Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.109
Filter
1.
Methods Mol Biol ; 2856: 71-78, 2025.
Article in English | MEDLINE | ID: mdl-39283447

ABSTRACT

Hi-C reads, which represent ligation events between different regions of the genome, must be processed into matrices of interaction frequencies for downstream analysis. Here, I describe a procedure for mapping Hi-C reads to the genome and conversion of mapped reads into the HOMER tag directory format and interaction matrix format for visualization with Juicebox. The method is demonstrated for the mouse composite X chromosome in which reads from the active and inactive X chromosomes are combined after mock DMSO treatment or targeted degradation of cohesin.


Subject(s)
X Chromosome , Animals , X Chromosome/genetics , Mice , Software , Cohesins , Chromosome Mapping/methods , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Computational Biology/methods
2.
Sci Rep ; 14(1): 20327, 2024 09 02.
Article in English | MEDLINE | ID: mdl-39223177

ABSTRACT

In female eutherian mammal development, X-chromosome inactivation (XCI) of one of the two X chromosomes is initiated early. Understanding the relationship between the initiation of XCI and cell fate is critical for understanding early female development and requires a system that can monitor XCI in single living cells. Traditional embryonic stem cells (ESCs) used for XCI studies often lose X chromosomes spontaneously during culture and differentiation, making accurate monitoring difficult. Additionally, most XCI assessment methods necessitate cell disruption, hindering cell fate tracking. We developed the Momiji (version 2) ESC line to address these difficulties, enabling real-time monitoring of X-chromosome activity via fluorescence. We inserted green and red fluorescent reporter genes and neomycin and puromycin resistance genes into the two X chromosomes of PGK12.1 ESCs, creating a female ESC line that retains two X chromosomes more faithfully during differentiation. Momiji (version 2) ESCs exhibit a more stable XX karyotype than other ESC lines, including the parental PGK12.1 line. This new tool offers valuable insights into the relationship between XCI and cell fate, improving our understanding of early female development.


Subject(s)
Time-Lapse Imaging , X Chromosome Inactivation , X Chromosome Inactivation/genetics , Animals , Female , Mice , Time-Lapse Imaging/methods , Cell Differentiation/genetics , Single-Cell Analysis/methods , Cell Line , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/cytology , X Chromosome/genetics , Genes, Reporter
3.
Science ; 385(6715): 1347-1354, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39298575

ABSTRACT

Long noncoding RNAs (lncRNAs) are essential regulatory elements of sex chromosomes that act to equalize gene expression levels between males and females. XIST, RSX, and roX2 regulate X chromosomes in placental mammals, marsupials, and Drosophila, respectively. Because the green anole (Anolis carolinensis) shows complete dosage compensation of its X chromosome, we tested whether a lncRNA was involved. We found an ancient lncRNA, MAYEX, that gained male-specific expression more than 89 million years ago. MAYEX evolved a notable association with the acetylated histone 4 lysine 16 (H4K16ac) epigenetic mark and the ability to loop its locus to the totality of the X chromosome to increase expression levels. MAYEX is the first lncRNA in reptiles linked to a dosage compensation mechanism that balances the expression of sex chromosomes.


Subject(s)
Dosage Compensation, Genetic , Lizards , RNA, Long Noncoding , X Chromosome , Animals , Female , Male , Acetylation , Epigenesis, Genetic , Evolution, Molecular , Histones/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , X Chromosome/genetics , Lizards/genetics
4.
Biochem Soc Trans ; 52(4): 1777-1784, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39149984

ABSTRACT

The non-Mendelian transmission of sex chromosomes during gametogenesis carries significant implications, influencing sex ratios and shaping evolutionary dynamics. Here we focus on known mechanisms that drive non-Mendelian inheritance of X chromosomes during spermatogenesis and their impact on population dynamics in species with different breeding systems. In Drosophila and mice, X-linked drivers targeting Y-bearing sperm for elimination or limiting their fitness, tend to confer unfavourable effects, prompting the evolution of suppressors to mitigate their impact. This leads to a complex ongoing evolutionary arms race to maintain an equal balance of males and females. However, in certain insects and nematodes with XX/X0 sex determination, the preferential production of X-bearing sperm through atypical meiosis yields wild-type populations with highly skewed sex ratios, suggesting non-Mendelian transmission of the X may offer selective advantages in these species. Indeed, models suggest X-meiotic drivers could bolster population size and persistence under certain conditions, challenging the conventional view of their detrimental effects. Furthering our understanding of the diverse mechanisms and evolutionary consequences of non-Mendelian transmission of X chromosomes will provide insights into genetic inheritance, sex determination, and population dynamics, with implications for fundamental research and practical applications.


