation (Table 2). H3.3 is deposited at regulatory and enhancer regions of MyoD, whereas H2A.Z is deposited around the transcription start site. While H2A.Z is required for the accumulation of activating marks, H3.3 is required for the removal of H1. H1b has the opposite function; its presence serves to repress MyoD expression. Unlike MyoD where deposition of variants occurs at different loci, multiple variants appear to deposited around the promoter resulting in Myog expression. Loss of either of these variants shows a reduction in the expression of Myog and other myogenic genes. However, given the relationship between H3.3 and H2A.Z, one can predict the existence of nucleosomes containing both these variants [37,126]. macroH2A is also required at enhancer regions (Table 2).
Table 2.
Role of histone variants in expression of MRFs
| MRF | RI Variant | Location | Deposition | Effect | References |
|---|---|---|---|---|---|
| MyoD | H3.3 | CER, PRR | Prior to onset of differentiation; increases during differentiation | Removal of H1; gene activation | 58 |
| H2A.Z | TSS | Prior to onset of differentiation | Accumulation of Ac marks; gene activation | 96 | |
| H1b | CER | - | Cooperates with MSX1; prevents activation | 123 | |
| Myog | H3.3 | Promoter | Prior to onset of differentiation; increases during differentiation | Accumulation of H3K4me3 marks; gene activation | 60 |
| H2A.Z | Promoter | Onset of differentiation | Gene activation | 97 | |
| macroH2A | Enhancer | Onset of differentiation | Accumulation of H3K27Ac marks; recruitment of Pbx1 and MyoD; gene activation | 116 |
The individual RI histone variants currently known to be involved in the in expression of two key myogenic regulatory factors, MyoD and Myog are shown here, along with location, time and effect of deposition.
While these studies have clearly started to shed light on their critical role in myogenesis, several questions remain: First, given their myriad roles in myogenesis, it is conceivable that the expression or function of histone variants may be deregulated in myopathies, but this has not been studied in any detail. Deregulation of the expression of certain variants, or their chaperone complexes, may have important pathological implications that are currently underappreciated. For instance, myogenesis is impaired in dystrophies, rhabdomyosarcoma, and age-dependent decline of muscle regenerative capacity. Congenital myopathy is characterized by impaired myotube formation as a result of loss of MyoD, Myog, dystrophin, and N-CAM expression. This defect is limited to skeletal muscle only [127]. Immunohistochemical staining of biopsies from patients with various forms of primary myopathies and neurogenic atrophy also showed varying degrees of loss of MyoD expression, mostly coinciding with the extent of atrophy [128]. It is unclear to what extent these defects are due to deregulation of histone variants that regulate MyoD expression and activity. The transcription factor Dux4, whose expression is deregulated in facioscapulohumeral dystrophy (FSHD), induces the expression of H3.X and H3.Y, which in turn are incorporated into the targets of Dux4, therefore enhancing a network of deregulated Dux4 target genes [129]. Other studies have shown the importance of deregulated histone modifications in certain myopathies [130]. Similarly, the age-related loss of muscle function is characterized by epigenetic changes that affect the ability of satellite cells to regenerate muscle tissue [131]. Small molecules targeting some epigenetic regulators that affect the differentiation process have been developed. For example, histone deacetylase (HDAC) inhibitors have been proposed as a therapeutic option for myopathies, but HDAC inhibitors often pose the problem of lack of specificity and off-target effects [132]. The requirement for certain histone variants in myogenic differentiation is somewhat overlooked. The Chaserr lncRNA inhibits Chd2 expression and thus targeting it could be a way to restore Chd2 levels [133] and Myog expression, as Myog is a specific target of the Chd2-HIRA-MyoD complex [60]. Similarly, it has been suggested that small-molecule inhibitors of the Asf1 histone chaperone may be used for therapeutic purposes in cancers, which harbour high levels of H3K56Ac marks and aberrant cell proliferation, or for treatment of fungal infections, as this histone mark is more abundant in fungal genomes [134,135]. The allosteric inhibitor, MK2206 can prevent the S650 phosphorylation of HIRA through inhibition of Akt phosphorylation. This is another potential, if somewhat complex, way to control HIRA and H3.3 deposition [71].
Second, while the expression of certain variants changes during myogenic differentiation, little is known about the upstream regulators of their expression. H3.3 is selectively incorporated into specific loci in skeletal muscle in response to exercise. It has been suggested that there is nucleosomal repackaging in response to exercise training. The kind and intensity of training are thought to determine target gene regulation [136–138]. However, the transcription factors and regulators involved are not yet clear. This is important if the expression of variants requires manipulation in the treatment of myopathies. Third, ChIP-sequencing and ATAC-sequencing have been done to study global changes upon perturbation of specific variants. Some targets genes such as MyoD and Myog have been identified. However, identification of additional downstream targets in physiological contexts as well as in myopathies may reveal novel genes that may be possible to target therapeutically in myopathies that are associated with a block of myogenic differentiation.
Several noncoding RNAs (ncRNAs), including microRNAs and lncRNAs have been shown to be critical to skeletal myogenesis (reviewed in [139–144]). However, the interaction between these ncRNAs and histone variants is unclear. It has been shown that Chro1 is important for H3.3 deposition [66]. H1.3 has also been previously shown to repress the expression of the h19 ncRNA in ovarian cancer [145], but the role of histone variants in either regulating expression of ncRNAs, or any interaction with ncRNAs, in the context of myogenesis, has not been shown. As these ncRNAs could also prove to be potential targets for regulating myogenesis, the interaction between ncRNAs and histone variants involved in skeletal myogenesis should be analysed. Similarly, DNA methylation is another important form of epigenetic regulation involved in myogenesis. Histone and DNA modifications work in conjunction to regulate gene expression [146–148]. DNA methylation has been shown to play an important role in skeletal myogenesis and response to exercise [149–152]. In addition, certain histone variants are known to regulate DNA methylation by regulating access to DNA methyltransferases and other modifiers (Table 3) [75,153–162]. However, the interaction between histone variants and DNA methylation has not been studied in the context of skeletal myogenesis. Such studies will shed light on mechanisms by which histone variants in combination with other epigenetic regulators, orchestrate chromatin landscaping during skeletal myogenesis.
Table 3.
