scispace - formally typeset
Search or ask a question

Showing papers on "Skewed X-inactivation published in 1997"


Journal ArticleDOI
TL;DR: The results indicate that the Xist RNA is required for female dosage compensation but plays no role in spermatogenesis.
Abstract: The X-linked Xist gene encodes a large untranslated RNA that has been implicated in mammalian dosage compensation and in spermatogenesis. To investigate the function of the Xist gene product, we have generated male and female mice that carry a deletion in the structural gene but maintain a functional Xist promoter. Mutant males were healthy and fertile. Females that inherited the mutation from their mothers were also normal and had the wild-type paternal X chromosome inactive in every cell. In contrast to maternal transmission, females that carry the mutation on the paternal X chromosome were severely growth-retarded and died early in embryogenesis. The wild-type maternal X chromosome was inactive in every cell of the growth-retarded embryo proper, whereas both X chromosomes were expressed in the mutant female trophoblast where X inactivation is imprinted. However, an XO mouse with a paternally inherited Xist mutation was healthy and appeared normal. The imprinted lethal phenotype of the mutant females is therefore due to the inability of extraembryonic tissue with two active X chromosomes to sustain the embryo. Our results indicate that the Xist RNA is required for female dosage compensation but plays no role in spermatogenesis.

695 citations


Journal ArticleDOI
05 Sep 1997-Cell
TL;DR: It is suggested that these developmentally modulated changes in Xist expression are regulated by several different mechanisms: factors that stabilize Xist transcripts at the Xi, an activity that blocks this stabilization at the Xa, and a mechanism that silences low level Xists expression from theXa.

308 citations


Journal ArticleDOI
TL;DR: A rare cytosine to guanine mutation in the XIST minimal promoter that underlies both epigenetic and functional differences between the two X chromosomes in nine females from two unrelated families is reported, suggesting that there is an association between alterations in the regulation of XIST expression and X-chromosome inactivation.
Abstract: X-chromosome inactivation is the process by which a cell recognizes the presence of two copies of an X chromosome early in the development of XX embryos and chooses one to be active and one to be inactive. Although it is commonly believed that the initiation of X inactivation is random, with an equal probability (50:50) that either X chromosome will be the inactive X in a given cell, significant variation in the proportion of cells with either X inactive is observed both in mice heterozygous for alleles at the Xce locus and among normal human females in the population. Families in which multiple females demonstrate extremely skewed inactivation patterns that are otherwise quite rare in the general population are thought to reflect possible genetic influences on the X-inactivation process. Here we report a rare cytosine to guanine mutation in the XIST minimal promoter that underlies both epigenetic and functional differences between the two X chromosomes in nine females from two unrelated families. All females demonstrate preferential inactivation of the X chromosome carrying the mutation, suggesting that there is an association between alterations in the regulation of XIST expression and X-chromosome inactivation.

278 citations


Journal ArticleDOI
20 Mar 1997-Nature
TL;DR: E ectopic Xic action resulted in chromosome-wide changes that are characteristic of the Xi: DNA replication was delayed, and histone H4 was markedly hypoacetylated, which suggest long-range cis effects on the autosome similar to those of X inactivation.
Abstract: In mammals, the X chromosome is unique in being capable of complete inactivation. Such X inactivation evolved to compensate for gene dosage differences between females with two X chromosomes and males with one. Transcriptional silencing of a single female X chromosome is controlled in cis by Xist, whose RNA product coats the inactive X chromosome (Xi), and the X-inactivation centre (Xic). A transgenic study limited the Xic to 450 kilobases including Xist, and demonstrated that it is sufficient to initiate X inactivation. Here we report that ectopic Xist RNA completely coats transgenic chromosome 12. Expression of genes over 50 centimorgans was reduced two-fold and was detected only from the normal homologue in fibroblasts. Moreover, ectopic Xic action resulted in chromosome-wide changes that are characteristic of the X(i): DNA replication was delayed, and histone H4 was markedly hypoacetylated. Our findings suggest long-range cis effects on the autosome similar to those of X inactivation, and imply that the Xic can both initiate X inactivation and drive heterochromatin formation. Thus, the potential for chromosome-wide gene regulation is not intrinsic to X-chromosome DNA, but can also occur on autosomes possessing the Xic.

