Hostname: page-component-848d4c4894-2pzkn Total loading time: 0 Render date: 2024-06-01T22:29:03.075Z Has data issue: false hasContentIssue false

X-Chromosome Inactivation and Related Diseases

Published online by Cambridge University Press:  01 January 2024

Zhuo Sun
Affiliation:
Xi’an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi’an Medical University, No. 1 XinWang Road, Weiyang District, Xi’an 710021, Shaanxi, China Institute of Basic Medical Sciences, Xi’an Medical University, No. 1 XinWang Road, 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 Road, Weiyang District, Xi’an 710021, Shaanxi, China
Yang Wang*
Affiliation:
Xi’an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi’an Medical University, No. 1 XinWang Road, Weiyang District, Xi’an 710021, Shaanxi, China
*
Correspondence should be addressed to Yang Wang; yang.wang@xiyi.edu.cn

Abstract

X-chromosome inactivation (XCI) is the form of dosage compensation in mammalian female cells to balance X-linked gene expression levels of the two sexes. Many diseases are related to XCI due to inactivation escape and skewing, and the symptoms and severity of these diseases also largely depend on the status of XCI. They can be divided into 3 types: X-linked diseases, diseases that are affected by XCI escape, and X-chromosome aneuploidy. Here, we review representative diseases in terms of their definition, symptoms, and XCI’s role in the pathogenesis of these diseases.

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 © 2022 Zhuo Sun et al.

1. X-Chromosome Inactivation, Escape, and Skewing

During XCI, many epigenetic events take place and ensure the compacted heterochromatin structure of the inactive X chromosome (X inactive, Xi) to silence most genes on the chromosome. XIST lncRNA is expressed and coats the Xi in cis [Reference Brown, Hendrich and Rupert1, Reference Brockdorff, Ashworth and Kay2], which starts a cascade of events including substitution of macroH2A [Reference Costanzi and Pehrson3], the removal of active histone modifications [Reference Jeppesen and Turner4, Reference Chadwick and Willard5], the addition of repressive histone modifications [Reference Boggs, Cheung, Heard, Spector, Chinault and Allis6Reference Silva, Mak and Zvetkova9], CpG islands methylation [Reference Pfeifer, Tanguay, Steigerwald and Riggs10], and heterochromatin protein recruitment [Reference Chadwick and Willard11, Reference Blewitt, Gendrel and Pang12]. As a result, the Xi is compacted into a rounder shape compared to the more flat structure of the active X chromosome (X active, Xa) [Reference Teller, Illner and Thamm13].

Not all genes on the Xi are in a repressed state. It is estimated that about 15%–30% of all the genes on the Xi escape gene repression and are expressed [Reference Carrel and Willard14, Reference Balaton and Brown15]. For different individuals, ages, and cell types, the gene escape patterns are diverse [Reference Carrel and Willard14, Reference Tukiainen, Villani and Villani16]. Some genes are found to be escaping in most cell types, others are highly variable depending on the origin of cells. The occurrence of escaped genes has been found to be essential in the pathogenesis of many diseases including autoimmune diseases [Reference Wang, Syrett, Kramer, Basu, Atchison and Anguera17] and cancer [Reference Pageau, Hall, Ganesan, Livingston and Lawrence18Reference Winham, Larson and Armasu21].

Skewed X-chromosome inactivation or X-chromosome inactivation skewing describes the phenomenon when more than 75% of cells in an individual chose the X chromosome from one parent as the Xi. This occurs since the choice of which X chromosome to silence is random and it takes place early in the gastrulation stage, so when the choice is at the tail end of the normal distribution, or if alleles of specific genes from one parent’s origin render the cells more robust, it may lead to skewing of the X-chromosome inactivation, instead of the 50% completely random choice [Reference Belmont22, Reference Migeon and Haisley-Royster23]. It is estimated that 1.5%–23% of females have skewed X inactivation [Reference Sharp, Robinson and Jacobs24Reference Lanasa, Hogge, Kubik, Blancato and Hoffman26]. The direction and degree of XCI skewing may influence the severity of some diseases including haemophilia B [Reference Orstavik, Orstavik and Schwartz27, Reference Okumura, Fujimori and Takagi28], dyskeratosis congenita [Reference Devriendt, Matthijs and Legius29], Duchenne muscular dystrophy [Reference Lupski, Garcia, Zoghbi, Hoffman and Fenwick30], myotubular myopathy [Reference Tanner, Ørstavik and Kristiansen31], and Fabry disease (FD) [Reference Morrone, Cavicchi and Bardelli32, Reference Dobrovolny, Dvorakova and Ledvinova33], which will be discussed more in detail in this review.

Many diseases have been found to be related to the XCI process. They can be roughly categorized to 3 types: (1) X-linked gene diseases, whose severity is greatly influenced by the direction and degree of X-inactivation skewing; here, we review FD that can be categorized to this type; (2) diseases that have higher occurrence in female population due to the presence of an extra pair of X chromosome and the possibilities to have escaped gene expression from the Xi: systemic lupus erythematosus (SLE) will be reviewed in this paper as an example; (3) X-chromosome aneuploidy: Turner syndrome (TS, 45, X), triple X syndrome (47, XXX), and Klinefelter syndrome (47, XXY) belong to this type. These diseases will be reviewed in terms of their clinical symptoms, pathogenic mechanism, and the role of XCI in the clinical presentation.

2. XCI-Related Diseases

2.1. X-Linked Diseases

For almost all X-linked diseases, clinical manifestation is more severe in males than in females. Female carriers are either asymptomatic or show milder phenotypes compared to males. Sex differences in these X-linked diseases are due to XCI [Reference Firth, Richards and Bevan34]. Males are hemizygous for most X-linked genes; thus, a male carrier of a mutant allele is usually affected with full presentation of the disease clinical features. Females have two types of cells in terms of their XCI status: those whose maternal X is active and those whose paternal X is active. Therefore, even if a female carries one copy of mutant allele, she probably still does not have full clinical presentation because a portion of the female cells has the mutant X inactivated. Data from OMIM show that there are more than 500 X-linked diseases that affect males more severely. X-linked retinitis pigmentosa [Reference Tsang and Sharma35, Reference Wang, Lu and Zhang36], Duchenne muscular dystrophy [Reference Abbadi, Philippe and Chery37], FD, and fragile X syndrome all belong to this category.

Among those X-linked diseases with higher male susceptibility, some rarely affect females, whereas others can present severe symptoms in female heterozygotes. There are mainly two factors behind whether female heterozygotes will have clinical manifestation: protein transfer and cell selection [Reference Migeon38]. Protein transfer is the process when variant cells could not make functional proteins but wildtype cells can transfer functional proteins into those deficient cells to make up for the loss. Cell selection is the process when there is a growth advantage for either the wildtype cells or mutant cells because of the mutant phenotype, the other cell population dies off gradually, making disease manifestation not evident or very severe, respectively.