Transcriptional properties of histone variants, PTMs, and associated epigenetic marks
| Variant | Transcriptional function | Associated Epigenetic Marks/Modifications | Associated ncRNA |
|---|---|---|---|
| H1.1 | Predominantly repressive | Multiple histone PTMs thought to be important for epigenetic regulation, including phosphorylation, methylation, acetylation, citrullination, ADP ribosylation, ubiquitination, carbonylation, formylation, denitration, crotonylation and lysine 2-hydroxyisobutyrylation[246,247] Enriched in regions of DNA methylation, recruit methyltransferases[161,162] |
|
| H1.2 | Activating[173], Repressive[174,175] | ||
| H1.3 | Repressive[145] | h19[145] | |
| H1.4 | Activating[174,177,178] | ||
| H1.5 | Predominantly repressive | ||
| H1.0 | Repressive | ||
| H1.X | |||
| H1t | - | ||
| H1T2 | - | ||
| H1oo | - | ||
| HILS | - | ||
| H2A | - | Multiple histone modifications, including acetylation, phosphorylation, methylation, demethylation, deamination, β-N-acetylglucosamine, ADP-ribosylation, ubiquitination and sumoylation[15,248] | |
| H2A.X | Predominantly repressive[249] | Specific histone residues include: K118Ub, K119Ub, K5Ac, K36Ac, K134me, K5Sumo, K9Sumo, K13Sumo, K15Sumo, K118Sumo, K119Sumo, K127Sumo, K133Sumo, K134Sumo[250] | |
| macroH2A.1.1 | Both activating and repressive[103,106,109,200,251] | Specific histone residues include: K17me, K122me2, K237me/me2, T128P, S137P, T101P, T136P, S139P, Y142P[250,252] | |
| macroH2A.1.2 | |||
| macroH2A2 | - | ||
| H2A.Z.2.1 | Activating[3] | Specific histone residues include: K4Ac, K7Ac, K11Ac, K13Ac, K15Ac, K120Ub, K121Ub, K125Ub, K4me, K7me, K101me2, K115Ub, K116Ub, K122Ub[250] Antagonizes DNA methylation[75] |
|
| H2A.Z.2.2 | Activating[201] | ||
| H2A.Z.1 | Predominantly activating[3,7596,205] | ||
| H2A.Bbd | Activating[207,253] | - | |
| H2B | - | Multiple histone modifications, including acetylation, phosphorylation, methylation, demethylation, deamination, β-N-acetylglucosamine, ADP-ribosylation, ubiquitination and sumoylation[15,248] Deubiquitination/deacetylation required for DNA methylation[159,160] |
|
| TSH2B | - | - | |
| H2BFWT | - | - | |
| H3.1 | Predominantly repressive[223,224] | Specific histone residues include: K27me3 [65,223,224], K9me2[254], K14Ac[246], K64me1[246] | |
| H3.2 | Predominantly repressive[224] | Specific residues include: K27me2/3, K36me1[246] | |
| H3.3 | Predominantly activating[34,35,37-40] | Specific histone residues include: K4me3, K36me3, K4Ac, K4me2, K9Ac, K27Ac, K79Ac, K14Ac, K18/K23Ac, K36me/me2 K79me/2[34,35,37-40,246,254] K27me3[41] Facilitates gene body and promoter DNA methylation[157,158] |
Chro1[66] |
| H3.1 t | - | - | |
| H3.5 | - | - | |
| H3.Y | - | - | |
| H3.X | - | - | |
| CENP-A | Unclear | Specific histone residues include: G1me3, S7P, S16P, S18P, S68P, K124Ub, K124Ac, K124me[255] | |
| H4 | - | Multiple histone modifications, including acetylation, phosphorylation, methylation, demethylation, deamination, β-N-acetylglucosamine, ADP-ribosylation, ubiquitination and sumoylation[15,248] |
The key transcriptional role of histone variants, along with the PTMs and epigenetic marks associated with each, are shown here. DNA methylation is an important regulatory mechanism in skeletal myogenesis and histone variants involved in changes in DNA methylation are shown here (under Epigenetic changes). ncRNAs, which also play an important role in skeletal myogenesis, may well work in conjunction with histone variants and require further study.
Funding Statement
This work was supported by Ministry of Education grant (MOE2019-T2-1-024);MOE [MOE2019-T2-1-024].
Disclosure statement
No potential conflict of interest.
References
- [1].Wang ZF, Krasikov T, Frey MR, et al. Characterization of the mouse histone gene cluster on chromosome 13: 45 histone genes in three patches spread over 1Mb. Genome Res. 1996;6(8):688–701.. [DOI] [PubMed] [Google Scholar]
- [2].Burgess RJ, Zhang Z.. Histone chaperones in nucleosome assembly and human disease. Nat Struct Mol Biol. 2013;20:14–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Buschbeck M, Hake SB.. Variants of core histones and their roles in cell fate decisions, development and cancer. Nat Rev Mol Cell Biol. 2017;18:299–314. [DOI] [PubMed] [Google Scholar]
- [4].Gurard-Levin ZA, Quivy J-P, Almouzni G. Histone chaperones: assisting histone traffic and nucleosome dynamics. Annu Rev Biochem. 2014;83:487–517. [DOI] [PubMed] [Google Scholar]
- [5].Venkatesh S, Workman JL. Histone exchange, chromatin structure and the regulation of transcription. Nat Rev Mol Cell Biol. 2015;16:178–189. [DOI] [PubMed] [Google Scholar]
- [6].Hergeth SP, Schneider R. The H1 linker histones: multifunctional proteins beyond the nucleosomal core particle. EMBO Rep. 2015;16:1439–1453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Izzo A, Kamieniarz K, Schneider R. The histone H1 family: specific members, specific functions? Biol Chem. 2008;389:333–343. [DOI] [PubMed] [Google Scholar]
- [8].Doenecke D, Albig W, Bouterfa H, et al. Organization and expression of H1 histone and H1 replacement histone genes. J Cell Biochem. 1994;54:423–431. [DOI] [PubMed] [Google Scholar]
- [9].Rudnicki MA, Jaenisch R. The MyoD family of transcription factors and skeletal myogenesis. BioEssays News Rev Mol Cell Dev Biol. 1995;17(3):203–209. [DOI] [PubMed] [Google Scholar]
- [10].Weintraub H, Davis R, Tapscott S, et al. The myoD gene family: nodal point during specification of the muscle cell lineage. Science. 1991;251(4995):761–766.. [DOI] [PubMed] [Google Scholar]
- [11].Weintraub H, Tapscott SJ, Davis RL, et al. Activation of muscle-specific genes in pigment, nerve, fat, liver, and fibroblast cell lines by forced expression of MyoD. Proc. Natl. Acad. Sci. U. S. A. 1989;86(14):5434–5438.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Le Grand F, Rudnicki MA. Skeletal muscle satellite cells and adult myogenesis. Curr. Opin. Cell Biol. 2007;19(6):628–633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Chal J, Pourquié O. Making muscle: skeletal myogenesis in vivo and in vitro. Development. 2017;144(12):2104–2122. [DOI] [PubMed] [Google Scholar]
- [14].Hernández-Hernández JM, García-González EG, Brun CE, et al. The myogenic regulatory factors, determinants of muscle development, cell identity and regeneration. Semin Cell Dev Biol. 2017;72:10–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell Res. 2011;21(3):381–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Giordani L, Puri PL. Epigenetic control of skeletal muscle regeneration. Febs J. 2013;280(17):4014–4025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Perdiguero E, Sousa-Victor P, Ballestar E, et al. Epigenetic regulation of myogenesis. Epigenetics. 2009;4(8):541–550. [DOI] [PubMed] [Google Scholar]
- [18].Saccone V, Puri PL. Epigenetic regulation of skeletal myogenesis. Organogenesis. 2010;6(1):48–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Sartorelli V, Juan AH. Sculpting chromatin beyond the double helix: epigenetic control of skeletal myogenesis. Curr Top Dev Biol. 2011;96:57–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Jin W, Peng J, Jiang S. The epigenetic regulation of embryonic myogenesis and adult muscle regeneration by histone methylation modification. Biochem Biophys Rep. 2016;6:209–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Hota SK, Bruneau BG. ATP-dependent chromatin remodeling during mammalian development. Dev Camb Engl. 2016;143:2882–2897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Toto PC, Puri PL, Albini S. SWI/SNF-directed stem cell lineage specification: dynamic composition regulates specific stages of skeletal myogenesis. Cell Mol Life Sci CMLS. 2016;73:3887–3896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Simone C, Forcales SV, Hill DA, et al. p38 pathway targets SWI-SNF chromatin-remodeling complex to muscle-specific loci. Nat. Genet. 2004;36:738–743. [DOI] [PubMed] [Google Scholar]
- [24].OhkawaY, Yoshimura S, Higashi C, et al. Myogenin and the SWI/SNF ATPase Brg1 maintain myogenic gene expression at different stages of skeletal myogenesis. J Biol Chem. 2007;282(9):6564-70. [DOI] [PubMed] [Google Scholar]