275 citations


Journal ArticleDOI
03 Oct 1997-Cell
TL;DR: It is demonstrated that increased stability of Xist RNA is the primary determinant of developmental up-regulation and discusses the implications of these findings in terms of models for the initiation of random and imprinted X inactivation.

262 citations


Journal ArticleDOI
20 Mar 1997-Nature
TL;DR: Xist, introduced onto an autosome, is sufficient by itself for inactivation in cis and that Xist RNA becomes localized close to the autosome into which the gene is integrated, suggesting that elements required for some aspects of chromosome counting are contained within the construct.
Abstract: X-chromosome inactivation is the process by which female mammals (with two X chromosomes) achieve expression of X-chromosomal genes equivalent to that of males (one X and one Y chromosome)1,2. This results in the transcriptional silencing of virtually all genes on one of the X chromosomes in female somatic cells. X-chromosome inactivation has been shown to act in cis and to initiate and spread from a single site on the X chromosome known as the X-inactivation centre (Xic)2,3. The Xic has been localized to a 450-kilobase region of the mouse X chromosome4. The Xist gene also maps to this region and is expressed exclusively from the inactive X chromosome3–7. Xist is unusual in that it appears not to code for a protein but produces a nuclear RNA which colocalizes with the inactive X chromosome4,8. The creation of a null allele of Xist in embryonic stem cells has demonstrated that this gene is required for X inactivation to occur in cis9. Here we show that Xist, introduced onto an autosome, is sufficient by itself for inactivation in cis and that Xist RNA becomes localized close to the autosome into which the gene is integrated. In addition, the presence of autosomal Xist copies leads to activation of the endogeneous Xist gene in some cells, suggesting that elements required for some aspects of chromosome counting are contained within the construct. Thus the Xist gene exhibits properties of the X-inactivation centre.

244 citations


Journal ArticleDOI
TL;DR: Examining the expression of 33 X- linked genes in eight mouse/human somatic-cell hybrids suggests that at least some X-linked genes may be under additional levels of epigenetic regulation not previously recognized and that somatic -cell hybrids may provide a useful approach for studying these chromosomal phenomena.
Abstract: X-chromosome inactivation results in the cis-limited inactivation of many, but not all, of the genes on one of the pair of X chromosomes in mammalian females. In addition to the genes from the pseudoautosomal region, which have long been anticipated to escape inactivation, genes from several other regions of the human X chromosome have now been shown to escape inactivation and to be expressed from both the active and inactive X chromosomes. The growing number of genes escaping inactivation emphasizes the need for a reliable system for assessing the inactivation status of X-linked genes. Since many features of the active or inactive X chromosome, including transcriptional activity, are maintained in rodent/human somatic-cell hybrids, such hybrids have been used to study the inactivation process and to determine the inactivation status of human X-linked genes. In order to assess the fidelity of inactivation status in such hybrids, we have examined the expression of 33 X-linked genes in eight mouse/human somatic-cell hybrids that contain either the human active (three hybrids) or inactive X (five hybrids) chromosome. Inactivation of nine of these genes had previously been demonstrated biochemically in human cells, and the expression of these genes only in hybrids retaining an active X, but not in those retaining an inactive X, confirms that expression in hybrids reflects expression in human cells. Although the majority of genes tested showed consistent patterns of expression among the active X hybrids or inactive X hybrids, surprisingly, 5 of the 33 genes showed heterogeneous expression among the hybrids, demonstrating a significantly higher rate of variability than previously reported for other genes in either human somatic cells or mouse/human somatic-cell hybrids. These data suggest that at least some X-linked genes may be under additional levels of epigenetic regulation not previously recognized and that somatic-cell hybrids may provide a useful approach for studying these chromosomal phenomena.

