scispace - formally typeset
Search or ask a question

Showing papers on "Dosage compensation published in 2013"


Journal ArticleDOI
TL;DR: New findings regarding the causes of sex- biased expression are highlighted, including sexual antagonism and incomplete dosage compensation, and how sex-biased expression can accelerate the evolution ofsex-linked genes are discussed.
Abstract: Females and males often differ extensively in their physical traits. This sexual dimorphism is largely caused by differences in gene expression. Recent advances in genomics, such as RNA sequencing (RNA-seq), have revealed the nature and extent of sex-biased gene expression in diverse species. Here we highlight new findings regarding the causes of sex-biased expression, including sexual antagonism and incomplete dosage compensation. We also discuss how sex-biased expression can accelerate the evolution of sex-linked genes.

315 citations


Journal ArticleDOI
TL;DR: It is shown that GC skew can distinguish four classes of promoters, including three types of CGI promoters, each associated with unique epigenetic and gene ontology signatures, and that nearly 2000 genes harbor GC skew at their 3' ends and that these genes are preferentially located in gene-dense regions and tend to be closely arranged.
Abstract: Strand asymmetry in the distribution of guanines and cytosines, measured by GC skew, predisposes DNA sequences toward R-loop formation upon transcription. Previous work revealed that GC skew and R-loop formation associate with a core set of unmethylated CpG island (CGI) promoters in the human genome. Here, we show that GC skew can distinguish four classes of promoters, including three types of CGI promoters, each associated with unique epigenetic and gene ontology signatures. In particular, we identify a strong and a weak class of CGI promoters and show that these loci are enriched in distinct chromosomal territories reflecting the intrinsic strength of their protection against DNA methylation. Interestingly, we show that strong CGI promoters are depleted from the X chromosome while weak CGIs are enriched, a property consistent with the acquisition of DNA methylation during dosage compensation. Furthermore, we identify a third class of CGI promoters based on its unique GC skew profile and show that this gene set is enriched for Polycomb group targets. Lastly, we show that nearly 2000 genes harbor GC skew at their 3' ends and that these genes are preferentially located in gene-dense regions and tend to be closely arranged. Genomic profiling of R-loops accordingly showed that a large proportion of genes with terminal GC skew form R-loops at their 3' ends, consistent with a role for these structures in permitting efficient transcription termination. Altogether, we show that GC skew and R-loop formation offer significant insights into the epigenetic regulation, genomic organization, and function of human genes.

289 citations


Journal ArticleDOI
15 Aug 2013-Nature
TL;DR: Down’s syndrome is a common disorder with enormous medical and social costs, caused by trisomy for chromosome 21, and the concept that gene imbalance across an extra chromosome can be de facto corrected by manipulating a single gene, XIST (the X-inactivation gene), is tested.
Abstract: Down's syndrome is a common disorder with enormous medical and social costs, caused by trisomy for chromosome 21. We tested the concept that gene imbalance across an extra chromosome can be de facto corrected by manipulating a single gene, XIST (the X-inactivation gene). Using genome editing with zinc finger nucleases, we inserted a large, inducible XIST transgene into the DYRK1A locus on chromosome 21, in Down's syndrome pluripotent stem cells. The XIST non-coding RNA coats chromosome 21 and triggers stable heterochromatin modifications, chromosome-wide transcriptional silencing and DNA methylation to form a 'chromosome 21 Barr body'. This provides a model to study human chromosome inactivation and creates a system to investigate genomic expression changes and cellular pathologies of trisomy 21, free from genetic and epigenetic noise. Notably, deficits in proliferation and neural rosette formation are rapidly reversed upon silencing one chromosome 21. Successful trisomy silencing in vitro also surmounts the major first step towards potential development of 'chromosome therapy'.

286 citations


Journal ArticleDOI
TL;DR: Analysis of the genomes and transcriptomes of snake species with homomorphic and heteromorphic sex chromosomes reveals the evolutionary dynamics of sex chromosome differentiation.
Abstract: Snakes exhibit genetic sex determination, with female heterogametic sex chromosomes (ZZ males, ZW females). Extensive cytogenetic work has suggested that the level of sex chromosome heteromorphism varies among species, with Boidae having entirely homomorphic sex chromosomes, Viperidae having completely heteromorphic sex chromosomes, and Colubridae showing partial differentiation. Here, we take a genomic approach to compare sex chromosome differentiation in these three snake families. We identify homomorphic sex chromosomes in boas (Boidae), but completely heteromorphic sex chromosomes in both garter snakes (Colubridae) and pygmy rattlesnake (Viperidae). Detection of W-linked gametologs enables us to establish the presence of evolutionary strata on garter and pygmy rattlesnake sex chromosomes where recombination was abolished at different time points. Sequence analysis shows that all strata are shared between pygmy rattlesnake and garter snake, i.e., recombination was abolished between the sex chromosomes before the two lineages diverged. The sex-biased transmission of the Z and its hemizygosity in females can impact patterns of molecular evolution, and we show that rates of evolution for Z-linked genes are increased relative to their pseudoautosomal homologs, both at synonymous and amino acid sites (even after controlling for mutational biases). This demonstrates that mutation rates are male-biased in snakes (male-driven evolution), but also supports faster-Z evolution due to differential selective effects on the Z. Finally, we perform a transcriptome analysis in boa and pygmy rattlesnake to establish baseline levels of sex-biased expression in homomorphic sex chromosomes, and show that heteromorphic ZW chromosomes in rattlesnakes lack chromosome-wide dosage compensation. Our study provides the first full scale overview of the evolution of snake sex chromosomes at the genomic level, thus greatly expanding our knowledge of reptilian and vertebrate sex chromosomes evolution.

