scispace - formally typeset
Search or ask a question

Showing papers on "Dosage compensation published in 2002"


Journal ArticleDOI
TL;DR: The authors are on the threshold of discovering the factors that regulate and interact with Xist to control X-inactivation, and closer to an understanding of the molecular mechanisms that underlie this complex process.
Abstract: ▪ Abstract Dosage compensation in mammals is achieved by the transcriptional inactivation of one X chromosome in female cells. From the time X chromosome inactivation was initially described, it was clear that several mechanisms must be precisely integrated to achieve correct regulation of this complex process. X-inactivation appears to be triggered upon differentiation, suggesting its regulation by developmental cues. Whereas any number of X chromosomes greater than one is silenced, only one X chromosome remains active. Silencing on the inactive X chromosome coincides with the acquisition of a multitude of chromatin modifications, resulting in the formation of extraordinarily stable facultative heterochromatin that is faithfully propagated through subsequent cell divisions. The integration of all these processes requires a region of the X chromosome known as the X-inactivation center, which contains the Xist gene and its cis-regulatory elements. Xist encodes an RNA molecule that plays critical roles in t...

463 citations


Journal ArticleDOI
TL;DR: Analysis of MAOA expression in bovine placentae from natural reproduction revealed imprinted XCI with preferential inactivation of the paternal X chromosome, and incomplete nuclear reprogramming may generate abnormal epigenetic marks on the X chromosomes of cloned cattle.
Abstract: In mammals, epigenetic marks on the X chromosomes are involved in dosage compensation. Specifically, they are required for X chromosome inactivation (XCI), the random transcriptional silencing of one of the two X chromosomes in female cells during late blastocyst development. During natural reproduction, both X chromosomes are active in the female zygote. In somatic-cell cloning, however, the cloned embryos receive one active (Xa) and one inactive (Xi) X chromosome from the donor cells. Patterns of XCIhave been reported normal in cloned mice, but have yet to be investigated in other species. We examined allele-specific expression of the X-linked monoamine oxidase type A (MAOA) gene and the expression of nine additional X-linked genes in nine cloned XX calves. We found aberrant expression patterns in nine of ten X-linked genes and hypomethylation of Xist in organs of deceased clones. Analysis of MAOA expression in bovine placentae from natural reproduction revealed imprinted XCI with preferential inactivation of the paternal X chromosome. In contrast, we found random XCI in placentae of the deceased clones but completely skewed XCI in that of live clones. Thus, incomplete nuclear reprogramming may generate abnormal epigenetic marks on the X chromosomes of cloned cattle, affecting both random and imprinted XCI.

314 citations



Journal ArticleDOI
TL;DR: Males can be rescued by roX cDNAs from autosomal transgenes, demonstrating the genetic separation of the chromatin entry and RNA‐encoding functions.
Abstract: The roX1 and roX2 genes of Drosophila produce male‐specific non‐coding RNAs that co‐localize with the Male‐Specific Lethal (MSL) protein complex. This complex mediates up‐regulation of the male X chromo some by increasing histone H4 acetylation, thus contributing to the equalization of X‐linked gene expression between the sexes. Both roX genes overlap two of ∼35 chromatin entry sites, DNA sequences proposed to act in cis to direct the MSL complex to the X chromosome. Although dosage compensation is essential in males, an intact roX1 gene is not required by either sex. We have generated flies lacking roX2 and find that this gene is also non‐essential. However, simultaneous removal of both roX RNAs causes a striking male‐specific reduction in viability accompanied by relocation of the MSL proteins and acetylated histone H4 from the X chromosome to autosomal sites and heterochromatin. Males can be rescued by roX cDNAs from autosomal transgenes, demonstrating the genetic separation of the chromatin entry and RNA‐encoding functions. Therefore, the roX1 and roX2 genes produce redundant, male‐specific lethal transcripts required for targeting the MSL complex.