190 citations


Journal Article
TL;DR: The finding that normal arteries may have large X inactivation patches raises the possibility that plaque monoclonality may arise by expanding a pre-existing clone of cells rather than generating a new clone by mutation or selection.
Abstract: Atherosclerotic plaques contain a large monoclonal population of cells. Monoclonality could arise by somatic mutation, selection of a pre-existing lineage, or expansion of a pre-existing (developmental) clone. To determine the monoclonal cell type in plaque and learn when monoclonality arises, we studied X chromosome inactivation patterns using methylation of the X-linked human androgen receptor gene. Assays based on polymerase chain reaction were performed on samples of known cellular composition, microdissected from histological sections of human arteries. In atherosclerotic vessels, the majority of medial samples (7/11 coronary and 2/3 aortic) showed balanced (paternal and maternal) patterns of X inactivation, indicating polyclonality. In contrast, most samples of plaque smooth muscle cells showed a single pattern of X inactivation (3/4 aortic plaques and 9/11 coronary plaques; P < 0.01 versus media), indicating that plaque smooth muscle cells are monoclonal. Samples of plaque containing inflammatory or endothelial cells showed balanced X inactivation, also demonstrating polyclonality. Multiple plaques from a given patient showed no bias toward one allele, indicating there was no X-linked selection of cells during plaque growth. To determine whether plaques might arise from pre-existing clones (large X inactivation patches), we then studied 10 normal coronaries with diffuse intimal thickening. Six of the ten coronaries showed skewed X inactivation patterns in normal media and intima, suggesting the patch size in normal arteries is surprisingly large. Thus, smooth muscle cells constitute the monoclonal population in atherosclerotic plaques. The finding that normal arteries may have large X inactivation patches raises the possibility that plaque monoclonality may arise by expanding a pre-existing clone of cells rather than generating a new clone by mutation or selection.

185 citations


Journal ArticleDOI
TL;DR: The first report of X inactivation patterns in purified subpopulations of blood cells in normal elderly women is reported and raises the possibility that clonal blood cell patterns may precede rather than follow mutations which subsequently give rise to myelodysplastic or myeloproliferative phenotypes.
Abstract: Studies of X chromosome inactivation patterns are central to many aspects of our understanding of the pathogenesis of haematological malignancies. In patients with myeloproliferative disorders and myelodysplastic syndromes the demonstration of skewed X inactivation patterns in multiple haemopoietic lineages has been taken to indicate a stem cell origin for these groups of diseases. However, stem cell depletion or selection pressures can also produce skewed X inactivation patterns and might increase with age. We have therefore used the HUMARA assay to study X inactivation patterns of elderly patients with myeloproliferative disorders together with an age-matched control group of normal elderly women. A clonal pattern (clonal granulocytes and polyclonal T cells) was observed in 23.1% of normal women and 63.4% of patients with myeloproliferative disorders. This is the first report of X inactivation patterns in purified subpopulations of blood cells in normal elderly women. These results have three significant implications. Firstly, the finding of clonal granulocytes and polyclonal T cells in normal elderly women is likely to reflect age-related stem cell depletion or selection pressures. Secondly, the demonstration of clonal granulocytes and polyclonal T cells is not a useful diagnostic marker for myeloproliferative disorders or myelodysplastic syndromes in elderly women. Thirdly, our data raise the possibility that clonal blood cell patterns may precede rather than follow mutations which subsequently give rise to myelodysplastic or myeloproliferative phenotypes.

166 citations


Journal ArticleDOI
TL;DR: This is the first gene-mapping study of abnormalities of X-inactivation patterns and the first association of a specific locus for recurrent spontaneous abortion in a cytogenetically normal family.
Abstract: We report a family ascertained for molecular diagnosis of muscular dystrophy in a young girl, in which preferential activation (> or = 95% of cells) of the paternal X chromosome was seen in both the proband and her mother. To determine the molecular basis for skewed X inactivation, we studied X-inactivation patterns in peripheral blood and/or oral mucosal cells from 50 members of this family and from a cohort of normal females. We found excellent concordance between X-inactivation patterns in blood and oral mucosal cell nuclei in all females. Of the 50 female pedigree members studied, 16 showed preferential use (> or = 95% cells) of the paternal X chromosome; none of 62 randomly selected females showed similarly skewed X inactivation was maternally inherited in this family. A linkage study using the molecular trait of skewed X inactivation as the scored phenotype localized this trait to Xq28 (DXS1108; maximum LOD score [Zmax] = 4.34, recombination fraction [theta] = 0). Both genotyping of additional markers and FISH of a YAC probe in Xq28 showed a deletion spanning from intron 22 of the factor VIII gene to DXS115-3. This deletion completely cosegregated with the trait (Zmax = 6.92, theta = 0). Comparison of clinical findings between affected and unaffected females in the 50-member pedigree showed a statistically significant increase in spontaneous-abortion rate in the females carrying the trait (P < .02). To our knowledge, this is the first gene-mapping study of abnormalities of X-inactivation patterns and is the first association of a specific locus for recurrent spontaneous abortion in a cytogenetically normal family. The involvement of this locus in cell lethality, cell-growth disadvantage, developmental abnormalities, or the X-inactivation process is discussed.