Hunter syndrome and FD are both X-linked lysosomal enzyme diseases. Hunter syndrome rarely affects females [Reference Fratantoni, Hall and Neufeld39], whereas FD can present severe symptoms in female patients. The difference in female susceptibility lies in the different ability for cells to share the lysosomal enzyme [Reference Pinto, Vieira, Giugliani and Schwartz40]. The enzyme iduronic sulfatase loss in Hunter syndrome can be readily supplied by nearby wildtype cells, whereas the mature form of ɑ-galactosidase A (ɑ-GAL A) is hard to uptake for mutant cells in FD [Reference Beck and Cox41].

There are X-linked diseases that only or mainly affect females. Those disease variants usually cause loss of an essential protein completely, so that males are lethal in utero, leaving females to be the major sex to be afflicted with these diseases. Cornelia de Lange 2 (with SMC1A truncating variants) [Reference Dowsett, Porras and Kruszka42] and CHILD syndrome [Reference Kaminska-Winciorek, Brzezinska-Wcisło, Jezela-Stanek, Krajewska-Walasek, Cunningham and Herman43] only affect females since male carriers are lethal in utero. Other diseases affect some males due to a milder form of mutant or mosaic. Rett syndrome [Reference Jan, Dooley and Gordon44], incontinentia pigmenti type 2 [Reference Arenas-Sordo Mde, Vallejo-Vega, Hernández-Zamora, Gálvez-Rosas and Montoya-Pérez45], and focal dermal hypoplasia [Reference Bostwick, Van den Veyver, Sutton and Adam46] all belong to this type.

In the next part, FD will be discussed in detail as an example to show how XCI is involved in the pathogenesis of X-linked diseases. FD is caused by mutations in the GLA gene which codes for the ɑ-GAL A enzyme. ɑ-GAL A breaks down globotriaosylceramide and glycosphingolipids in the lysosome for recycling in cell metabolism, and decreased activity or loss of ɑ-GAL A leads to buildup of those molecules in the lysosome which can cause multisystemic effects in patients [Reference Sweeley and Klionsky47]. ɑ-GAL A is abundant in the kidney and vascular tissues, and key manifestation of FD includes malfunction of the kidney and heart. The estimated incidence of FD is 1 in 117,000 [Reference Meikle, Hopwood, Clague and Carey48].

The disease phenotype depends on residual enzyme activity: less than 1% of normal activity results in classic FD, and levels between 1% and 30% leads to atypical forms of FD (also called late-onset FD). Classic FD mainly affects males. Classic FD patients usually present symptoms early in childhood that include acute pain in extremities and fatigue, hypohidrosis [Reference Orteu, Jansen and Lidove49], neuropathic pain in the hands and feet, angiokeratomas in the lower abdomen and bathing trunk area [Reference Zampetti, Orteu and Antuzzi50], gastrointestinal problems, and cornea verticillata [Reference Sher, Letson and Desnick51, Reference Desnick, Brady and Barranger52]. Because of the residual ɑ-GAL A activity, atypical FD patients develop symptoms much later in life. Some develop multiple symptoms as young adults, while others only show signs of FD in specific organs such as the heart and kidneys. Heterozygous females have 0–100% of normal plasma ɑ-GAL A activity and can have symptoms that range from mild to severe depending on their skewing of XCI [Reference MacDermot, Holmes and Miners53].

Males are usually severely affected, whereas clinical presentation in female patients is more variable [Reference MacDermot, Holmes and Miners53, Reference MacDermot, Holmes and Miners54]. Females usually develop symptoms in their adulthood which is much later than males, and symptoms are usually milder [Reference Arends, Wanner and Hughes55]. This can lead to misdiagnosis in female patients. Since male has one copy of X chromosome, defect in the GLA gene can cause FD in males, whereas female has two X chromosomes and depending on X chromosome being inactivated and skewing of XCI, female mutant gene carriers can have a spectrum of clinical presentation from completely nonsymptomatic to severe symptoms as seen in males [Reference Echevarria, Benistan and Toussaint56].

In female FD patients, both random XCI and skewed XCI are observed. In a study that evaluated XCI pattern of four different tissues from female FD patients, random XCI is observed in 71% of samples and skewed XCI in 29% of samples [Reference Echevarria, Benistan and Toussaint56]. Other studies have found similar ratios [Reference Dobrovolny, Dvorakova and Ledvinova33, Reference Maier, Osterrieder and Whybra57]. For patients with random XCI, disease presentation usually worsens severely with age, which is partially due to inefficient protein transfer between wildtype and affected cells. For patients with skewed XCI, predominant expression of the mutant GLA allele usually results in early onset and rapid progression in FD, whereas the favored expression of the wildtype GLA allele is associated with mild phenotype and little progression over time.

For male patients, diagnosis can be confirmed by low ɑ-GAL A activity in leukocytes, whereas for female patients, ɑ-GAL A activity can range from very low to normal levels. Therefore, gene sequencing is the gold standard for diagnosis in females [Reference Zarate and Hopkin58]. It has been shown that level of ɑ-GAL A cannot predict the severity of symptoms well in female patients, and some researchers argue that the enzyme deficiency may result in other pathogenetic mechanisms [Reference Shen, Meng and Moore59Reference Tuttolomondo, Simonetta and Duro61].

It was believed that skewed XCI is the main reason for phenotype variability in female heterozygotes [Reference Morrone, Cavicchi and Bardelli32, Reference Dobrovolny, Dvorakova and Ledvinova33, Reference Echevarria, Benistan and Toussaint56, Reference Redonnet-Vernhet, Ploos van Amstel, Jansen, Wevers, Salvayre and Levade62], but recently some studies have come at the conclusions that skewed XCI could not explain all cases of severe FD and is not the main factor in the variable clinical presentation of FD in females [Reference Maier, Osterrieder and Whybra57, Reference Rossanti, Nozu and Fukunaga63, Reference Elstein, Schachamorov, Beeri and Altarescu64].

One factor that might have complicated the results is the tissues chosen for analyzing the XCI skewing. Most studies chose easily accessible leukocytes, urinary cells, and buccal epithelia, rather than the tissues from affected organs, such as cardiac and renal tissues. Also, XCI skewing pattern could be different in different tissues from the same subject [Reference Viggiano and Politano65]. Examining samples from affected tissues would give the best perspective on XCI skewing’s role in phenotype variability in female patients, but would require invasive biopsies.