- [25].Doynova MD, Markworth JF, Cameron-Smith D, et al. Linkages between changes in the 3D organization of the genome and transcription during myotube differentiation in vitro. Skelet Muscle. 2017;7:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Harada A, Ohkawa Y, Imbalzano AN. Temporal regulation of chromatin during myoblast differentiation. Semin Cell Dev Biol. 2017;72:77–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Neems DS, Garza-Gongora AG, Smith ED, et al. Topologically associated domains enriched for lineage-specific genes reveal expression-dependent nuclear topologies during myogenesis. Proc Natl Acad Sci U S A. 2016;113:E1691–1700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].McKinsey TA, Zhang CL, Olson EN. Signaling chromatin to make muscle. Curr Opin Cell Biol. 2002;14:763–772. [DOI] [PubMed] [Google Scholar]
- [29].Franklin SG, Zweidler A. Non-allelic variants of histones 2a, 2b and 3 in mammals. Nature. 1977;266(5599):273–275. [DOI] [PubMed] [Google Scholar]
- [30].Goldberg AD,Banaszynski LA, Noh KM, et al. Distinct factors control histone variant H3.3 localization at specific genomic regions. Cell. 2010;140:678–691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Deaton AM, Gómez-Rodríguez M, Mieczkowski J, et al. Enhancer regions show high histone H3.3 turnover that changes during differentiation. eLife. 2016 Jun 15;5:e15316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Kaludov NK, Pabón-Peña L, Seavy M, et al. A mouse histone H1 variant, H1b, binds preferentially to a regulatory sequence within a mouse H3.2 replication-dependent histone gene. J Biol Chem. 1997;272:15120–15127. [DOI] [PubMed] [Google Scholar]
- [33].Braunschweig U, Hogan GJ, Pagie L, et al. Histone H1 binding is inhibited by histone variant H3.3. Embo J. 2009;28:3635–3645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Ahmad K, Henikoff S. The histone variant H3.3 marks active chromatin by replication-independent nucleosome assembly. Mol Cell. 2002;9:1191–1200. [DOI] [PubMed] [Google Scholar]
- [35].Chen P, Zhao J, Wang Y, et al. H3.3 actively marks enhancers and primes gene transcription via opening higher-ordered chromatin. Genes Dev. 2013;27(19):2109–2124.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Harada A, Maehara K, Ono Y, et al. Histone H3.3 sub-variant H3mm7 is required for normal skeletal muscle regeneration. Nat Commun. 2018;9:1400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Jin C, Zang C, Wei G, et al. H3.3/H2A.Z double variant-containing nucleosomes mark ‘nucleosome-free regions’ of active promoters and other regulatory regions. Nat Genet. 2009;41:941–945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Loyola A, Bonaldi T, Roche D, et al. PTMs on H3 variants before chromatin assembly potentiate their final epigenetic state. Mol Cell. 2006;24:309–316. [DOI] [PubMed] [Google Scholar]
- [39].Mito Y, Henikoff JG, Henikoff S. Genome-scale profiling of histone H3.3 replacement patterns. Nat Genet. 2005;37:1090–1097. [DOI] [PubMed] [Google Scholar]
- [40].Wirbelauer C, Bell O, Schübeler D. Variant histone H3.3 is deposited at sites of nucleosomal displacement throughout transcribed genes while active histone modifications show a promoter-proximal bias. Genes Dev. 2005;19:1761–1766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Ng RK, Gurdon JB. Epigenetic memory of an active gene state depends on histone H3.3 incorporation into chromatin in the absence of transcription. Nat Cell Bio. 2008;10:102–109. [DOI] [PubMed] [Google Scholar]
- [42].Fang HT,El Farran CA, Xing QR, et al. Global H3.3 dynamic deposition defines its bimodal role in cell fate transition. Nat Commun. 2018,(1):1537. [DOI] [PMC free article] [PubMed]
- [43].Jang C-W, Shibata Y, Starmer J, et al. Histone H3.3 maintains genome integrity during mammalian development. Genes Dev. 2015;29(13):1377–1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Tang MCW, Jacobs SA, Mattiske DM, et al. Contribution of the two genes encoding histone variant H3.3 to viability and fertility in mice. PLOS Genet. 2015;11(2):e1004964.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Bush KM, Yuen BT, Barrilleaux BL, et al. Endogenous mammalian histone H3.3 exhibits chromatin-related functions during development. Epigenetics Chromatin. 2013;6(1):7.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Szenker E, Lacoste N, Almouzni G. A developmental requirement for HIRA-dependent H3.3 deposition revealed at gastrulation in Xenopus. Cell Rep. 2012;1(6):730–740. [DOI] [PubMed] [Google Scholar]
- [47].Sitbon D, Boyarchuk E, Dingli F, et al. Histone variant H3.3 residue S31 is essential for Xenopus gastrulation regardless of the deposition pathway. Nat Commun. 2020;11(1):1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Bachu M, Tamura T, Chen C, et al. A versatile mouse model of epitope-tagged histone H3.3 to study epigenome dynamics. J Biol Chem. 2019;294:1904–1914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Evano B, Khalilian S, Le Carrou G, et al. Dynamics of asymmetric and symmetric divisions of muscle stem cells in vivo and on artificial niches. Cell Rep. 2020;30(3195–3206.e7). DOI: 10.1016/j.celrep.2020.01.097 [DOI] [PubMed] [Google Scholar]
- [50].Banaszynski LA, Wen D, Dewell S, et al. Hira-dependent histone H3.3 deposition facilitates PRC2 recruitment at developmental loci in ES cells. Cell. 2013.155(1):107–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Adam S, Polo SE, Almouzni G. Transcription Recovery after DNA damage requires chromatin priming by the H3.3 histone chaperone HIRA. Cell. 2013;155(1):94–106. [DOI] [PubMed] [Google Scholar]
- [52].Dutta D, Ray S, Home P, et al. Regulation of angiogenesis by histone chaperone HIRA-mediated incorporation of lysine 56-acetylated histone H3.3 at chromatin domains of endothelial genes. J Biol Chem. 2010;285:41567–41577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Lewis PW, Elsaesser SJ, Noh K-M, et al. Daxx is an H3.3-specific histone chaperone and cooperates with ATRX in replication-independent chromatin assembly at telomeres. Proc Natl Acad Sci U S A. 2010;107:14075–14080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Drané P, Ouararhni K, Depaux A, et al. The death-associated protein DAXX is a novel histone chaperone involved in the replication-independent deposition of H3.3. Genes Dev. 2010;24:1253–1265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Fromental-Ramain C, Ramain P, Hamiche A. The drosophila DAXX-like protein (DLP) Cooperates with ASF1 for H3.3 deposition and heterochromatin formation. Mol Cell Biol. 2017;37. DOI: 10.1128/MCB.00597-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Konev AY, Tribus M, Park SY, et al. CHD1 motor protein is required for deposition of histone variant H3.3 into chromatin in vivo. Science. 2007;317:1087–1090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Wunsch AM, Lough J. Modulation of histone H3 variant synthesis during the myoblast-myotube transition of chicken myogenesis. Dev Biol. 1987;119:94–99. [DOI] [PubMed] [Google Scholar]
- [58].Yang J-H, Song Y, Seol J-H, et al. Myogenic transcriptional activation of MyoD mediated by replication-independent histone deposition. Proc Natl Acad Sci. 2011;108(1):85–90.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Song TY, Yang JH, Park JY, et al. The role of histone chaperones in osteoblastic differentiation of C2C12 myoblasts. Biochem Biophys Res Commun. 2012. 423(4):726–32 [DOI] [PubMed] [Google Scholar]
- [60].Harada A, Okada S, Konno D, et al. Chd2 interacts with H3.3 to determine myogenic cell fate. Embo J. 2012;31(13):2994–3007.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Azuara V, Perry P, Sauer S, et al. Chromatin signatures of pluripotent cell lines. Nat. Cell Biol. 2006;8:532–538. [DOI] [PubMed] [Google Scholar]
- [62].Bernstein BE, Mikkelsen TS, Xie X, et al.. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell. 2006. 125(2):315-26. [DOI] [PubMed] [Google Scholar]
