Fact-checked by Grok 2 weeks ago

X-inactivation

X-chromosome inactivation (XCI) is an epigenetic dosage compensation mechanism in female mammals that transcriptionally silences one of the two , ensuring equivalent expression of X-linked genes between XX females and males. This process prevents overexpression of X-linked genes in females, who would otherwise have double the dosage compared to males possessing a single . First proposed by geneticist Mary Lyon in 1961 based on observations of X-linked coat color variegation in mice, XCI was later confirmed through cytological evidence of a condensed, inactive X chromosome (the Barr body) in female somatic cells. XCI occurs early in embryonic development and manifests in two primary forms: random XCI, where either the maternal or paternal X is chosen for inactivation with approximately equal probability in the inner cell mass and epiblast of placental mammals like humans and mice; and imprinted XCI, which preferentially silences the paternal X in marsupials and the extraembryonic tissues of rodents. These forms highlight evolutionary adaptations to achieve balanced gene dosage across mammalian lineages. The molecular initiation of XCI is orchestrated by the X-inactivation center (Xic), a genomic locus on the X chromosome that regulates the process through long non-coding RNAs. In eutherian mammals, the lncRNA Xist is upregulated on the future inactive X (Xi), coating it in cis and recruiting protein complexes that induce chromatin remodeling, including deposition of repressive histone marks like H3K27me3 and H2AK119ub. Key factors such as SPEN and hnRNP K mediate Xist function by bridging RNA to chromatin and promoting gene silencing. In marsupials, the analogous Rsx RNA performs a similar role but with distinct repeat structures. Maintenance of XCI throughout cell divisions relies on stable epigenetic modifications, including at CpG islands and the formation of a condensed structure characterized by the absence of topologically associating domains (TADs) and the presence of mega-domains. Recent advances reveal that Xist-driven compartmentalization reorganizes the Xi into repressive territories, while proteins like SmcHD1 ensure uniform late replication timing to stabilize silencing. Although most X-linked genes are silenced, approximately 15-25% escape inactivation in humans, contributing to and disease susceptibility. Dysregulation or skewing of XCI can influence the manifestation of X-linked disorders, such as in females, and contribute to the development of certain cancers, and its study has illuminated broader principles of epigenetic regulation, chromatin organization, and control. Ongoing research continues to uncover therapeutic potentials, including strategies to reactivate the for treating X-linked conditions.

Overview

Definition and Purpose

X-inactivation is an epigenetic process in which one of the two X chromosomes in each of mammals is randomly silenced to achieve dosage compensation, ensuring that XX females express X-linked genes at levels comparable to males. This phenomenon, proposed in the Lyon hypothesis, posits that the inactivation of one X chromosome occurs early in embryonic development and is maintained throughout the cell's lifetime, resulting in a of across female tissues. The primary purpose of X-inactivation is to prevent the overexpression of X-linked genes in females, which would otherwise lead to an imbalance in relative to males and potentially cause developmental or abnormalities. By transcriptionally silencing the majority of genes on the inactive , although approximately 15-25% of X-linked genes escape inactivation, this mechanism balances the output of X-linked products essential for cellular function and organismal viability. X-inactivation evolved in therian mammals, encompassing both marsupials and placental mammals, as a strategy for dosage compensation, though it exhibits variations in randomness and developmental timing between these groups. In placental mammals, the process is typically random, with either the maternal or paternal X chromosome inactivated with equal probability, whereas in marsupials, it is predominantly imprinted, favoring inactivation of the paternal X. This inactivation is achieved through a series of epigenetic modifications that propagate along the chromosome.

Timing and Species Specificity

X-inactivation occurs early in mammalian embryogenesis to achieve dosage compensation between sexes. In mice, imprinted paternal X-inactivation initiates at the 2- to 4-cell stage, with RNA coating the paternal , and progresses through the stage (embryonic days E3.5 to E5.5). In humans, the process begins during preimplantation , with evidence of a silent in female embryos as early as the 8-cell stage, though random inactivation is not fully established until later stages, around day 12 post-fertilization.00200-6) This early timing ensures balanced X-linked as the embryo differentiates into distinct lineages. Species-specific variations in X-inactivation reflect evolutionary adaptations in dosage compensation. In marsupials, such as the tammar wallaby, inactivation is strictly imprinted, targeting the paternal early in embryogenesis and maintaining it throughout life in all female tissues.00262-2) In like mice, imprinted paternal X-inactivation occurs in extraembryonic tissues, such as the trophectoderm and primitive endoderm, while the (epiblast) undergoes random inactivation of either parental X.00101-X) By contrast, humans exhibit random X-inactivation in both embryonic and extraembryonic lineages, including the , differing from the imprinted pattern seen in murine extraembryonic tissues. These differences highlight how imprinted mechanisms predominate in more primitive mammals, while random choice evolved in eutherians for equitable dosage in cells.00320-8) A notable exception to stable inactivation involves temporary reactivation during germ cell development in both sexes. In mice, the inactive X chromosome reactivates in female primordial germ cells between embryonic days E8.5 and E12.5, restoring biallelic expression to reset the germline epigenome. This reactivation also occurs in male germ cells, where the single X achieves balanced expression through upregulation, ensuring proper gametogenesis. X-inactivation is a conserved process essential for female viability in mammals (marsupials and placentals) but is absent in and egg-laying mammals like the . employ a ZW system with dosage compensation via partial Z upregulation in males, without inactivation. In monotremes such as the , which possess a chain of multiple homologous to Z and mammalian X, no equivalent X-inactivation mechanism operates, relying instead on other regulatory strategies for dosage balance.00241-8.pdf) This absence underscores X-inactivation's evolution within mammals as a specialized response to XX dosage imbalance.

Mechanism of Inactivation

Selection of the Inactive X Chromosome

In female cells of placental mammals, the process of X-inactivation begins with the random selection of one of the two X chromosomes for silencing, ensuring dosage compensation between XX females and XY males. This stochastic choice assigns each X chromosome an approximately equal probability—around 50%—of becoming the inactive X (Xi), a mechanism first proposed based on variegated coat color patterns in heterozygous female mice. The randomness of this selection, confirmed in human cells through analysis of glucose-6-phosphate dehydrogenase (G6PD) mosaicism in heterozygous females, results in a mosaic pattern of gene expression across tissues, where subpopulations of cells express either the maternal or paternal X chromosome. To guarantee that exactly one X chromosome remains active per cell despite the presence of two, mammalian cells employ a dosage-sensing or "counting" mechanism centered on the X-inactivation center (Xic), a critical genomic region on the . This process involves X-linked signal elements that interact with limiting autosomal factors, allowing the to tally the number of X chromosomes and initiate inactivation on all but one. One influential model suggests that a scarce autosomal activator is titrated by X-linked repressors at the Xic; in XX cells, this competition results in one X escaping repression while the other is targeted for silencing, whereas cells lack sufficient X-linked elements to trigger inactivation. This counting occurs early in embryonic development, synchronizing with the initial waves of upregulation. While the default selection is random and balanced, various factors can lead to , where one X is preferentially inactivated, deviating from the 50:50 ratio—for instance, resulting in ratios like 70:30 or more extreme imbalances. variation due to the small number of founder cells during early embryogenesis can cause such skewing purely by chance, particularly in specific tissues. Age-related skewing becomes prominent after 55–60 years, with up to 60% of women exhibiting moderate to extreme skewing (defined as ≥80:20), likely driven by clonal expansion of hematopoietic stem cells favoring one X lineage. Additionally, mutations in X-linked genes, such as those underlying X-linked mental retardation or , can impose selective pressure, biasing inactivation toward the mutant X to mitigate deleterious effects, though this often manifests as secondary skewing post-initial choice. In contrast to the random process in embryonic lineages, X-inactivation in extraembryonic tissues of placental mammals like mice exhibits an imprinted form with a strong paternal bias, where the paternal X is preferentially silenced due to pre-existing epigenetic marks established during . This non-random choice, initiated at the 2–4 cell stage, ensures dosage compensation in and other extraembryonic structures without mosaicism. The distinction highlights lineage-specific regulation, with the paternal bias linked to parent-of-origin effects at the Xic that protect the maternal X from inactivation.

