Hostname: page-component-848d4c4894-hfldf Total loading time: 0 Render date: 2024-06-01T15:02:15.672Z Has data issue: false hasContentIssue false

trans-Acting Factors and cis Elements Involved in the Human Inactive X Chromosome Organization and Compaction

Published online by Cambridge University Press:  01 January 2024

Zhuo Sun
Affiliation:
Institute of Basic Medical Sciences, Xi’an Medical University, No. 1 XinWang Rd, Weiyang District, Xi’an 710021, Shaanxi, China Xi’an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi’an Medical University, No. 1 XinWang Rd, Weiyang District, Xi’an 710021, Shaanxi, China
Jinbo Fan
Affiliation:
Xi’an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi’an Medical University, No. 1 XinWang Rd, Weiyang District, Xi’an 710021, Shaanxi, China
Yufeng Zhao*
Affiliation:
Institute of Basic Medical Sciences, Xi’an Medical University, No. 1 XinWang Rd, Weiyang District, Xi’an 710021, Shaanxi, China
*
Correspondence should be addressed to Yufeng Zhao; yufeng.zhao@xiyi.edu.cn

Abstract

During X chromosome inactivation, many chromatin changes occur on the future inactive X chromosome, including acquisition of a variety of repressive covalent histone modifications, heterochromatin protein associations, and DNA methylation of promoters. Here, we summarize trans-acting factors and cis elements that have been shown to be involved in the human inactive X chromosome organization and compaction.

Type
Review Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © 2021 Zhuo Sun et al.

1. Introduction

X chromosome inactivation (XCI) is the form of dosage compensation used by female cells to balance X-linked gene expression levels between the sexes in mammals [Reference Lyon1]. As a result, the inactive X (Xi) is compacted, taking on a rounder and slightly tighter configuration compared to the more flat and extended structure of the active X (Xa) [Reference Teller, Illner and Thamm2]. This compacted structure of the Xi is thought to limit the access of transcription machinery. The Xi is a classic example of developmentally induced heterochromatin, or facultative heterochromatin, which can be readily detected by DNA dyes in human cells [Reference Emil3] as a densely stained mass usually found at the periphery of the nucleus known as the Barr body [Reference Barr and Bertram4]. Heterochromatin is typically considered transcriptionally silent [Reference Brown5], and heterochromatic regions of the genome are thought of as “condensed” and therefore less accessible to transcriptional machinery.

The initiation of XCI is dependent on a region of the X chromosome known as the X inactivation center (XIC). Inside the XIC, the X-inactive specific transcript (XIST) gene encodes for the long noncoding RNA XIST, which is expressed from the future Xi, coating it in cis [Reference Brockdorff, Ashworth and Kay6]. Yin Yang 1 (YY1) is an important transcription activator for XIST [Reference Makhlouf, Ouimette and Oldfield7], also serving to tether XIST RNA to its own locus [Reference Jeon and Lee8].

XIST induces many epigenetic changes on the Xi, including depletion of euchromatic histone modifications such as histone acetylation [Reference Jeppesen and Turner9] and histone H3 dimethylation at lysine 4 (H3K4me2) [Reference Boggs, Cheung, Heard, Spector, Chinault and Allis10]. Other epigenetic changes are gained including the acquisition of the histone variant macroH2A [Reference Costanzi and Pehrson11], and the deposition of repressive histone modifications including trimethylation of histone H3 at lysine 9 (H3K9me3) [Reference Boggs, Cheung, Heard, Spector, Chinault and Allis10] and 27 (H3K27me3) [Reference Plath12]. It is known that H3K27me3 at the Xi is mediated via enhancer of zeste 2 (EZH2) [Reference Plath12], a part of the polycomb repressive complex 2 (PRC2) [Reference Cao, Wang and Wang13], but it is not known which histone lysine methyltransferase (HMTase) is responsible for Xi H3K9me3 marks. Furthermore, there is DNA CpG island methylation [Reference Pfeifer, Tanguay, Steigerwald and Riggs14] and recruitment of heterochromatin proteins such as heterochromatin protein 1(HP1) [Reference Chadwick and Willard15], structural maintenance of chromosomes flexible hinge domain-containing protein 1 (SMCHD1) [Reference Blewitt, Gendrel and Pang16] and ligand-dependent nuclear receptor-interacting factor 1 (LRIF1), also known as HP1-binding protein (HBiX1) [Reference Nozawa, Nagao and Igami17].

In addition to chromatin changes, there is also a delay of Xi DNA replication during the S-phase, such that it replicates asynchronously relative to the Xa [Reference Gilbert, Muldal, Lajtha and Rowley18], with the DNA underlying the bands of H3K27me3 replicating during the midlate S-phase and the bands of H3K9me3 replicating after H3K27me3 replication is complete [Reference Chadwick and Willard19]. Collectively, these changes are likely responsible for shutting down most gene expressions originating from the Xi [Reference Carrel and Willard20].

After the Xi is established, XCI enters the maintenance stage. It is difficult to reverse the effect of gene repression once this stage has been attained, evidenced by the high degree of effort required to reactivate genes on a large scale in somatic cells [Reference Mohandas, Sparkes and Shapiro21]. Several mechanisms are in place to work synergistically to ensure the repressive state of the Xi. These include continued XIST RNA expression, DNA methylation, histone hypoacetylation [Reference Csankovszki, Nagy and Jaenisch22], and the acquisition of macroH2A [Reference Hernandez-Munoz, Lund and van der Stoop23]. DNA methylation is a robust way to keep genes in a repressive state, and the reactivation of genes has been linked with DNA hypomethylation in promoter regions [Reference Cotton, Price, Jones, Balaton, Kobor and Brown24]. Additionally, the attainment of DNA methylation may be linked to histone modifications such as H3K9me3, as suggested by a recent study that elucidates the Mbd1-Atf7ip-Setdb1 pathway in the maintenance of XCI [Reference Minkovsky, Sahakyan, Rankin-Gee, Bonora, Patel and Plath25]. Atf7ip acts synergistically with methyl-DNA binding protein Mbd1 and H3K9 methyltransferase Setdb1, which links DNA methylation with H3K9me3, and this pathway is essential in maintaining the silent state of Xi in somatic cells.