Since some female patients with severe symptoms have random XCI, it points to factors other than XCI skewing that contribute to FD disease severity in heterozygous females. The nature of mutation could be essential in determining disease severity. Mutations that result in complete loss of the functional protein result in severe phenotypes, whereas missense mutations could result in mild phenotype or late-onset presentation. Even for family members with the same mutation, there could be vastly different clinical expression [Reference Tuttolomondo, Simonetta and Duro61, Reference Rigoldi, Concolino and Morrone66]. It is likely that besides XCI skewing and nature of mutation, the variability in female patients is dependent on other genetic, epigenetic, and environment factors as well [Reference Zarate and Hopkin58].

2.2. Diseases That Are Affected by XCI Escape or X-Chromosome Dosage Effect

Another type of diseases that are affected by XCI are due to X-inactivation escape. The escape and thus overexpression of some genes in female cells could be related to disease presentation and sex bias in those diseases. These include autoimmune diseases (SLE and autoimmune thyroid diseases [Reference Santiwatana, Mahachoklertwattana and Limwongse67, Reference Chabchoub, Uz and Maalej68]) and some psychiatric disorders (bipolar disorder and major depression [Reference Ji, Higa, Kelsoe and Zhou69]).

Here, SLE will be discussed in detail as an example to show how XCI is involved in the pathogenesis of diseases affected by XCI escape and X-chromosome dosage effect. Many autoimmune diseases have sex bias where number of female patients is significantly higher than that of male patients. Many factors contribute to the sex bias including difference in innate immunity, immune response intensity [Reference Jaillon, Berthenet and Garlanda70], and hormones [Reference Moulton71]. Many genetic risk loci have been discovered to be associated with SLE predisposition, some of which are X-linked. The extra pair of X chromosome, the possibility for these genes to escape, and a higher amount of the gene product in females than males contribute to sex bias of this disease.

SLE is a chronic autoimmune disease which leads to variable clinical presentations depending on the major organ affected. The word erythematosus refers to the rash that patients usually have on their skin. SLE incidence ranges from 2.2 to 23.1/100,000 person-years globally [Reference López, Mozo, Gutiérrez and Suárez72, Reference Feldman, Hiraki and Liu73], with the highest estimated incidence in North America. Women have higher prevalence of SLE [Reference Zhu, Liang and Liany74, Reference Jacob, Zhu and Armstrong75], and people of African ethnicity have higher incidence and prevalence than Caucasians [Reference Rees, Doherty, Grainge, Lanyon and Zhang76].

For people with predisposition for SLE, antinuclear antibodies are produced and form immune complexes with nuclear antigen, which then deposit in tissues and cause inflammation. Antibodies against red blood cells and white blood cells could also be produced and result in type II hypersensitivity. There are more than 80 genetic predispositions that have been discovered to date, such as TREX1 [Reference Namjou, Kothari and Kelly77], C8orf13-BLK [Reference Hom, Graham and Modrek78], ITGAM-ITGAX [Reference Hom, Graham and Modrek78], IL10 [Reference Gateva, Sandling and Hom79], TNIP1 [Reference Gateva, Sandling and Hom79], and IKZF1 [Reference Han, Zheng and Cui80] (the abovementioned gene loci are not X-linked). For most patients, SLE is caused by mutations in several genes rather than a single locus.

Besides genetic factors, there are also epigenetic factors that are essential in the disease pathogenesis. There is around 70%–75% discordance of SLE incidence between identical twins [Reference Stohl, Elliott, Hamilton, Deapen, Mack and Horwitz81, Reference Huang, Parfitt, Grennan and Manolios82]; this could be due to the different epigenetic landscape for these twins and also different X-inactivation pattern [Reference Kast83]. X-linked genes that are related to onset of SLE might be differentially inactivated among different cells, tissues, and individuals, contributing to the different onset of SLE in identical twins [Reference Stewart84].

Symptoms of SLE include fatigue, fever, painful joints, rashes (especially butterfly-shaped rash on the cheek), and sensitivity to sun [Reference Kiriakidou and Ching85]. Since there are a variety of general and specific symptoms, diagnosis of SLE is difficult. Patients are diagnosed by adding scores from 10 clinical domains: constitutional, cutaneous, arthritis, neurological, serositis, haematological, renal, antiphospholipid antibodies, complement proteins, and highly specific antibodies [Reference Dörner and Furie86]. SLE is characterized by periods of flare-ups and remittance, and treatments mainly involve immunomodulation and immunosuppression drugs and are targeted at preventing and limiting the severity of flare-ups.

Ratio of women with SLE to men is estimated to be 9 : 1 to 11 : 1. There could be several factors that contribute to the gender difference in disease susceptibility. These factors are hormones [Reference Laffont and Guéry87, Reference Sekigawa, Naito and Hira88], X-chromosome dosage factor, and X-linked gene overexpression.

It is shown that sex steroids can regulate autoimmune regulator (AIRE) locus expression, which in turn affects susceptibility to autoimmunity diseases [Reference Dragin, Bismuth and Cizeron-Clairac89, Reference Zhu, Bakhru and Conley90]. The fact that there is big increase of SLE incidence and prevalence in postpubertal females than males and prepubertal females also implies that sex hormones play an important role in the onset of SLE [Reference Rees, Doherty, Grainge, Davenport, Lanyon and Zhang91, Reference Arnaud, Fagot, Mathian, Paita, Fagot-Campagna and Amoura92].

The hypothesis that X-chromosome dosage is a contributing factor in SLE sex bias comes from the observations of SLE incidence in X-chromosome aneuploidies. Women with TS (45, X) are underrepresented compared to karyotypically normal women (46, XX) in SLE [Reference Cooney, Bruner and Aberle93]. The risk of SLE in Klinefelter syndrome (47, XXY) males is 14-fold higher than karyotypically normal men (46, XY) [Reference Scofield, Bruner and Namjou94]. The estimated prevalence of SLE in women with (47, XXX) is 2.5 times higher than women with normal karyotype and is 25 times higher than men [Reference Liu, Kurien and Zimmerman95], suggesting that dosage of X chromosome could be related to disease pathogenesis. Evidence from mouse models also indicates that an extra X chromosome is an important contributing factor in female bias in autoimmunity [Reference Whitacre96, Reference Smith-Bouvier, Divekar and Sasidhar97]. It is important to note that chromosome dosage factor could be in part due to X-linked gene escape [Reference Harris, Koelsch and Kurien98], which is the factor that is discussed next.