282 citations


Journal ArticleDOI
18 Jul 2013-Nature
TL;DR: The results reveal several puzzling aspects of Drosophila dot chromosome biology to be possible remnants of its former life as a sex chromosome, such as its minor feminizing role in sex determination or its targeting by a chromosome-specific regulatory mechanism.
Abstract: Although transitions of sex-determination mechanisms are frequent in species with homomorphic sex chromosomes, heteromorphic sex chromosomes are thought to represent a terminal evolutionary stage owing to chromosome-specific adaptations such as dosage compensation or an accumulation of sex-specific mutations. Here we show that an autosome of Drosophila, the dot chromosome, was ancestrally a differentiated X chromosome. We analyse the whole genome of true fruitflies (Tephritidae), flesh flies (Sarcophagidae) and soldier flies (Stratiomyidae) to show that genes located on the dot chromosome of Drosophila are X-linked in outgroup species, whereas Drosophila X-linked genes are autosomal. We date this chromosomal transition to early drosophilid evolution by sequencing the genome of other Drosophilidae. Our results reveal several puzzling aspects of Drosophila dot chromosome biology to be possible remnants of its former life as a sex chromosome, such as its minor feminizing role in sex determination or its targeting by a chromosome-specific regulatory mechanism. We also show that patterns of biased gene expression of the dot chromosome during early embryogenesis, oogenesis and spermatogenesis resemble that of the current X chromosome. Thus, although sex chromosomes are not necessarily evolutionary end points and can revert back to an autosomal inheritance, the highly specialized genome architecture of this former X chromosome suggests that severe fitness costs must be overcome for such a turnover to occur.

178 citations


Journal ArticleDOI
TL;DR: The status of dosage compensation is surveyed to answer questions about what sorts of genes are likely to be dosage compensated, how dosage compensation evolves, and why complete dosage compensation appears to be limited to male heterogametic species.

174 citations


Journal ArticleDOI
15 Nov 2013-Science
TL;DR: It is shown that the acquisition of dozens of MSL binding sites on evolutionarily new X chromosomes was facilitated by the independent co-option of a mutant helitron TE that attracts the MSL complex (TE domestication) and fine-tuning of the cis-regulatory element supplied by the TE and erosion of nonfunctional regions.
Abstract: Transposable elements (TEs) may contribute to evolutionary innovations through the rewiring of networks by supplying ready-to-use cis regulatory elements. Genes on the Drosophila X chromosome are coordinately regulated by the male specific lethal (MSL) complex to achieve dosage compensation in males. We show that the acquisition of dozens of MSL binding sites on evolutionarily new X chromosomes was facilitated by the independent co-option of a mutant helitron TE that attracts the MSL complex (TE domestication). The recently formed neo-X recruits helitrons that provide dozens of functional, but suboptimal, MSL binding sites, whereas the older XR chromosome has ceased acquisition and appears to have fine-tuned the binding affinities of more ancient elements for the MSL complex. Thus, TE-mediated rewiring of regulatory networks through domestication and amplification may be followed by fine-tuning of the cis-regulatory element supplied by the TE and erosion of nonfunctional regions.

154 citations


Journal ArticleDOI
TL;DR: It is proposed that repetitive structural motifs in lncRNAs could provide plasticity during multiprotein complex assemblies to ensure efficient targeting in cis or in trans along chromosomes.

147 citations


Journal ArticleDOI
18 Jun 2013-eLife
TL;DR: It is found that C. elegans equalizes X-chromosome expression between the sexes, to a level equivalent to autosomes, by reducing Pol II recruitment to promoters of hermaphrodite X-linked genes using a chromosome-restructuring condensin complex.
Abstract: The X-chromosome gene regulatory process called dosage compensation ensures that males (1X) and females (2X) express equal levels of X-chromosome transcripts. The mechanism in Caenorhabditis elegans has been elusive due to improperly annotated transcription start sites (TSSs). Here we define TSSs and the distribution of transcriptionally engaged RNA polymerase II (Pol II) genome-wide in wild-type and dosage-compensation-defective animals to dissect this regulatory mechanism. Our TSS-mapping strategy integrates GRO-seq, which tracks nascent transcription, with a new derivative of this method, called GRO-cap, which recovers nascent RNAs with 5' caps prior to their removal by co-transcriptional processing. Our analyses reveal that promoter-proximal pausing is rare, unlike in other metazoans, and promoters are unexpectedly far upstream from the 5' ends of mature mRNAs. We find that C. elegans equalizes X-chromosome expression between the sexes, to a level equivalent to autosomes, by reducing Pol II recruitment to promoters of hermaphrodite X-linked genes using a chromosome-restructuring condensin complex. DOI:http://dx.doi.org/10.7554/eLife.00808.001.