2. Proteins Involved in XCI

2.1. MacroH2A

MacroH2A is a variant of H2A that has an extensive C terminal tail that makes up two-thirds of the protein [Reference Pehrson and Fried26]. MacroH2A is enriched on the Xi and by immunofluorescence can be seen as an intensely staining structure, called a macrochromatin body, which also coincides with the Barr body in human cells [Reference Costanzi and Pehrson11]. On a human retinal pigment epithelial cell (RPE1) metaphase Xi chromosome, macroH2A deposition overlaps with H3K27me3 marks, forming an alternating banding pattern with H3K9me3 territories. The accumulation of macroH2A on the Xi occurs in the late stages of Xi establishment.

2.2. PRC1 and PRC2

Polycomb complexes PRC1 and PRC2 carry out histone modifications that are integral to the process of XCI. PRC2 complex catalyzes H3K27me3 [Reference Zhao, Sun, Erwin, Song and Lee27] and PRC1 complex catalyzes ubiquitination of histone H2A at lysine 119 (H2AK119u1) [Reference Cao, Wang and Wang13]. PRC2 complex’ recruitment to XIST RNA on the Xi is mediated through the SWI/SNF family helicase/ATPase alpha-thalassemia/mental retardation X-linked (ATRX) [Reference Sarma, Cifuentes-Rojas and Ergun28]. It has been shown that there are around 150 strong binding sites for PRC2 along the Xi, which are mostly within bivalent domains, serving as seeding sites for propagation of PRC2 binding [Reference Pinter, Sadreyev and Yildirim29]. Bivalent domains are regions of the chromosome exhibiting both H3K27me3-repressive and H3K4me3-active histone marks and are thought to be the signature of developmentally poised genes. PRC2 recruitment to nearby loci mostly within nonbivalent domains is then observed around these seeding sites, laying down H3K27me3 marks in a concentration gradient [Reference Pinter, Sadreyev and Yildirim29].

There are two pathways by which the PRC1 complex can be recruited to the Xi, either dependent on PRC2 or independent of the presence of PRC2 [Reference Cao, Wang and Wang13, Reference Schoeftner, Sengupta and Kubicek30Reference Almeida, Pintacuda and Masui32]. Early evidence has shown that the PRC2 complex is directly recruited to XIST RNA and lays down H3K27me3 [Reference Zhao, Sun, Erwin, Song and Lee27], which could then be recognized by PRC1 (referred to as canonical PRC1) to lay down H2AK119u1 [Reference Cao, Wang and Wang13]. More recently, another class of PRC1 complexes called noncanonical PRC1 has been found, whose recruitment to the Xi is independent of the H3K27me3 mark [Reference Schoeftner, Sengupta and Kubicek30]. Examples of noncanonical PRC1 complexes include RING1-YY1-binding protein (RYBP-PRC1) [Reference Tavares, Dimitrova and Oxley31] and polycomb group RING finger 3/5-PRC1 (PCGF3/5-PRC1) which can recruit other noncanonical PRC1 complexes and PRC2 complex to establish H3K27me3 modification chromosome-wide on the Xi [Reference Almeida, Pintacuda and Masui32].

The recruitment of polycomb complexes occurs in the early stages of Xi establishment. Polycomb complexes’ enrichment on the Xi is readily detectable when Xist is induced and the enrichment is lost when Xist expression is inhibited [Reference Mak33, Reference Kohlmaier, Savarese, Lachner, Martens, Jenuwein and Wutz34].

2.3. HP1

HP1 is another protein thought to be important for the establishment and maintenance of the Xi heterochromatin. In humans, there are three isoforms of HP1: HP1-alpha, HP1-beta, and HP1-gamma, all of which contain 3 domains: a chromodomain that can recognize and be recruited to the H3K9me3 on the Xi, a hinge domain, and a chromo shadow domain that is important for dimerization and enrichment of HP1 proteins [Reference Lachner, O'Carroll, Rea, Mechtler and Jenuwein35Reference Nishibuchi, Machida and Osakabe37]. It has been shown that all three isoforms of HP1 can be detected at the human interphase Xi [Reference Chadwick and Willard15]. HP1 can recognize the H3K9me3 modifications and help to maintain the heterochromatin structure and gene silencing on the human Xi and it is generally considered a maintenance factor for the Xi.

2.4. hnRNP U/SAF-A and SHARP/SPEN

Although the exact mechanism of how Xist mediates the chromosome-wide inactivation and gene silencing is still largely unknown, several factors have been identified to interact with Xist RNA and are essential for Xist-mediated gene silencing. These factors include hnRNP U/SAF-A [Reference Hasegawa, Brockdorff, Kawano, Tsutui, Tsutui and Nakagawa38], SHARP/SPEN [Reference Chu, Zhang and da Rocha39Reference McHugh, Chen and Chow42], and hnRNP K [Reference Chu, Zhang and da Rocha39].

The nuclear matrix binding protein hnRNP U/SAF-A is essential for the anchoring of Xist on the Xi [Reference Hasegawa, Brockdorff, Kawano, Tsutui, Tsutui and Nakagawa38]. hnRNP (heterogeneous nuclear ribonucleoproteins) are a family of RNA-binding proteins that have important functions in gene transcription regulation. hnRNP U/SAF-A is widely distributed in the nucleus but is concentrated on the Xi [Reference Helbig and Fackelmayer43, Reference Pullirsch, Härtel, Kishimoto, Leeb, Steiner and Wutz44]. hnRNP U/SAF-A consists of three domains: the DNA-binding SAF domain, SPRY domain, and RNA-binding RGG domain. The SAF and RGG domains are important for the recruitment and localization of Xist [Reference Hasegawa, Brockdorff, Kawano, Tsutui, Tsutui and Nakagawa38]. Loss of hnRNP U/SAF-A results in delocalization of Xist RNA from the X chromosome, and hnRNP U/SAF-A is essential for establishing the Xi during ES cell differentiation [Reference Hasegawa, Brockdorff, Kawano, Tsutui, Tsutui and Nakagawa38]. It is also shown that hnRNP U/SAF-A is required for Xist-mediated gene silencing [Reference McHugh, Chen and Chow42].