- [63].Mikkelsen TS, Ku M, Jaffe DB, et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature. 2007;448(7153):553–560.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Vastenhouw NL, Zhang Y, Woods IG, et al. Chromatin signature of embryonic pluripotency is established during genome activation. Nature. 2010;464(7290):922–926.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Harada A, Maehara K, Sato Y, et al. Incorporation of histone H3.1 suppresses the lineage potential of skeletal muscle. Nucleic Acids Res. 2015. January;43(2):775-86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Park J, Lee H, Han N, et al. Long non-coding RNA ChRO1 facilitates ATRX/DAXX-dependent H3.3 deposition for transcription-associated heterochromatin reorganization. Nucleic Acids Res. 2018;46(22):11759–11775.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Morgan JE, Partridge TA. Muscle satellite cells. Int J Biochem Cell Biol. 2003;35:1151–1156. [DOI] [PubMed] [Google Scholar]
- [68].Yablonka-Reuveni Z. The skeletal muscle satellite cell. J Histochem Cytochem. 2011;59:1041–1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Frontiers | satellite cells in human skeletal muscle plasticity | physiology. https://www.frontiersin.org/articles/10.3389/fphys.2015.00283/full. [DOI] [PMC free article] [PubMed]
- [70].Maehara K,Harada A, Sato Y, et al.. Tissue-specific expression of histone H3 variants diversified after species separation. Epigenetics Chromatin. 2015. 8:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Yang JH,Song TY, Jo C, et al.. Differential regulation of the histone chaperone HIRA during muscle cell differentiation by a phosphorylation switch. Exp Mol Med. 2016. August 12;48(8):e252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Valenzuela N, Soibam B, Li L, et al.HIRA deficiency in muscle fibers causes hypertrophy and susceptibility to oxidative stress. J Cell Sci. 2017;130:2551–2563. [DOI] [PubMed] [Google Scholar]
- [73].Yang JH,Choi JH, Jang H, et al.. Histone chaperones cooperate to mediate Mef2-targeted transcriptional regulation during skeletal myogenesis. Biochem Biophys Res Commun. 2011. April 15;407(3):541-7. [DOI] [PubMed] [Google Scholar]
- [74].Huh MS,Price O'Dea T, Ouazia D, et al.. Compromised genomic integrity impedes muscle growth after Atrx inactivation. J Clin Invest. 2012. December;122(12):4412-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Giaimo BD, Ferrante F, Herchenröther A, et al. The histone variant H2A.Z in gene regulation. Epigenetics Chromatin. 2019;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Weber CM, Henikoff S. Histone variants: dynamic punctuation in transcription. Genes Dev. 2014;28:672–682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Zlatanova J, Thakar A. H2A.Z: view from the top. Structure. 2008;16:166–179. [DOI] [PubMed] [Google Scholar]
- [78].Faast R, Thonglairoam V, Schulz TC, et al.. Histone variant H2A.Z is required for early mammalian development. Current Biology: : CB. 2001. Aug;11(15):1183–1187 [DOI] [PubMed] [Google Scholar]
- [79].Adam M, Robert F, Larochelle M, et al. H2A.Z is required for global chromatin integrity and for recruitment of RNA polymerase II under specific conditions. Mol Cell Biol. 2001;21:6270–6279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Barski A,Cuddapah S, Cui K, et al.. High-resolution profiling of histone methylations in the human genome. Cell. 2007;129(4):823-837. [DOI] [PubMed] [Google Scholar]
- [81].Raisner RM,Hartley PD, Meneghini MD,et al. Histone variant H2A.Z marks the 5' ends of both active and inactive genes in euchromatin. Cell. 2005. October 21;123(2):233-48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Brickner DG,Cajigas I, Fondufe-Mittendorf Y, et al. H2A.Z-mediated localization of genes at the nuclear periphery confers epigenetic memory of previous transcriptional state. Version 2. PLoS Biol. 2007. April;5(4):e81. [DOI] [PMC free article] [PubMed]
- [83].Rangasamy D, Greaves I, Tremethick DJ. RNA interference demonstrates a novel role for H2A.Z in chromosome segregation. Nat Struct Mol Biol. 2004;11:650–655. [DOI] [PubMed] [Google Scholar]
- [84].Rangasamy D, Berven L, Ridgway P, et al. Pericentric heterochromatin becomes enriched with H2A.Z during early mammalian development. Embo J. 2003;2:1599–1607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Meneghini MD, Wu M, Madhani HD. Conserved histone variant H2A.Z protects euchromatin from the ectopic spread of silent heterochromatin. Cell. 2003;112:725–736. [DOI] [PubMed] [Google Scholar]
- [86].Subramaniam V,Mazumder A, Surface LE, et al. H2A.Z acidic patch couples chromatin dynamics to regulation of gene expression programs during ESC differentiation. PLoS Genet. 2013;9(8):e1003725. [DOI] [PMC free article] [PubMed]
- [87].van Daal A, Elgin SC. A histone variant, H2AvD, is essential in Drosophila melanogaster. Mol Biol Cell. 1992;3:593–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Shen T, Ji F, Wang Y, et al. Brain-specific deletion of histone variant H2A.z results in cortical neurogenesis defects and neurodevelopmental disorder. Nucleic Acids Res. 2018;46:2290–2307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Rispal J, Baron L, Beaulieu JF, et al. The H2A.Z histone variant integrates Wnt signaling in intestinal epithelial homeostasis. Nat Commun. 2019. Apr 23;10(1):1827 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Zhao B, Chen Y, Jiang N, et al . Znhit1 controls intestinal stem cell maintenance by regulating H2A.Z incorporation. Nat Commun. 2019;10:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Matsuda R, Matsuda R, Hori T, et al. Identification and characterization of the two isoforms of the vertebrate H2A.Z histone variant. Nucleic Acids Res. 2010;38:4263–4273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Wunsch AM, Reinhardt K, Lough J. Normal transitions in synthesis of replacement histones H2A.Z and H3.3 during differentiation of dystrophic myotube cells. A brief note. Mech Ageing Dev. 1991;59:299–305. [DOI] [PubMed] [Google Scholar]
- [93].Draker R, Sarcinella E, Cheung P. USP10 deubiquitylates the histone variant H2A.Z and both are required for androgen receptor-mediated gene activation. Nucleic Acids Res. 2011;39:3529–3542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Ishibashi T, Dryhurst D, Rose KL, et al. Acetylation of vertebrate H2A.Z and its effect on the structure of the nucleosome. Biochemistry. 2009;48:5007–5017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Sarcinella E, Zuzarte PC, Lau PNI, et al. Monoubiquitylation of H2A.Z distinguishes its association with euchromatin or facultative heterochromatin. Mol Cell Biol. 2007;27:6457–6468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Law C, Cheung P. Expression of non-acetylatable H2A.Z in myoblast cells blocks myoblast differentiation through disruption of MyoD expression. J Biol Chem. 2015;290:13234–13249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Cuadrado A, Corrado N, Perdiguero E, et al. Essential role of p18Hamlet/SRCAP-mediated histone H2A.Z chromatin incorporation in muscle differentiation. Embo J. 2010;29:2014–2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Belotti E, Lacoste N, Simonet T, et al. H2A.Z is dispensable for both basal and activated transcription in post-mitotic mouse muscles. Nucleic Acids Res. 2020. May 21;48(9):4601-4613. doi: 10.1093/nar/gkaa157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Chakravarthy S, Luger K. The histone variant macro-H2A preferentially forms “Hybrid Nucleosomes”. J Biol Chem. 2006;281:25522–25531. [DOI] [PubMed] [Google Scholar]
- [100].Pehrson JR, Fuji RN. Evolutionary conservation of histone macroH2A subtypes and domains. Nucleic Acids Res. 1998;26:2837–2842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Chakravarthy S, Gundimella SK, Caron C, et al. Structural characterization of the histone variant macroH2A. Mol Cell Biol. 2005;25:7616–7624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Kustatscher G, Hothorn M, Pugieux C, et al. Splicing regulates NAD metabolite binding to histone macroH2A. Nat Struct Mol Biol. 2005;12:624–625. [DOI] [PubMed] [Google Scholar]
- [103].Buschbeck M, Uribesalgo I, Wibowo I, et al. The histone variant macroH2A is an epigenetic regulator of key developmental genes. Nat Struct Mol Biol. 2009;16:1074–1079. [DOI] [PubMed] [Google Scholar]