132 citations


Journal ArticleDOI
TL;DR: A methylation difference near a polymorphism in the X-linked androgen-receptor gene was used to investigate the possibility that nonrandom X inactivation is increases in fetuses and newborns that are associated with confined placental mosaicism (CPM) involving an autosomal trisomy.
Abstract: The inactivation of one X chromosome in females is normally random with regard to which X is inactivated. However, exclusive or almost-exclusive inactivation of one X may be observed in association with some X-autosomal rearrangements, mutations of the XIST gene, certain X-linked diseases, and MZ twinning. In the present study, a methylation difference near a polymorphism in the X-linked androgen-receptor gene was used to investigate the possibility that nonrandom X inactivation is increases in fetuses and newborns that are associated with confined placental mosaicism (CPM) involving an autosomal trisomy. Extreme skewing was observed in 7 (58%) of 12 cases with a meiotic origin of the trisomy, but in none of 10 cases examined with a somatic origin of the trisomy, and in only 1 (4%) of 27 control adult females. In addition, an extremely skewed X-inactivation pattern was observed in 3 of 10 informative cases of female uniparental disomy (UPD) of chromosome 15. This may reflect the fact that a proportion of UPD cases arise by "rescue" of a chromosomally abnormal conceptus and are therefore associated with CPM. A skewed pattern of X inactivation in CPM cases is hypothesized to result from a reduction in the size of the early-embryonic cell pool, because of either poor early growth or subsequent selection against the trisomic cells. Since approximately 2% of pregnancies detected by chorionic villus sampling are associated with CPM, this is likely a significant contributor to both skewed X inactivation observed in the newborn population and the expression of recessive X-linked diseases in females.

01 Jan 1997
TL;DR: In this article, an extremely skewed X-inactiva-tion pattern was observed in 3 of 10 informative cases offemaleuniparental disomy (UPD)ofchromosome15.
Abstract: Summary IntroductionThe inactivation of one Xchromosome in females isnormallyrandomwithregardtowhichXis inactivated.However, exclusive or almost-exclusive inactivation ofone Xmay be observed in association with some X-autosomal rearrangements, mutations ofthe XIST gene,certainX-linked diseases, andMZ twinning. Inthepres-ent study, a methylationdifference neara polymorphism in the X-linked androgen-receptor genewasusedto in-vestigate the possibility that nonrandomXinactivationis increases in fetuses andnewbornsthat are associatedwith confined placental mosaicism (CPM)involving anautosomal trisomy. Extreme skewing wasobservedin 7(58%) of 12 cases with a meiotic origin ofthe trisomy,butin noneof10cases examinedwithasomaticoriginof the trisomy, and in only 1 (4%) of27control adultfemales. In addition, an extremely skewed X-inactiva-tion pattern was observed in 3 of 10 informative casesoffemaleuniparental disomy (UPD)ofchromosome15.Thismayreflect thefact that aproportionofUPDcasesarise by "rescue" of a

Journal Article
TL;DR: It is concluded that X inactivation allows the excessive gain of X chromosomes found in germ-cell-derived tumors of the adult testis and offers an interesting model to study the fundamental mechanisms of these processes.
Abstract: In female mammalian cells, inactivation of one of the X chromosomes compensates the increased dosage of X-linked genes as compared with their male counterparts This process is initiated by the X-inactive specific transcripts of the xist/XIST gene in cis, resulting in methylation of specific sites of genes to be silenced However, in male germ cells, X inactivation is established by xist/XIST expression only We investigated the X inactivation pattern in human testicular tumors of different histogenesis by analysis of XIST expression and methylation of the androgen receptor gene XIST was expressed only in tumors derived from the germ cell lineage with supernumerical X chromosomes: seminomas, nonseminomas, and spermatocytic seminomas Although low expression was present in testicular parenchyma with spermatogenesis, XIST was expressed at a higher level in parenchyma with carcinoma in situ, the precursor lesion of seminomas and nonseminomas Despite the consistent expression of XIST in germ-cell-derived tumors with gain of X chromosomes, methylation of the androgen receptor gene was present in all differentiated but only in a proportion of the undifferentiated nonseminomas This differential pattern of methylation was also found in a number of representative cell lines Our data indicate that the counting mechanism resulting in X inactivation is functional in testicular cancers of different histogenesis Moreover, the differentiation-dependent pattern of X inactivation as reported during normal development in the case of multiple X chromosomes by methylation is retained in these tumors We conclude therefore that X inactivation allows the excessive gain of X chromosomes found in germ-cell-derived tumors of the adult testis In addition, this offers an interesting model to study the fundamental mechanisms of these processes