RNA-binding protein SHARP/SPEN is important for Xist-mediated gene silencing [Reference Chu, Zhang and da Rocha39Reference McHugh, Chen and Chow42], which has recently been verified in preimplantation embryos [Reference Dossin, Pinheiro and Żylicz45]. SPEN is recruited to enhancers and promoters of active genes on the X chromosome once Xist is upregulated and quickly dissociates once the gene silencing is established [Reference Dossin, Pinheiro and Żylicz45]. Xist recruits HDAC3 through interaction with SHARP/SPEN, and HDAC3 removes acetylation modification from histones [Reference Chu, Zhang and da Rocha39, Reference Moindrot, Cerase and Coker40, Reference McHugh, Chen and Chow42]. SHARP/SPEN expels Pol II [Reference Dossin, Pinheiro and Żylicz45, Reference Nesterova, Wei and Coker46] and serves as a bridge between Xist and transcription machinery/histone modifiers.

2.5. SMCHD1 and LRIF1

SMCHD1 was first described in an N-ethyl-N-nitrosourea (ENU) mutagenesis screen to identify genes involved in epigenetic regulation and gene silencing [Reference Blewitt, Vickaryous and Hemley47]. SMCHD1 belongs to the structural maintenance of chromosome (SMC) domain family of proteins, which also include cohesin and condensin [Reference Haering and Gruber48], but is different in that its SMC hinge domain is not in the middle of the protein but at the C-terminus. On the Xi, SMCHD1 has been shown to be important for the maintenance phase of silencing [Reference Blewitt, Gendrel and Pang16], heterochromatin compaction [Reference Nozawa, Nagao and Igami17], and the methylation of CpG islands [Reference Gendrel, Apedaile and Coker49].

SMCHD1 is crucial for both random XCI in the embryo and imprinted XCI in the placenta [Reference Blewitt, Gendrel and Pang16]. The loss of SMCHD1 does not interfere with the accumulation of Xist or H3K27me3 modification on the Xi, which suggests that SMCHD1 is not involved in the initiation phase of XCI, but the maintenance phase. This is also implied by the fact that, in interphase nuclei, SMCHD1 is enriched on the Xi [Reference Blewitt, Gendrel and Pang16]. In the absence of SMCHD1, decompaction of the Xi territory is observed [Reference Nozawa, Nagao and Igami17]. It is suggested that SMCHD1 is enriched on the H3K27me3 territory, whereas LRIF1/HBiX1 is enriched on the H3K9me3 territory through the interaction with HP1, and the interaction between SMCHD1 and LRIF1/HBiX1 brings together the H3K27me3 territory and H3K9me3 territory to form a compacted structure [Reference Nozawa, Nagao and Igami17]. Notably, PRC2 and H3K27me3 are dispensable for this compaction process, whereas XIST is required for the correct localization of SMCHD1 and LRIF1/HBiX1 to the Xi [Reference Gendrel, Apedaile and Coker49, Reference Gendrel, Tang and Suzuki50].

HBiX1/LRIF1 was first identified as a novel nuclear matrix transcription repressor known as ligand-dependent nuclear receptor-interacting factor 1(LRIF1, RIF1, or C1orf103) [Reference Li, Haque, Chen and Mendelsohn51]. It is an HP1-interacting protein that is enriched on the Xi in interphase nuclei. It has been shown to be essential for the compaction of the Xi chromatin [Reference Nozawa, Nagao and Igami17, Reference Brideau, Coker and Gendrel52]. HBiX1/LRIF1 interacts with SMCHD1 through its coiled-coil domain with the hinge domain on SMCHD1 [Reference Nozawa, Nagao and Igami17, Reference Brideau, Coker and Gendrel52].

2.6. SETDB1

SET domain bifurcated 1 (SETDB1), an H3-K9 histone methyltransferase with the highest activity for lysine 9 trimethylation [Reference Schultz53, Reference Yang, Xia and Wu54], has been shown to be important for establishing H3K9me3 [Reference Keniry, Gearing and Jansz55] and maintaining gene silencing on the mouse Xi [Reference Minkovsky, Sahakyan, Rankin-Gee, Bonora, Patel and Plath25]. Recently, it is shown that loss of SETDB1 does not lead to large-scale H3K9me3 changes on the Xi but results in decompaction of the human Xi territory [Reference Sun and Chadwick56]. Although the pathway of SETDB1 recruitment to KRAB-zinc finger proteins through TRIM28 [Reference Friedman, Fredericks and Jensen57] to silence gene expression is well characterized for some autosomal regions [Reference Peng, Ivanov, Oh, Lau and Rauscher58], it is not known how SETDB1 is recruited to the Xi.

To summarize the process of XCI in regard to the proteins involved and their order of recruitment, hnRNP U/SAF-A is essential for the localization of Xist to the future Xi; Xist then spreads and coats the whole chromosome. Xist recruits polycomb complexes PRC2 and PRC1 and lays down repressive histone modifications. SHARP/SPEN expels RNA polymerase and inhibits transcription. MacroH2A is then recruited and CpG islands are methylated, and at this point, the establishment stage of XCI is concluded. Heterochromatin proteins such as HP1, SMCHD1, LRIF1, and SETDB1 exert their function in the maintenance stage of XCI and work together to maintain the heterochromatin structure and gene silencing on the Xi.

3. lncRNAs Involved in X Chromosome Inactivation

The XIC is the minimal region on the X chromosome that is both necessary and sufficient to initiate XCI [Reference Russell and Montgomery59, Reference Rastan60]. The XIC contains some protein-coding genes and some noncoding genes. Essential to the XIC is the region encoding for the XIST lncRNA and TSIX lncRNA, which is the antisense transcript of XIST that can mediate the repression of XIST [Reference Lee and Lu61].