251 citations


Journal ArticleDOI
TL;DR: It is shown that blocking H4K16 acetylation suppresses the X chromosome defects resulting from loss of ISWI function in males and directly counteracts chromatin compaction mediated by the ISWI ATPase.
Abstract: Mutations in Drosophila ISWI, a member of the SWI2/SNF2 family of chromatin remodeling ATPases, alter the global architecture of the male X chromosome. The transcription of genes on this chromosome is increased 2-fold relative to females due to dosage compensation, a process involving the acetylation of histone H4 at lysine 16 (H4K16). Here we show that blocking H4K16 acetylation suppresses the X chromosome defects resulting from loss of ISWI function in males. In contrast, the forced acetylation of H4K16 in ISWI mutant females causes X chromosome defects indistinguishable from those seen in ISWI mutant males. Increased expression of MOF, the histone acetyltransferase that acetylates H4K16, strongly enhances phenotypes resulting from the partial loss of ISWI function. Peptide competition assays revealed that H4K16 acetylation reduces the ability of ISWI to interact productively with its substrate. These findings suggest that H4K16 acetylation directly counteracts chromatin compaction mediated by the ISWI ATPase.

244 citations


Journal ArticleDOI
TL;DR: It is shown that Eed and a second Polycomb group protein, Enx1, are directly localized to the inactive X chromosome in XX trophoblast stem (TS) cells, suggesting a mechanism for the maintenance of imprinted X inactivation in these cells.

228 citations


Journal ArticleDOI
TL;DR: In the house fly, Sex-lethal is not involved in sex determination, and dosage compensation, if existent at all, is not coupled with sexual differentiation, which allows for more adaptive plasticity in the housefly system.
Abstract: The genetic cascades regulating sex determination of the housefly, Musca domestica, and the fruitfly, Drosophila melanogaster, appear strikingly different. The bifunctional switch gene doublesex, however, is present at the bottom of the regulatory cascades of both species, and so is transformer-2, one of the genetic elements required for the sex-specific regulation of doublesex. The upstream regulators are different: Drosophila utilizes Sex-lethal to coordinate the control of sex determination and dosage compensation, i.e., the process that equilibrates the difference of two X chromosomes in females versus one X chromosome in males. In the housefly, Sex-lethal is not involved in sex determination, and dosage compensation, if existent at all, is not coupled with sexual differentiation. This allows for more adaptive plasticity in the housefly system. Accordingly, natural housefly populations can vary greatly in their mechanism of sex determination, and new types can be generated in the laboratory.

132 citations


Journal ArticleDOI
22 Nov 2002-Science
TL;DR: MSL protein abundance is defined as a determinant of whether the MSL complex will spread in cis from an autosomalroX transgene and a model in which MSL proteins assemble into active complexes by binding nascent roXtranscripts is suggested.
Abstract: The untranslated roX1 and roX2 RNAs are components of the Drosophila male-specific lethal (MSL) complex, which modifies histones to up-regulate transcription of the male X chromosome. roX genes are normally located on the X chromosome, and roX transgenes can misdirect the dosage compensation machinery to spread locally on other chromosomes. Here we define MSL protein abundance as a determinant of whether the MSL complex will spread in cis from an autosomal roX transgene. The number of expressed roX genes in a nucleus was inversely correlated with spreading from roX transgenes. We suggest a model in which MSL proteins assemble into active complexes by binding nascent roX transcripts. When MSL protein/ roX RNA ratios are high, assembly will be efficient, and complexes may be completed while still tethered to the DNA template. We propose that this local production of MSL complexes determines the extent of spreading into flanking chromatin.

131 citations


Journal ArticleDOI
TL;DR: It is demonstrated that three TTN genes encode chromosome scaffold proteins of the condensin (SMC2) and cohesin (sMC1 and SMC3) classes, which have been studied extensively in yeast and animal systems and should provide clues to chromosome mechanics in plants and insights into the regulation of nuclear activity during endosperm development.
Abstract: The titan (ttn) mutants of Arabidopsis exhibit striking alterations in chromosome dynamics and cell division during seed development. Endosperm defects include aberrant mitoses and giant polyploid nuclei. Mutant embryos differ in cell size, morphology and viability, depending on the locus involved. Here we demonstrate that three TTN genes encode chromosome scaffold proteins of the condensin (SMC2) and cohesin (SMC1 and SMC3) classes. These proteins have been studied extensively in yeast and animal systems, where they modulate chromosome condensation, chromatid separation, and dosage compensation. Arabidopsis contains single copies of SMC1 and SMC3 cohesins. We used forward genetics to identify duplicate T-DNA insertions in each gene. These mutants (ttn7 and ttn8) have similar titan phenotypes: giant endosperm nuclei and arrested embryos with a few small cells. A single SMC2 knockout (ttn3) was identified and confirmed by molecular complementation. The weak embryo phenotype observed in this mutant may result from expression of a related gene (AtSMC2) with overlapping functions. Further analysis of titan mutants and the SMC gene family in Arabidopsis should provide clues to chromosome mechanics in plants and insights into the regulation of nuclear activity during endosperm development.