Initiation and Propagation in Embryonic Development

X-inactivation initiates at the X-inactivation center (XIC), a master regulatory locus on the that orchestrates the choice and silencing of the future inactive X in female mammalian embryos. This process begins with the upregulation of regulatory non-coding RNAs at the XIC on the selected X chromosome, which coat the chromosome in cis and trigger the onset of silencing specifically from this locus. The XIC ensures that inactivation occurs on only one X per cell, integrating signals for chromosome counting and choice prior to the spreading of the repressive signal. Once initiated, silencing spreads from the XIC across the entire X chromosome by exploiting its three-dimensional architecture, involving the sequential domain-by-domain accumulation of repressive marks via cis-limited diffusion on the coated chromosome. Recent biophysical models describe this propagation as a reaction-diffusion process facilitated by Xist RNA condensates, which is efficient and typically completes within a few cell divisions while confining the effect to the originating X, preventing trans effects on the active X chromosome. In embryonic development, X-inactivation proceeds in two distinct waves: the first is imprinted, occurring in the trophectoderm lineage shortly after fertilization, where the paternal X is preferentially inactivated; the second is random, taking place later in the that gives rise to the proper. This imprinted wave is maintained in extraembryonic tissues, while the random wave in the establishes the pattern observed in cells. The timing of X-inactivation exhibits cell-to-cell variability, with asynchronous initiation across individual s in the epiblast, leading to a heterogeneous where each independently inactivates one X . This stochastic asynchrony results in the characteristic mosaic phenotype in female tissues, where neighboring cells may express different parental X s.

Molecular Regulators

Xist RNA Function

is a (lncRNA) approximately 17-19 kilobases in length, transcribed from the X-inactivation center (XIC) on the . It plays a pivotal role in X-chromosome inactivation by accumulating specifically on the chromosome destined to become inactive, forming distinct foci that coat the entire territory of the inactive X chromosome (). This coating was first observed through , revealing Xist RNA's localization as a dense, cloud-like structure enveloping the Xi during , thereby marking it for silencing. The mechanism of Xist-mediated silencing involves the recruitment of repressive protein complexes through specific repeat elements within its sequence. Xist contains multiple tandem repeats, including the A-repeats, which are crucial for initiating gene repression by facilitating the binding of (PRC2); deletion studies in mouse models demonstrated that A-repeats are necessary for efficient early silencing and PRC2 enrichment along the . Additionally, the B and C repeats mediate the recruitment of via interactions with proteins like hnRNPK, stabilizing the repressive state; experiments using engineered Xist mutants showed that B/C repeats are essential for chromosome-wide Polycomb deposition without which silencing is impaired. Xist spreads in cis along the from its transcription site, a process that excludes active transcription factors and from the coated region, creating a transcriptionally repressive nuclear compartment. Expression of is dynamically regulated during X-inactivation. In female mammalian cells, transcription is upregulated specifically from the future inactive , with low basal levels present on both s prior to inactivation but rapidly stabilized and accumulated only on the chosen to persist throughout development.80012-X) This upregulation coincides with increased stability, extending its half-life from minutes to hours, ensuring sustained coating and maintenance of ; in contrast, is absent or rapidly degraded on the active . Recent studies have elucidated that Xist RNA participates in liquid-liquid phase separation (LLPS) to form condensates, concentrating silencing factors in phase-separated domains that facilitate efficient Xi encapsulation and spreading. In a 2025 biophysical analysis, Xist was shown to interact with hnRNPK to drive LLPS, forming dynamic droplets that pull into a condensed state while limiting trans-chromosomal , providing a mechanistic basis for cis-specific action.01417-X)

Tsix RNA Role and Regulation

Tsix is a gene located within the X-inactivation center (XIC) on the , transcribed in the antisense orientation relative to the gene. It initiates approximately 15 kb downstream of the Xist 3' terminus and extends upstream to span the entire ~17 kb Xist locus, producing a ~40 kb primary transcript that is processed into a mature ~7-10 kb . Like Xist, Tsix is non-coding and predominantly nuclear, localizing to the XIC region where it forms an RNA domain that overlaps with Xist expression. The primary role of Tsix is to repress Xist expression in on the active , thereby preventing inappropriate Xist RNA coating and ensuring that only one X remains active per . Tsix transcription through the Xist locus actively blocks Xist RNA accumulation by interfering with its promoter activity and stabilizing a repressive state at the Xist locus. Experimental evidence from gain-of-function studies, where Tsix expression was enhanced using an EF-1α promoter, demonstrated a ~3-fold increase in Tsix levels that suppressed Xist upregulation by ~70% during , confirming that Tsix acts as a negative regulator of Xist coating without altering chromosome counting. Tsix regulation establishes between the two X chromosomes during the choice phase of X-inactivation, with high Tsix expression maintained on the future active X to inhibit , while Tsix is downregulated on the future inactive X, allowing upregulation. This arises stochastically from random differences in promoter activation probabilities at the Tsix locus, creating a competitive dynamic where the X with higher initial Tsix expression represses more effectively, ensuring monoallelic expression and one active X per cell. Downregulation of Tsix on the inactive X involves transcriptional silencing and degradation mechanisms that precede activation by several hours. Mutations disrupting Tsix function lead to defective X-chromosome choice and severe developmental consequences in mice. Targeted deletion of a 3.7 kb region containing the Tsix CpG island and promoter abolishes Tsix expression in cis, resulting in non-random inactivation where the mutant X is preferentially silenced in >97% of cells due to unchecked expression. In female embryonic stem cells, such mutations cause patterns and massive cell lethality during differentiation, with viable cells reduced by over 10,000-fold compared to wild-type, highlighting Tsix's essential role in maintaining cellular viability through proper dosage compensation. Homozygous Tsix mutants exhibit embryonic lethality at mid-gestation, underscoring its non-redundant function in random X-inactivation.

Epigenetic Silencing

Chromatin Modifications

X-chromosome inactivation (XCI) involves a series of modifications that establish and maintain the repressive state of the inactive X chromosome (), forming a facultative structure that is reversible, such as during reprogramming. These modifications include the enrichment of repressive marks, loss of active marks, DNA hypermethylation, and incorporation of specific histone variants, which collectively compact the Xi and silence . A key repressive modification is the trimethylation of at 27 (), a Polycomb group (PcG) mark deposited by the PRC2 complex, which accumulates on the Xi approximately 8 hours after Xist RNA coating and is essential for stable across broad domains. Another repressive mark, trimethylation of at 9 (), forms in intergenic regions of the Xi and is mediated by the SETDB1, contributing to compaction, though it emerges later and overlaps less with promoters compared to H3K27me3. Concurrently, active histone marks are depleted: trimethylation of H3 at 4 () and acetylation at H3 9 (H3K9ac) are lost chromosome-wide within 4-8 hours of Xist induction, particularly at promoters of silenced genes, facilitating the transition to a repressive state via recruitment of histone deacetylases like HDAC3. DNA hypermethylation at CpG islands (CGIs) of Xi genes occurs later in XCI, primarily at promoters, and is catalyzed by de novo DNA methyltransferases such as DNMT3A and DNMT3B, which establish stable silencing independent of initial Xist coating but dependent on upstream factors like SMCHD1 for certain loci. Additionally, the histone variant macroH2A, including subtypes macroH2A1 and macroH2A2, is incorporated into Xi nucleosomes, replacing canonical H2A and forming macrochromatin bodies that correlate with heterochromatic domains, though its depletion does not disrupt overall silencing. This combination of marks defines the Xi as facultative heterochromatin, which can be reactivated in primordial germ cells through erasure of repressive modifications, ensuring proper dosage compensation across generations.