XIST was first discovered to be important for the XCI in mice and humans in 1991 [Reference Borsani, Tonlorenzi and Simmler62Reference Brown, Ballabio and Rupert64]. XIST lncRNA is 17 kb in length; within XIST are several tandem repeat regions that are conserved. The most highly conserved A repeats were found to be essential for gene silencing during XCI [Reference Wutz, Rasmussen and Jaenisch65]. The upregulation of XIST expression is the first step of inactivation along the future Xi. The localization of XIST to the future Xi is dependent on the transcription factor YY1 and starts with a nucleation center within exon 1 of the XIST locus [Reference Jeon and Lee8]. It has been shown that the spreading of XIST follows a stepwise mechanism. It targets gene-rich regions before spreading to intervening gene-poor regions [Reference Simon, Pinter and Fang66]. XIST then recruits polycomb proteins PRC1 and PRC2 to set up repressive histone marks for the maintenance of the inactive chromatin structure [Reference Zhao, Sun, Erwin, Song and Lee27].

While the expression of XIST is unique to the Xi, the lncRNA TSIX dictates which X chromosome will be the Xa. TSIX is the antisense transcript of XIST and has an antagonistic role in XIST expression. TSIX controls XIST expression by modifying the chromatin state and DNA methylation of the XIST promoter [Reference Sado, Hoki and Sasaki67, Reference Sun, Deaton and Lee68]. Before the choice of which X chromosome is going to be the future Xa or Xi, chromosome pairing between the two X chromosomes takes place. The pairing might be important for the redistribution of transcription activators to only allow the transient expression of TSIX on the future Xa [Reference Xu, Tsai and Lee69Reference Scialdone and Nicodemi72].

A few other lncRNAs have been discovered to be important activators for XIST expression: RepA [Reference Zhao, Sun, Erwin, Song and Lee27], Jpx [Reference Tian, Sun and Lee73, Reference Sun, Del Rosario and Szanto74], and Ftx [Reference Chureau, Chantalat and Romito75].

4. cis Elements Involved in Xi Organization

At interphase in human RPE1 cells, the Xi forms a bipartite structure, with all H3K9me3 bands aggregating together towards the nuclear periphery and the H3K27me3 bands more towards the interior of the nucleus [Reference Chadwick and Willard19]. How and why the Xi arranges itself into these two compartments is not clear. Possible factors that might contribute to this arrangement include the various chromatin proteins that bind to H3K27me3 and H3K9me3 marks, respectively, and self-aggregate. An alternative or additional factor that may contribute to this arrangement is potential DNA folding elements. Several large tandem repeat DNA sequences (TRs) have been identified that are unique to the X chromosome, including the macrosatellite DXZ4, and the TRs X56 and X130. These TRs adopt a Xi-specific euchromatic configuration that is bound by the epigenetic organizer protein CCCTC-binding factor (CTCF) [Reference Chadwick76]. DXZ4, X56, and X130 have been shown to interact with each other over millions of bases, exclusively from the Xi alleles, potentially acting as epigenetically regulated DNA folding elements [Reference Horakova, Moseley, McLaughlin, Tremblay and Chadwick77, Reference Darrow, Huntley and Dudchenko78] forming massive chromosome loops restricted to the Xi [Reference Rao, Huntley and Durand79]. Each TR is located at the intersection between the H3K9me3 and H3K27me3 bands and could contribute to the compartmentalization of the bipartite structure [Reference Chadwick and Willard19].

5. Impact of X Chromosome Inactivation on Disease

With the exception of the pseudoautosomal regions located at the tips of Xp and Xq, most genes on the X chromosome have been lost on the Y chromosome. As such, males are hemizygous for most X-linked genes. As a consequence, inheritance of an X-linked recessive mutant allele in males will act as dominant and disease onset will be unavoidable. Females are afforded some protection from X-linked recessive disorders due to XCI.

Because XCI occurs at a multicell stage, and the choice of which X to silence is random and that decision is made independently in each cell, females are a mosaic, where some cells express the mutant allele, and others express the wild-type allele. If the mutant allele impacts cell growth or survival, these cells will be outgrown giving the appearance of skewing toward the wild-type active X. Skewing of XCI occurs naturally, which can affect disease severity. Fabry disease is one example where there is a mutation in the X-linked lysosomal alpha-galactosidase. The peak of the normal distribution represents equal numbers of cells with either the paternal or maternal X chromosome chosen as the Xi. At either tail end of the normal distribution is the phenomenon called skewed XCI, where at one tail end, it might be asymptomatic, but for the other end of the distribution, there is a severe representation of disease. The direction and degree of skewed XCI influence the phenotype and progression of Fabry disease in female patients [Reference Echevarria, Benistan and Toussaint80].

XCI serves as a mechanism for balancing the difference in expression between different sexes in mammalian cells, but there is a certain level of escape from gene repression in both normal and disease cells. A recent systematic survey integrating transcriptomes from 449 individuals across 29 tissues has shown that, besides 683 X-linked genes that are consistently inactivated, there is heterogeneity in expression patterns among different individuals and different tissues [Reference Tukiainen, Villani and Yen81]. X-linked gene reactivation has also been observed in aged tissue [Reference Wareham, Lyon, Glenister and Williams82Reference Sharp, Robinson and Jacobs84], autoimmune diseases [Reference Wang, Syrett, Kramer, Basu, Atchison and Anguera85], and cancer [Reference Pageau, Hall, Ganesan, Livingston and Lawrence86Reference Kang, Lee and Kim88]. There is evidence that, in ovarian cancer cells, there is a unique profile of X-linked gene expression and escape, suggesting that XCI may play a role in the development of ovarian cancer [Reference Winham, Larson and Armasu89].

6. Concluding Remarks

X chromosome inactivation is the classic model system to study epigenetic questions. What we learn under the X chromosome inactivation context could also be useful to unravel unsolved problems in autosomes and developmental contexts as well. Research and more mechanistic insight for X chromosome inactivation could assist in developing strategies and therapy for X-linked diseases.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Acknowledgments

This work was supported by the Ph. D starting fund from Xi’an Medical University (Program nos. 2020DOC14 and 2020DOC17) and by the Natural Science Basic Research Plan in Shaanxi Province of China (Program nos. 2021JQ-776 and 2021JQ-774).