128 citations


Journal ArticleDOI
TL;DR: It is shown that human TSIX antisense transcripts are unable to repress XIST, and serves as a mutant for mouse Tsix, providing insights into features responsible for antisense activity in imprinted X inactivation.
Abstract: Transcriptional silencing of the human inactive X chromosome is induced by the XIST gene within the human X-inactivation center. The XIST allele must be turned off on one X chromosome to maintain its activity in cells of both sexes. In the mouse placenta, where X inactivation is imprinted (the paternal X chromosome is always inactive), the maternal Xist allele is repressed by a cis-acting antisense transcript, encoded by the Tsix gene. However, it remains to be seen whether this antisense transcript protects the future active X chromosome during random inactivation in the embryo proper. We recently identified the human TSIX gene and showed that it lacks key regulatory elements needed for the imprinting function of murine Tsix. Now, using RNA FISH for cellular localization of transcripts in human fetal cells, we show that human TSIX antisense transcripts are unable to repress XIST. In fact, TSIX is transcribed only from the inactive X chromosome and is coexpressed with XIST. Also, TSIX is not maternally imprinted in placental tissues, and its transcription persists in placental and fetal tissues, throughout embryogenesis. Therefore, the repression of Xist by mouse Tsix has no counterpart in humans, and TSIX is not the gene that protects the active X chromosome from random inactivation. Because human TSIX cannot imprint X inactivation in the placenta, it serves as a mutant for mouse Tsix, providing insights into features responsible for antisense activity in imprinted X inactivation.

126 citations


Journal ArticleDOI
TL;DR: In this article, evolutionary aspects of escape from X inactivation, in relation to the divergence of the sex chromosomes, are discussed, including their developmental regulation and the implications of chromatin domains along the X chromosome in modeling the escape process.
Abstract: Although the process of X inactivation in mammalian cells silences the majority of genes on the inactivated X chromosome, some genes escape this chromosome-wide silencing. Genes that escape X inactivation present a unique opportunity to study the process of silencing and the mechanisms that protect some genes from being turned off. In this review, we will discuss evolutionary aspects of escape from X inactivation, in relation to the divergence of the sex chromosomes. Molecular characteristics, expression, and epigenetic modifications of genes that escape will be presented, including their developmental regulation and the implications of chromatin domains along the X chromosome in modeling the escape process.

Journal ArticleDOI
01 Dec 2002-Genesis
TL;DR: Examination of expression from the Xist locus during spermatogenesis in wild‐type mice and detected sense (Xist), but not antisense (Tsix) transcripts indicates that a functional Xist gene is not required for X‐chromosome inactivation during s permatogenesis and that this process is therefore regulated by a different mechanism than that which regulates X‐ Chromosome Inactivation in female embryonic cells.
Abstract: Transcriptional inactivation of the single X chromosome occurs in spermatogenic cells during male meiosis in mammals and has been shown to be coincident with expression of the Xist gene in spermatogonia and spermatocytes in mice. However, male mice carrying an ablated Xist gene show normal fertility. Here we examined expression from the Xist locus during spermatogenesis in wild-type mice and detected sense (Xist), but not antisense (Tsix) transcripts. In addition, we examined expression and chromatin conformation of X-linked structural genes in meiotic and postmeiotic spermatogenic cells from wild-type and Xist(-) mice and found no differences associated with the absence of a functional Xist gene. These results, along with the formation of a morphologically normal XY body in primary spermatocytes in Xist(-) mice, indicate that a functional Xist gene is not required for X-chromosome inactivation during spermatogenesis and that this process is therefore regulated by a different mechanism than that which regulates X-chromosome inactivation in female embryonic cells.