Silencing Cascade and Maintenance

The silencing cascade in X-chromosome inactivation (XCI) begins with the coating of the future inactive () by RNA, which spreads in cis along the to initiate widespread gene repression. This coating recruits Polycomb repressive complexes (PRCs), particularly PRC2, leading to the deposition of repressive marks that compact and further silence gene expression. Subsequent steps involve the establishment of at promoter regions of silenced genes, which reinforces transcriptional repression, and tethering of the to the , promoting and spatial compartmentalization that stabilizes the inactive state. This ordered cascade ensures efficient, chromosome-wide silencing during early embryonic development, with each modification building upon the previous to achieve stable dosage compensation. Maintenance of XCI relies on a self-perpetuating epigenetic loop that sustains silencing across cell divisions, independent of continuous Xist expression in certain contexts. Once established, the repressive marks, including and DNA modifications, are propagated semi-conservatively during , creating a bistable system where the Xi remains silenced while the active X (Xa) stays expressed. In mice, this stability persists without Xist RNA after the initial phase, as demonstrated by conditional Xist deletion in embryonic fibroblasts, where endures through multiple divisions due to the entrenched epigenetic landscape. However, Xist may contribute to in specific lineages or genes, highlighting - and context-dependent variations in this process. Reactivation of the Xi can occur in specific cellular reprogramming events, such as the generation of induced pluripotent stem (iPS) cells, where pluripotency factors erode repressive marks, leading to partial X-chromosome reactivation (XCR) and biallelic expression. Similarly, recent studies have identified age-related escape from XCI, with increased Xi gene expression in aging female mouse and human tissues, particularly in the hippocampus and brain, driven by progressive loss of epigenetic barriers and linked to cognitive decline. These reactivation contexts underscore the dynamic nature of XCI maintenance, where external factors can disrupt the epigenetic loop. To prevent the spread of silencing into adjacent autosomal regions, insulator elements, such as CTCF-bound DNA sequences, act as boundaries that block heterochromatin propagation and protect transgenes or escapee genes from ectopic repression. This barrier function ensures that XCI remains confined to the Xi, maintaining genomic integrity across the nuclear landscape.

Inactive X Chromosome Features

Barr Body Formation

The Barr body, also known as the sex , represents the condensed form of the inactive X chromosome (Xi) in female mammalian somatic cells, serving as a cytological hallmark of X-chromosome inactivation (XCI). This dense, heterochromatic structure arises through progressive compaction following the initiation of XCI, where the Xi folds into a compact domain enriched with repressive modifications such as and macroH2A1.2, distinguishing it from the transcriptionally active X chromosome. The condensation process transforms the linear Xi into a looped configuration, often with a bipartite featuring two superdomains separated by a hinge region at the DXZ4 locus, as revealed by high-resolution imaging and techniques. Visualization of the Barr body was first achieved through light microscopy in the late 1940s, when observations of darkly staining nuclear inclusions in female cat neurons led to the identification of one such body per somatic cell in XX individuals, contrasting with its absence in XY males. Subsequent studies using electron microscopy and fluorescence in situ hybridization (FISH) confirmed its identity as the Xi, appearing as an irregularly shaped, electron-dense mass approximately 0.5–1 μm in diameter. In human and mouse cells, the Barr body is typically singular in diploid female nuclei, with the number correlating to the excess of X chromosomes beyond one (e.g., two Barr bodies in XXX cells). Barr body formation occurs after XCI initiation during early embryonic development, becoming morphologically evident by the stage in the epiblast lineage of the proper. In mice, random XCI in the leads to appearance around embryonic day 5.5–6.5, while imprinted XCI in extraembryonic tissues precedes this slightly earlier. The structure's size and precise positioning vary by cell type; for instance, in fibroblasts, it measures about 1 μm and localizes near the , whereas in lymphocytes, it may adopt a more central position. This variability reflects cell-specific nuclear architecture and interactions with the via proteins like lamin B receptor (LBR). Notably, Barr bodies are absent in germ cells and pre-implantation embryos due to the reactivation of the , ensuring biallelic X-linked expression during and early cleavage stages to support and initial . In primordial germ cells, for example, the reactivates around embryonic day 7.5 in mice, eliminating the condensed structure to achieve dosage equivalence without inactivation.

Escapee Genes and Partial Activity

In X-chromosome inactivation (XCI), approximately 15-25% of genes on the human X chromosome escape silencing and continue to be expressed from the inactive X chromosome (Xi), leading to biallelic expression in female cells. This incomplete silencing contrasts with the majority of X-linked genes that are robustly repressed to achieve dosage compensation. Notable examples of escapee genes include XIST itself, which is paradoxically expressed from the Xi to maintain inactivation of other genes, and the steroid sulfatase gene (STS), located near the pseudoautosomal region 1 (PAR1), which shows consistent biallelic expression across tissues. The pseudoautosomal regions (PAR1 and PAR2) represent hotspots of escape, where nearly all genes fully evade XCI due to their homology with the Y chromosome and functional pairing requirements during meiosis. Mechanisms underlying escape from XCI involve several epigenetic and structural features that prevent or limit the spread of silencing. A primary factor is the absence of Xist RNA coating on escapee loci, as observed for genes like KDM5C and KDM6A, which lack recruitment of repressive complexes. Boundary elements, such as CTCF-binding sites, act as insulators to demarcate escape domains and block the propagation of from silenced regions. Additionally, escapee genes often reside in open chromatin environments characterized by active histone marks like and reduced , maintaining an euchromatic state conducive to transcription. These features collectively insulate escapees from the broader repressive landscape of the Xi. The biallelic expression of escapee genes results in 1.5-2-fold higher levels in females compared to males, contributing to sexually dimorphic traits and phenotypes. For instance, elevated expression of PAR genes like SHOX influences height differences between sexes, while other escapees may affect immune responses or metabolic processes. Recent high-resolution mapping efforts have refined our understanding of escape variability. In 2024, single-cell analyses quantified escape for ~23% of X-linked genes across diverse types, revealing tissue-specific patterns. Similarly, a sex-aware profile of T development identified dynamic escape maps, with certain genes like CXCR3 showing increased biallelic activity upon activation, highlighting lineage-specific regulation.

Biological Implications

Dosage Compensation Effects

X-inactivation serves as the primary mechanism of dosage compensation in female mammals, equalizing X-linked between XX females and XY males by silencing one of the two X chromosomes. According to Ohno's hypothesis, proposed in 1967, the X chromosome in both sexes underwent upregulation to approximately twice the level of autosomal genes to compensate for the degeneration of the , ensuring balanced expression prior to the evolution of X-inactivation in females.00736-2) This two-step process—X upregulation followed by inactivation of one X in females—maintains dosage parity for most X-linked genes across sexes. In humans, approximately 85% of X-linked genes are subject to X-inactivation, resulting in expression levels from the inactive X that are comparable to those from the single active X in males, thereby achieving overall dosage balance for the majority of X-linked transcripts. The remaining 15% of genes escape inactivation to varying degrees, leading to approximately twofold higher expression in females compared to males for these loci. This incomplete silencing contributes to sex-specific dosage differences that are biologically significant. The random of X-inactivation generates cellular mosaicism in females, where individual cells express either the maternal or paternal , averaging out to balanced expression across tissues but introducing variability at the single-cell level. This mosaicism underlies subtle sex biases in effects, particularly for escapee genes, which can amplify differences in cellular responses. Evolutionarily, the persistence of escapee genes represents a , as their overexpression in females drives in traits such as immune responses; for instance, escape of the X-linked TLR7 enhances antiviral immunity in females but may increase risk.

Manifestation of X-Linked Traits in Females

In females heterozygous for X-linked mutations, random X-chromosome inactivation during early embryonic development results in cellular mosaicism, where individual cells randomly silence either the maternal or paternal , leading to a mixture of cells expressing either the mutant or wild-type . This mosaicism typically produces milder or patchy phenotypes compared to hemizygous males, as approximately half the cells produce functional protein from the normal , compensating for the defective one in the other half. For instance, female carriers of hemophilia A or B often exhibit subclinical or mild bleeding tendencies, with or IX levels varying based on the proportion of cells expressing the normal . Extreme skewing of X-inactivation, where inactivation favors one X chromosome in over 90% of cells, can amplify symptoms by reducing the proportion of cells producing functional protein from the normal allele. This bias, which occurs in about 7% of females, may result in near-complete expression of the mutant allele, mimicking male hemizygote phenotypes in affected tissues. Classic examples illustrate this mosaic manifestation. In calico cats, the X-linked orange fur color gene produces patchy pigmentation patterns due to random X-inactivation in melanocytes, with black and orange fur patches corresponding to cells expressing different alleles. Similarly, female carriers of X-linked anhidrotic ectodermal dysplasia display mosaic skin patterns along Blaschko's lines, with regions of normal sweating alternating with hypohidrotic areas lacking sweat glands, detectable via starch-iodine tests. Detection of these Lyonization patterns—referring to the effects of X-inactivation—involves analyzing tissue-specific X-chromosome inactivation ratios, often using methylation-sensitive assays on polymorphic markers like the gene to quantify the proportion of cells with active maternal versus paternal X chromosomes. This approach reveals variable expression across tissues, aiding in the diagnosis of carrier status for X-linked traits.