Overexpression of several X-linked genes has been shown to be involved in SLE pathogenesis, such as CD40LG [Reference Desai-Mehta, Lu, Ramsey-Goldman and Datta99], CXCR3 [Reference Oghumu, Varikuti and Stock100], KDM6A [Reference Itoh, Golden and Itoh101], CXorf21 [Reference Harris, Koelsch and Kurien98], MECP2 [Reference Sawalha, Webb and Han102, Reference Kaufman, Zhao and Kelly103], IRAK1 [Reference Jacob, Zhu and Armstrong75, Reference Kaufman, Zhao and Kelly103], TLR7(Toll-like receptor 7) [Reference Shen, Fu and Deng104, Reference García-Ortiz, Velázquez-Cruz, Espinosa-Rosales, Jiménez-Morales, Baca and Orozco105], GPR173 [Reference Zhang, Zhang and Wang106], and PRPS2. Several of them are immunity genes, including CD40LG, CXCR3, CXorf21, IRAK1, and TLR7. There could be 3 different scenarios of these X-linked gene overexpression, whether transcription from the active X allele is enhanced or there is escape from the inactive X allele or both. For the scenarios where XCI escape is involved in overexpression, it contributes to sex bias. Females by having the extra X chromosome have chances of XCI escape, whereas males do not.

Several studies have looked at the overexpression origin. TLR7 binds RNA in endosomes and activates the interferon response. TLR7 overexpression is observed in male patients, which is due to a SNP that increased transcription of the gene on the active X chromosome [Reference Shen, Fu and Deng104]. Another study found that there is biallelic expression of TLR7 in both female normal and SLE patient B cell lines, which means TLR7 is an escape even in healthy individuals [Reference Wang, Syrett, Kramer, Basu, Atchison and Anguera17]. The abovementioned studies show that TLR7 overexpression can come from both enhancing Xa allele expression and escape from Xi. CXorf21 is another immunity-related gene that shows XCI escape and has female-biased expression [Reference Harris, Koelsch and Kurien98]. These are two examples of X-linked immunity gene escape contributing to sex bias in SLE.

A few other of these X-linked genes show overexpression only in female SLE patients but not in males, such as CD40LG [Reference Lu, Wu, Tesmer, Ray, Yousif and Richardson107, Reference Hewagama, Gorelik and Patel108] and CXCR3 [Reference Hewagama, Gorelik and Patel108], which means the overexpression comes from the Xi, rather than Xa. Demethylation of the promoter region is also observed, which suggests that overexpression originates from XCI escape.

It has recently been observed that inactive X chromosome in SLE patient B cells have dramatic reduction in heterochromatic modifications, predisposing X-linked immunity gene escape [Reference Pyfrom, Paneru and Knox109]. This has provided further mechanistic insight as to how X-linked genes might contribute to SLE pathogenesis and sex bias.

2.3. X-Chromosome Aneuploidy

X-chromosome aneuploidy results in disease phenotypes in human: TS (45, X), Klinefelter syndrome (47, XXY), and triple X syndrome (47, XXX). For both Klinefelter syndrome and triple X syndrome, only one X chromosome remains active and all extra pairs of X chromosomes are inactivated. It is hypothesized that overexpression of escape genes results in the phenotypic abnormalities seen in those diseases [Reference Geschwind, Boone, Miller and Swerdloff110]. The expression would be lower in TS due to haploinsufficiency. Indeed, SHOX gene, which escapes XCI, has been associated with tall stature in Klinefelter syndrome and triple X syndrome and short stature in TS [Reference Ottesen, Aksglaede and Garn111, Reference Rao, Weiss and Fukami112]. On the other hand, increased expression is also observed for some X-linked genes with decreasing X-chromosome dosage [Reference Raznahan, Parikshak and Chandran113], indicating a compensatory mechanism in the complex relationship between X-chromosome dosage and X-linked gene expression level.

There is complex and diverse comorbidity associated with X-chromosome aneuploidy diseases, and identifying causal genes for different phenotypes has been difficult [Reference Trolle, Nielsen and Skakkebæk114]. However, recent studies of genome-wide DNA methylation profile and transcriptome in patients revealed that there is DNA hypermethylation associated with Klinefelter syndrome and DNA hypomethylation associated with TS, which also shed light upon several candidate genes [Reference Trolle, Nielsen and Skakkebæk114, Reference Skakkebæk, Nielsen and Trolle115].

3. Future Directions

X-linked diseases affect female patients differently resulting in a wide range of phenotype depending on their X-chromosome inactivation pattern. As seen in the review above, most of the studies mentioned are case studies and larger sample size could benefit exploration of the relationship between skewed X inactivation and phenotype severity. It is also important to note the cell type used when analyzing XCI skewing pattern, since it varies between different cell types even in the same individual. Also, it would be the best to analyze samples from affected organs to explore the relationship between XCI skewing and phenotype severity in female heterozygotes. Targeted reactivation of normal allele on the Xi could be further explored to support the development of more treatment options, as shown in studies in rodent models and cell lines [Reference Guy, Gan, Selfridge, Cobb and Bird116, Reference Carrette, Wang and Wei117].

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Acknowledgments

This work was supported by the PhD starting fund from Xi’an Medical University (no. 2020DOC14), funding from the Innovative Group of Xi’an Medical University (no. 2021TD01), Natural Science Basic Research Plan in Shaanxi Province of China (no. 2021JQ-776), Specialized Research Fund of Department of Education in Shaanxi Province (nos. 21JK0896 and 21JK0886), and National Natural Science Foundation of China (no. 32070069).