Journal ArticleDOI
TL;DR: It is proposed that deleting both Tsix alleles results in chaotic choice and that randomness in XΔXΔ survivors reflects a fortuitous selection of distinct X chromosomes as active and inactive.
Abstract: Tsix controls X-chromosome inactivation (XCI) by blocking the accumulation of Xist RNA on the future active X chromosome. Deleting Tsix on one X chromosome (X(Delta)X) skews XCI toward the mutated X chromosome in the female soma. Here I have generated homozygous Tsix-null mice (X(Delta)X(Delta)) to test how deleting the second allele affects the choice of XCI. Homozygosity leads to extremely low fertility and reveals two previously unknown non-mendelian patterns of inheritance. First, the sex ratio is skewed against female births so that one daughter is born for every two to three sons. Second, the pattern of XCI unexpectedly returns to random in surviving X(Delta)X(Delta) mice. Thus, with respect to choice, mutation of Tsix yields a phenotypic abnormality in heterozygotes but not homozygotes. To reconcile the paradox of female loss with apparent reversion to random choice, I propose that deleting both Tsix alleles results in chaotic choice and that randomness in X(Delta)X(Delta) survivors reflects a fortuitous selection of distinct X chromosomes as active and inactive.

Journal ArticleDOI
TL;DR: The origin of the human X, and the evolution of dosage compensation and gene content, is discussed, in the light of recent demonstrations that particular functions in sex and reproduction and cognition have accumulated on it.
Abstract: In humans, as in other mammals, sex is determined by an XX female/XY male chromosome system. Most attention has focused on the small, degenerate Y chromosome, which bears the male-dominant gene SRY. The X, in contrast, has been considered a well-behaved and immaculately conserved element that has hardly changed since the pre-mammal days when it was just another autosome pair. However, the X, uniquely in the genome, is present in two copies in females and only one in males. This has had dire consequences genetically on the evolution of its activity--and now it appears, on its gene content and/or the function of its genes. Here we will discuss the origin of the human X, and the evolution of dosage compensation and gene content, in the light of recent demonstrations that particular functions in sex and reproduction and cognition have accumulated on it.

Journal ArticleDOI
TL;DR: It is shown here that SDC-2 recruits the entire dosage compensation complex to her-1, directing this X-chromosome repression machinery to silence an individual, autosomal gene.
Abstract: Gene-specific and chromosome-wide mechanisms of transcriptional regulation control development in multicellular organisms. SDC-2, the determinant of hermaphrodite fate in Caenorhabditis elegans, is a paradigm for both modes of regulation. SDC-2 represses transcription of X chromosomes to achieve dosage compensation, and it also represses the male sex-determination gene her-1 to elicit hermaphrodite differentiation. We show here that SDC-2 recruits the entire dosage compensation complex to her-1, directing this X-chromosome repression machinery to silence an individual, autosomal gene. Functional dissection of her-1 in vivo revealed DNA recognition elements required for SDC-2 binding, recruitment of the dosage compensation complex, and transcriptional repression. Elements within her-1 differed in location, sequence, and strength of repression, implying that the dosage compensation complex may regulate transcription along the X chromosome using diverse recognition elements that play distinct roles in repression.

Journal ArticleDOI
Hans Ellegren1
TL;DR: A recently identified hypermethylated region on the Z chromosome, with similarities to the X inactivation centre on the mammalian X chromosome, might play a part in this process or have a role in avian sex determination.

Journal ArticleDOI
TL;DR: None of the silencing proteins identified so far is unique to X chromosome inactivation, the specificity must partly reside in Xist RNA whose spread along the X orchestrates general silencing factors for this specific task.