Clinical and Pathological Aspects

Skewed X-Inactivation and Disorders

refers to a non-random pattern where one is preferentially inactivated over the other in a significant proportion of s, often exceeding an 80:20 ratio, which can lead to imbalanced and phenotypic variability in females. This skewing can arise through primary mechanisms, such as inherited genetic factors including rare mutations in the gene that disrupt the inactivation process, or secondary mechanisms driven by cell selection where cells expressing a disadvantageous (e.g., carrying deleterious mutations) are selectively eliminated during development or tissue differentiation. Aging also contributes to acquired skewing, as cumulative cellular turnover may favor survival of cells with a particular active , leading to progressive imbalance in older individuals. In disorders like Rett syndrome, caused by mutations in the MECP2 gene on the X chromosome, skewed X-inactivation plays a critical role in disease manifestation and severity. Females with Rett syndrome are typically mosaic due to random inactivation, but when skewing favors inactivation of the wild-type X chromosome, a higher proportion of cells express the mutant MECP2, resulting in more severe neurological symptoms such as regression, seizures, and motor impairments; studies report that approximately 8-10% of affected females exhibit such skewed patterns, often due to selective pressures against cells with the mutant active X. Similarly, in fragile X premutation carriers (55-200 CGG repeats in FMR1), skewed X-inactivation is associated with fragile X-associated primary ovarian insufficiency (FXPOI), where preferential inactivation of the normal X chromosome exacerbates ovarian dysfunction, leading to premature ovarian failure in up to 20% of carriers; this skewing correlates with higher CGG repeat sizes and may amplify FMR1 toxicity through imbalanced expression. Diagnosis of skewed X-inactivation typically involves quantifying the inactivation ratio through methylation-sensitive assays targeting polymorphic markers, such as the CAG repeat in the () gene, where differential distinguishes active from inactive X chromosomes after digestion with enzymes like HpaII; skewing is defined as >85% inactivation of one X. Advanced methods, including single-cell sequencing, enable precise assessment at the cellular level by analyzing allele-specific expression and escape from inactivation, particularly useful for tissue-specific skewing in disorders. These approaches are essential for identifying carriers or predicting disease risk in X-linked conditions. Recent advances in 2025 have explored to address in by reactivating the healthy silenced . In a mouse model, researchers used a DNA-based "sponge" delivered via AAV vector to inhibit microRNA-106a (miR-106a), a regulator of XIST-mediated silencing, resulting in partial reactivation of the wild-type MECP2 gene, improved survival, motor function, and breathing patterns without toxicity. This approach highlights potential for targeted reactivation in females with unfavorable skewing, offering a strategy to restore dosage balance beyond traditional gene replacement.

Escape from Inactivation and Disease Risks

Escape from X-chromosome inactivation (XCI) can become pathological when it extends beyond the normal subset of constitutively escaping genes, leading to aberrant biallelic expression that disrupts dosage compensation. Recent studies have shown that aging promotes widespread reactivation of the inactive (), particularly in specific tissues such as the and . In female mice, the proportion of X-linked genes escaping inactivation increases from an average of 3.5% in adults to 6.6% in aged individuals (approximately 1.5 years old), representing nearly a doubling of escape rates and involving up to 31 age-specific escapee genes across organs. This age-related escape is concentrated at distal chromosome regions and correlates with enhanced accessibility at regulatory elements, potentially contributing to cellular dysfunction in elderly females. Similarly, in the female mouse , aging activates select Xi genes, which may influence cognitive resilience but also raises risks for imbalance if dysregulated. Overexpression of genes escaping XCI has been linked to heightened disease risks, particularly in conditions with female bias. In autoimmunity, biallelic expression of immune-related escapees, such as those involved in signaling, contributes to the female predominance in systemic lupus erythematosus (SLE), where women are affected 9 times more often than men. For instance, escape from XCI in immune cells amplifies dosage of pro-inflammatory X-linked genes like TLR7, promoting hyperactive responses that drive SLE pathogenesis. This mechanism extends to other autoimmune disorders, with the inactive X chromosome acting as a genetic driver of female-biased through incomplete silencing. In cancer, biallelic expression of tumor-suppressor escapees reduces protective effects, contributing to sex-biased tumor development; for example, escape of genes like UTX is associated with increased female risk in certain hematologic malignancies. Defective XCI, arising from mutations in key factors such as XIST or associated chromatin regulators, further exacerbates cancer predisposition by allowing additive risks from incomplete silencing. A 2025 analysis of The Cancer Genome Atlas data revealed that defective XCI exceeding 10% of X-linked genes carries a 40% attributable risk across 12 cancer types, with mutations in XCI machinery compounding oncogenic potential through unchecked biallelic activity. This defect integrates with other genetic hits, elevating overall malignancy rates in affected females. Lineage-specific variations in escape amplify these risks, notably in immune cells where incomplete XCI is more prevalent. In T cells, a high-resolution map indicates stable XCI overall during development, with approximately 12-16% of X-linked genes showing partial escape (up to 20% for heterozygous SNPs), though with minimal sex-biased expression; altered XCI in T cells may contribute to immune dysregulation underlying female susceptibility to autoimmune diseases like SLE and Sjögren's syndrome. A whole-organism analysis confirms that tissue-specific escape patterns may drive sex differences in disease severity, particularly in immunity-related pathologies.

Experimental Applications

Research Tools and Models

chimeras have been instrumental in modeling X-chromosome inactivation (XCI) mosaicism, allowing researchers to study the functional consequences of heterogeneous populations . In these models, female heterozygous for X-linked mutations produce mosaic tissues where express either the mutant or wild-type due to random XCI, revealing patterns of selection and tissue-specific effects. For instance, analysis of mosaic carrying mutations in GPI-linked protein genes demonstrated that selection favors expressing functional alleles, highlighting the role of XCI in developmental viability. Similarly, studies in Flna heterozygous female confirmed normal XCI mosaicism in corneal epithelia, providing a platform to investigate X-linked disorders like periventricular heterotopia. Induced pluripotent stem (iPS) cells serve as a key model for investigating XCI reactivation, particularly during cellular reprogramming. Reprogramming female somatic cells to iPSCs reverses XCI, reactivating the inactive X chromosome (Xi) and providing insights into the epigenetic steps involved. A seminal study mapped X-chromosome reactivation dynamics in mouse iPSCs, identifying sequential stages of Xist RNA depletion, H3K27me3 loss, and gene re-expression that occur over reprogramming. In human iPSCs, stable reactivation of the Xi has been observed, with erosion of XCI in long-term cultures leading to biallelic X-linked expression, which underscores the plasticity of XCI maintenance. These models enable dissection of reactivation triggers, such as interferon γ pathway activation, which accelerates pluripotency and X-reactivation during reprogramming. A 2025 study further characterized XCI erosion in human iPSCs, showing progressive gene reactivation on the Xi and its implications for modeling X-linked diseases. Xist transgenes represent a foundational tool for manipulating XCI initiation and maintenance in experimental systems. Transgenic expression of Xist cDNA in mouse embryonic stem cells induces reversible XCI upon differentiation, allowing controlled timing of inactivation to study its progression from initiation to stability. In human cells, inducible XIST transgenes in somatic lines localize XIST RNA to the X chromosome, triggering transcriptional silencing and H3K27me3 enrichment, thus recapitulating XCI in non-native contexts. Yeast artificial chromosome (YAC) transgenes carrying human XIST demonstrate partial X-inactivation in transgenic mice, confirming the cis-acting role of XIST in silencing. CRISPR-Cas9 editing of the X-inactivation center (XIC) has emerged as a precise tool to probe XCI regulatory elements. Targeted deletions in the XIC, such as removal of the , disrupt choice and lead to non-random XCI in mouse cells, validating Tsix's role in Xist repression. Editing the promoter in human iPSCs restores XIST expression and prevents XCI erosion, mitigating dosage imbalances during prolonged culture. Similarly, CRISPR-mediated of Xist repeat D impairs XCI establishment, reducing Xist levels and chromosome-wide silencing in cells. Full XIC deletions in mice abolish XCI, resulting in embryonic for XX embryos and confirming the XIC's essentiality. Live imaging techniques have advanced the visualization of Xist RNA dynamics during XCI. Double-tagged Xist reporters in cells enable real-time tracking of Xist cloud formation, revealing its accumulation on the and association with silencing factors like SPEN. These methods show Xist spreading in cis within nuclear territories, with confinement to the silenced X, and have been used to dissect defects upon Xist repeat B deletion. In differentiating female cells, live imaging captures nascent Xi formation, highlighting temporal asynchrony in Xist coating and assembly. Stem cell differentiation assays provide applications for monitoring XCI timing and fidelity in vitro. In mouse embryonic stem cells, differentiation protocols recapitulate random XCI, with Xist upregulation and Xi silencing occurring post-pluripotency exit, allowing assays of escapee gene expression. Human iPSCs exhibit variable XCI upon differentiation, with single-cell RNA sequencing revealing asynchrony between XCI, pluripotency loss, and lineage commitment. These assays have mapped XCI erosion in hiPSCs, linking it to autosomal gene dysregulation. Clonal lineage tracing leverages XCI mosaicism to track developmental contributions. X-linked polymorphisms in heterozygous females enable clonality assessment via methylation-sensitive assays, distinguishing monoclonal from polyclonal expansions in tissues. Single-cell tracing in human iPSCs uses XCI to infer somatic mutations, enhancing resolution of lineage hierarchies in development. In T cell models, XCI patterns reveal clonal heterogeneity, with competition between Xi-active and Xi-inactive lineages shaping immune repertoires. Recent advancements from 2023-2024 include workshops and high-resolution XCI maps in specialized models. The EMBO Workshop on X-chromosome inactivation in (June 2023) gathered experts to discuss 60 years of insights, emphasizing molecular regulators and relevance. In organoids, placental models display imprinted XCI patterns akin to early embryos, enabling study of reactivation during trophoblast . For T cells, single-cell atlases from 2024 map XCI escape during thymic development, identifying stage-specific escapees like that influence maturation. Activation studies confirm XCI maintenance post-stimulation, with no widespread reactivation in mature T cells.