144 citations


Journal ArticleDOI
TL;DR: The discovery of a long noncoding RNA, XACT, that is expressed from and coats the active X chromosome specifically in human pluripotent cells, suggesting a unique role for XACT in the control of human XCI initiation.
Abstract: X-chromosome inactivation (XCI) in mammals relies on XIST, a long noncoding transcript that coats and silences the X chromosome in cis. Here we report the discovery of a long noncoding RNA, XACT, that is expressed from and coats the active X chromosome specifically in human pluripotent cells. In the absence of XIST, XACT is expressed from both X chromosomes in humans but not in mice, suggesting a unique role for XACT in the control of human XCI initiation.

132 citations


Journal ArticleDOI
TL;DR: It is demonstrated that a previously uncharacterized zinc finger protein, CLAMP (chromatin-linked adaptor for MSL proteins), functions as the first link between the MSL complex and the X chromosome, providing new insights into how subnuclear domains of coordinate gene regulation are formed within metazoan genomes.
Abstract: The Drosophila male-specific lethal (MSL) dosage compensation complex increases transcript levels on the single male X chromosome to equal the transcript levels in XX females. However, it is not known how the MSL complex is linked to its DNA recognition elements, the critical first step in dosage compensation. Here, we demonstrate that a previously uncharacterized zinc finger protein, CLAMP (chromatin-linked adaptor for MSL proteins), functions as the first link between the MSL complex and the X chromosome. CLAMP directly binds to the MSL complex DNA recognition elements and is required for the recruitment of the MSL complex. The discovery of CLAMP identifies a key factor required for the chromosome-specific targeting of dosage compensation, providing new insights into how subnuclear domains of coordinate gene regulation are formed within metazoan genomes.

Journal ArticleDOI
Hans Ellegren1
TL;DR: Avian sex chromosome evolution, representing the best characterized ZW system to date, follows patterns seen in other organisms but has the notable exception of incomplete dosage compensation.
Abstract: Rapidly accumulating genome sequence information in birds, which show several unique genomic features, provides novel insights into evolutionary genomics. The avian karyotype with numerous microchromosomes has remained stable during evolution, although frequent intrachromosomal inversions have occurred. Avian sex chromosome evolution, representing the best characterized ZW system to date, follows patterns seen in other organisms but has the notable exception of incomplete dosage compensation. Recombination is unevenly distributed in the avian genome; it occurs at very high rates in microchromosomes, a consequence of an obligate crossing over in even small chromosomes, and has highly elevated rates near chromosome ends. Moreover, a heterogeneous landscape of recombination feeds significant heterogeneity in base composition via GC-biased gene conversion. A uniform molecular clock is not applicable to birds, and ample evidence for substitution rate heterogeneity both among lineages and within genomes exists....

Journal ArticleDOI
TL;DR: Sexual differentiation at the molecular level is established in chicken early in embryogenesis, before gonadal sex differentiation, indicating that molecular sexual differentiation is tissue specific.
Abstract: Birds have a ZZ male: ZW female sex chromosome system and while the Z-linked DMRT1 gene is necessary for testis development, the exact mechanism of sex determination in birds remains unsolved. This is partly due to the poor annotation of the W chromosome, which is speculated to carry a female determinant. Few genes have been mapped to the W and little is known of their expression. We used RNA-seq to produce a comprehensive profile of gene expression in chicken blastoderms and embryonic gonads prior to sexual differentiation. We found robust sexually dimorphic gene expression in both tissues pre-dating gonadogenesis, including sex-linked and autosomal genes. This supports the hypothesis that sexual differentiation at the molecular level is at least partly cell autonomous in birds. Different sets of genes were sexually dimorphic in the two tissues, indicating that molecular sexual differentiation is tissue specific. Further analyses allowed the assembly of full-length transcripts for 26 W chromosome genes, providing a view of the W transcriptome in embryonic tissues. This is the first extensive analysis of W-linked genes and their expression profiles in early avian embryos. Sexual differentiation at the molecular level is established in chicken early in embryogenesis, before gonadal sex differentiation. We find that the W chromosome is more transcriptionally active than previously thought, expand the number of known genes to 26 and present complete coding sequences for these W genes. This includes two novel W-linked sequences and three small RNAs reassigned to the W from the Un_Random chromosome.