Journal ArticleDOI
TL;DR: It is proposed that even autosomal chromatin that had been inactivated earlier in development may undergo a stepwise loss of inactivation hallmarks, beginning with XIST RNA, which may be a primary cause of incomplete or unstable autosomal inactivation.
Abstract: Whether XIST RNA is indifferent to the sequence content of the chromosome is fundamental to understanding its mechanism of chromosomal inactivation. Transgenic Xist RNA appears to associate with and inactivate an entire autosome. However, the behavior of XIST RNA on naturally occurring human X;autosome translocations has not been thoroughly investigated. Here, the relationship of human XIST RNA to autosomal chromatin is investigated in cells from two patients carrying X;autosome translocations in the context of almost complete trisomy for the involved autosome. Since trisomies of either 14 or 9 are lethal in early development, the lack of serious phenotypic consequences of the trisomy demonstrates that the translocated autosomes had been inactivated. Surprisingly, our analyses show that in primary fibroblasts from adult patients, XIST RNA does not associate with most of the involved autosome even though the bulk of it exhibits other hallmarks of inactivation beyond the region associated with XIST RNA. While results show that XIST RNA can associate with human autosomal chromatin to some degree, several observations indicate that this interaction may be unstable, with progressive loss over time. Thus, even where autosomal inactivation is selected for rather than against, there is a fundamental difference in the affinity of XIST RNA for autosomal versus X chromatin. Based on these results we propose that even autosomal chromatin that had been inactivated earlier in development may undergo a stepwise loss of inactivation hallmarks, beginning with XIST RNA. Hence compromised interaction with XIST RNA may be a primary cause of incomplete or unstable autosomal inactivation.

Journal ArticleDOI
01 Aug 2002-Genomics
TL;DR: A novel gene is described, Enox (expressed neighbor of Xist), that maps to an unmethylated CpG island 10 kb upstream of Xists, which is antisense relative to Xist, highly heterogeneous, and apparently noncoding.

Journal ArticleDOI
TL;DR: It is shown that the pachytene germline X chromosomes in both sexes lack Me(K4)H3 when compared with autosomes, consistent with their being transcriptionally inactive, and that an evolutionarily conserved mechanism for silencing the X chromosome specifically in the male germline is detected.

Book ChapterDOI
TL;DR: This chapter focuses on dosage compensation in Drosophila, in which most X-linked genes are upregulated by a male-specific ribonucleoprotein complex, which is thought to recognize the X chromosome through approximately 35 dispersed chromatin entry sites and then spread in cis to dosage compensate most genes on theX chromosome.
Abstract: Just as homology can trigger a chain of events as described in many of the chapters of this volume, sometimes a lack of homology causes a crisis of a different sort. So it is for the single X chromosome in XY males in many species. Divergent sex chromosome pairs, such as the X and Y chromosomes in mammals and in fruit flies, are thought to have evolved from homologous autosomes. During evolution, the Y chromosome has retained little coding capacity, leaving the male with reduced gene dosage for many functions encoded by the X chromosome. In this chapter we focus on dosage compensation in Drosophila, in which most X-linked genes are upregulated by a male-specific ribonucleoprotein complex. This complex is thought to recognize the X chromosome through approximately 35 dispersed chromatin entry sites and then spread in cis to dosage compensate most genes on the X chromosome.

Journal ArticleDOI
TL;DR: The maintenance methylase model is discussed and updated to consider methylation patterns in cell populations that have occasional, stochastic methylation changes by de novo methylation or demethylation, either active or passive.
Abstract: X chromosome inactivation and DNA methylation are reviewed, with emphasis on the contributions of Susumu Ohno and the predictions made in my 1975 paper (Riggs, 1975), in which I proposed the “maintena

Journal ArticleDOI
TL;DR: Comparison between expression levels in males and females by real-time quantitative PCR suggested that expression was compensated for the CHD-Z gene but not for the B4GALT1 gene.
Abstract: In birds, females are heterogametic (ZW), while males are homogametic (ZZ). It has been proposed that there is no dosage compensation for the expression of Z-linked genes in birds. In order to examine