Therapeutic Strategies

Therapeutic strategies targeting X-inactivation primarily focus on reactivating the inactive () to restore in X-linked disorders, particularly those affecting females like . One prominent approach involves the use of small-molecule (DNMT) inhibitors, such as 5-azacytidine (5-Aza), combined with antisense oligonucleotides (ASOs) to downregulate RNA, the that coats and silences the . This mixed-modality strategy has demonstrated synergistic effects in reactivating silenced genes, achieving up to 100-fold higher expression levels compared to single agents in cellular models of X-linked disorders. In the context of , caused by mutations in the X-linked MECP2 gene, recent efforts have targeted X-inactivation mechanisms to reactivate the healthy allele on the Xi. A 2025 study from UC Davis Health developed a approach that modulates microRNA-dependent control of X-chromosome inactivation, leading to significant phenotypic improvements in Rett syndrome mouse models, including enhanced motor function and reduced neurological symptoms. Strategies to modulate X-inactivation skewing in female carriers of X-linked diseases seek to preferentially inactivate the mutant , potentially alleviating symptoms through epigenetic interventions. While specific drugs remain under development, therapeutic modulation of the epigenome has been proposed as a viable approach for conditions like deficiency, where skewing towards the normal could mitigate disease manifestation. Despite these advances, therapeutic targeting of X-inactivation faces significant challenges, including off-target effects that could disrupt global epigenetic balance and unintended reactivation of non-therapeutic genes. Achieving tissue-specific delivery remains difficult, as may not uniformly affect all relevant cell types, such as neurons in neurological disorders. Additionally, ethical concerns arise with interventions in embryonic stages, where altering X-inactivation could have heritable implications and raise questions about and long-term safety. Looking ahead, disrupting condensates—phase-separated structures formed by and protein interactors—offers promising prospects for selective reactivation. Recent reviews highlight small molecules or PROTACs that could target these condensates to destabilize scaffolding, enabling precise reactivation without broad off-target impacts, with ongoing preclinical evaluations for X-linked diseases.

Historical Development

Early Discoveries

The discovery of the , also known as the sex mass, marked the initial observation pivotal to understanding X-inactivation. In 1949, Murray L. Barr and Ewart G. Bertram identified a distinct body in the nuclei of female somatic cells while studying nerve cells in cats, noting its absence in male nuclei. This heterochromatic structure, typically located near the nuclear membrane or nucleolus, was later recognized as the condensed, inactive in females. Building on this, early genetic studies in the and provided evidence for a mechanism ensuring dosage compensation between sexes. Researchers observed variegated coat color patterns in female mice heterozygous for X-linked genes, such as the tabby or mottled mutants, where individual hairs or patches displayed alternating wild-type and mutant phenotypes, suggesting expression due to the inactivation of one X per . These findings indicated that the inactivation process occurs early in embryonic and is stable through cell divisions, leading to clonal patches of . Mary F. Lyon formalized these observations in her 1961 hypothesis, proposing that in female mammals, one of the two s is randomly inactivated in each to achieve dosage equivalence with males, who possess a single . Drawing from the mouse coat color data, Lyon suggested that this inactivation is heritable and results in a functional hemizygosity, explaining the variegated phenotypes as a consequence of random choice between maternal and paternal s. Her model predicted that approximately 50% of cells would express each in heterozygotes, a later confirmed in various . Susumu Ohno further contextualized these ideas in 1967 by linking X-inactivation to broader principles of compensation, arguing that it maintains balanced expression of X-linked genes relative to the single X and autosomes in both sexes. Ohno's work emphasized evolutionary conservation, noting that similar mechanisms might operate in other organisms to prevent overexpression from , thus integrating the and into a unified framework for sex-specific gene regulation.

Key Molecular Insights

The identification of the X-inactivation center (Xic) marked a crucial advance in elucidating the genetic control of X-inactivation in mammals. In the early 1990s, genetic mapping studies in mice pinpointed the Xic to a discrete locus on the , approximately 450 kb in size, which orchestrates chromosome counting, choice, and silencing initiation in cis.80079-3) This region was defined through analysis of X-chromosome rearrangements and transgenic insertions that disrupted inactivation patterns, confirming its necessity for the process. A major breakthrough came with the cloning of the Xist gene within the Xic in 1991. Independent studies by Brockdorff et al. and Borsani et al. identified as a gene expressed exclusively from the inactive , producing a 15-17 kb transcript that localizes to the without encoding a conserved open reading frame.90519-I) In 1992, the human ortholog was cloned by et al., revealing conserved repeats and nuclear localization, establishing Xist as the key effector RNA that coats the to initiate silencing.90520-7) Functional tests in embryonic stem cells demonstrated that Xist upregulation is essential for inactivation, as transgenes carrying Xist could induce silencing on autosomes. The discovery of the antisense regulator Tsix in 1999 further refined the molecular model of Xic function. Jeannie T. Lee and colleagues identified Tsix as a 40-kb transcribed in the opposite direction to , originating 15 kb downstream within the Xic. Tsix expression represses on the future active , ensuring random choice and preventing ectopic inactivation; targeted deletion of Tsix led to non-random, female-biased X-inactivation without affecting counting.80061-6) Recent advances from 2023 to 2025 have deepened insights into mechanisms, leveraging single-cell and imaging to dissect dynamics and elements. Studies revealed that forms biomolecular condensates via its A-repeat domain, driving and early compaction during inactivation initiation.00697-6) Single-cell analyses have mapped from inactivation at high resolution, showing tissue-specific heterogeneity in ~15-25% of X-linked genes and identifying cis-regulatory elements that evade silencing.00229-5) Additionally, investigations into turnover via m6A modifications and nuclear exosome complexes have highlighted dynamic regulation, with implications for maintaining long-term silencing. These findings underscore 's role in condensate-mediated and precise control, informed by high-impact models like CRISPR-edited human iPSCs.