Journal ArticleDOI
TL;DR: Individual‐based simulations of a Muller's ratchet process are used to test how the relevant parameters (effective population size, strength and dominance of deleterious mutations, size of nonrecombining segment, and strength of sexually antagonistic selection) are expected to affect the rate of turnovers.
Abstract: In sharp contrast with mammals and birds, many cold-blooded vertebrates present homomorphic sex chromosomes. Empirical evidence supports a role for frequent turnovers, which replace nonrecombining sex chromosomes before they have time to decay. Three main mechanisms have been proposed for such turnovers, relying either on neutral processes, sex-ratio selection, or intrinsic benefits of the new sex-determining genes (due, e.g., to linkage with sexually antagonistic mutations). Here, we suggest an additional mechanism, arising from the load of deleterious mutations that accumulate on nonrecombining sex chromosomes. In the absence of dosage compensation, this load should progressively lower survival rate in the heterogametic sex. Turnovers should occur when this cost outweighs the benefits gained from any sexually antagonistic genes carried by the nonrecombining sex chromosome. We use individual-based simulations of a Muller's ratchet process to test this prediction, and investigate how the relevant parameters (effective population size, strength and dominance of deleterious mutations, size of nonrecombining segment, and strength of sexually antagonistic selection) are expected to affect the rate of turnovers.

Journal ArticleDOI
TL;DR: How different states of XIST expression define three classes of female human pluripotent stem cells are described and progress in discovering the reasons for these variations and how they might be countered is explored.
Abstract: Reprogramming somatic cells to derive induced pluripotent stem cells (iPSCs) has provided a new method to model disease and holds great promise for regenerative medicine. Although genetically identical to their donor somatic cells, iPSCs undergo substantial changes in the epigenetic landscape during reprogramming. One such epigenetic process, X chromosome inactivation (XCI), has recently been shown to vary widely in human female iPSCs and embryonic stem cells (ESCs). XCI is a form of dosage compensation whose chief regulator is the noncoding RNA Xist. In mouse iPSCs and ESCs, Xist expression and XCI strictly correlate with the pluripotent state, but no such correlation exists in humans. Lack of XIST expression in human cells is linked to reduced developmental potential and an altered transcriptional profile, including upregulation of genes associated with cancer, which has therefore led to concerns about the safety of pluripotent stem cells for use in regenerative medicine. In this review, we describe how different states of XIST expression define three classes of female human pluripotent stem cells and explore progress in discovering the reasons for these variations and how they might be countered.

Journal ArticleDOI
TL;DR: This comprehensive analysis provides a new level of information on different interaction modes of a multiprotein complex at distinct regions within the genome.
Abstract: Genes on the single X chromosome in Drosophila melanogaster males are subjected to transcriptional enhancement in order to meet the levels of expression product in females that carry two X chromosomes. This process is referred to as dosage compensation (DC). Even though similar compensatory processes can be observed in several unrelated heterogametic organisms, major principles and mechanisms differ substantially (Straub and Becker 2007; Mank 2009). In Drosophila, a ribonucleoprotein complex called Dosage Compensation Complex (DCC) or Male-Specific-Lethal (MSL) complex (MSL-DCC) constitutes specifically in males where it targets X-chromosomal genes (Larsson and Meller 2006; Gelbart and Kuroda 2009; Lucchesi 2009; Conrad and Akhtar 2011). Genetic screenings for male-specific lethality identified MSL-1, MSL-2, MSL-3, the histone acetyl transferase MOF, and the RNA/DNA helicase MLE as protein subunits. Two redundant noncoding RNAs—roX1 and roX2—complete the complex. MOF acetylates histone H4 at lysine 16 (H4K16ac), a modification that is expected to promote the unfolding of the chromatin fiber (Shogren-Knaak et al. 2006), boosting gene expression via enhanced transcriptional elongation (Larschan et al. 2011). Correct targeting of the MSL-DCC poses a major challenge, as ∼1000 active genes on the X chromosome must be selectively identified. Based on a multitude of genetic and biochemical studies, a two-step model has been proposed (for reviews, see Gelbart and Kuroda 2009; Conrad and Akhtar 2011; Straub and Becker 2011): First, the dosage compensation machinery is attracted to ∼100 initiation sites along the X, termed high-affinity sites (HAS) or chromosomal entry sites (CES). In a second step the complex is disseminated to active target genes in the vicinity of these sites. Genetic analyses of the MSL genes point to a crucial role of MSL-2 and MSL-1 in the identification of HAS/CES as these two factors can bind these selected sites in the absence of all other dosage compensation components (Lyman et al. 1997). HAS targeting most likely involves specific DNA sequence motifs. A GA-rich motif is highly enriched in these regions and contributes to complex recruitment (Alekseyenko et al. 2008; Straub et al. 2008). Conceivably, a core complex consisting of MSL-2 and MSL-1 is involved in recognizing this sequence, since MSL-2 is a DNA binding protein (Fauth et al. 2010). The distribution of the MSL-DCC to active gene targets requires the enzymatic activities of MLE and MOF (Gu et al. 2000; Morra et al. 2008), the presence of MSL-3, and at least one of the two roX RNAs (Kelley et al. 1999; Meller and Rattner 2002). It has been proposed that the contact with transcribed chromatin is established by recognition of H3 trimethylated on lysine 36 (H3K36me3) through MSL-3 (Larschan et al. 2007). Complex assembly is triggered by male-specific expression of MSL-2. Importantly, all other MSL proteins are expressed in females, suggesting their involvement in processes outside the realm of dosage compensation. Given the male-specific lethal phenotype of loss-of-function mutations, however, these functions are probably not essential. MLE is required for the editing of a Na+-channel mRNA (Reenan et al. 2000). MOF is part of the so-called “Non-Specific-Lethal” (NSL) complex, which preferentially binds promoters of some housekeeping genes in both sexes, most likely serving a role in transcription initiation (Prestel et al. 2010; Raja et al. 2010; Feller et al. 2012). Functions for MSL-1 and MSL-3 outside of the dosage compensation system are not known even though both are expressed at low levels in females. During recent years, genome-wide mapping studies have revealed in great details the global binding pattern of the MSL proteins and roX RNAs (Straub and Becker 2011). These studies confirm the overwhelming enrichment of the complex on the X chromosome in males. The MSL proteins studied so far (MSL-1, MSL-2, MSL-3, MOF) preferentially bind the bodies of active genes, in many cases with clear 3′ enrichment. Even though the binding patterns of the different MSLs show some variation, current models assume that all MSL proteins, in the context of a well-defined MSL-DCC, are involved in all steps of targeting and dissemination (Gelbart and Kuroda 2009; Conrad and Akhtar 2011; Straub and Becker 2011). We present here the first comprehensive description of the MLE binding pattern. Comparing ChIP-chip with ChIP-seq profiles (in the former assay the ChIP material is used to probe DNA microarrays, whereas in the latter the recovered DNA is determined by deep sequencing) revealed striking differences. A systematic analysis of the phenomenon showed that the chromatin shearing protocol we employed allowed us to visualize the primary contacts of the MSL proteins at different chromatin targets. The data reveal different modes of MSL interactions at HAS and within genes, demonstrate an unexpected contribution of MLE to a novel HAS definition, and point to a novel function of MSL-1 and MOF outside the compensation process. Our experimental strategy allowed the assessment of the topology of large protein complexes at distinct classes of chromosomal interaction sites and it may be applied to other regulatory processes outside of the dosage compensation system.