Journal Article
TL;DR: The case of a karyotypically normal female affected with DMD as a result of homozygosity for a deletion of exon 50 of the dystrophin gene is reported, demonstrating that uniparental isodisomy of the X chromosome is an additional mechanism for the expression of X-linked recessive disorders.
Abstract: Duchenne muscular dystrophy (DMD) is a severe, progressive, X-linked muscle-wasting disorder with an incidence of approximately 1/3,500 male births. Females are also affected, in rare instances. The manifestation of mild to severe symptoms in female carriers of dystrophin mutations is often the result of the preferential inactivation of the X chromosome carrying the normal dystrophin gene. The severity of the symptoms is dependent on the proportion of cells that have inactivated the normal X chromosome. A skewed pattern of X inactivation is also responsible for the clinical manifestation of DMD in females carrying X;autosome translocations, which disrupt the dystrophin gene. DMD may also be observed in females with Turner syndrome (45,X), if the remaining X chromosome carries a DMD mutation. We report here the case of a karyotypically normal female affected with DMD as a result of homozygosity for a deletion of exon 50 of the dystrophin gene. PCR analysis of microsatellite markers spanning the length of the X chromosome demonstrated that homozygosity for the dystrophin gene mutation was caused by maternal isodisomy for the entire X chromosome. This finding demonstrates that uniparental isodisomy of the X chromosome is an additional mechanism for the expression of X-linked recessive disorders. The proband's clinical presentation is consistent with the absence of imprinted genes (i.e., genes that are selectively expressed based on the parent of origin) on the X chromosome.

Journal Article
TL;DR: A critical role for the DC gene in bone marrow-cell and fibroblast-cell proliferation is suggested and highly skewed X inactivation was observed in white blood cells, cultured skin fibroblasts, and buccal mucosa from female carriers of DC in this family.
Abstract: In this study, we report on a family with X-linked dyskeratosis congenita (DC). Linkage analysis with markers in the factor VIII gene at Xq28 yielded a LOD score of 2 at a recombination of 0. Clinical manifestations of DC, such as skin lesions following the Blaschko lines, were present in two obligate carrier females. Highly skewed X inactivation was observed in white blood cells, cultured skin fibroblasts, and buccal mucosa from female carriers of DC in this family. This suggests a critical role for the DC gene in bone marrow-cell and fibroblast-cell proliferation.

Journal ArticleDOI
TL;DR: The expression of the human homologue, XIST, in human preimplantation embryos from the 5- to 10-cell stage onwards consistent with its role in the initiation of inactivation is reported.
Abstract: In the somatic cells of female mammals, either the maternally or paternally derived X chromosome (X(M) or X(P)) is randomly inactivated to achieve dosage compensation for X-linked genes. In early mouse development, however, selective inactivation of X(P) occurs first in extraembryonic lineages at the blastocyst stage around the time of implantation before later random inactivation in the embryonic ectoderm from which the fetus is derived. Xist, a gene mapping to the X-inactivation centre (Xic), is exclusively expressed from the inactive X-chromosome and is thought to be involved in the initiation of X-inactivation. Consistent with this, Xist is first expressed at the 4-to 8-cell stages, prior to functional inactivation at the blastocyst stage, exclusively from X(P) in female embryos. This also suggests that genomic imprinting may influence the earliest expression of Xist resulting in selective inactivation of X(P) and a candidate methylation site in the promoter region has recently been described. Here we report the expression of the human homologue, XIST, in human preimplantation embryos from the 5- to 10-cell stage onwards consistent with its role in the initiation of inactivation. In contrast to the mouse, however, transcripts were detected in both male and female embryos demonstrating XIST expression from the X(M) in male embryos (X(M)Y).