Subject(s)
Population Dynamics , Sex Ratio , X Chromosome , Animals , X Chromosome/genetics , Male , Female , Sex Determination Processes , Spermatogenesis/genetics , Breeding , Mice , Meiosis/genetics , Drosophila/genetics , Humans , Biological Evolution
5.
Genome Res ; 34(7): 997-1007, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39103228

ABSTRACT

We present the first chromosome-level genome assembly of the grasshopper, Locusta migratoria, one of the largest insect genomes. We use coverage differences between females (XX) and males (X0) to identify the X Chromosome gene content, and find that the X Chromosome shows both complete dosage compensation in somatic tissues and an underrepresentation of testis-expressed genes. X-linked gene content from L. migratoria is highly conserved across seven insect orders, namely Orthoptera, Odonata, Phasmatodea, Hemiptera, Neuroptera, Coleoptera, and Diptera, and the 800 Mb grasshopper X Chromosome is homologous to the fly ancestral X Chromosome despite 400 million years of divergence, suggesting either repeated origin of sex chromosomes with highly similar gene content, or long-term conservation of the X Chromosome. We use this broad conservation of the X Chromosome to test for temporal dynamics to Fast-X evolution, and find evidence of a recent burst evolution for new X-linked genes in contrast to slow evolution of X-conserved genes.


Subject(s)
Evolution, Molecular , Genome, Insect , Grasshoppers , X Chromosome , Animals , X Chromosome/genetics , Male , Female , Grasshoppers/genetics , Genes, X-Linked , Chromosomes, Insect/genetics , Locusta migratoria/genetics , Dosage Compensation, Genetic
6.
Mol Biol Evol ; 41(8)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39136558

ABSTRACT

Sex chromosomes display remarkable diversity and variability among vertebrates. Compared with research on the X/Y and Z/W chromosomes, which have long evolutionary histories in mammals and birds, studies on the sex chromosomes at early evolutionary stages are limited. Here, we precisely assembled the genomes of homozygous XX female and YY male Lanzhou catfish (Silurus lanzhouensis) derived from an artificial gynogenetic family and a self-fertilized family, respectively. Chromosome 24 (Chr24) was identified as the sex chromosome based on resequencing data. Comparative analysis of the X and Y chromosomes showed an approximate 320 kb Y-specific region with a Y-specific duplicate of anti-Mullerian hormone type II receptor (amhr2y), which is consistent with findings in 2 other Silurus species but on different chromosomes (Chr24 of Silurus meridionalis and Chr5 of Silurus asotus). Deficiency of amhr2y resulted in male-to-female sex reversal, indicating that amhr2y plays a male-determining role in S. lanzhouensis. Phylogenetic analysis and comparative genomics revealed that the common sex-determining gene amhr2y was initially translocated to Chr24 of the Silurus ancestor along with the expansion of transposable elements. Chr24 was maintained as the sex chromosome in S. meridionalis and S. lanzhouensis, whereas a sex-determining region transition triggered sex chromosome turnover from Chr24 to Chr5 in S. asotus. Additionally, gene duplication, translocation, and degeneration were observed in the Y-specific regions of Silurus species. These findings present a clear case for the early evolutionary trajectory of sex chromosomes, including sex-determining gene origin, repeat sequence expansion, gene gathering and degeneration in sex-determining region, and sex chromosome turnover.


Subject(s)
Catfishes , Sex Determination Processes , Animals , Male , Female , Catfishes/genetics , Evolution, Molecular , Phylogeny , Sex Chromosomes/genetics , Y Chromosome/genetics , Genome , X Chromosome/genetics , Receptors, Peptide , Receptors, Transforming Growth Factor beta
7.
Sci Adv ; 10(32): eadj8862, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39110794

ABSTRACT

Reprogramming somatic cells into induced pluripotent stem cells (iPSCs) requires activation of the pluripotency network and resetting of the epigenome by erasing the epigenetic memory of the somatic state. In female mouse cells, a critical epigenetic reprogramming step is the reactivation of the inactive X chromosome. Despite its importance, a systematic understanding of the regulatory networks linking pluripotency and X-reactivation is missing. Here, we reveal important pathways for pluripotency acquisition and X-reactivation using a genome-wide CRISPR screen during neural precursor to iPSC reprogramming. In particular, we discover that activation of the interferon γ (IFNγ) pathway early during reprogramming accelerates pluripotency acquisition and X-reactivation. IFNγ stimulates STAT3 signaling and the pluripotency network and leads to enhanced TET-mediated DNA demethylation, which consequently boosts X-reactivation. We therefore gain a mechanistic understanding of the role of IFNγ in reprogramming and X-reactivation and provide a comprehensive resource of the molecular networks involved in these processes.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells , Interferon-gamma , Signal Transduction , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Animals , Interferon-gamma/metabolism , Cellular Reprogramming/genetics , Mice , Female , X Chromosome/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Epigenesis, Genetic , DNA Methylation
8.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 155-160, 2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39097881