References

Lyon, M. F., “Gene action in the X-chromosome of the mouse (mus musculus L.),Nature, vol. 190, no. 4773, pp. 372373, 1961.CrossRefGoogle ScholarPubMed
Teller, K., Illner, D., Thamm, S. et al., A top-down analysis of Xa- and Xi-territories reveals differences of higher order structure at ≥ 20 Mb genomic length scales,Nucleus, vol. 2, no. 5, pp. 465477, 2011.CrossRefGoogle ScholarPubMed
Emil, H., “Das heterochromatin der Moose,Jahrbücher für Wissenschaftliche Botanik, vol. 69, pp. 762818, 1928.Google Scholar
Barr, M. L., Bertram, E. G., “A morphological distinction between neurones of the male and female, and the behaviour of the nucleolar satellite during accelerated nucleoprotein synthesis,Nature, vol. 163, no. 4148, pp. 676677, 1949.CrossRefGoogle ScholarPubMed
Brown, S. W., “Heterochromatin,Science, vol. 151, no. 3709, pp. 417425, 1966.CrossRefGoogle ScholarPubMed
Brockdorff, N., Ashworth, A., Kay, G. F. et al., The product of the mouse XIST gene is a 15 kb inactive X-specific transcript containing no conserved ORF and located in the nucleus,Cell, vol. 71, no. 3, pp. 515526, 1992.CrossRefGoogle ScholarPubMed
Makhlouf, M., Ouimette, J.-F., Oldfield, A. et al., A prominent and conserved role for YY1 in XIST transcriptional activation,Nature Communications, vol. 5, 2014.CrossRefGoogle ScholarPubMed
Jeon, Y., Lee, J. T., “YY1 tethers XIST RNA to the inactive X nucleation center,Cell, vol. 146, no. 1, pp. 119133, 2011.CrossRefGoogle Scholar
Jeppesen, P. and Turner, B. M., “The inactive X chromosome in female mammals is distinguished by a lack of histone H4 acetylation, a cytogenetic marker for gene expression,Cell, vol. 74, no. 2, pp. 281289, 1993.CrossRefGoogle ScholarPubMed
Boggs, B. A., Cheung, P., Heard, E., Spector, D. L., Chinault, A. C., and Allis, C. D., “Differentially methylated forms of histone H3 show unique association patterns with inactive human X chromosomes,Nature Genetics, vol. 30, no. 1, pp. 7376, 2002.CrossRefGoogle ScholarPubMed
Costanzi, C. and Pehrson, J. R., “Histone macroH2A1 is concentrated in the inactive X chromosome of female mammals,Nature, vol. 628, pp. 19971999, 1998.Google Scholar
Plath, K., “Role of histone H3 lysine 27 methylation in X inactivation,Science, vol. 300, no. 5616, pp. 131135, 2003.CrossRefGoogle ScholarPubMed
Cao, R., Wang, L., Wang, H. et al., Role of histone H3 lysine 27 methylation in polycomb-group silencing,Science, vol. 298, no. 5595, pp. 10391043, 2002.CrossRefGoogle ScholarPubMed
Pfeifer, G. P., Tanguay, R. L., Steigerwald, S. D., and Riggs, A. D., “In vivo footprint and methylation analysis by PCR-aided genomic sequencing: comparison of active and inactive X chromosomal DNA at the CpG island and promoter of human PGK-1,Genes & Development, vol. 4, no. 8, pp. 12771287, 1990.CrossRefGoogle ScholarPubMed
Chadwick, B. P. and Willard, H. F., “Chromatin of the barr body: histone and non-histone proteins associated with or excluded from the inactive X chromosome,Human Molecular Genetics, vol. 12, no. 17, pp. 21672178, 2003.CrossRefGoogle ScholarPubMed
Blewitt, M. E., Gendrel, A.-V., Pang, Z. et al., SmcHD1, containing a structural-maintenance-of-chromosomes hinge domain, has a critical role in X inactivation,Nature Genetics, vol. 40, no. 5, pp. 663669, 2008.CrossRefGoogle Scholar
Nozawa, R.-S., Nagao, K., Igami, K.-T. et al., Human inactive X chromosome is compacted through a PRC2-independent SMCHD1-HBiX1 pathway,Nature Structural & Molecular Biology, vol. 20, no. 5, pp. 566573, 2013.CrossRefGoogle ScholarPubMed
Gilbert, C. W., Muldal, S., Lajtha, L. G., and Rowley, J., “Time-sequence of human chromosome duplication,Nature, vol. 195, no. 4844, pp. 869873, 1962.CrossRefGoogle ScholarPubMed
Chadwick, B. P. and Willard, H. F., “Multiple spatially distinct types of facultative heterochromatin on the human inactive X chromosome,Proceedings of the National Academy of Sciences, vol. 101, no. 50, pp. 1745017455, 2004.CrossRefGoogle ScholarPubMed
Carrel, L. and Willard, H. F., “X-inactivation profile reveals extensive variability in X-linked gene expression in females,Nature, vol. 434, no. 7031, pp. 400404, 2005.CrossRefGoogle ScholarPubMed
Mohandas, T., Sparkes, R., and Shapiro, L., “Reactivation of an inactive human X chromosome: evidence for X inactivation by DNA methylation,Science, vol. 211, no. 4480, pp. 393396, 1981.CrossRefGoogle ScholarPubMed
Csankovszki, G., Nagy, A., and Jaenisch, R., “Synergism of XIST RNA, DNA methylation, and histone hypoacetylation in maintaining X chromosome inactivation,Journal of Cell Biology, vol. 153, no. 4, pp. 773784, 2001.CrossRefGoogle ScholarPubMed
Hernandez-Munoz, I., Lund, A. H., van der Stoop, P. et al., Stable X chromosome inactivation involves the PRC1 polycomb complex and requires histone MACROH2A1 and the CULLIN3/SPOP ubiquitin E3 ligase,Proceedings of the National Academy of Sciences, vol. 102, no. 21, pp. 76357640, 2005.CrossRefGoogle ScholarPubMed