Journal ArticleDOI
TL;DR: Fluctuations in the rate of variant females in field populations and in laboratory colonies of Akodon depend on the balance between the appearance of new variant females (s–/s–, XY* specimens) and the extinction of sex reversed specimens due to imprinting escape.
Abstract: The existence of fertile A. azarae females with a chromosome sex pair indistinguishable from that of males was reported more than 35 years ago. These heterogametic females were initially thought to occur due to an extreme process of dosage compensation in which X inactivation was restricted to Xp and complemented by a deletion of Xq (Xx females). Later on, a C-banding analysis of A. mollis variant females showed that these specimens were in fact XY* sex reversed and not Xx females. The finding of positive testing for Zfy and Sry multiple-copy genes in Akodon males and heterogametic females confirmed the XY* assumption. At the present time, XY* sex reversed females have been found to exist in nine Akodon species. Akodon heterogametic females produce X and Y* oocytes, which upon sperm fertilization give rise to viable XX (female), XY* (female), and XY (male) embryos, and to non-viable Y*Y zygotes. Heterozygous females exhibit a better reproductive performance than XX females in order to compensate the Y*Y zygote wastage. XY* sex reversed females are assumed to occur due to a deficient Sry expression resulting in the development of ovaries instead of testes. Moreover, the appearance of Y* elements is a highly recurrent event. It is proposed that homozygosity for an autosomal or pseudoautosomal recessive mutation (s-) inhibits Sry expression giving rise to XY* embryos with ovary development. Location of the Y* chromosome in the female germ cell lineage produces an ovary-specific imprinting of the Sry* gene maintaining its defective expression through generations independently from the presence or absence of s- homozygosity. By escaping the ovary-specific methylation some Y* chromosomes turn back to normal Ys producing Y oocytes capable of generating normal male embryos when fertilized by an X sperm. Fluctuations in the rate of variant females in field populations and in laboratory colonies of Akodon depend on the balance between the appearance of new variant females (s-/s-, XY* specimens) and the extinction of sex reversed specimens due to imprinting escape.

Journal ArticleDOI
TL;DR: The pressures that drove the evolution of sex and the mechanisms by which it occurred are discussed, discussing the various hypotheses proposed and the evidence supporting them.
Abstract: Mammalian sex chromosomes appear, behave and function differently than the autosomes, passing on their genes in a unique sex-linked manner. The publishing of Ohno's hypothesis provided a framework for discussion of sex chromosome evolution, allowing it to be developed and challenged numerous times. In this report we discuss the pressures that drove the evolution of sex and the mechanisms by which it occurred. We concentrate on how the sex chromosomes evolved in mammals, discussing the various hypotheses proposed and the evidence supporting them.

Journal ArticleDOI
TL;DR: This review compares XCI in mouse and human, and discusses how much of the murine data can be extrapolated to humans.
Abstract: Mammals perform dosage compensation of X-linked gene products between XY males and XX females by transcriptionally silencing all but one X chromosome per diploid cell, a process called X chromosome inactivation (XCI). XCI involves counting X chromosomes in a cell, random or imprinted choice of one X to remain active, initiation and spread of the inactivation signal in cis throughout the other X chromosomes, and maintenance of the inactive state of those X chromosomes during cell divisions thereafter. Most of what is known of the molecular mechanisms involved in the different steps of XCI has been studied in the mouse. In this review we compare XCI in mouse and human, and discuss how much of the murine data can be extrapolated to humans.

Journal ArticleDOI
TL;DR: The case of a girl at the age of 32 months with dysmorphic features, including general muscular hypotonia, developmental delay and mental retardation is presented, with the exception of the previously observed break‐point region which revealed an early replicating pattern with strong fluorescent signal, similar to the pattern of the active X chromosome.
Abstract: In this paper we present the case of a girl at the age of 32 months with dysmorphic features, including general muscular hypotonia, developmental delay and mental retardation. The cytogenetic analysis revealed de novo partial duplication of Xp: 46,X,dup(X)(p11.23-->p22.33: :p11.23-->p22.33). To characterize the duplication, X painting, Kallman (KAL), yeast artificial chromosomes (YACs) and bacterial artificial chromosomes (BACs) covering Xp11.23-->Xp22.33 region were used. Selective inactivation of the abnormal X chromosome using HpaII digestion of the AR gene was evident. After BrdU incorporation the abnormal X was late-replicating in all lymphocytes examined. There was one peculiar exception observed: the break-point region was consistently early replicating. The replicating pattern of this region corresponded to the active X chromosome. Methylation pattern of late replicating X chromosome was studied also using antibodies against 5-methylcytosine. The pattern corresponded to the normally inactive X chromosome, with the exception of the previously observed break-point region which revealed an early replicating pattern with strong fluorescent signal, similar to the pattern of the active X chromosome. The observed phenomenon could lead to the abnormal phenotype of the patient, with some normally inactive genes of the break-point region escaping the inactivation process. The abnormal clinical findings could also be due to tissue-dependent differences in the inactivation pattern.