- [104].Pehrson JR, Changolkar LN, Costanzi C, et al. Mice without MacroH2A Histone Variants. Mol Cell Biol. 2014;34:4523–4533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Costanzi C, Pehrson JR. Histone macroH2A1 is concentrated in the inactive X chromosome of female mammals. Nature. 1998;393:599–601. [DOI] [PubMed] [Google Scholar]
- [106].Gaspar-Maia A, Qadeer ZA, Hasson D, et al. MacroH2A histone variants act as a barrier upon reprogramming towards pluripotency. Nat Commun. 2013;4:1565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Pasque V, Radzisheuskaya A, Gillich A, et al. Histone variant macroH2A marks embryonic differentiation in vivo and acts as an epigenetic barrier to induced pluripotency. J Cell Sci. 2012;125:6094–6104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Kozlowski M, Corujo D, Hothorn M, et al. MacroH2A histone variants limit chromatin plasticity through two distinct mechanisms. EMBO Rep. 2018. October;19(10):e44445. doi: 10.15252/embr.201744445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Changolkar LN, Costanzi C, Leu NA, et al. Developmental changes in histone macroH2A1-mediated gene regulation. Mol Cell Biol. 2007;27:2758–2764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Douet J, Corujo D, Malinverni R, et al. MacroH2A histone variants maintain nuclear organization and heterochromatin architecture. J Cell Sci. 2017;130:1570–1582. [DOI] [PubMed] [Google Scholar]
- [111].Posavec Marjanović M, Hurtado-Bagès S, Lassi M, et al. MacroH2A1.1 regulates mitochondrial respiration by limiting nuclear NAD+ consumption. Nat Struct Mol Biol. 2017;24:902–910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Oláh G, Szczesny B, Brunyánszki A, et al. Differentiation-associated downregulation of Poly(ADP-Ribose) polymerase-1 expression in myoblasts serves to increase their resistance to oxidative stress. Plos One. 2015;10:e0134227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Ouararhni K, Hadj-Slimane R, Ait-Si-Ali S, et al. The histone variant mH2A1.1 interferes with transcription by down-regulating PARP-1 enzymatic activity. Genes Dev. 2006;20:3324–3336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Berkes CA, Bergstrom DA, Penn BH, et al. Pbx marks genes for activation by MyoD indicating a role for a homeodomain protein in establishing myogenic potential. Mol Cell. 2004;14:465–477. [DOI] [PubMed] [Google Scholar]
- [115].Maves L, Waskiewicz AJ, Paul B, et al. Pbx homeodomain proteins direct Myod activity to promote fast-muscle differentiation. Development. 2007;134:3371–3382. [DOI] [PubMed] [Google Scholar]
- [116].Dell'Orso S, Wang AH, Shih HY, et al. The histone variant macroH2A1.2 is necessary for the activation of muscle enhancers and recruitment of the transcription factor Pbx1. Cell Rep. 2016;14:1156–1168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [117].Zhou BR, Feng H, Kato H, et al. Structural insights into the histone H1-nucleosome complex. Proc Natl Acad Sci. 2013;110:19390–19395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Fan Y, Sirotkin A, Russell RG, et al. Individual somatic H1 subtypes are dispensable for mouse development even in mice lacking the H10 replacement subtype. Mol Cell Biol. 2001;21:7933–7943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [119].Fan Y, Nikitina T, Morin-Kensicki EM, et al. H1 linker histones are essential for mouse development and affect nucleosome spacing in vivo. Mol Cell Biol. 2003;23:4559–4572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Sirotkin AM, Edelmann W, Cheng G, et al. Mice develop normally without the H1(0) linker histone. Proc Natl Acad Sci. 1995;92:6434–6438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Lu X, Wontakal SN, Emelyanov AV, et al. Linker histone H1 is essential for Drosophila development, the establishment of pericentric heterochromatin, and a normal polytene chromosome structure. Genes Dev. 2009;23:452–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Steinbach OC, Wolffe AP, Rupp RAW. Somatic linker histones cause loss of mesodermal competence in Xenopus. Nature. 1997;389:395–399. [DOI] [PubMed] [Google Scholar]
- [123].Lee H, Habas R, Abate-Shen C. MSX1 cooperates with histone H1b for inhibition of transcription and myogenesis. Science. 2004;304:1675–1678. [DOI] [PubMed] [Google Scholar]
- [124].Larraín J, Alvarez J, Hassell JR, et al. Expression of perlecan, a proteoglycan that binds myogenic inhibitory basic fibroblast growth factor, is down regulated during skeletal muscle differentiation. Exp Cell Res. 1997;234:405–412. [DOI] [PubMed] [Google Scholar]
- [125].Henriquez JP, Casar JC, Fuentealba L, et al. Extracellular matrix histone H1 binds to perlecan, is present in regenerating skeletal muscle and stimulates myoblast proliferation. J Cell Sci. 2002;115:2041–2051. [DOI] [PubMed] [Google Scholar]
- [126].Wang Y, Long H, Yu J et al. Histone variants H2A.Z and H3.3 coordinately regulate PRC2-dependent H3K27me3 deposition and gene expression regulation in mES cells. BMC Biol. 2018;16:107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [127].Ketelsen UP, Brand-Saberi B, Uhlenberg B, et al. Congenital myopathy with arrest of myogenesis prior to formation of myotubes. Neuropediatrics. 2005;36:246–251. [DOI] [PubMed] [Google Scholar]
- [128].Parham DM, Dias P, Bertorini T, et al. Immunohistochemical analysis of the distribution of MyoD1 in muscle biopsies of primary myopathies and neurogenic atrophy. Acta Neuropathol (Berl.). 1994;87:605–611. [DOI] [PubMed] [Google Scholar]
- [129].Resnick R, Wong CJ, Hamm DC, et al. DUX4-induced histone variants H3.X and H3.Y Mark DUX4 Target Genes for Expression. Cell Rep. 2019;29:1812–1820.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Majumder A, Behera J, Jeremic N, et al. Hypermethylation: causes and consequences in skeletal muscle myopathy. J Cell Biochem. 2017;118:2108–2117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [131].Gensous N, Bacalini MG, Pirazzini C, et al. The epigenetic landscape of age-related diseases: the geroscience perspective. Biogerontology. 2017;18:549–559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Sincennes M-C, Brun CE, Rudnicki MA. Concise review: epigenetic regulation of myogenesis in health and disease. Stem Cells Transl Med. 2016;5:282–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Rom A, Melamed L, Gil N, et al. Regulation of CHD2 expression by the Chaserr long noncoding RNA gene is essential for viability. Nat Commun. 2019;10:1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [134].Miknis GF, Stevens SJ, Smith LE, et al. Development of novel Asf1-H3/H4 inhibitors. Bioorg Med Chem Lett. 2015;25:963–968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Seol JH, Song TY, Oh SE, et al. Identification of small molecules that inhibit the histone chaperone Asf1 and its chromatin function. BMB Rep. 2015;48:685–690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Lim C, Shimizu J, Kawano F, et al. Adaptive responses of histone modifications to resistance exercise in human skeletal muscle. Plos One. 2020;15:e0231321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Nakamura K, Ohsawa I, Masuzawa R, et al. Running training experience attenuates disuse atrophy in fast-twitch skeletal muscles of rats. J Appl Physiol. 2017;123:902–913. [DOI] [PubMed] [Google Scholar]
- [138].Ohsawa I, Konno R, Masuzawa R, et al. Amount of daily exercise is an essential stimulation to alter the epigenome of skeletal muscle in rats. J Appl Physiol. 2018;125:1097–1104. [DOI] [PubMed] [Google Scholar]