Journal ArticleDOI
TL;DR: The mechanism of X inactivation in somatic cells of mammalian females, resulting in male/female dosage compensation, has been recruited from the Xist-activated chromosome condensation machinery that developed in male meiosis earlier in evolution.
Abstract: In eutherian mammals, X inactive-specific transcripts (Xist) are expressed in somatic cells possessing more than one X chromosome, and in germline cells of males, in which the single X chromosome is transcriptionally inactive. In early meiosis of males the sex chromosomes form an inactive XY nuclear compartment (XY body). We show by in situ reverse-transcribed polymerase chain reaction that Xist RNA is concentrated in the XY body. This fine localization suggests that Xist RNA is involved in inactivation of the male X chromosome, and that it has spreading capability, not only in cis but also in a quasi-cis mode, to juxtaposed non-X chromosomes. A hypothetical scheme links the evolution of heteromorphic sex chromosomes to the development of X condensation/inactivation in the male. The mechanism of X inactivation in somatic cells of mammalian females, resulting in male/female dosage compensation, has been recruited from the Xist-activated chromosome condensation machinery that developed in male meiosis earlier in evolution.

Journal ArticleDOI
TL;DR: The highly skewed X-inactivation pattern of the grandmother suggests that her NDI phenotype is caused by dominant methylation of the normal allele of V2R gene.
Abstract: Most cases of congenital nephrogenic diabetes insipidus (NDI) are inherited in an X-linked manner, which is due to the mutations of the vasopressin type 2 receptor (V2R) gene. However, recent reports have presented female NDI patients with heterozygote V2R gene mutations. The mechanism of inheritance was thought to be skewed X-inactivation. We present a family with congenital NDI. Three male members were diagnosed with NDI, and examination of their V2R gene revealed a G inserted at nucleotide 804 of the open reading frame. Three female individuals display different degrees of symptoms of NDI, and all of them possess both the normal and abnormal genes. The X-inactivation patterns of the female members were investigated via the detection of methylated trinucleotide repeat in the human androgen receptor gene. The grandmother showed extremely skewed methylation of one X chromosome, and the mother revealed moderately skewed methylation. The daughter of the grandmother's sister, who has no symptoms of NDI, showed random methylation. The highly skewed X-inactivation pattern of the grandmother suggests that her NDI phenotype is caused by dominant methylation of the normal allele of V2R gene.

Journal ArticleDOI
15 Sep 1997-Blood
TL;DR: Investigation of the pattern of X-inactivation in the peripheral blood of carriers of DC using the methylation-sensitive Hpa II site in the androgen receptor gene (HUMARA) indicates that, in the hematopoiesis of heterozygous females, cells expressing the normal DC allele have a growth advantage over cells that express the mutant allele.

Journal ArticleDOI
TL;DR: It is concluded that there is a differential germline strategy for maintenance of X-inactivation and that H4 underacetylation, though associated with the long-term marking of inactive X chromosomes in the female soma, is not always essential for the transcriptional down-regulation ofX-linked genes.

Journal ArticleDOI
TL;DR: Differences are seen in the evolution of the sex chromosome locations of IL9R and SYBL1, which may be correlated with the longer history of X linkage, and at a more centromeric chromosomal position during evolution.
Abstract: The recently discovered second pseudoautosomal region (XqPAR) contains at least two genes, IL9R and SYBL1. Recent findings show that, like XpPAR genes, IL9R escapes X inactivation and its Y allele is also expressed, but SYBL1 seems to act like an X-linked gene, expressed from the active X chromosome but not from the inactive X or Y. Here we show that differences are also seen in the evolution of the sex chromosome locations of IL9R and SYBL1. IL9R is known to be autosomal in mice, and is X-linked only in primates. SYBL1, however, has been found to be on the X chromosome in all mammals tested, from marsupials to humans. Both genes were duplicated on the Y homologue of the terminal portion of the X chromosome during the evolution of Homo sapiens from other higher primates. The inactivation pattern of SYBL1 may be correlated with its longer history of X linkage, and at a more centromeric chromosomal position during evolution; the more recent X linkage and more telomeric position of the IL9R gene may explain its autosomal, ‘uninactivated’ transcriptional status.