References

  1. [1]
    Mechanisms of Choice in X-Chromosome Inactivation - PMC
    Here, we review our current understanding of the process of choice during X-chromosome inactivation and its implications.
  2. [2]
    X chromosome inactivation in mammals: general principles and ...
    Jun 19, 2025 · X chromosome inactivation (XCI) is a mammalian dosage compensation mechanism that ensures balanced expression of X-linked genes between males and females.
  3. [3]
    X-chromosome inactivation: the gift that keeps on giving - Nature
    Aug 18, 2023 · In the orange patches, the X chromosome with the allele that results in brown color is inactivated, while in the brown patches, the X ...
  4. [4]
    Dosage compensation in mammals: fine-tuning the expression of ...
    The inactivation of one X chromosome in females equalizes gene expression between the sexes. This process of X-chromosome inactivation (XCI) is a remarkable ...
  5. [5]
    Dosage Compensation in Mammals - PMC - NIH
    The X inactivation hypothesis has continued to provide an explanation for the peculiarities of X-linked gene expression in female cells and has remained ...
  6. [6]
  7. [7]
    Epigenetic modifications on X chromosomes in marsupial and ...
    Sep 22, 2010 · In mammals, X inactivation has evolved to solve the difference in X chromosome gene dosage between homogametic female mammals and heterogametic ...
  8. [8]
    Early X chromosome inactivation during human preimplantation ...
    Sep 7, 2017 · In mice, the process starts at 2- to 4-cell stage female embryos, where the long non-coding Xist RNA is first observed coating the paternal X in ...
  9. [9]
  10. [10]
    Concurrent X chromosome inactivation and upregulation during non ...
    May 5, 2021 · Studies in human embryos revealed that random X chromosome inactivation starts at the preimplantation stage and is not complete by day 12 of development.
  11. [11]
    Dosage compensation in the process of inactivation/reactivation ...
    Jun 16, 2017 · First, the inactivated X chromosome is reactivated between embryonic day (E) 8.5 and E12.5 during the primordial germ cell (PGC) development in ...
  12. [12]
    Expression of Xist in mouse germ cells correlates with X ... - Nature
    Nov 1, 1992 · A transiently inactive X chromosome is also found in germ cells, specifically in premeiotic oogenic cells and in meiotic and postmeiotic ...
  13. [13]
    Multi-layered dosage compensation of the avian Z chromosome by ...
    Oct 13, 2025 · While therian mammals (XX/XY system) achieve near-perfect balance of X-chromosome mRNAs through X-upregulation and X-inactivation, birds (ZW/ZZ ...
  14. [14]
    The origin and evolution of vertebrate sex chromosomes ... - Nature
    Nov 16, 2011 · It is currently unknown which epigenetic changes are associated with the partial X inactivation system in platypus. Rens et al.
  15. [15]
  16. [16]
  17. [17]
  18. [18]
  19. [19]
    Skewness of X-chromosome inactivation increases with age and ...
    Feb 22, 2021 · Mosaicism in blood varies with age, and cross-sectional studies indicate that for women, skewness of X-chromosomal mosaicism increases with age.
  20. [20]
  21. [21]
    Conversion of random X-inactivation to imprinted X ... - eLife
    Apr 2, 2019 · Imprinted X-inactivation results in the silencing of genes exclusively on the paternal X-chromosome and initiates during preimplantation ...
  22. [22]
  23. [23]
  24. [24]
    Genetic analysis of the mouse X inactivation center defines an 80-kb ...
    In response to developmental cues, the Xic orchestrates events of X inactivation, including chromosome counting and choice, initiation, spread, and ...
  25. [25]
    Stabilization of Xist RNA Mediates X Chromosome Inactivation
    The XIC is required both for initiation of X inactivation early in development ... In early mouse embryogenesis, initiation of X inactivation occurs in two waves.
  26. [26]
  27. [27]
    Dynamic changes in paternal X-chromosome activity during ... - PNAS
    Mar 31, 2009 · In mice, X inactivation is initially imprinted, with inactivation of the paternal X (Xp) chromosome occurring during preimplantation development.
  28. [28]
    Xist RNA in action: Past, present, and future - PMC - PubMed Central
    Sep 19, 2019 · Xist was initially proven to be indispensable for XCI by targeted mutagenesis and transgenesis in mouse embryonic stem cells (ESCs) and in mice.Fig 1. Xist. A Multi-Tasking... · Rbm15 And Wtap · Hnrnp K
  29. [29]
    XIST RNA paints the inactive X chromosome at interphase - NIH
    The XIST gene is implicated in X chromosome inactivation, yet the RNA contains no apparent open reading frame. An accumulation of XIST RNA is observed near ...
  30. [30]
    Xist RNA Exhibits a Banded Localization on the Inactive X ...
    We show that Xist RNA exhibits a banded pattern on the inactive X and is excluded from regions of constitutive heterochromatin.
  31. [31]
    Xist Repeats A and B account for two distinct phases of X ... - NIH
    Repeat A initiates Polycomb recruitment and gene silencing, whereas Repeat B stabilizes them. Surprisingly, X-inactivation can initiate without Repeat B.
  32. [32]
    Article hnRNPK Recruits PCGF3/5-PRC1 to the Xist RNA B-Repeat ...
    Dec 7, 2017 · The Polycomb-repressive complexes PRC1 and PRC2 play a key role in chromosome silencing induced by the non-coding RNA Xist.
  33. [33]
    New Xist-Interacting Proteins in X-Chromosome Inactivation
    Apr 25, 2016 · Following upregulation from one X chromosome, Xist spreads in cis ... transcription factors and RNA polymerase II are excluded and active histone ...
  34. [34]
    X Chromosome Inactivation Is Mediated by Xist RNA Stabilization
    Low level Xist expression can be detected from both active X chromosomes (Xa) in female embryonic stem cells prior to X inactivation.Results · Xist Expression In... · Half-Life Of Xist Rna...
  35. [35]
    A functional role for Tsix transcription in blocking Xist RNA ...
    The expression pattern of Tsix is consistent with a role in regulating Xist (13). All active X chromosomes express Tsix before differentiation, a time when Xist ...
  36. [36]
  37. [37]
    Chromatin-mediated silencing on the inactive X chromosome
    Nov 22, 2023 · It is unknown how early H3K9 methylation accumulates on the Xi; however, it is clear that H3K9me2/3 forms domains unoccupied by H3K27me3 in both ...Initial Chromatin Changes On... · Polycomb Chromatin Changes... · 3d Chromatin Structure And...
  38. [38]
    Histone modification patterns associated with the human X ... - NIH
    Our analysis of two X-linked chromosomal regions showed that H3K9me3 is present within active coding regions, whereas H3K27me3 is an abundant mark within ...Missing: H3K9me | Show results with:H3K9me
  39. [39]
    The role of Xist‐mediated Polycomb recruitment in the initiation of X ...
    Xist RNA has been established as the master regulator of X‐chromosome inactivation (XCI) in female eutherian mammals, but its mechanism of action remains ...
  40. [40]
    Histone H2A variants and the inactive X chromosome - PubMed
    May 1, 2001 · MacroH2A1 is an unusual variant of the core histone H2A which is enriched in chromatin on the inactive X chromosome of female mammals.
  41. [41]
    The Molecular and Nuclear Dynamics of X-Chromosome Inactivation
    Xist coating triggers a cascade of chromosome-wide changes occurring at the levels of transcription, chromatin composition, chromosome structure, and spatial ...
  42. [42]
    Recruitment of PRC1 function at the initiation of X inactivation ...
    We show that PRC1 recruitment by Xist RNA is independent of gene silencing. We find that Eed is required for the recruitment of the canonical PRC1 proteins.Xist Mediated H2a... · Xist Recruits Prc1... · Eed And H3k27me3 Are Not...Missing: cascade lamina<|control11|><|separator|>
  43. [43]
    X Inactivation and Escape: Epigenetic and Structural Features
    Sep 30, 2019 · This review focuses first on mechanisms that govern X chromosome structure and nuclear location in relation to XCI, with a specific emphasis ...
  44. [44]
    A Chromosomal Memory Triggered by Xist Regulates Histone ... - NIH
    Our results suggest that this silencing-independent chromosomal memory has important implications for the maintenance of X inactivation, where previously self- ...
  45. [45]
    Synergism of Xist Rna, DNA Methylation, and Histone ... - NIH
    However, several lines of evidence indicate that after X inactivation has been established, Xist is no longer required for maintenance. In studies using mouse ...