Cotton, A. M., Price, E. M., Jones, M. J., Balaton, B. P., Kobor, M. S., and Brown, C. J., “Landscape of DNA methylation on the X chromosome reflects CpG density, functional chromatin state and X-chromosome inactivation,Human Molecular Genetics, vol. 24, no. 6, pp. 15281539, 2015.CrossRefGoogle ScholarPubMed
Minkovsky, A., Sahakyan, A., Rankin-Gee, E., Bonora, G., Patel, S., and Plath, K., “The Mbd1-Atf7ip-Setdb1 pathway contributes to the maintenance of X chromosome inactivation,Epigenetics & Chromatin, vol. 7, no. 1, p. 12, 2014.CrossRefGoogle Scholar
Pehrson, J. R. and Fried, V., “MacroH2A, a core histone containing a large nonhistone region,Science, vol. 257, no. 5075, pp. 13981400, 1992.CrossRefGoogle ScholarPubMed
Zhao, J., Sun, B. K., Erwin, J. A., Song, J.-J., and Lee, J. T., “Polycomb proteins targeted by a short repeat RNA to the mouse X chromosome,Science, vol. 322, no. 5902, pp. 750756, 2008.CrossRefGoogle Scholar
Sarma, K., Cifuentes-Rojas, C., Ergun, A. et al., ATRX directs binding of PRC2 to XIST RNA and polycomb targets,Cell, vol. 159, no. 4, pp. 869883, 2014.CrossRefGoogle ScholarPubMed
Pinter, S. F., Sadreyev, R. I., Yildirim, E. et al., Spreading of X chromosome inactivation via a hierarchy of defined Polycomb stations,Genome Research, vol. 22, no. 10, pp. 18641876, 2012.CrossRefGoogle Scholar
Schoeftner, S., Sengupta, A. K., Kubicek, S. et al., Recruitment of PRC1 function at the initiation of X inactivation independent of PRC2 and silencing,The EMBO Journal, vol. 25, no. 13, pp. 31103122, 2006.CrossRefGoogle ScholarPubMed
Tavares, L., Dimitrova, E., Oxley, D. et al., RYBP-PRC1 complexes mediate H2A ubiquitylation at polycomb target sites independently of PRC2 and H3K27me3,Cell, vol. 148, no. 4, pp. 664678, 2012.CrossRefGoogle ScholarPubMed
Almeida, M., Pintacuda, G., Masui, O. et al., PCGF3/5-PRC1 initiates polycomb recruitment in X chromosome inactivation,Science, vol. 356, no. 6342, pp. 10811084, 2017.CrossRefGoogle ScholarPubMed
Mak, W., “Reactivation of the paternal X chromosome in early mouse embryos,Science, vol. 303, no. 5658, pp. 666669, 2004.CrossRefGoogle ScholarPubMed
Kohlmaier, A., Savarese, F., Lachner, M., Martens, J., Jenuwein, T., and Wutz, A., “A chromosomal memory triggered by XIST regulates histone methylation in X inactivation,PLoS Biology, vol. 2, no. 7, p. E171, 2004.CrossRefGoogle ScholarPubMed
Lachner, M., O'Carroll, D., Rea, S., Mechtler, K., and Jenuwein, T., “Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins,Nature, vol. 410, no. 6824, pp. 116120, 2001.CrossRefGoogle ScholarPubMed
Lomberk, G., Bensi, D., Fernandez-Zapico, M. E., and Urrutia, R., “Evidence for the existence of an HP1-mediated subcode within the histone code,Nature Cell Biology, vol. 8, no. 4, pp. 407415, 2006.CrossRefGoogle ScholarPubMed
Nishibuchi, G., Machida, S., Osakabe, A. et al., N-terminal phosphorylation of HP1α increases its nucleosome-binding specificity,Nucleic Acids Research, vol. 42, no. 20, pp. 1249812511, 2014.CrossRefGoogle ScholarPubMed
Hasegawa, Y., Brockdorff, N., Kawano, S., Tsutui, K., Tsutui, K., and Nakagawa, S., “The matrix protein hnRNP U is required for chromosomal localization of XIST RNA,Developmental Cell, vol. 19, no. 3, pp. 469476, 2010.CrossRefGoogle ScholarPubMed
Chu, C., Zhang, Q. C., da Rocha, S. T. et al., Systematic discovery of XIST RNA binding proteins,Cell, vol. 161, no. 2, pp. 404416, 2015.CrossRefGoogle ScholarPubMed
Moindrot, B., Cerase, A., Coker, H. et al., A pooled shRNA screen identifies Rbm15, spen, and wtap as factors required for XIST RNA-mediated silencing,Cell Reports, vol. 12, no. 4, pp. 562572, 2015.CrossRefGoogle ScholarPubMed
Monfort, A., Di Minin, G., Postlmayr, A. et al., Identification of spen as a crucial factor for XIST function through forward genetic screening in haploid embryonic stem cells,Cell Reports, vol. 12, no. 4, pp. 554561, 2015.CrossRefGoogle ScholarPubMed
McHugh, C. A., Chen, C.-K., Chow, A. et al., The XIST lncRNA interacts directly with SHARP to silence transcription through HDAC3,Nature, vol. 521, no. 7551, pp. 232236, 2015.CrossRefGoogle ScholarPubMed
Helbig, R. and Fackelmayer, F. O., “Scaffold attachment factor A (SAF-A) is concentrated in inactive X-chromosome territories through its RGG domain,Chromosoma, vol. 112, no. 4, pp. 173182, 2003.CrossRefGoogle ScholarPubMed
Pullirsch, D., Härtel, R., Kishimoto, H., Leeb, M., Steiner, G., and Wutz, A., “The trithorax group protein Ash2l and Saf-A are recruited to the inactive X chromosome at the onset of stable X inactivation,Development, vol. 137, no. 6, pp. 935943, 2010.CrossRefGoogle Scholar
Dossin, F., Pinheiro, I., Żylicz, J. J. et al., SPEN integrates transcriptional and epigenetic control of X-inactivation,Nature, vol. 578, no. 7795, pp. 455460, 2020.CrossRefGoogle ScholarPubMed