Journal ArticleDOI
TL;DR: This study shows how young sex chromosomes have altered their chromatin structure in Drosophila, and what genomic changes have led to silencing of the Y, and hyper-transcription of the X.
Abstract: Sex chromosomes originated from autosomes but have evolved a highly specialized chromatin structure. Drosophila Y chromosomes are composed entirely of silent heterochromatin, while male X chromosomes have highly accessible chromatin and are hypertranscribed as a result of dosage compensation. Here, we dissect the molecular mechanisms and functional pressures driving heterochromatin formation and dosage compensation of the recently formed neo-sex chromosomes of Drosophila miranda. We show that the onset of heterochromatin formation on the neo-Y is triggered by an accumulation of repetitive DNA. The neo-X has evolved partial dosage compensation and we find that diverse mutational paths have been utilized to establish several dozen novel binding consensus motifs for the dosage compensation complex on the neo-X, including simple point mutations at pre-binding sites, insertion and deletion mutations, microsatellite expansions, or tandem amplification of weak binding sites. Spreading of these silencing or activating chromatin modifications to adjacent regions results in massive mis-expression of neo-sex linked genes, and little correspondence between functionality of genes and their silencing on the neo-Y or dosage compensation on the neo-X. Intriguingly, the genomic regions being targeted by the dosage compensation complex on the neo-X and those becoming heterochromatic on the neo-Y show little overlap, possibly reflecting different propensities along the ancestral chromosome that formed the sex chromosome to adopt active or repressive chromatin configurations. Our findings have broad implications for current models of sex chromosome evolution, and demonstrate how mechanistic constraints can limit evolutionary adaptations. Our study also highlights how evolution can follow predictable genetic trajectories, by repeatedly acquiring the same 21-bp consensus motif for recruitment of the dosage compensation complex, yet utilizing a diverse array of random mutational changes to attain the same phenotypic outcome.

Journal ArticleDOI
TL;DR: The evolution of dosage compensation and how it is controlled during embryogenesis of mammals is discussed and the potential role of X chromosome number during early development is summarized.

Journal ArticleDOI
TL;DR: Recent advances in the understanding of Xi reactivation during development and reprogramming are reviewed and potential clinical applications are illustrated.
Abstract: In mammals, one of the two X chromosomes of female cells is inactivated for dosage compensation between the sexes. X chromosome inactivation is initiated in early embryos by the noncoding Xist RNA. Subsequent chromatin modifications on the inactive X chromosome (Xi) lead to a remarkable stability of gene repression in somatic cell lineages. In mice, reactivation of genes on the Xi accompanies the establishment of pluripotent cells of the female blastocyst and the development of primordial germ cells. Xi reactivation also occurs when pluripotency is established during the reprogramming of somatic cells to induced pluripotent stem cells. The mechanism of Xi reactivation has attracted increasing interest for studying changes in epigenetic patterns and for improving methods of cell reprogramming. Here, we review recent advances in the understanding of Xi reactivation during development and reprogramming and illustrate potential clinical applications.