  46. [46]
    Incomplete X-inactivation initiated by a hypomorphic Xist allele in ...
    Jul 1, 2011 · Interestingly, conditional deletion of Xist revealed that X-inactivation is maintained even in the absence of Xist RNA in embryonic fibroblasts ...Results · Xist Is Unique Among Xist... · Xist Produces Rna That...
  47. [47]
    Xist exerts gene-specific silencing during XCI maintenance and ...
    Aug 1, 2022 · We conclude that Xist is necessary for gene-specific silencing during XCI maintenance and impacts lineage-specific cell differentiation and proliferation ...
  48. [48]
    Aging activates escape of the silent X chromosome in the female ...
    Mar 5, 2025 · In female mammals with two X's, one is silenced through X chromosome inactivation (XCI), resulting in an active (Xa) and inactive X (Xi) (24).
  49. [49]
    Aging promotes reactivation of the Barr body at distal chromosome ...
    May 1, 2025 · We found that aging led to a substantial increase in escape rates, concentrated at distal chromosome regions.
  50. [50]
    A DNA insulator prevents repression of a targeted X-linked ...
    One possible escape mechanism is that heterochromatization during X inactivation can be blocked by boundary elements. DNA insulators are candidates for ...
  51. [51]
    Structural aspects of the inactive X chromosome - PMC
    Sep 25, 2017 · The Barr body is a looped X chromosome formed by telomere association. Proc. Natl Acad. Sci. USA 88, 6191–6195. ( 10.1073/pnas.88.14.6191) ...
  52. [52]
  53. [53]
  54. [54]
    X Chromosome Inactivation and Embryonic Stem Cells - NCBI - NIH
    X chromosome inactivation (XCI) is a process required to equalize the dosage of X-encoded genes between female and male cells.
  55. [55]
  56. [56]
    Reactivation of the inactive X chromosome in development ... - NIH
    X chromosome inactivation is initiated in early embryos by the noncoding Xist RNA. Subsequent chromatin modifications on the inactive X chromosome (Xi) lead to ...
  57. [57]
    Escape from X inactivation in mice and humans | Genome Biology
    Jun 24, 2010 · While Xist RNA coating is important in the initiation of X inactivation, many other epigenetic modifications follow to silence the X and ...
  58. [58]
    Quantification of escape from X chromosome inactivation with single ...
    Aug 14, 2024 · Several X-linked genes escape from X chromosome inactivation (XCI), while differences in escape across cell types and tissues are still ...
  59. [59]
    Genes that escape from X inactivation - PMC - NIH
    Genes that escape X inactivation remain expressed from both active and inactive X alleles, unlike most genes silenced by X inactivation.
  60. [60]
    Landscape of X chromosome inactivation across human tissues
    Oct 12, 2017 · X chromosome inactivation (XCI) silences transcription from one of the two X chromosomes in female mammalian cells to balance expression ...
  61. [61]
    A landscape of X-inactivation during human T cell development
    Dec 4, 2024 · We present a sex-aware expression profile of T cell development and generate a high-resolution map of escape from X-chromosome inactivation (XCI).Missing: percentage examples
  62. [62]
    Human active X-specific DNA methylation events showing stability ...
    Apr 9, 2014 · The process of XCI is incomplete, with approximately 15% of genes on human X chromosomes 'escaping' inactivation, potentially leading to gene ...
  63. [63]
    Skewed X-inactivation is common in the general female population
    Dec 14, 2018 · X-chromosome inactivation is the example of an extraordinary epigenetic silencing mechanism spreading across the entire human ~160 Mbp ...
  64. [64]
    Heritability of skewed X-inactivation in female twins is tissue-specific ...
    Nov 25, 2019 · This random inactivation results in a mosaic of cells within an individual, where overall, a balanced expression (50:50) of both parental X- ...Missing: asynchronous | Show results with:asynchronous
  65. [65]
    ImmGen report: sexual dimorphism in the immune system ... - Nature
    Sep 20, 2019 · For example, the X-linked gene Tlr7 plays a role in innate immune response and displays higher expression in females compared to males, ...
  66. [66]
    X-linked diseases: susceptible females | Genetics in Medicine - Nature
    Apr 14, 2020 · X-inactivation silences all X chromosomes but one; therefore, both males and females have a single active X. For 46 XY males, that X is the only ...
  67. [67]
    Mosaics and haemophilia - KASPER - 2009 - Wiley Online Library
    Oct 26, 2009 · Non-random inactivation appears to be the major reason. A carrier with non-random X-inactivation, in whom the X chromosome bearing a mutant ...
  68. [68]
    Skewed X inactivation in Lesch–Nyhan disease carrier females
    Sep 14, 2017 · Extreme skewing of XCI is the preferential inactivation of one X chromosome in ⩾90 of cells or ⩾95% of cells. In several X-linked disorders, ...
  69. [69]
    Recent Advances in X-Chromosome Inactivation - PMC
    The mosaic coat color of the calico cat exemplifies X-chromosome inactivation (Lyon, 1961). Calico cats are almost exclusively female and comprise of two ...
  70. [70]
    Clinical Findings in Mosaic Carriers of Hypohidrotic Ectodermal ...
    Dental abnormalities, mild hypohidrosis, and mild hypotrichosis are the most commonly described signs in female carriers of X-linked HED. The reported studies ...
  71. [71]
    Nonrandom X Chromosome Inactivation Detection - Caylor - 2023
    Apr 19, 2023 · X chromosome inactivation patterns may be clinically useful in assessing tumor clonality, determining carrier status for certain X-linked disorders.
  72. [72]
    X Chromosome–Inactivation Patterns of 1,005 Phenotypically ... - NIH
    X-chromosome inactivation is widely believed to be random in early female development and to result in a mosaic distribution of cells, approximately half with ...
  73. [73]
    A skewed view of X chromosome inactivation - JCI
    Dec 20, 2007 · Most of the skewing observed in humans results from secondary events rather than being due to an inherited tendency to inactivate a particular X chromosome.Missing: factors | Show results with:factors
  74. [74]
    Molecular Mechanisms of Skewed X-Chromosome Inactivation in ...
    Aug 23, 2021 · It may also be caused by mutations in the XIST gene, the gene responsible for the inactivation process [17], although such mutations are rare ...3. Results · 3.1. Patient 1 · 3.3. Patient 3
  75. [75]
    Age acquired skewed X chromosome inactivation is associated with ...
    Nov 22, 2022 · Our study demonstrates that age acquired XCI-skew captures changes to the haematopoietic stem cell population and has clinical potential as a unique biomarker ...
  76. [76]
    Increased skewing of X chromosome inactivation in Rett syndrome ...
    Jul 5, 2006 · A skewed X inactivation pattern may arise due to chance, to inherited factors or to selective forces acting on dividing cells with particular ...
  77. [77]
    Analysis of X‐inactivation status in a Rett syndrome natural history ...
    Mar 23, 2022 · XCI skewing occurs when one X chromosome is preferentially inactivated over the other in a nonrandom manner. ... Previous reports have found ~8.8% ...
  78. [78]
    Skewed X‐chromosome inactivation is associated with primary but ...
    Apr 12, 2007 · Skewed X-chromosome inactivation is associated with primary but not secondary ovarian failure ... Fragile X premutation is a significant ...
  79. [79]
    Association of skewed X-chromosome inactivation with FMR1 CGG ...
    Apr 28, 2017 · Premature ovarian failure and fragile X female premutation carriers: no evidence for a skewed X-chromosome inactivation pattern. Menopause.
  80. [80]
    X-Chromosome Inactivation Analysis | Test Fact Sheet - ARUP Consult
    Jun 27, 2024 · XCI patterns may differ among tissues. XCI ratio reported is for the tissue type tested, with a standard deviation (SD) of 0.08 for XCI ...
  81. [81]
    Assessment of XCI skewing and demonstration of XCI escape ...
    Jan 31, 2025 · To assess XCI escape and skewing, it is challenging to differentiate using bulk sequencing. Therefore, single-cell RNA sequencing (scRNA-seq) is ...
  82. [82]
    Targeting microRNA-dependent control of X chromosome ... - Nature
    Jul 4, 2025 · Targeting microRNA-dependent control of X chromosome inactivation improves the Rett Syndrome phenotype. Nat Commun 16, 6169 (2025). https ...
  83. [83]
    Escape from X Chromosome Inactivation and the Female ...