References

Brown, C. J., Hendrich, B. D., Rupert, J. L. et al., “The human XIST gene: analysis of a 17 kb inactive X-specific RNA that contains conserved repeats and is highly localized within the nucleus,Cell, vol. 71, no. 3, pp. 527542, 1992.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
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
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
Chadwick, B. P. and Willard, H. F., “Cell cycle-dependent localization of macroH2A in chromatin of the inactive X chromosome,Journal of Cell Biology, vol. 157, no. 7, pp. 11131123, 2002.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
Peters, A. H. F. M., Mermoud, J. E., O’Carroll, D. et al., “Histone H3 lysine 9 methylation is an epigenetic imprint of facultative heterochromatin,Nature Genetics, vol. 30, no. 1, pp. 7780, 2002.CrossRefGoogle ScholarPubMed
Plath, K., Fang, J., Mlynarczyk-Evans, S. K. et al., “Role of histone H3 lysine 27 methylation in X inactivation,Science, vol. 300, no. 5616, pp. 131135, 2003.CrossRefGoogle ScholarPubMed
Silva, J., Mak, W., Zvetkova, I. et al., “Establishment of histone H3 methylation on the inactive X chromosome requires transient recruitment of Eed-Enx1 polycomb group complexes,Developmental Cell, vol. 4, no. 4, pp. 481495, 2003.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
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
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
Balaton, B. P. and Brown, C. J., “Escape artists of the X chromosome,Trends in Genetics, vol. 32, no. 6, pp. 348359, 2016.CrossRefGoogle ScholarPubMed
Tukiainen, T., Villani, A. C., Villani, A.-C. et al., “Landscape of X chromosome inactivation across human tissues,Nature, vol. 550, no. 7675, pp. 244248, 2017.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 of the United States of America, 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., Lee, J. J., and Maeng, L.-S., “Dysregulation of X Chromosome inactivation in high grade ovarian serous adenocarcinoma,PloS One, vol. 10, no. 3, 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, 2019.CrossRefGoogle Scholar
Belmont, J. W., “Genetic control of X inactivation and processes leading to X-inactivation skewing,American Journal of Human Genetics, vol. 58, no. 6, pp. 11011108, 1996.Google ScholarPubMed
Migeon, B. R. and Haisley-Royster, C., “Familial skewed X inactivation and X-linked mutations: unbalanced X inactivation is a powerful means to ascertain X-linked genes that affect cell proliferation,The American Journal of Human Genetics, vol. 62, no. 6, pp. 15551557, 1998.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
Busque, L., Mio, R., Mattioli, J. et al., “Nonrandom X-inactivation patterns in normal females: lyonization ratios vary with age,Blood, vol. 88, no. 1, pp. 5965, 1996.CrossRefGoogle ScholarPubMed
Lanasa, M. C., Hogge, W. A., Kubik, C., Blancato, J., and Hoffman, E. P., “Highly skewed X-chromosome inactivation is associated with idiopathic recurrent spontaneous abortion,American Journal of Human Genetics, vol. 65, no. 1, pp. 252254, 1999.CrossRefGoogle ScholarPubMed
Orstavik, K. H., Orstavik, R. E., and Schwartz, M., “Skewed X chromosome inactivation in a female with haemophilia B and in her non-carrier daughter: a genetic influence on X chromosome inactivation?,Journal of Medical Genetics, vol. 36, no. 11, pp. 865866, 1999.Google Scholar
Okumura, K., Fujimori, Y., Takagi, A. et al., “Skewed X chromosome inactivation in fraternal female twins results in moderately severe and mild haemophilia B,Haemophilia, vol. 14, no. 5, pp. 10881093, 2008.CrossRefGoogle ScholarPubMed
Devriendt, K., Matthijs, G., Legius, E. et al., “Skewed X-chromosome inactivation in female carriers of dyskeratosis congenita,American Journal of Human Genetics, vol. 60, no. 3, pp. 581587, 1997.Google ScholarPubMed
Lupski, J. R., Garcia, C. A., Zoghbi, H. Y., Hoffman, E. P., and Fenwick, R. G., “Discordance of muscular dystrophy in monozygotic female twins: evidence supporting asymmetric splitting of the inner cell mass in a manifesting carrier of duchenne dystrophy,American Journal of Medical Genetics, vol. 40, no. 3, pp. 354364, 1991.CrossRefGoogle Scholar
Tanner, S. M., Ørstavik, K. H., Kristiansen, M. et al., “Skewed X-inactivation in a manifesting carrier of X-linked myotubular myopathy and in her non-manifesting carrier mother,Human Genetics, vol. 104, no. 3, pp. 249253, 1999.CrossRefGoogle Scholar
Morrone, A., Cavicchi, C., Bardelli, T. et al., “Fabry disease: molecular studies in Italian patients and X inactivation analysis in manifesting carriers,Journal of Medical Genetics, vol. 40, no. 8, p. e103, 2003.CrossRefGoogle Scholar
Dobrovolny, R., Dvorakova, L., Ledvinova, J. et al., “Relationship between X-inactivation and clinical involvement in fabry heterozygotes. eleven novel mutations in the α-galactosidase a gene in the czech and slovak population,Journal of Molecular Medicine, vol. 83, no. 8, pp. 647654, 2005.CrossRefGoogle ScholarPubMed
Firth, H. V., Richards, S. M., Bevan, A. P. et al., “DECIPHER: database of chromosomal imbalance and phenotype in humans using ensembl resources,American Journal of Human Genetics, vol. 84, no. 4, pp. 524533, 2009.CrossRefGoogle ScholarPubMed
Tsang, S. H. and Sharma, T., “X-linked retinitis pigmentosa,Advances in Experimental Medicine and Biology, vol. 1085, pp. 3135, 2018.CrossRefGoogle ScholarPubMed
Wang, Y., Lu, L., Zhang, D. et al., “A novel mutation of the RPGR gene in a Chinese X-linked retinitis pigmentosa family and possible involvement of X-chromosome inactivation,Eye, vol. 35, no. 6, pp. 16881696, 2021.CrossRefGoogle Scholar
Abbadi, N., Philippe, C., Chery, M. et al., “Additional case of female monozygotic twins discordant for the clinical manifestations of duchenne muscular dystrophy due to opposite X-chromosome inactivation,American Journal of Medical Genetics, vol. 52, no. 2, pp. 198206, 1994.CrossRefGoogle ScholarPubMed
Migeon, B. R., “X-linked diseases: susceptible females,Genetics in Medicine, vol. 22, no. 7, pp. 11561174, 2020.CrossRefGoogle ScholarPubMed