Journal ArticleDOI
TL;DR: Birds (relative to eutherian mammals) are expected to show more widespread cell‐autonomous sex determination in non‐gonadal tissues, because of ineffective sex chromosome dosage compensation mechanisms.
Abstract: Background: The classic model of sex determination in mammals states that the sex of the individual is determined by the type of gonad that develops, which in turn determines the gonadal hormonal milieu that creates sex differences outside of the gonads. However, XX and XY cells are intrinsically different because of the cell-autonomous sex-biasing action of X and Y genes. Results: Recent studies of mice, in which sex chromosome complement is independent of gonadal sex, reveal that sex chromosome complement has strong effects contributing to sex differences in phenotypes such as metabolism. Adult mice with two X chromosomes (relative to mice with one X chromosome) show dramatically greater increases in body weight and adiposity after gonadectomy, irrespective of their gonadal sex. When fed a high-fat diet, XX mice develop striking hyperinsulinemia and fatty liver, relative to XY mice. The sex chromosome effects are modulated by the presence of gonadal hormones, indicating an interaction of the sex-biasing effects of gonadal hormones and sex chromosome genes. Conclusions: Other cell-autonomous sex chromosome effects are detected in mice in many phenotypes. Birds (relative to eutherian mammals) are expected to show more widespread cell-autonomous sex determination in non-gonadal tissues, because of ineffective sex chromosome dosage compensation mechanisms. Developmental Dynamics 242:371–379, 2013. © 2013 Wiley Periodicals, Inc.

Posted Content
TL;DR: In this article, the authors dissected the molecular mechanisms and functional pressures driving heterochromatin formation and dosage compensation of the recently formed neo-sex chromosomes of Drosophila miranda.
Abstract: Drosophila Y chromosomes are composed entirely of silent heterochromatin, while male X chromosomes have highly accessible chromatin and are hypertranscribed due to dosage compensation. Here, we dissect the molecular mechanisms and functional pressures driving heterochromatin formation and dosage compensation of the recently formed neo-sex chromosomes of Drosophila miranda. We show that the onset of heterochromatin formation on the neo-Y is triggered by an accumulation of repetitive DNA. The neo-X has evolved partial dosage compensation and we find that diverse mutational paths have been utilized to establish several dozen novel binding consensus motifs for the dosage compensation complex on the neo-X, including simple point mutations at pre-binding sites, insertion and deletion mutations, microsatellite expansions, or tandem amplification of weak binding sites. Spreading of these silencing or activating chromatin modifications to adjacent regions results in massive mis-expression of neo-sex linked genes, and little correspondence between functionality of genes and their silencing on the neo-Y or dosage compensation on the neo-X. Intriguingly, the genomic regions being targeted by the dosage compensation complex on the neo-X and those becoming heterochromatic on the neo-Y show little overlap, possibly reflecting different propensities along the ancestral chromosome to adopt active or repressive chromatin configurations. Our findings have broad implications for current models of sex chromosome evolution, and demonstrate how mechanistic constraints can limit evolutionary adaptations. Our study also highlights how evolution can follow predictable genetic trajectories, by repeatedly acquiring the same 21-bp consensus motif for recruitment of the dosage compensation complex, yet utilizing a diverse array of random mutational changes to attain the same phenotypic outcome.

Journal ArticleDOI
TL;DR: Using linear discriminant analysis, it is shown that X-inactivation status can successfully classify 90% of X-linked genes into those with functional or nonfunctional Y homologs, and support and expand upon the hypothesis that X inactivation is primarily driven by gene loss on the Y.
Abstract: Y chromosomes have long been dismissed as “graveyards of genes,” but there is still much to be learned from the genetic relics of genes that were once functional on the human Y. We identified human X-linked genes whose gametologs have been pseudogenized or completely lost from the Y chromosome and inferred which evolutionary forces may be acting to retain genes on the Y. Although gene loss appears to be largely correlated with the suppression of recombination, we observe that X-linked genes with functional Y homologs evolve under stronger purifying selection and are expressed at higher levels than X-linked genes with nonfunctional Y homologs. Additionally, we support and expand upon the hypothesis that X inactivation is primarily driven by gene loss on the Y. Using linear discriminant analysis, we show that X-inactivation status can successfully classify 90% of X-linked genes into those with functional or nonfunctional Y homologs.

Journal ArticleDOI
TL;DR: A model of compensation occurring on a gene-by-gene basis is supported, supported by an absence of clustering of genes on the Z chromosome with respect to the extent of compensation, which suggests male-biased expression of Z-linked genes is a derived trait after avian sex chromosome divergence.
Abstract: Sex chromosome divergence, which follows the cessation of recombination and degeneration of the sex-limited chromosome, can cause a reduction in expression level for sex-linked genes in the heterozygous sex, unless some mechanisms of dosage compensation develops to counter the reduction in gene dose. Because large-scale perturbations in expression levels arising from changes in gene dose might have strong deleterious effects, the evolutionary response should be strong. However, in birds and in at least some other female heterogametic organisms, wholesale sex chromosome dosage compensation does not seem to occur. Using RNA-seq of multiple tissues and individuals, we investigated male and female expression levels of Z-linked and autosomal genes in the collared flycatcher, a bird for which a draft genome sequence recently has been reported. We found that male expression of Z-linked genes was on average 50% higher than female expression, although there was considerable variation in the male-to-female ratio among genes. The ratio for individual genes was well correlated among tissues and there was also a correlation in the extent of compensation between flycatcher and chicken orthologs. The relative excess of male expression was positively correlated with expression breadth, expression level, and number of interacting proteins (protein connectivity), and negatively correlated with variance in expression. These observations lead to a model of compensation occurring on a gene-by-gene basis, supported by an absence of clustering of genes on the Z chromosome with respect to the extent of compensation. Equal mean expression level of autosomal and Z-linked genes in males, and 50% higher expression of autosomal than Z-linked genes in females, is compatible with that partial compensation is achieved by hypertranscription from females’ single Z chromosome. A comparison with male-to-female expression ratios in orthologous Z-linked genes of ostriches, where Z–W recombination still occurs, suggests that male-biased expression of Z-linked genes is a derived trait after avian sex chromosome divergence.