Nesterova, T. B., Wei, G., Coker, H. et al., Systematic allelic analysis defines the interplay of key pathways in X chromosome inactivation,Nature Communications, vol. 10, no. 1, p. 3129, 2019.CrossRefGoogle ScholarPubMed
Blewitt, M. E., Vickaryous, N. K., Hemley, S. J. et al., An N-ethyl-N-nitrosourea screen for genes involved in variegation in the mouse,Proceedings of the National Academy of Sciences, vol. 102, no. 21, pp. 76297634, 2005.CrossRefGoogle ScholarPubMed
Haering, C. H. and Gruber, S., “SnapShot: SMC protein complexes part I,Cell 2016 164 1-2326CrossRefGoogle ScholarPubMed
Gendrel, A.-V., Apedaile, A., Coker, H. et al., Smchd1-dependent and -independent pathways determine developmental dynamics of CpG island methylation on the inactive X chromosome,Developmental Cell, vol. 23, no. 2, pp. 265279, 2012.CrossRefGoogle ScholarPubMed
Gendrel, A.-V., Tang, Y. A., Suzuki, M. et al., Epigenetic functions of Smchd1 repress gene clusters on the inactive X chromosome and on autosomes,Molecular and Cellular Biology, vol. 33, no. 16, pp. 31503165, 2013.CrossRefGoogle ScholarPubMed
Li, H. J., Haque, Z. K., Chen, A., and Mendelsohn, M., “RIF-1, a novel nuclear receptor corepressor that associates with the nuclear matrix,Journal of Cellular Biochemistry, vol. 102, no. 4, pp. 10211035, 2007.CrossRefGoogle ScholarPubMed
Brideau, N. J., Coker, H., Gendrel, A.-V. et al., Independent mechanisms target SMCHD1 to trimethylated histone H3 lysine 9-modified chromatin and the inactive X chromosome,Molecular and Cellular Biology, vol. 35, no. 23, pp. 40534068, 2015.CrossRefGoogle ScholarPubMed
Schultz, D. C., “SETDB1: a novel KAP-1-associated histone H3, lysine 9-specific methyltransferase that contributes to HP1-mediated silencing of euchromatic genes by KRAB zinc-finger proteins,Genes & Development, vol. 16, no. 8, pp. 919932, 2002.CrossRefGoogle ScholarPubMed
Yang, L., Xia, L., Wu, D. Y. et al., Molecular cloning of ESET, a novel histone h3-specific methyltransferase that interacts with erg transcription factor,Oncogene, vol. 21, no. 1, pp. 148152, 2002.CrossRefGoogle Scholar
Keniry, A., Gearing, L. J., Jansz, N. et al., Setdb1-mediated H3K9 methylation is enriched on the inactive X and plays a role in its epigenetic silencing,Epigenetics & Chromatin vol. 9, p. 16, 2016.CrossRefGoogle Scholar
Sun, Z. and Chadwick, B. P., “Loss of SETDB1 decompacts the inactive X chromosome in part through reactivation of an enhancer in the IL1RAPL1 gene,Epigenetics and Chromatin, vol. 11, no. 1, 2018.CrossRefGoogle ScholarPubMed
Friedman, J. R., Fredericks, W. J., Jensen, D. E. et al., KAP-1, a novel corepressor for the highly conserved KRAB repression domain,Genes & Development, vol. 10, no. 16, pp. 20672078, 1996.CrossRefGoogle ScholarPubMed
Peng, H., Ivanov, A. V., Oh, H. J., Lau, Y.-F. C., and Rauscher, F. J., “Epigenetic gene silencing by the SRY protein is mediated by a KRAB-O protein that recruits the KAP1 co-repressor machinery,Journal of Biological Chemistry, vol. 284, no. 51, pp. 3567035680, 2009.CrossRefGoogle ScholarPubMed
Russell, L. B. and Montgomery, C. S., “Comparative studies on X-autosome translocations in the mouse. Ii. Inactivation of autosomal loci, segregation, and mapping of autosomal breakpoints in five T(x;1)'S,Genetics, vol. 64, no. 2, pp. 281312, 1970.CrossRefGoogle ScholarPubMed
Rastan, S., “Non-random X-chromosome inactivation in mouse X-autosome translocation embryos-location of the inactivation centre,Development, vol. 78, no. 1, pp. 122, 1983.CrossRefGoogle ScholarPubMed
Lee, J. T. and Lu, N., “Targeted mutagenesis of Tsix leads to nonrandom X inactivation,Cell, vol. 99, no. 1, pp. 4757, 1999.CrossRefGoogle ScholarPubMed
Borsani, G., Tonlorenzi, R., Simmler, M. C. et al., Characterization of a murine gene expressed from the inactive X chromosome,Nature, vol. 351, no. 6324, pp. 325329, 1991.CrossRefGoogle ScholarPubMed
Brockdorff, N., Ashworth, A., Kay, G. F. et al., Conservation of position and exclusive expression of mouse XIST from the inactive X chromosome,Nature, vol. 351, no. 6324, pp. 329331, 1991.CrossRefGoogle ScholarPubMed
Brown, C. J., Ballabio, A., Rupert, J. L. et al., A gene from the region of the human X inactivation centre is expressed exclusively from the inactive X chromosome,Nature, vol. 349, no. 6304, pp. 3844, 1991.CrossRefGoogle ScholarPubMed
Wutz, A., Rasmussen, T. P., and Jaenisch, R., “Chromosomal silencing and localization are mediated by different domains of XIST RNA,Nature Genetics, vol. 30, no. 2, pp. 167174, 2002.CrossRefGoogle ScholarPubMed
Simon, M. D., Pinter, S. F., Fang, R. et al., High-resolution XIST binding maps reveal two-step spreading during X-chromosome inactivation,Nature, vol. 504, no. 7480, pp. 465469, 2013.CrossRefGoogle ScholarPubMed