Journal ArticleDOI
TL;DR: The finding that the paternally derived X chromosome is eliminated in females suggests that late DNA replication may provide the imprint for paternal X inactivation and the elimination of sex chromosomes in bandicoots.
Abstract: Cytogenetic studies have shown that bandicoots (family Peramelidae) eliminate one X chromosome in females and the Y chromosome in males from some somatic tissues at different stages during development. The discovery of a polymorphism for X-linked phosphoglycerate kinase (PGK-1) in a population of Isoodon obesulus from Mount Gambier, South Australia, has allowed us to answer a number of long standing questions relating to the parental source of the eliminated X chromosome, X chromosome inactivation and reactivation in somatic and germ cells of female bandicoots. We have found no evidence of paternal PGK-1 allele expression in a wide range of somatic tissues and cell types from known female heterozygotes. We conclude that paternal X chromosome inactivation occurs in bandicoots as in other marsupial groups and that it is the paternally derived X chromosome that is eliminated from some cell types of females. The absence of PGK-1 paternal activity in somatic cells allowed us to examine the state of X chromosome activity in germ cells. Electrophoresis of germ cells from different aged pouch young heterozygotes showed only maternal allele expression in oogonia whereas an additional paternally derived band was observed in pre-dictyate oocytes. We conclude that reactivation of the inactive X chromosome occurs around the onset of meiosis in female bandicoots. As in other mammals, late replication is a common feature of the Y chromosome in male and the inactive X chromosome in female bandicoots. The basis of sex chromosome loss is still not known; however later timing of DNA synthesis is involved. Our finding that the paternally derived X chromosome is eliminated in females suggests that late DNA replication may provide the imprint for paternal X inactivation and the elimination of sex chromosomes in bandicoots.

Journal ArticleDOI
TL;DR: Loss of gene control due to hypomethylation and chromatin disruption by polyamines or other factors can include loss of dosage compensation from the inactive X chromosome for spermine synthase and spermidine/spermine N(1)-acetyltransferase at Xp22.1.

Journal ArticleDOI
TL;DR: The extent of underreplication of tena DNA in IH region 11A6–9 negatively correlates with the amount of MSL complex, which is found to reflect the different levels of transcription and chromosome compaction due to dosage compensation.
Abstract: Regions of intercalary heterochromatin (IH) are dispersed in the euchromatic arms of polytene chromosomes and share the main properties of heterochromatin, namely chromosome constrictions resulting from DNA underreplication. These constrictions are frequent on the paired X chromosomes of females, but are practically absent from the single X chromosome of males. These sex-specific differences have been proposed to reflect the different levels of transcription and chromosome compaction due to dosage compensation, which in turn may affect the degree of underreplication in IH regions. To test this hypothesis, we induced dosage compensation in females by ectopic expression of MSL-2 protein. We then measured the extent of underreplication in IH regions by determining frequencies of constrictions, or by Southern blot analysis using a fragment of the ten a gene which is located in IH region 11A6–9. Females transheterozygous for Sxl fhv1 /Sxl f1 or carrying a constitutive msl-2 transgene are known to hypertranscribe their X chromosomes. In such females, both the frequency of constrictions and DNA underreplication were reduced. Suppression of underreplication occurs only when a complete functional MSL complex assembles on the X chromosomes. We also used three strains that carried constitutive transgenes of msl-2 with mutations in the 5′ untranslated regions. These strains produced normal levels of SXL protein, but variable levels of MSL-2 protein. The SXL protein did not prevent the formation of an MSL complex in these transgenic females. We found that the extent of underreplication of ten a DNA in IH region 11A6–9 negatively correlates with the amount of MSL complex.