    Jan 23, 2021 · ... autoimmunity, in particular SLE and Sjögren syndrome. However ... Tumor-suppressor genes that escape from X-inactivation contribute to cancer sex ...
  84. [84]
    Xist ribonucleoproteins promote female sex-biased autoimmunity: Cell
    Feb 1, 2024 · Specific X-linked genes, such as TLR7, that can escape X inactivation have been nominated as contributors to specific autoimmune diseases.<|control11|><|separator|>
  85. [85]
    The Inactive X Chromosome: A Genetic Driver of Female‐Biased ...
    May 26, 2025 · In humans and mice, the number of X chromosomes, rather than sex‐steroid hormones, is associated with a higher risk to develop autoimmunity, ...
  86. [86]
    Defective X-chromosome inactivation and cancer risk in women
    Feb 22, 2025 · Defective XCI of more than 10% has an attributable risk of 40% among 12 different cancers from The Cancer Genome Atlas. Validations between ...
  87. [87]
    Altered X-chromosome inactivation in T cells may promote sex ...
    Apr 4, 2019 · Altered X-chromosome inactivation in T cells may promote sex-biased autoimmune diseases ... X-linked genes that escape XCI for T and B cell ...
  88. [88]
    A whole-organism landscape of X-inactivation in humans - eLife
    Jul 17, 2025 · During embryonic development in humans, female cells (46, XX) inactivate a single X-chromosome to balance the dosage of X-linked gene ...
  89. [89]
    X inactivation and somatic cell selection rescue female mice ... - PNAS
    Because of X chromosome inactivation, female offspring are mosaic for cells that express or lack GPI-linked proteins. Analysis of mosaic mice showed that in ...
  90. [90]
    Normal X-inactivation mosaicism in corneas of heterozygous Flna ...
    Aug 6, 2025 · The aim of this study was to determine whether X-inactivation mosaicism in the corneal epithelia of FlnaDilp2/+ mice was affected in any way ...
  91. [91]
    Stable X Chromosome Reactivation in Female Human Induced ...
    Jan 29, 2015 · To investigate whether reprogramming of female human fibroblasts into induced pluripotent stem cells (iPSCs) leads to reactivation of the inactive X chromosome ...
  92. [92]
    The interferon γ pathway enhances pluripotency and X-chromosome ...
    We discover that activation of the interferon γ (IFNγ) pathway early during reprogramming accelerates pluripotency acquisition and X-reactivation.
  93. [93]
    A shift from reversible to irreversible X inactivation is ... - PubMed - NIH
    To study the initiation of X inactivation, we have generated a full-length mouse Xist cDNA transgene and an inducible expression system facilitating controlled ...
  94. [94]
    Inducible XIST-dependent X-chromosome inactivation in human ...
    We now present a model system for human XIST RNA function in which induction of an XIST cDNA in somatic cells results in localized XIST RNA and transcriptional ...
  95. [95]
    Human XIST yeast artificial chromosome transgenes show partial X ...
    Initiation of X chromosome inactivation requires the presence, in cis, of the X inactivation center (XIC). The Xist gene, which lies within the XIC region ...
  96. [96]
    Gene reactivation upon X chromosome inactivation erosion in hiPSCs
    May 13, 2025 · Targeting XIST promoter region by CRISPR-Cas9 gene editing can restore XIST expression and overcome erosion; however, this depends on the ...
  97. [97]
    D-repeat in the XIST gene is required for X chromosome inactivation
    (C) CRISPR/Cas9 mediated D-repeat knockout resulted in the downregulated expression of XIST, which lead to the compromise of X-inactivation.Missing: papers | Show results with:papers
  98. [98]
    Revisiting the consequences of deleting the X inactivation center
    Jun 14, 2021 · This process, known as “X chromosome inactivation” (XCI), requires a master switch within the X inactivation center (Xic). The Xic spans several ...
  99. [99]
    Live-Cell Imaging and Functional Dissection of Xist RNA Reveal ...
    Oct 26, 2018 · We double-tagged Xist (inactivated X chromosome-specific transcript), a prototype long non-coding RNA pivotal for X chromosome inactivation (XCI).
  100. [100]
    Deletion of Xist repeat B disrupts cell cycle and asymmetric cell ...
    Mar 5, 2025 · During X chromosome inactivation (XCI), Xist RNA establishes silencing by coating the chromosome in cis and binding diverse proteins to promote ...
  101. [101]
    Live Cell Imaging of the Nascent Inactive X Chromosome during the ...
    Random X-chromosome inactivation ensures dosage compensation in mammals through the transcriptional silencing of one of the two X chromosomes present in ...Missing: techniques | Show results with:techniques
  102. [102]
    Single-cell analyses of X Chromosome inactivation dynamics and ...
    Our study sheds light on the dynamics of XCI progression and the asynchronicity between pluripotency, differentiation, and XCI.
  103. [103]
    X-chromosome inactivation in human iPSCs provides insight into X ...
    May 31, 2024 · The uncommon and variable reactivation of X chromosome genes in female hiPSCs can provide insight into X chromosome's role in regulating gene ...
  104. [104]
    X-linked clonality testing: interpretation and limitations - PMC - NIH
    We discuss clonality testing in general, emphasizing X-chromosome inactivation pattern (XCIP)–based assays and the pitfalls in their interpretation.
  105. [105]
    Inferring clonal somatic mutations directed by X chromosome ...
    Aug 9, 2024 · Here, we show that analysis of mitochondrial mutations in single cells is dramatically improved in females when using X chromosome inactivation to select ...
  106. [106]
    Competition shapes the landscape of X-chromosome-linked genetic ...
    Jul 26, 2024 · ... genes that escape X-inactivation in humans. Ninety percent of X chromosome pairs harbored at least 101 missense variants. This analysis ...
  107. [107]
    X-chromosome inactivation: New insights on its 60th anniversary
    This EMBO Workshop will bring together a diverse range of experts across the world working on different aspects of the X-inactivation process.
  108. [108]
    Modeling X-chromosome inactivation and reactivation during human ...
    Sep 30, 2025 · These organoid cultures display clonal X chromosome inactivation patterns previously described in the human placenta. We further demonstrate ...
  109. [109]
    Maintenance of X chromosome inactivation after T cell activation ...
    Oct 4, 2024 · For example, Cxcr3 escapes XCI in stimulated T cells during Leishmania infection and correlates with enhanced T cell effector function (25).<|control11|><|separator|>
  110. [110]
    Genetic and epigenetic determinants of reactivation of Mecp2 and ...
    Mar 8, 2022 · If Xist was knocked down with ASOs in combination with larger amounts of 5-Aza, reactivation was synergistic and 100-fold higher compared with ...
  111. [111]
    A mixed modality approach towards Xi reactivation for Rett ...
    Dec 27, 2017 · We develop a mixed modality approach that combines a small-molecule inhibitor of DNA methylation and an antisense oligonucleotide against Xist RNA.Missing: DNMT | Show results with:DNMT
  112. [112]
    X chromosome switch offers hope for girls with Rett syndrome
    Jul 23, 2025 · Researchers developed a promising gene therapy that reactivates healthy but silent genes and could treat Rett syndrome.
  113. [113]
    Recapitulation of Skewed X-Inactivation in Female Ornithine ...
    ... X-linked diseases such as ornithine transcarbamylase (OTC) deficiency (OMIM no. 311250) are also potentially curable by therapeutic modulation of the epigenome.
  114. [114]
    Selective Xi Reactivation and Alternative Methods to Restore ...
    X-reactivation may be achieved by drugging Xist RNA and DNA ... Xist RNA, DNA methylation, and histone hypoacetylation in maintaining X chromosome inactivation.
  115. [115]
    Xist condensates: perspectives for therapeutic intervention
    Jul 21, 2025 · Xist is a 15–17-kb long noncoding RNA (lncRNA) transcribed from the X-Inactivation Centre (XIC), a complex locus that determines the number and ...
  116. [116]
    Genetic analysis of the mouse X inactivation center defines an 80-kb ...
    In response to developmental cues, the Xic orchestrates events of X inactivation, including chromosome counting and choice, initiation, spread, and ...
  117. [117]
    Xist has properties of the X-chromosome inactivation centre - Nature
    Mar 20, 1997 · Xist is unusual in that it appears not to code for a protein but produces a nuclear RNA which colocalizes with the inactive X chromosome.Missing: discovery | Show results with:discovery