Journal ArticleDOI
TL;DR: Findings in a girl with Hunter disease of the severe type suggest that the severe form of Hunter disease in this girl was the result of selective expression of the maternal allele carrying the missense mutation R468Q, which in turn resulted from skewed X inactivation of the paternal nonmutant X chromosome.
Abstract: Hunter disease is an X-linked recessive mucopolysaccharide storage disorder caused by iduronate-2-sulfatase deficiency and is rare in females. We describe here findings in a girl with Hunter disease of the severe type. She had a normal karyotype but a marked deficiency of iduronate-2-sulfatase activity in lymphocytes and cultured fibroblasts. In a sequence analysis of the iduronate-2-sulfatase gene, evidence was obtained for the R468Q (G1403 to A) mutation, a common one in Hunter disease. RT-PCR showed her cDNA to represent only the R468Q allele, although at the genomic level she was a heterozygote with one normal allele. Her brother had the R468Q mutation, and their mother was a carrier of this mutation. The fusion products of CHO (TG(R),Neo(R)) with patient's fibroblasts cultured in HAT/G418 selective medium, carried only the maternal allele. However, in genomic DNA from the patient's fibroblasts, only the paternal allele of the androgen receptor gene, a gene subjected to differential methylation of the inactive X-chromosome, was methylated. These findings strongly suggest that the severe form of Hunter disease in this girl was the result of selective expression of the maternal allele carrying the missense mutation R468Q, which in turn resulted from skewed X inactivation of the paternal nonmutant X chromosome.

Journal ArticleDOI
TL;DR: Mouse EC cells used in this study were capable of altering methylation status of the human XIST gene in a manner lacking consistency and unable to repress its transcription, and failed to obtain any positive evidence for the occurrence of X chromosome inactivation in differentiating monochromosome EC hybrids.

Journal ArticleDOI
TL;DR: The results showed an unexpected random X-inactivation pattern in these patients, unlike most other known X-linked disorders inherited in this fashion, Aicardi syndrome patients have a normal (i.e., random) X- inactivation pattern.
Abstract: Aicardi syndrome is a rare disorder characterized by absent corpus callosum, infantile spasms, and chorioretinal lacunae. It is sporadic in nature and affects only females, resulting in severe mental and physical handicap. It has been suggested that the disease is caused by a dominant X-linked mutation which occurs de novo in females, and is lethal in hemizygous male embryos. This mode of inheritance has been observed in a number of other rare syndromes. In these syndromes, when X inactivation is studied, a non-random pattern is usually found. We have studied the X inactivation pattern in ten female patients with Aicardi syndrome and their parents using the highly polymorphic, differentially methylated androgen receptor gene. The results showed an unexpected random X-inactivation pattern in these patients. Previous clinical and cytogenetic evidence suggests that Aicardi syndrome is caused by an X-linked dominant mutation, de novo in females and lethal in males. However, unlike most other known X-linked disorders inherited in this fashion, Aicardi syndrome patients have a normal (i.e., random) X-inactivation pattern. A number of possible explanations is proposed for this apparently contradictory evidence.

Journal ArticleDOI
TL;DR: It is shown, using an X-linked lacZ transgenic mouse line, that loss of beta-galactosidase activity occurs in certain somatic cells of the developing male urogenital ridge, which indicates that sex determination might also be influenced by a gene-dosage compensation mechanism, in addition to a dominant action of the Sry gene.
Abstract: In the mouse, the activity of Sry (sex-determining gene on the Y chromosome) initiates the transformation of the indifferent gonad into a testis. In humans, a partial Xp21 duplication leads to the development of ovaries instead of testes in XY individuals. This observation indicates that sex determination might also be influenced by a gene-dosage compensation mechanism, in addition to a dominant action of the Sry gene. In female mammals, the regulation of X-linked gene dosage at early embryogenesis is achieved through the inactivation of one of the two X chromosomes. Here we have investigated the possibility that inactivation of the X chromosome may play a role in male sex determination. We have shown, using an X-linked lacZ transgenic mouse line, that loss of beta-galactosidase activity occurs in certain somatic cells of the developing male urogenital ridge. When changes associated with apoptosis of mesonephric tubules in the developing urogenital ridges are taken into account, expression of the Xist (X inactive specific transcript) gene correlates with X inactivation revealed by loss of beta-galactosidase activity in very early mesonephric tubule epithelial cells, gonadal interstitial mesenchymal cells and coelomic epithelial cells.