Journal ArticleDOI
TL;DR: Evidence is found for the creation of new CESs, with the analogous sequence and spacing as in D. melanogaster, providing strong support for the spreading model in the establishment of dosage compensation.
Abstract: Dosage compensation has arisen in response to the evolution of distinct male (XY) and female (XX) karyotypes. In Drosophila melanogaster, the MSL complex increases male X transcription approximately twofold. X-specific targeting is thought to occur through sequence-dependent binding to chromatin entry sites (CESs), followed by spreading in cis to active genes. We tested this model by asking how newly evolving sex chromosome arms in Drosophila miranda acquired dosage compensation. We found evidence for the creation of new CESs, with the analogous sequence and spacing as in D. melanogaster, providing strong support for the spreading model in the establishment of dosage compensation.

Journal ArticleDOI
TL;DR: The stronger selection on CUB on the X chromosome leads to a lower rate of synonymous site divergence compared with the autosomes, which will cause a stronger upward bias for X than A in estimates of the proportion of nonsynonymous mutations fixed by positive selection, for methods based on the McDonald–Kreitman test.
Abstract: Codon usage bias (CUB) in Drosophila is higher for X-linked genes than for autosomal genes. One possible explanation is that the higher effective recombination rate for genes on the X chromosome compared with the autosomes reduces their susceptibility to Hill-Robertson effects, and thus enhances the efficacy of selection on codon usage. The genome sequence of D. melanogaster was used to test this hypothesis. Contrary to expectation, it was found that, after correcting for the effective recombination rate, CUB remained higher on the X than on the autosomes. In contrast, an analysis of polymorphism data from a Rwandan population showed that mean nucleotide site diversity at 4-fold degenerate sites for genes on the X is approximately three-quarters of the autosomal value after correcting for the effective recombination rate, compared with approximate equality before correction. In addition, these data show that selection for preferred versus unpreferred synonymous variants is stronger on the X than the autosomes, which accounts for the higher CUB of genes on the X chromosome. This difference in the strength of selection does not appear to reflect the effects of dominance of mutations affecting codon usage, differences in gene expression levels between X and autosomes, or differences in mutational bias. Its cause therefore remains unexplained. The stronger selection on CUB on the X chromosome leads to a lower rate of synonymous site divergence compared with the autosomes; this will cause a stronger upward bias for X than A in estimates of the proportion of nonsynonymous mutations fixed by positive selection, for methods based on the McDonald-Kreitman test.

Journal ArticleDOI
TL;DR: This review highlights and discusses imprinted and random XCI from such a comparative viewpoint and indicates clear differences in the form and timing of XCI.

Journal ArticleDOI
TL;DR: A long noncoding RNA, XACT, is reported that is expressed from and coats the active X chromosome specifically in human pluripotent cells, suggesting a unique role for XACT in the control of human XCI initiation.
Abstract: X-chromosome inactivation (XCI), the dosage compensation process that equalizes X-linked gene expression between sexes, has mostly been studied in the mouse, where the central role for the non-coding RNA Xist in the initiation and spreading of the process was demonstrated. Although Xist is conserved in humans [1], very little is known concerning its regulation and function in this species. Several lines of evidence moreover suggest that different strategies have been adopted in the human to control XCI as compared to the mouse. In particular, in human pre-implantation development, XIST RNA coats the X chromosome(s) in both male and female embryos without inducing X-chromosome silencing [2]. This indicates that XIST expression and X-inactivation can be uncoupled during human embryogenesis and that other elements likely participate to the control of X chromosome activity in humans. XCI is established early during embryonic development, and embryonic stem cells can be used to decipher the kinetics and the molecular actors of the process. Human female embryonic stem cells (hESC) can be found in different configurations regarding XIST expression: most female hESC have already undergone XCI but tend to spontaneously lose XIST expression [3]. In the course of an RNA-seq analysis of female hESC, we identified an extended and un-annotated transcribed region producing a long unspliced, likely non-coding nuclear RNA. RNA-FISH analysis reveals that this transcript is expressed from, and coats the active X chromosome. We called this transcript XACT, for X-active coating transcript. In female hESC in which XIST is repressed, XACT is expressed from and coats both Xs, and this correlates with significant reactivation of the inactive X chromosome. Expression of XACT appears to be specific for pluripotent cells as its expression decreases during differentiation. Finally, we provide evidence that XACT is not conserved in the mouse. In conclusion, we have identified XACT as the first long ncRNA that coats the active X chromosome in human. Given its expression profile and lack of conservation, it is tempting to speculate that XACT is involved in the peculiar control of XCI initiation in human.