- [139].Gonçalves TJM, Armand A-S. Non-coding RNAs in skeletal muscle regeneration. Non-Coding RNA Res. 2017;2:56–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Martone J, Mariani D, Desideri F, et al. Non-coding RNAs Shaping Muscle. Front Cell Dev Biol. 2019;7:394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [141].Zhao Y, Chen M, Lian D, et al. Non-Coding RNA Regulates the Myogenesis of Skeletal Muscle Satellite Cells, Injury Repair and Diseases. Cells. 2019;8(9):988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Wang S, Jin J, Xu Z, et al. Functions and regulatory mechanisms of lncRNAs in skeletal myogenesis, muscle disease and meat production. Cells. 2019;8(9):1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].Nie M, Deng Z-L, Liu J, et al. Noncoding RNAs, emerging regulators of skeletal muscle development and diseases. Biomed Res Int. 2015:e676575. https://www.hindawi.com/journals/bmri/2015/676575/(2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [144].Sweta S, Dudnakova T, Sudheer S, et al. Importance of long non-coding RNAs in the development and disease of skeletal muscle and cardiovascular lineages. Front Cell Dev Biol. 2019;7:228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Medrzycki M, Zhang Y, Zhang W, et al. Histone h1.3 suppresses h19 noncoding RNA expression and cell growth of ovarian cancer cells. Cancer Res. 2014;74:6463–6473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [146].Cheng X. Structural and functional coordination of DNA and histone methylation. Cold Spring Harb Perspect Biol. 2014;6:a018747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [147].Cheung P, Lau P. Epigenetic regulation by histone methylation and histone variants. Mol Endocrinol. 2005;19:563–573. [DOI] [PubMed] [Google Scholar]
- [148].Miller JL, Grant PA. The role of DNA methylation and histone modifications in transcriptional regulation in humans. Subcell Biochem. 2013;61:289–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [149].Carrió E, Suelves M. DNA methylation dynamics in muscle development and disease. Front Aging Neurosci. 2015;7:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Brown WM. Exercise-associated DNA methylation change in skeletal muscle and the importance of imprinted genes: a bioinformatics meta-analysis. Br J Sports Med. 2015;49:1567–1578. [DOI] [PubMed] [Google Scholar]
- [151].Laker RC, Ryall JG. DNA methylation in skeletal muscle stem cell specification, proliferation, and differentiation. Stem Cells Int. 2016:e5725927. https://www.hindawi.com/journals/sci/2016/5725927/(2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [152].Carrió E, Díez-Villanueva A, Lois S, et al. Deconstruction of DNA methylation patterns during myogenesis reveals specific epigenetic events in the establishment of the skeletal muscle lineage. Stem Cells. 2015;33:2025–2036. [DOI] [PubMed] [Google Scholar]
- [153].Miyata K, Miyata T, Nakabayashi K, et al. DNA methylation analysis of human myoblasts during in vitro myogenic differentiation: de novo methylation of promoters of muscle-related genes and its involvement in transcriptional down-regulation. Hum Mol Genet. 2015;24:410–423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [154].Bourguet P, Picard CL, Yelagandula R, et al. The histone variant H2A.W promotes heterochromatin accessibility for efficient DNA methylation in Arabidopsis. bioRxiv. 2020. 2020.03.19.998609. DOI: 10.1101/2020.03.19.998609. [DOI] [Google Scholar]
- [155].Wollmann H, Stroud H, Yelagandula R, et al. The histone H3 variant H3.3 regulates gene body DNA methylation in Arabidopsis thaliana. Genome Biol. 2017;18:94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [156].Madakashira B, Corbett L, Zhang C, et al. Variant histone H2afv reprograms DNA methylation during early zebrafish development. Epigenetics. 2017;12:811–824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [157].Delbarre E, Jacobsen BM, Reiner AH, et al.Chromatin environment of histone variant H3.3 revealed by quantitative imaging and genome-scale chromatin and DNA immunoprecipitation. Mol Biol Cell. 2010;21:1872–1884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [158].Wollmann H, Stroud H, Yelagandula R, et al. The histone H3 variant H3.3 regulates gene body DNA methylation in Arabidopsis thaliana. Genome Biol. 2017;18:94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [159].Sridhar VV, Kapoor A, Zhang K, et al. Control of DNA methylation and heterochromatic silencing by histone H2B deubiquitination. Nature. 2007;447:735–738. [DOI] [PubMed] [Google Scholar]
- [160].Smith KM, Dobosy JR, Reifsnyder JE, et al. H2B- and H3-specific histone deacetylases are required for DNA methylation in Neurospora crassa. Genetics. 2010;186:1207–1216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [161].Choi J, Lyons DB, Kim MY, et al. DNA methylation and histone H1 jointly repress transposable elements and aberrant intragenic transcripts. Mol Cell. 2020;77:310–323.e7. [DOI] [PubMed] [Google Scholar]
- [162].Yang S-M, Kim BJ, Toro LN, et al. H1 linker histone promotes epigenetic silencing by regulating both DNA methylation and histone H3 methylation. Proc Natl Acad Sci. 1708–1713;110:2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [163].Kato K, Okuwaki M, Nagata K. Role of Template Activating Factor-I as a chaperone in linker histone dynamics. J Cell Sci. 2011;124:3254–3265. [DOI] [PubMed] [Google Scholar]
- [164].Kajitani K, Kato K, Nagata K. Histone H1 chaperone activity of TAF-I is regulated by its subtype-dependent intramolecular interaction. Genes Cells. 2017;22(4):334–347. [DOI] [PubMed] [Google Scholar]
- [165].Zhang Q, Giebler HA, Isaacson MK, et al. Eviction of linker histone H1 by NAP-family histone chaperones enhances activated transcription. Epigenetics Chromatin. 2015;8(1):30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [166].Kepert JF, Mazurkiewicz J, Heuvelman GL, et al. NAP1 modulates binding of linker histone H1 to chromatin and induces an extended chromatin fiber conformation. J Biol Chem. 2005;280:34063–34072. [DOI] [PubMed] [Google Scholar]
- [167].Gadad SS, Senapati P, Syed SH, et al. The multifunctional protein nucleophosmin (NPM1) is a human linker histone H1 chaperone. Biochemistry. 2011;50(14):2780–2789.. [DOI] [PubMed] [Google Scholar]
- [168].George EM, Brown DT. Prothymosin alpha is a component of a linker histone chaperone. FEBS Lett. 2010;584:2833–2836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [169].Karetsou Z, Sandaltzopoulos R, Frangou-Lazaridis M, et al. Prothymosin alpha modulates the interaction of histone H1 with chromatin. Nucleic Acids Res. 1998;26(13):3111–3118.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [170].Kondili K, Tsolas O, Papamarcaki T. Selective interaction between parathymosin and histone H1. Eur J Biochem. 1996;242:67–74. [DOI] [PubMed] [Google Scholar]
- [171].Erard MS, Belenguer P, Caizergues-Ferrer M, et al. A major nucleolar protein, nucleolin, induces chromatin decondensation by binding to histone H1. Eur J Biochem. 1988;175:525–530. [DOI] [PubMed] [Google Scholar]
- [172].Bhan S, May W, Warren SL, et al. Global gene expression analysis reveals specific and redundant roles for H1 variants, H1c and H1°, in gene expression regulation. Gene. 2008;414:10–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [173].Kim K, Choi J, Heo K, et al. Isolation and characterization of a novel H1.2 complex that acts as a repressor of p53-mediated transcription. J Biol Chem. 2008;283:9113–9126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [174].Zheng Y, John S, Pesavento JJ, et al. Histone H1 phosphorylation is associated with transcription by RNA polymerases I and II. J Cell Biol. 2010;189:407–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [175].Kim K, Lee B, Kim J, et al. Linker Histone H1.2 cooperates with Cul4A and PAF1 to drive H4K31 ubiquitylation-mediated transactivation. Cell Rep. 2013;5:1690–1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [176].Sancho M, Diani E, Beato M, et al. Depletion of human histone H1 variants uncovers specific roles in gene expression and cell growth. PLOS Genet. 2008;4:e1000227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [177].Trojer P, Zhang J, Yonezawa M, et al. Dynamic histone H1 Isotype 4 methylation and demethylation by histone lysine methyltransferase G9a/KMT1C and the Jumanji domain-containing JMJD2/KDM4 proteins. J Biol Chem. 2009;284:8395–8405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [178].Kamieniarz K, Izzo A, Dundr M, et al. A dual role of linker histone H1.4 Lys 34 acetylation in transcriptional activation. Genes Dev. 2012;26:797–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [179].Zlatanova J, Doenecke D. Histone H1 zero: a major player in cell differentiation? Faseb J. 1994;8:1260–1268. [DOI] [PubMed] [Google Scholar]
- [180].Takata H, Matsunaga S, Morimoto A et al. H1.X with different properties from other linker histones is required for mitotic progression. FEBS Lett. 2007;581:3783–3788. [DOI] [PubMed] [Google Scholar]