Sado, T., Hoki, Y., and Sasaki, H., “Tsix silences XIST through modification of chromatin structure,Developmental Cell, vol. 9, no. 1, pp. 159165, 2005.CrossRefGoogle ScholarPubMed
Sun, B. K., Deaton, A. M., and Lee, J. T., “A transient heterochromatic state in XIST preempts X inactivation choice without RNA stabilization,Molecular Cell, vol. 21, no. 5, pp. 617628, 2006.CrossRefGoogle Scholar
Xu, N., Tsai, C.-L., and Lee, J. T., “Transient homologous chromosome pairing marks the onset of X inactivation,Science, vol. 311, no. 5764, pp. 11491152, 2006.CrossRefGoogle ScholarPubMed
Nicodemi, M. and Prisco, A., “Self-assembly and DNA binding of the blocking factor in X chromosome inactivation,PLoS Computational Biology, vol. 3, no. 11, pp. 21352142, 2007.CrossRefGoogle ScholarPubMed
Xu, N., Donohoe, M. E., Silva, S. S., and Lee, J. T., “Evidence that homologous X-chromosome pairing requires transcription and CTCF protein,Nature Genetics, vol. 39, no. 11, pp. 13901396, 2007.CrossRefGoogle ScholarPubMed
Scialdone, A. and Nicodemi, M., “Mechanics and dynamics of X-chromosome pairing at X inactivation,PLoS Computational Biology, vol. 4, >2008.CrossRefGoogle ScholarPubMed
Tian, D., Sun, S., and Lee, J. T., “The long noncoding RNA, JPX, Is a molecular switch for X chromosome inactivation,Cell, vol. 143, no. 3, pp. 390403, 2010.CrossRefGoogle Scholar
Sun, S., Del Rosario, B. C., Szanto, A. et al., XJpx RNA activates XIST by evicting CTCF,Cell, vol. 153, 2013.CrossRefGoogle ScholarPubMed
Chureau, C., Chantalat, S., Romito, A. et al., FTX is a non-coding RNA which affects XIST expression and chromatin structure within the X-inactivation center region,Human Molecular Genetics, vol. 20, no. 4, pp. 705718, 2011.CrossRefGoogle ScholarPubMed
Chadwick, B. P., “DXZ4 chromatin adopts an opposing conformation to that of the surrounding chromosome and acquires a novel inactive X-specific role involving CTCF and antisense transcripts,Genome Research, vol. 18, no. 8, pp. 12591269, 2008.CrossRefGoogle ScholarPubMed
Horakova, A. H., Moseley, S. C., McLaughlin, C. R., Tremblay, D. C., and Chadwick, B. P., “The macrosatellite DXZ4 mediates CTCF-dependent long-range intrachromosomal interactions on the human inactive X chromosome,Human Molecular Genetics, vol. 21, no. 20, pp. 43674377, 2012.CrossRefGoogle ScholarPubMed
Darrow, E. M., Huntley, M. H., Dudchenko, O. et al., Deletion of DXZ4 on the human inactive X chromosome alters higher-order genome architecture,Proceedings of the National Academy of Sciences, vol. 113, no. 31, pp. E4504E4512, 2016.CrossRefGoogle ScholarPubMed
Rao, S. S. P., Huntley, M. H., Durand, N. C. et al., A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping,Cell, vol. 159, no. 7, pp. 16651680, 2014.CrossRefGoogle ScholarPubMed
Echevarria, L., Benistan, K., Toussaint, A. et al., X-chromosome inactivation in female patients with Fabry disease,Clinical Genetics, vol. 89, no. 1, pp. 4454, 2016.CrossRefGoogle ScholarPubMed
Tukiainen, T., Villani, A.-C., Yen, A. et al., Landscape of X chromosome inactivation across human tissues,Nature, vol. 550, no. 7675, pp. 244248, 2017.CrossRefGoogle ScholarPubMed
Wareham, K. A., Lyon, M. F., Glenister, P. H., and Williams, E. D., “Age related reactivation of an X-linked gene,Nature, vol. 327, no. 6124, pp. 725727, 1987.CrossRefGoogle ScholarPubMed
Migeon, B. R., Axelman, J., Beggs, A. H., “Effect of ageing on reactivation of the human X-linked HPRT locus,Nature, vol. 335, no. 6185, pp. 9396, 1988.CrossRefGoogle ScholarPubMed
Sharp, A., Robinson, D., and Jacobs, P., “Age- and tissue-specific variation of X chromosome inactivation ratios in normal women,Human Genetics, vol. 107, no. 4, pp. 343349, 2000.CrossRefGoogle ScholarPubMed
Wang, J., Syrett, C. M., Kramer, M. C., Basu, A., Atchison, M. L., and Anguera, M. C., “Unusual maintenance of X chromosome inactivation predisposes female lymphocytes for increased expression from the inactive X,Proceedings of the National Academy of Sciences, vol. 113, no. 14, pp. E2029E2038, 2016.Google ScholarPubMed
Pageau, G. J., Hall, L. L., Ganesan, S., Livingston, D. M., and Lawrence, J. B., “The disappearing Barr body in breast and ovarian cancers,Nature Reviews Cancer, vol. 7, no. 8, pp. 628633, 2007.CrossRefGoogle ScholarPubMed
Chaligné, R., Popova, T., Mendoza-Parra, M.-A. et al., The inactive X chromosome is epigenetically unstable and transcriptionally labile in breast cancer,Genome Research, vol. 25, no. 4, pp. 488503, 2015.CrossRefGoogle ScholarPubMed
Kang, J., Lee, H. J., Kim, J. et al., Dysregulation of X chromosome inactivation in high grade ovarian serous adenocarcinoma,PLoS One, vol. 10, Article ID e0118927, 2015.Google ScholarPubMed
Winham, S. J., Larson, N. B., Armasu, S. M. et al., Molecular signatures of X chromosome inactivation and associations with clinicaloutcomes in epithelial ovarian cancer,Human Molecular Genetics, vol. 28, no. 8, pp. 13311342, 2019.CrossRefGoogle Scholar