Journal ArticleDOI
TL;DR: This clinical case shows that a somatic and germline mosaicism for a single-gene disorder led to an unusual pattern of X-linked inheritance in the family, and all three daughters in thefamily died of OTC deficiency.
Abstract: A Leu148Phe substitution of the ornithine transcarbamylase (OTC) gene was identified in a 2-year-old girl with OTC deficiency (14% of control). Her two elder sisters died in childhood of hyperammonemia, and the patient also died of OTC deficiency. Enzyme activity in Cos1 cells transfected by the mutant cDNA was undetectable, thereby indicating a definite pathogenic mutation. Familial gene analysis showed that the mother had wild-type OTC alleles on both X-chromosomes and the father was a mosaic for the mutant allele in his lymphocytes and spermatozoa. This clinical case shows that a somatic and germline mosaicism for a single-gene disorder led to an unusual pattern of X-linked inheritance in the family, and all three daughters in the family died of OTC deficiency. The possibility that inherited factors will lead to skewed X-inactivation needs to be considered.

Journal ArticleDOI
TL;DR: There was an inverse correlation between loss of heterozygosity (LOH) for chromosome 12 and inactivation of the p16ink4 gene, which may suggest that a leukaemogenic event on chromosome 12p affects the same pathway of cell‐cycle control as p16inks4 inactivation.
Abstract: Seventy-four patients with acute lymphoblastic leukaemia (ALL) were analysed with limited allelotyping to detect loss of heterozygosity on chromosome segments 6q, 9p, 12p and 13q in order to detect patterns of genetic alteration. In the case of chromosome 9, analyses were also performed to detect inactivation of the p15ink4B and p16ink4 genes by Southern blot and sequencing techniques. The deletion data from these chromosomes were correlated to each other and to clinical features including prognosis. Allelic loss of these chromosomal regions could be detected in 24% (6q), 15% (12p) and 10% (13q) of the patients respectively, whereas aberrations involving 9p were detected in approximately 50% of the cases. There was an inverse correlation between loss of heterozygosity (LOH) for chromosome 12 and inactivation of the p16ink4 gene. This finding may suggest that a leukaemogenic event on chromosome 12p affects the same pathway of cell-cycle control as p16ink4 inactivation or, alternatively, reflects the fact that these mutations tend to occur in cells of different lineages.

Journal ArticleDOI
TL;DR: Analysis of X inactivation patterns in manifesting carriers of familial and sporadic Duchenne muscular dystrophy or unaffected carriers of DMD by analysis of the methylation of HpaII sites in the first exon of the human androgen-receptor gene (HUMARA) from peripheral blood samples suggests that in carriers ofDMD, both affected and unaffected, it is valuable to analyze the pattern of skewed X in activation because it provides important prognostic information.

01 Jan 1997
TL;DR: Differential histone H4 acetylation between the X chromosomes of a female marsupial, Macropus eugenii, is demonstrated, indicating that histone underacetylation was a feature of dosage compensation in a common mammalian ancestor, and is likely to play a central role in X chromosome inactivation in all mammals.
Abstract: Underacetylation of histone H4 is thought tobe involved in the molecular mechanism of mammalian Xchromosome inactivation, which is an important model sys-tem for large-scale genetic control in eukaryotes. However, ithas not been established whether histone underacetylationplays a critical role in the multistep inactivation pathway.Here we demonstrate differential histone H4 acetylation be-tween the X chromosomes of a female marsupial, Macropuseugenii . Histone underacetylation is the only molecular aspectof X inactivation known to be shared by marsupial andeutherian mammals. Its strong evolutionary conservationimplies that, unlike DNA methylation, histone underacetyla-tion was a feature of dosage compensation in a commonmammalian ancestor, and is therefore likely to play a centralrole in X chromosome inactivation in all mammals. The inactive X hypothesis states that genes on all but a singleX chromosome are inactivated in the somatic cells of femalemammals (1). Inactivation results in transcriptional repression(2) and serves to equalize X chromosome gene expressionbetween male (XY) and female (XX) mammals. The inactiveX chromosome (Xi) is late replicating and forms a hetero-chromatic body (sex chromatin) on the periphery of thenucleus (3). In eutherian (‘‘placental’’) mammals, inactivationoccurs at random in early embryogenesis and involves countingof X chromosomes, initiation, spreading, and maintenance ofthe inactive state (4). X inactivation involves changes to DNA(5), and it is well established that cytosine methylation isinvolved in the maintenance of X inactivation in humans andmice (6, 7), although it is not the only, or necessarily theprimary, change (8, 9).