- [181].Drabent B, Kardalinou E, Doenecke D. Structure and expression of the human gene encoding testicular H1 histone (H1t). Gene. 1991;103:263–268. [DOI] [PubMed] [Google Scholar]
- [182].Martianov I, Brancorsini S, Catena R, et al. Polar nuclear localization of H1T2, a histone H1 variant, required for spermatid elongation and DNA condensation during spermiogenesis. Proc Natl Acad Sci. 2005;102:2808–2813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [183].Tanaka H, Iguchi N, Isotani A, et al. HANP1/H1T2, a novel histone H1-like protein involved in nuclear formation and sperm fertility. Mol Cell Biol. 2005;25:7107–7119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [184].Tanaka Y, Kato S, Tanaka M, et al. Structure and expression of the human oocyte-specific histone H1 gene elucidated by direct RT-nested PCR of a single oocyte. Biochem Biophys Res Commun. 2003;304:351–357. [DOI] [PubMed] [Google Scholar]
- [185].Mishra LN, Shalini V, Gupta N et al. Spermatid-specific linker histone HILS1 is a poor condenser of DNA and chromatin and preferentially associates with LINE-1 elements. Epigenetics Chromatin. 2018;11:43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [186].Yan W, Ma L, Burns KH, et al. HILS1 is a spermatid-specific linker histone H1-like protein implicated in chromatin remodeling during mammalian spermiogenesis. Proc Natl Acad Sci. 2003;100:10546–10551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [187].Belotserkovskaya R, Oh S, Bondarenko VA, et al. FACT facilitates transcription-dependent nucleosome alteration. Science. 2003;301:1090–1093. [DOI] [PubMed] [Google Scholar]
- [188].Andrews AJ, Chen X, Zevin A, et al. The histone chaperone Nap1 promotes nucleosome assembly by eliminating nonnucleosomal histone DNA interactions. Mol Cell. 2010;37:834–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [189].Mosammaparast N, Ewart CS, Pemberton LF. A role for nucleosome assembly protein 1 in the nuclear transport of histones H2A and H2B. Embo J. 2002;21:6527–6538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [190].Okuwaki M, Kato K, Nagata K. Functional characterization of human nucleosome assembly protein 1-like proteins as histone chaperones. Genes Cells. 2010;15:13–27. [DOI] [PubMed] [Google Scholar]
- [191].Park Y-J, Chodaparambil JV, Bao Y, et al. Nucleosome assembly protein 1 exchanges histone H2A-H2B dimers and assists nucleosome sliding. J Biol Chem. 2005;280:1817–1825. [DOI] [PubMed] [Google Scholar]
- [192].Kuryan BG, Kim J, Tran NN, et al. Histone density is maintained during transcription mediated by the chromatin remodeler RSC and histone chaperone NAP1 in vitro. Proc Natl Acad Sci U S A. 2012;109:1931–1936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [193].Angelov D, Bondarenko VA, Almagro S, et al. Nucleolin is a histone chaperone with FACT-like activity and assists remodeling of nucleosomes. Embo J. 2006;25:1669–1679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [194].Bönisch C, Hake SB. Histone H2A variants in nucleosomes and chromatin: more or less stable? Nucleic Acids Res. 2012;40:10719–10741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [195].Piquet S, Le Parc F, Bai SK, et al. The histone chaperone FACT coordinates H2A.X-dependent signaling and repair of DNA damage. Mol Cell. 2018;72:888–901.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [196].Celeste A, Petersen S, Romanienko PJ, et al. Genomic instability in mice lacking histone H2AX. Science. 2002;296:922–927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [197].Turinetto V, Orlando L, Sanchez-Ripoll Y, et al. High basal γH2AX levels sustain self-renewal of mouse embryonic and induced pluripotent stem cells. Stem Cells Dayt Ohio. 2012;30:1414–1423. [DOI] [PubMed] [Google Scholar]
- [198].Rogakou EP, Pilch DR, Orr AH, et al. DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J Biol Chem. 1998;273:5858–5868. [DOI] [PubMed] [Google Scholar]
- [199].Ratnakumar K, Duarte LF, LeRoy G, et al. ATRX-mediated chromatin association of histone variant macroH2A1 regulates α-globin expression. Genes Dev. 2012;26:433–438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [200].Creppe C, Janich P, Cantariño N, et al. MacroH2A1 regulates the balance between self-renewal and differentiation commitment in embryonic and adult stem cells. Mol Cell Biol. 2012;32:1442–1452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [201].Bönisch C, Schneider K, Pünzeler S, et al. H2A.Z.2.2 is an alternatively spliced histone H2A.Z variant that causes severe nucleosome destabilization. Nucleic Acids Res. 2012;40:5951–5964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [202].Gursoy-Yuzugullu O, Ayrapetov MK, Price BD. Histone chaperone Anp32e removes H2A.Z from DNA double-strand breaks and promotes nucleosome reorganization and DNA repair. Proc Natl Acad Sci U S A. 2015;112:7507–7512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [203].Alatwi HE, Downs JA. Removal of H2A.Z by INO80 promotes homologous recombination. EMBO Rep. 2015;16:986–994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [204].Ruhl DD, Jin J, Cai Y, et al. Purification of a human SRCAP complex that remodels chromatin by incorporating the histone variant H2A.Z into nucleosomes. Biochemistry. 2006;45:5671–5677. [DOI] [PubMed] [Google Scholar]
- [205].Wong MM, Cox LK, Chrivia JC. The chromatin remodeling protein, SRCAP, is critical for deposition of the histone variant H2A.Z at promoters. J Biol Chem. 2007;282:26132–26139. [DOI] [PubMed] [Google Scholar]
- [206].Hu G, Cui K, Northrup D, et al. H2A.Z facilitates access of active and repressive complexes to chromatin in embryonic stem cell self-renewal and differentiation. Cell Stem Cell. 2013;12:180–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [207].Bao Y, Konesky K, Park YJ, et al. Nucleosomes containing the histone variant H2A.Bbd organize only 118 base pairs of DNA. Embo J. 2004;23:3314–3324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [208].Sansoni V, Casas-Delucchi CS, Rajan M, et al. The histone variant H2A.Bbd is enriched at sites of DNA synthesis. Nucleic Acids Res. 2014;42:6405–6420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [209].Tolstorukov MY, Goldman JA, Gilbert C, et al. Histone variant H2A.Bbd is associated with active transcription and mRNA processing in human cells. Mol Cell. 2012;47:596–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [210].Govin J, Khochbin S. Histone variants and sensing of chromatin functional states. Nucleus. 2013;4:438–442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [211].Shinagawa T, Takagi T, Tsukamoto D, et al. Histone variants enriched in oocytes enhance reprogramming to induced pluripotent stem cells. Cell Stem Cell. 2014;14:217–227. [DOI] [PubMed] [Google Scholar]
- [212].Boulard M, Gautier T, Mbele GO, et al. The NH2 tail of the novel histone variant H2BFWT exhibits properties distinct from conventional H2B with respect to the assembly of mitotic chromosomes. Mol Cell Biol. 2006;26:1518–1526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [213].Eckey M, Hong W, Papaioannou M, et al. The nucleosome assembly activity of NAP1 is enhanced by alien. Mol Cell Biol. 2007;27:3557–3568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [214].Vlijm R, Lee M, Ordu O, et al Comparing the assembly and handedness dynamics of (H3.3-H4)2 tetrasomes to canonical tetrasomes. PLoS ONE. 2015;10(10): e0141267.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [215].Bowman A, Ward R, Wiechens N, et al. The histone chaperones Nap1 and Vps75 bind histones H3 and H4 in a tetrameric conformation. Mol Cell. 2011;41:398–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [216].English CM, Adkins MW, Carson JJ, et al. Structural basis for the histone chaperone activity of Asf1. Cell. 2006;127:495–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [217].English CM, Maluf NK, Tripet B, et al. ASF1 binds to a heterodimer of histones H3 and H4: a two-step mechanism for the assembly of the H3–H4 heterotetramer on DNA. Biochemistry. 2005;44:13673–13682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [218].Clément C, Orsi GA, Gatto A et al. High-resolution visualization of H3 variants during replication reveals their controlled recycling. Nat Commun. 2018;9:3181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [219].Cook AJL, Gurard-Levin ZA, Vassias I, et al. A specific function for the histone chaperone NASP to fine-tune a reservoir of soluble H3-H4 in the histone supply chain. Mol Cell. 2011;44:918–927. [DOI] [PubMed] [Google Scholar]