Fratantoni, J. C., Hall, C. W., and Neufeld, E. F., “Hurler and hunter syndromes: mutual correction of the defect in cultured fibroblasts,Science, vol. 162, no. 3853, pp. 570572, 1968.CrossRefGoogle ScholarPubMed
Pinto, L. L., Vieira, T. A., Giugliani, R., and Schwartz, I. V., “Expression of the disease on female carriers of X-linked lysosomal disorders: a brief review,Orphanet Journal of Rare Diseases, vol. 5, no. 1, p. 14, 2010.CrossRefGoogle ScholarPubMed
Beck, M. and Cox, T. M., “Comment: why are females with fabry disease affected?,Molecular Genetics and Metabolism Reports, vol. 21, Article ID 100529, 2019.CrossRefGoogle ScholarPubMed
Dowsett, L., Porras, A. R., Kruszka, P. et al., “Cornelia de Lange syndrome in diverse populations,American Journal of Medical Genetics, Part A, vol. 179, no. 2, pp. 150158, 2019.CrossRefGoogle ScholarPubMed
Kaminska-Winciorek, G., Brzezinska-Wcisło, L., Jezela-Stanek, A., Krajewska-Walasek, M., Cunningham, D., and Herman, G. E., “CHILD syndrome: clinical picture and diagnostic procedures,Journal of the European Academy of Dermatology and Venereology, vol. 21, no. 5, pp. 715716, 2007.CrossRefGoogle ScholarPubMed
Jan, M. M. S., Dooley, J. M., and Gordon, K. E., “Male Rett syndrome variant: application of diagnostic criteria,Pediatric Neurology, vol. 20, no. 3, pp. 238240, 1999.CrossRefGoogle ScholarPubMed
Arenas-Sordo Mde, L., Vallejo-Vega, B., Hernández-Zamora, E., Gálvez-Rosas, A., and Montoya-Pérez, L. A., “Incontinentia pigmenti (IP2): familiar case report with affected men. literature review,Medicina Oral, Patologia Oral y Cirugia Bucal, vol. 10, no. Suppl 2, pp. E122E129, 2005.Google ScholarPubMed
Bostwick, B., Van den Veyver, I. B., and Sutton, V. R., “Focal dermal hypoplasia,” in GeneReviews(®), Adam, M. P., Ed., University of Washington, Seattle, WA, USA, 1993.Google Scholar
Sweeley, C. C. and Klionsky, B., “FABRY’S disease: classification as a sphingolipidosis and partial characterization of a novel glycolipid,Journal of Biological Chemistry, vol. 238, pp. 31483150, 1963.CrossRefGoogle ScholarPubMed
Meikle, P. J., Hopwood, J. J., Clague, A. E., and Carey, W. F., “Prevalence of lysosomal storage disorders,JAMA, vol. 281, no. 3, pp. 249254, 1999.CrossRefGoogle ScholarPubMed
Orteu, C. H., Jansen, T., Lidove, O. et al., “Fabry disease and the skin: data from FOS, the fabry outcome survey,British Journal of Dermatology, vol. 157, no. 2, pp. 331337, 2007.CrossRefGoogle ScholarPubMed
Zampetti, A., Orteu, C. H., Antuzzi, D. et al., “Angiokeratoma: decision-making aid for the diagnosis of fabry disease,British Journal of Dermatology, vol. 166, no. 4, pp. 712720, 2012.CrossRefGoogle ScholarPubMed
Sher, N. A., Letson, R. D., and Desnick, R. J., “The ocular manifestations in fabry’s disease,Archives of Ophthalmology, vol. 97, no. 4, pp. 671676, 1979.CrossRefGoogle ScholarPubMed
Desnick, R. J., Brady, R., Barranger, J. et al., “Fabry disease, an under-recognized multisystemic disorder: expert recommendations for diagnosis, management, and enzyme replacement therapy,Annals of Internal Medicine, vol. 138, no. 4, pp. 338346, 2003.CrossRefGoogle ScholarPubMed
MacDermot, K. D., Holmes, A., and Miners, A. H., “Anderson-Fabry disease: clinical manifestations and impact of disease in a cohort of 60 obligate carrier females,Journal of Medical Genetics, vol. 38, no. 11, pp. 769775, 2001.CrossRefGoogle Scholar
MacDermot, K. D., Holmes, A., and Miners, A. H., “Anderson-Fabry disease: clinical manifestations and impact of disease in a cohort of 98 hemizygous males,Journal of Medical Genetics, vol. 38, no. 11, pp. 750760, 2001.CrossRefGoogle Scholar
Arends, M., Wanner, C., Hughes, D. et al., “Characterization of classical and nonclassical fabry disease: a multicenter study,Journal of the American Society of Nephrology, vol. 28, no. 5, pp. 16311641, 2017.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
Maier, E. M., Osterrieder, S., Whybra, C. et al., “Disease manifestations and X inactivation in heterozygous females with fabry disease,Acta Paediatrica (Oslo, Norway: 1992) Supplement, vol. 95, no. 451, pp. 3038, 2006.CrossRefGoogle Scholar
Zarate, Y. A. and Hopkin, R. J., “Fabry’s disease,Lancet, vol. 372, no. 9647, pp. 14271435, 2008.CrossRefGoogle ScholarPubMed
Shen, J.-S., Meng, X.-L., Moore, D. F. et al., “Globotriaosylceramide induces oxidative stress and up-regulates cell adhesion molecule expression in fabry disease endothelial cells,Molecular Genetics and Metabolism, vol. 95, no. 3, pp. 163168, 2008.CrossRefGoogle ScholarPubMed
Del Pinto, R. and Ferri, C., “The role of immunity in fabry disease and hypertension: a review of a novel common pathway,High Blood Pressure & Cardiovascular Prevention, vol. 27, no. 6, pp. 539546, 2020.CrossRefGoogle ScholarPubMed
Tuttolomondo, A., Simonetta, I., Duro, G. et al., “Inter-familial and intra-familial phenotypic variability in three sicilian families with Anderson-fabry disease,Oncotarget, vol. 8, no. 37, pp. 6141561424, 2017.CrossRefGoogle ScholarPubMed
Redonnet-Vernhet, I., Ploos van Amstel, J. K., Jansen, R. P., Wevers, R. A., Salvayre, R., and Levade, T., “Uneven X inactivation in a female monozygotic twin pair with fabry disease and discordant expression of a novel mutation in the alpha-galactosidase a gene,Journal of Medical Genetics, vol. 33, no. 8, pp. 682688, 1996.CrossRefGoogle Scholar
Rossanti, R., Nozu, K., Fukunaga, A. et al., “X-chromosome inactivation patterns in females with fabry disease examined by both ultra-deep RNA sequencing and methylation-dependent assay,Clinical and Experimental Nephrology, vol. 25, no. 11, pp. 12241230, 2021.CrossRefGoogle ScholarPubMed
Elstein, D., Schachamorov, E., Beeri, R., and Altarescu, G., “X-inactivation in fabry disease,Gene, vol. 505, no. 2, pp. 266268, 2012.CrossRefGoogle ScholarPubMed
Viggiano, E. and Politano, L., “X chromosome inactivation in carriers of fabry disease: review and meta-analysis,International Journal of Molecular Sciences, vol. 22, no. 14, 2021.CrossRefGoogle ScholarPubMed