Journal ArticleDOI
TL;DR: It is concluded that Xcea is either a derived allele that arose concurrently with the domestication of fancy mice but prior the derivation of most classical inbred strains or a rare allele in the wild, and that each mouse taxa examined has a different functional Xce allele.
Abstract: X chromosome inactivation (XCI) is the mammalian mechanism of dosage compensation that balances X-linked gene expression between the sexes. Early during female development, each cell of the embryo proper independently inactivates one of its two parental X-chromosomes. In mice, the choice of which X chromosome is inactivated is affected by the genotype of a cis-acting locus, the X-chromosome controlling element (Xce). Xce has been localized to a 1.9 Mb interval within the X-inactivation center (Xic), yet its molecular identity and mechanism of action remain unknown. We combined genotype and sequence data for mouse stocks with detailed phenotyping of ten inbred strains and with the development of a statistical model that incorporates phenotyping data from multiple sources to disentangle sources of XCI phenotypic variance in natural female populations on X inactivation. We have reduced the Xce candidate 10-fold to a 176 kb region located approximately 500 kb proximal to Xist. We propose that structural variation in this interval explains the presence of multiple functional Xce alleles in the genus Mus. We have identified a new allele, Xcee present in Mus musculus and a possible sixth functional allele in Mus spicilegus. We have also confirmed a parent-of-origin effect on X inactivation choice and provide evidence that maternal inheritance magnifies the skewing associated with strong Xce alleles. Based on the phylogenetic analysis of 155 laboratory strains and wild mice we conclude that Xcea is either a derived allele that arose concurrently with the domestication of fancy mice but prior the derivation of most classical inbred strains or a rare allele in the wild. Furthermore, we have found that despite the presence of multiple haplotypes in the wild Mus musculus domesticus has only one functional Xce allele, Xceb. Lastly, we conclude that each mouse taxa examined has a different functional Xce allele.

Journal ArticleDOI
TL;DR: It is found that the majority of the X-linked genes in metafemales exhibit dosage compensation with an expression level similar to that of normal diploid females, providing evidence for an inverse dosage component to X chromosome compensation.
Abstract: Dosage compensation, the equalized X chromosome gene expression between males and females in Drosophila, has also been found in triple X metafemales. Inverse dosage effects, produced by genomic imbalance, are believed to account for this modulated expression, but they have not been studied on a global level. Here, we show a global expression comparison of metafemales (XXX; AA) with normal females (XX; AA) with high-throughput RNA-sequencing. We found that the majority of the X-linked genes in metafemales exhibit dosage compensation with an expression level similar to that of normal diploid females. In parallel, most of the autosomal genes were expressed at about two-thirds the level of normal females, the ratio of inverse dosage effects produced by the extra X chromosome. Both compensation and inverse effects were further confirmed by combination of X-linked and autosomally located miniwhite reporter genes in metafemales and relative quantitative PCR of selected genes. These data provide evidence for an inverse dosage component to X chromosome compensation.

Journal ArticleDOI
TL;DR: Analysis shows that the high number of paralogous gene families on the mammalian X chromosome relative to autosomes contributes to the ambiguity in RXE calculations, and RNA-seq analysis that takes into account that single- and multi-copy genes are compensated differently supports the conclusion that, in many somatic tissues, the mammalianX is up-regulated compared to the autosomes.
Abstract: An enduring question surrounding sex chromosome evolution is whether effective hemizygosity in the heterogametic sex leads inevitably to dosage compensation of sex-linked genes, and whether this compensation has been observed in a variety of organisms. Incongruence in the conclusions reached in some recent reports has been attributed to different high-throughput approaches to transcriptome analysis. However, recent reports each utilizing RNA-seq to gauge X-linked gene expression relative to autosomal gene expression also arrived at diametrically opposed conclusions regarding X chromosome dosage compensation in mammals. Here we analyze RNA-seq data from X-monosomic female human and mouse tissues, which are uncomplicated by genes that escape X-inactivation, as well as published RNA-seq data to describe relative X expression (RXE). We find that the determination of RXE is highly dependent upon a variety of computational, statistical and biological assumptions underlying RNA-seq analysis. Parameters implemented in short-read mapping programs, choice of reference genome annotation, expression data distribution, tissue source for RNA and RNA-seq library construction method have profound effects on comparing expression levels across chromosomes. Our analysis shows that the high number of paralogous gene families on the mammalian X chromosome relative to autosomes contributes to the ambiguity in RXE calculations, RNA-seq analysis that takes into account that single- and multi-copy genes are compensated differently supports the conclusion that, in many somatic tissues, the mammalian X is up-regulated compared to the autosomes.