- [220].Nabeel-Shah S, Ashraf K, Pearlman RE, et al. Molecular evolution of NASP and conserved histone H3/H4 transport pathway. BMC Evol Biol. 2014;14:139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [221].Liu WH, Roemer SC, Zhou Y, et al. The Cac1 subunit of histone chaperone CAF-1 organizes CAF-1-H3/H4 architecture and tetramerizes histones. eLife;5:e18023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [222].Verreault A, Kaufman PD, Kobayashi R, et al. Nucleosome Assembly by a Complex of CAF-1 and Acetylated Histones H3/H4. Cell. 1996;87:95–104. [DOI] [PubMed] [Google Scholar]
- [223].Tagami H, Ray-Gallet D, Almouzni G, et al. Histone H3.1 and H3.3 complexes mediate nucleosome assembly pathways dependent or independent of DNA synthesis. Cell. 2004;116:51–61. [DOI] [PubMed] [Google Scholar]
- [224].Hake SB, Allis CD. Histone H3 variants and their potential role in indexing mammalian genomes: the ‘H3 barcode hypothesis’. Proc Natl Acad Sci U S A. 2006;103:6428–6435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [225].Horard B, Sapey-Triomphe L, Bonnefoy E, et al. ASF1 is required to load histones on the HIRA complex in preparation of paternal chromatin assembly at fertilization. Epigenetics Chromatin. 2018;11:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [226].Wong LH, McGhie JD, Sim M, et al. ATRX interacts with H3.3 in maintaining telomere structural integrity in pluripotent embryonic stem cells. Genome Res. 2010;20:351–360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [227].Hoelper D, Huang H, Jain AY, et al. Structural and mechanistic insights into ATRX-dependent and -independent functions of the histone chaperone DAXX. Nat Commun. 2017;8:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [228].Jang C-W, Shibata Y, Starmer J, et al. Histone H3.3 maintains genome integrity during mammalian development. Genes Dev. 2015;29:1377–1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [229].Elsässer SJ, Huang H, Lewis PW, et al. DAXX envelops a histone H3.3-H4 dimer for H3.3-specific recognition. Nature. 2012;491:560–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [230].Shi L, Wen H, Shi X. The histone variant H3.3 in transcriptional regulation and human disease. J Mol Biol. 2017;429:1934–1945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [231].Ray-Gallet D, Ricketts MD, Sato Y, et al. Functional activity of the H3.3 histone chaperone complex HIRA requires trimerization of the HIRA subunit. Nat Commun. 2018;9:1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [232].Xiong C, Wen Z, Yu J, et al. UBN1/2 of HIRA complex is responsible for recognition and deposition of H3.3 at cis-regulatory elements of genes in mouse ES cells. BMC Biol. 2018;16:110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [233].Ray-Gallet D, Quivy JP, Scamps C, et al. HIRA is critical for a nucleosome assembly pathway independent of DNA synthesis. Mol Cell. 2002;9:1091–1100. [DOI] [PubMed] [Google Scholar]
- [234].Zhang H, Gan H, Wang Z, et al. RPA Interacts with HIRA and regulates H3.3 deposition at gene regulatory elements in mammalian cells. Mol Cell. 2017;65:272–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [235].Siggens L, Cordeddu L, Rönnerblad M, et al. Transcription-coupled recruitment of human CHD1 and CHD2 influences chromatin accessibility and histone H3 and H3.3 occupancy at active chromatin regions. Epigenetics Chromatin.2015;8:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [236].Zhang K, Rajput SK, et al. CHD1 regulates deposition of histone variant H3.3 during bovine early embryonic development. Biol Reprod. 2016;94(6):1-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [237].Ivanauskiene K, Delbarre E, McGhie JD, et al. The PML-associated protein DEK regulates the balance of H3.3 loading on chromatin and is important for telomere integrity. Genome Res. 2014;24(10):1584–1594.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [238].Pradhan SK, Su T, Yen L, et al. EP400 Deposits H3.3 into Promoters and Enhancers during Gene Activation. Mol Cell. 2016;61(1):27–38.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [239].Shiraishi K, Shindo A, Harada A, et al. Roles of histone H3.5 in human spermatogenesis and spermatogenic disorders. Andrology. 2018;6:158–165. [DOI] [PubMed] [Google Scholar]
- [240].Urahama T, Harada A, Maehara K, et al. Histone H3.5 forms an unstable nucleosome and accumulates around transcription start sites in human testis. Epigenetics Chromatin. 2016;9(1):2.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [241].Kujirai T, Horikoshi N, Sato K, et al. Structure and function of human histone H3.Y nucleosome. Nucleic Acids Res. 2016;44(13):6127–6141.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [242].Wiedemann SM, Mildner SN, Bönisch C, et al. Identification and characterization of two novel primate-specific histone H3 variants, H3.X and H3.Y. J Cell Biol. 2010;190(5):777–791.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [243].Dunleavy EM, Roche D, Tagami H, et al. HJURP is a cell-cycle-dependent maintenance and deposition factor of CENP-A at centromeres. Cell. 2009;137(3):485–497.. [DOI] [PubMed] [Google Scholar]
- [244].Foltz DR, Jansen LET, Bailey AO, et al. Centromere-specific assembly of CENP-a nucleosomes is mediated by HJURP. Cell. 2009;137(3):472–484.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [245].Howman EV, Fowler KJ, Newson AJ, et al. Early disruption of centromeric chromatin organization in centromere protein A (Cenpa) null mice. Proc. Natl. Acad. Sci. U. S. A. 2000;97(3):1148–1153.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [246].Wisniewski JR, Zougman A, Krüger S, et al. Mass spectrometric mapping of linker histone H1 variants reveals multiple acetylations, methylations, and phosphorylation as well as differences between cell culture and tissue. Mol Cell Proteomics MCP. 2007;6:72–87. [DOI] [PubMed] [Google Scholar]
- [247].Izzo A, Schneider R. The role of linker histone H1 modifications in the regulation of gene expression and chromatin dynamics. Biochim Biophys Acta BBA - Gene Regul Mech. 2016;1859:486–495. [DOI] [PubMed] [Google Scholar]
- [248].Tessarz P, Kouzarides T. Histone core modifications regulating nucleosome structure and dynamics. Nat Rev Mol Cell Biol. 2014;15:703–708. [DOI] [PubMed] [Google Scholar]
- [249].Turinetto V, Giachino C. Multiple facets of histone variant H2AX: a DNA double-strand-break marker with several biological functions. Nucleic Acids Res. 2015;43:2489–2498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [250].Corujo D, Buschbeck M. Post-translational modifications of H2A histone variants and their role in cancer. Cancers (Basel). 2018;10(3):59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [251].Gamble MJ, Kraus WL. Multiple facets of the unique histone variant macroH2A: from genomics to cell biology. Cell Cycle. 2010;9:2568–2574. [DOI] [PubMed] [Google Scholar]
- [252].Chu F, Nusinow DA, Chalkley RJ, et al. Mapping post-translational modifications of the histone variant MacroH2A1 using tandem mass spectrometry. Mol Cell Proteomics MCP. 2006;5:194–203. [DOI] [PubMed] [Google Scholar]
- [253].Zhou J, Fan JY, Rangasamy D, et al. The nucleosome surface regulates chromatin compaction and couples it with transcriptional repression. Nat Struct Mol Biol. 2007;14:1070–1076. [DOI] [PubMed] [Google Scholar]
- [254].McKittrick E, Gafken PR, Ahmad K, et al. Histone H3.3 is enriched in covalent modifications associated with active chromatin. Proc Natl Acad Sci U S A. 2004;101:1525–1530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [255].Srivastava S, Foltz DR. Posttranslational modifications of CENP-A: marks of distinction. Chromosoma. 2018;127:279–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