Rigoldi, M., Concolino, D., Morrone, A. et al., “Intrafamilial phenotypic variability in four families with Anderson-fabry disease,Clinical Genetics, vol. 86, no. 3, pp. 258263, 2014.CrossRefGoogle ScholarPubMed
Santiwatana, S., Mahachoklertwattana, P., Limwongse, C. et al., “Skewed X chromosome inactivation in girls and female adolescents with autoimmune thyroid disease,Clinical Endocrinology, vol. 89, no. 6, pp. 863869, 2018.CrossRefGoogle ScholarPubMed
Chabchoub, G., Uz, E., Maalej, A. et al., “Analysis of skewed X-chromosome inactivation in females with rheumatoid arthritis and autoimmune thyroid diseases,Arthritis Research & Therapy, vol. 11, no. 4, p. R106, 2009.CrossRefGoogle ScholarPubMed
Ji, B., Higa, K. K., Kelsoe, J. R., and Zhou, X., “Over-expression of XIST, the master gene for X chromosome inactivation, in females with major affective disorders,EBioMedicine, vol. 2, no. 8, pp. 909918, 2015.CrossRefGoogle ScholarPubMed
Jaillon, S., Berthenet, K., and Garlanda, C., “Sexual dimorphism in innate immunity,Clinical Reviews in Allergy & Immunology, vol. 56, no. 3, pp. 308321, 2019.CrossRefGoogle ScholarPubMed
Moulton, V. R., “Sex hormones in acquired immunity and autoimmune disease,Frontiers in Immunology, vol. 9, p. 2279, 2018.CrossRefGoogle ScholarPubMed
López, P., Mozo, L., Gutiérrez, C., and Suárez, A., “Epidemiology of systemic lupus erythematosus in a northern Spanish population: gender and age influence on immunological features,Lupus, vol. 12, no. 11, pp. 860865, 2003.CrossRefGoogle Scholar
Feldman, C. H., Hiraki, L. T., Liu, J. et al., “Epidemiology and sociodemographics of systemic lupus erythematosus and lupus nephritis among US adults with Medicaid coverage, 2000-2004,Arthritis and Rheumatism, vol. 65, no. 3, pp. 753763, 2013.CrossRefGoogle ScholarPubMed
Zhu, Z., Liang, Z., Liany, H. et al., “Discovery of a novel genetic susceptibility locus on X chromosome for systemic lupus erythematosus,Arthritis Research & Therapy, vol. 17, no. 1, p. 349, 2015.CrossRefGoogle ScholarPubMed
Jacob, C. O., Zhu, J., Armstrong, D. L. et al., “Identification of IRAK1 as a risk gene with critical role in the pathogenesis of systemic lupus erythematosus,Proceedings of the National Academy of Sciences, vol. 106, no. 15, pp. 62566261, 2009.CrossRefGoogle ScholarPubMed
Rees, F., Doherty, M., Grainge, M. J., Lanyon, P., and Zhang, W., “The worldwide incidence and prevalence of systemic lupus erythematosus: a systematic review of epidemiological studies,Rheumatology, vol. 56, no. 11, pp. 19451961, 2017.CrossRefGoogle ScholarPubMed
Namjou, B., Kothari, P. H., Kelly, J. A. et al., “Evaluation of the TREX1 gene in a large multi-ancestral lupus cohort,Genes & Immunity, vol. 12, no. 4, pp. 270279, 2011.CrossRefGoogle Scholar
Hom, G., Graham, R. R., Modrek, B. et al., “Association of systemic lupus erythematosus withC8orf13-BLKandITGAM-ITGAX,New England Journal of Medicine, vol. 358, no. 9, pp. 900909, 2008.CrossRefGoogle Scholar
Gateva, V., Sandling, J. K., Hom, G. et al., “A large-scale replication study identifies TNIP1, PRDM1, JAZF1, UHRF1BP1 and IL10 as risk loci for systemic lupus erythematosus,Nature Genetics, vol. 41, no. 11, pp. 12281233, 2009.CrossRefGoogle ScholarPubMed
Han, J.-W., Zheng, H.-F., Cui, Y. et al., “Genome-wide association study in a Chinese Han population identifies nine new susceptibility loci for systemic lupus erythematosus,Nature Genetics, vol. 41, no. 11, pp. 12341237, 2009.CrossRefGoogle Scholar
Stohl, W., Elliott, J. E., Hamilton, A. S., Deapen, D. M., Mack, T. M., and Horwitz, D. A., “Impaired recovery and cytolytic function of CD56+T and non-T cells in systemic lupus erythematosus following in vitro polyclonal T cell stimulation. Studies in unselected patients and monozygotic disease-discordant twins,Arthritis & Rheumatism, vol. 39, no. 11, pp. 18401851, 1996.CrossRefGoogle ScholarPubMed
Huang, Q., Parfitt, A., Grennan, D. M., and Manolios, N., “X-chromosome inactivation in monozygotic twins with systemic lupus erythematosus,Autoimmunity, vol. 26, no. 2, pp. 8593, 1997.CrossRefGoogle ScholarPubMed
Kast, R. E., “Predominance of autoimmune and rheumatic diseases in females,Journal of Rheumatology, vol. 4, no. 3, pp. 288292, 1977.Google ScholarPubMed
Stewart, J. J., “The female X-inactivation mosaic in systemic lupus erythematosus,Immunology Today, vol. 19, no. 8, pp. 352357, 1998.CrossRefGoogle ScholarPubMed
Kiriakidou, M. and Ching, C. L., Systemic Lupus Erythematosus. Annals of Internal Medicine, vol. 172, no. 11, pp. Itc81itc96, 2020.CrossRefGoogle Scholar
Dörner, T. and Furie, R., “Novel paradigms in systemic lupus erythematosus,Lancet (North American Edition), vol. 393, no. 10188, pp. 23442358, 2019.Google ScholarPubMed
Laffont, S. and Guéry, J.-C., “Deconstructing the sex bias in allergy and autoimmunity: from sex hormones and beyond,Advances in Immunology, vol. 142, pp. 3564, 2019.CrossRefGoogle ScholarPubMed
Sekigawa, I., Naito, T., Hira, K. et al., “Possible mechanisms of gender bias in SLE: a new hypothesis involving a comparison of SLE with atopy,Lupus, vol. 13, no. 4, pp. 217222, 2004.CrossRefGoogle ScholarPubMed
Dragin, N., Bismuth, J., Cizeron-Clairac, G. et al., “Estrogen-mediated downregulation of AIRE influences sexual dimorphism in autoimmune diseases,Journal of Clinical Investigation, vol. 126, no. 4, pp. 15251537, 2016.CrossRefGoogle ScholarPubMed
Zhu, M.-L., Bakhru, P., Conley, B. et al., “Sex bias in CNS autoimmune disease mediated by androgen control of autoimmune regulator,Nature Communications, vol. 7, no. 1, Article ID 11350, 2016.CrossRefGoogle ScholarPubMed
Rees, F., Doherty, M., Grainge, M., Davenport, G., Lanyon, P., and Zhang, W., “The incidence and prevalence of systemic lupus erythematosus in the UK, 1999-2012,Annals of the Rheumatic Diseases, vol. 75, no. 1, pp. 136141, 2016.CrossRefGoogle ScholarPubMed
Arnaud, L., Fagot, J.-P., Mathian, A., Paita, M., Fagot-Campagna, A., and Amoura, Z., “Prevalence and incidence of systemic lupus erythematosus in France: a 2010 nation-wide population-based study,Autoimmunity Reviews, vol. 13, no. 11, pp. 10821089, 2014.CrossRefGoogle Scholar
Cooney, C. M., Bruner, G. R., Aberle, T. et al., “46, X, del (X) (q13) Turner’s syndrome women with systemic lupus erythematosus in a pedigree multiplex for SLE,Genes & Immunity, vol. 10, no. 5, pp. 478481, 2009.CrossRefGoogle Scholar
Scofield, R. H., Bruner, G. R., Namjou, B. et al., “Klinefelter’s syndrome (47, XXY) in male systemic lupus erythematosus patients: support for the notion of a gene-dose effect from the X chromosome,Arthritis & Rheumatism, vol. 58, no. 8, pp. 25112517, 2008.CrossRefGoogle ScholarPubMed
Liu, K., Kurien, B. T., Zimmerman, S. L. et al., “X chromosome dose and sex bias in autoimmune diseases: increased prevalence of 47, XXX in systemic lupus erythematosus and sjögren’s syndrome,Arthritis & Rheumatology, vol. 68, no. 5, pp. 12901300, 2016.CrossRefGoogle ScholarPubMed
Whitacre, C. C., “Sex differences in autoimmune disease,Nature Immunology, vol. 2, no. 9, pp. 777780, 2001.CrossRefGoogle ScholarPubMed
Smith-Bouvier, D. L., Divekar, A. A., Sasidhar, M. et al., “A role for sex chromosome complement in the female bias in autoimmune disease,Journal of Experimental Medicine, vol. 205, no. 5, pp. 10991108, 2008.CrossRefGoogle ScholarPubMed
Harris, V. M., Koelsch, K. A., Kurien, B. T. et al., “Characterization of cxorf21 provides molecular insight into female-bias immune response in SLE pathogenesis,Frontiers in Immunology, vol. 10, p. 2160, 2019.CrossRefGoogle ScholarPubMed
Desai-Mehta, A., Lu, L., Ramsey-Goldman, R., and Datta, S. K., “Hyperexpression of CD40 ligand by B and T cells in human lupus and its role in pathogenic autoantibody production,Journal of Clinical Investigation, vol. 97, no. 9, pp. 20632073, 1996.CrossRefGoogle Scholar
Oghumu, S., Varikuti, S., Stock, J. C. et al., “Cutting edge: CXCR3 escapes X chromosome inactivation in T cells during infection: potential implications for sex differences in immune responses,Journal of Immunology, vol. 203, no. 4, pp. 789794, 2019.CrossRefGoogle Scholar
Itoh, Y., Golden, L. C., Itoh, N. et al., “The X-linked histone demethylase Kdm6a in CD4+ T lymphocytes modulates autoimmunity,Journal of Clinical Investigation, vol. 129, no. 9, pp. 38523863, 2019.CrossRefGoogle ScholarPubMed
Sawalha, A. H., Webb, R., Han, S. et al., “Common variants within MECP2 confer risk of systemic lupus erythematosus,PloS One, vol. 3, no. 3, Article ID e1727, 2008.CrossRefGoogle ScholarPubMed
Kaufman, K. M., Zhao, J., Kelly, J. A. et al., “Fine mapping of Xq28: bothMECP2 and IRAK1contribute to risk for systemic lupus erythematosus in multiple ancestral groups,Annals of the Rheumatic Diseases, vol. 72, no. 3, pp. 437444, 2013.CrossRefGoogle ScholarPubMed
Shen, N., Fu, Q., Deng, Y. et al., “Sex-specific association of X-linked Toll-like receptor 7 (TLR7) with male systemic lupus erythematosus,Proceedings of the National Academy of Sciences, vol. 107, no. 36, pp. 1583815843, 2010.CrossRefGoogle ScholarPubMed
García-Ortiz, H., Velázquez-Cruz, R., Espinosa-Rosales, F., Jiménez-Morales, S., Baca, V., and Orozco, L., “Association of TLR7 copy number variation with susceptibility to childhood-onset systemic lupus erythematosus in Mexican population,Annals of the Rheumatic Diseases, vol. 69, no. 10, pp. 18611865, 2010.CrossRefGoogle ScholarPubMed
Zhang, H., Zhang, Y., Wang, Y.-F. et al., “Meta-analysis of GWAS on both Chinese and European populations identifies GPR173 as a novel X chromosome susceptibility gene for SLE,Arthritis Research & Therapy, vol. 20, no. 1, p. 92, 2018.CrossRefGoogle ScholarPubMed
Lu, Q., Wu, A., Tesmer, L., Ray, D., Yousif, N., and Richardson, B., “Demethylation of CD40LG on the inactive X in T cells from women with lupus,Journal of Immunology, vol. 179, no. 9, pp. 63526358, 2007.CrossRefGoogle Scholar
Hewagama, A., Gorelik, G., Patel, D. et al., “Overexpression of X-linked genes in T cells from women with lupus,Journal of Autoimmunity, vol. 41, pp. 6071, 2013.CrossRefGoogle Scholar
Pyfrom, S., Paneru, B., Knox, J. J. et al., “The dynamic epigenetic regulation of the inactive X chromosome in healthy human B cells is dysregulated in lupus patients,Proceedings of the National Academy of Sciences of the United States of America, vol. 118, no. 24, 2021.Google Scholar
Geschwind, D. H., Boone, K. B., Miller, B. L., and Swerdloff, R. S., “Neurobehavioral phenotype of klinefelter syndrome,Mental Retardation and Developmental Disabilities Research Reviews, vol. 6, no. 2, pp. 107116, 2000.3.0.CO;2-2>CrossRefGoogle ScholarPubMed
Ottesen, A. M., Aksglaede, L., Garn, I. et al., “Increased number of sex chromosomes affects height in a nonlinear fashion: a study of 305 patients with sex chromosome aneuploidy,American Journal of Medical Genetics, Part A, vol. 152a, no. 5, pp. 12061212, 2010.CrossRefGoogle Scholar
Rao, E., Weiss, B., Fukami, M. et al., “Pseudoautosomal deletions encompassing a novel homeobox gene cause growth failure in idiopathic short stature and turner syndrome,Nature Genetics, vol. 16, no. 1, pp. 5463, 1997.CrossRefGoogle ScholarPubMed
Raznahan, A., Parikshak, N. N., Chandran, V. et al., “Sex-chromosome dosage effects on gene expression in humans,Proceedings of the National Academy of Sciences, vol. 115, no. 28, pp. 73987403, 2018.CrossRefGoogle ScholarPubMed
Trolle, C., Nielsen, M. M., Skakkebæk, A. et al., “Widespread DNA hypomethylation and differential gene expression in Turner syndrome,Scientific Reports, vol. 6, no. 1, Article ID 34220, 2016.CrossRefGoogle ScholarPubMed
Skakkebæk, A., Nielsen, M. M., Trolle, C. et al., “DNA hypermethylation and differential gene expression associated with klinefelter syndrome,Scientific Reports, vol. 8, no. 1, Article ID 13740, 2018.CrossRefGoogle ScholarPubMed
Guy, J., Gan, J., Selfridge, J., Cobb, S., and Bird, A., “Reversal of neurological defects in a mouse model of rett syndrome,Science, vol. 315, no. 5815, pp. 11431147, 2007.CrossRefGoogle Scholar
Carrette, L. L. G., Wang, C. Y., Wei, C. et al., “A mixed modality approach towards Xi reactivation for Rett syndrome and other X-linked disorders,Proceedings of the National Academy of Sciences of the United States of America, vol. 115, no. 4, pp. E668e675, 2018.Google ScholarPubMed