ABSTRACT

Goats are considered the leading farm animal that has a substantial role in the agricultural sector in the Kurdistan Region of Iraq. No cytological examination has been carried out on them. This experiment aims to identify the Karyotype of the local breeds of domestic goats. This experiment was conducted on the Karyotype and prepared the ideogram of Meriz goats. The determination of the relative length and centromeric index arm ratio of the chromosomes in the breed was achieved by the production of karyotypes. A total of (30)Meriz goats, consisting of (10) males and (20) females, were selected to collect blood samples for a short-term lymphocyte culture. The diploid chromosome count was observed to be (60), consisting of (29) pairs of acrocentric autosomes and one pair of allosomes, specifically the X and Y chromosomes. The acrocentric nature of the X-chromosome and the sub-metacentric nature of the Y-chromosome were identified through scientific investigation. The study observed a variation in the relative length of autosomal chromosomes in Meriz goats, with females ranging from 4.49% to 1.89% and males ranging from (4.53%) to (1.75%). The X-chromosome had a relative length of 3.96 in females, while the Y-chromosome displayed a relative length of (5.05). The findings of this karyological investigation suggest that the chromosomal composition seen in the Meriz goats under examination was within the expected range of normalcy. It is recommended that more cytogenetic analyses be conducted at the population level in order to identify individuals within the Meriz breed population who possesses numerical and/or structural chromosome abnormalities. This research is crucial for enhancing the efficiency of production and reproduction in this breed.


Subject(s)
Breeding , Goats , Karyotyping , Animals , Goats/genetics , Female , Iraq , Male , Karyotype , Cytogenetic Analysis , Y Chromosome/genetics , X Chromosome/genetics
9.
Development ; 151(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39140247

ABSTRACT

Changes in gene dosage can have tremendous evolutionary potential (e.g. whole-genome duplications), but without compensatory mechanisms, they can also lead to gene dysregulation and pathologies. Sex chromosomes are a paradigmatic example of naturally occurring gene dosage differences and their compensation. In species with chromosome-based sex determination, individuals within the same population necessarily show 'natural' differences in gene dosage for the sex chromosomes. In this Review, we focus on the mammalian X chromosome and discuss recent new insights into the dosage-compensation mechanisms that evolved along with the emergence of sex chromosomes, namely X-inactivation and X-upregulation. We also discuss the evolution of the genetic loci and molecular players involved, as well as the regulatory diversity and potentially different requirements for dosage compensation across mammalian species.


Subject(s)
Dosage Compensation, Genetic , Mammals , X Chromosome Inactivation , X Chromosome , Animals , Humans , X Chromosome/genetics , Mammals/genetics , X Chromosome Inactivation/genetics , Gene Dosage , Evolution, Molecular
10.
PLoS Genet ; 20(8): e1011238, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39146391

ABSTRACT

Sex determination in the nematode C. elegans is controlled by the master regulator XOL-1 during embryogenesis. Expression of xol-1 is dependent on the ratio of X chromosomes and autosomes, which differs between XX hermaphrodites and XO males. In males, xol-1 is highly expressed and in hermaphrodites, xol-1 is expressed at very low levels. XOL-1 activity is known to be critical for the proper development of C. elegans males, but its low expression was considered to be of minimal importance in the development of hermaphrodite embryos. Our study reveals that XOL-1 plays an important role as a regulator of developmental timing during hermaphrodite embryogenesis. Using a combination of imaging and bioinformatics techniques, we found that hermaphrodite embryos have an accelerated rate of cell division, as well as a more developmentally advanced transcriptional program when xol-1 is lost. Further analyses reveal that XOL-1 is responsible for regulating the timing of initiation of dosage compensation on the X chromosomes, and the appropriate expression of sex-biased transcriptional programs in hermaphrodites. We found that xol-1 mutant embryos overexpress the H3K9 methyltransferase MET-2 and have an altered H3K9me landscape. Some of these effects of the loss of xol-1 gene were reversed by the loss of met-2. These findings demonstrate that XOL-1 plays an important role as a developmental regulator in embryos of both sexes, and that MET-2 acts as a downstream effector of XOL-1 activity in hermaphrodites.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Embryonic Development , Gene Expression Regulation, Developmental , Sex Determination Processes , X Chromosome , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Male , Female , Embryonic Development/genetics , X Chromosome/genetics , Sex Determination Processes/genetics , Histones/metabolism , Histones/genetics , Dosage Compensation, Genetic , Embryo, Nonmammalian/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Proc Natl Acad Sci U S A ; 121(36): e2412185121, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39190362

ABSTRACT

X chromosome inactivation (XCI) is an epigenetic process that results in the transcriptional silencing of one X chromosome in the somatic cells of females. This phenomenon is common to both eutherian and marsupial mammals, but there are fundamental differences. In eutherians, the X chosen for silencing is random. DNA methylation on the eutherian inactive X is high at transcription start sites (TSSs) and their flanking regions, resulting in universally high DNA methylation. This contrasts XCI in marsupials where the paternally derived X is always silenced, and in which DNA methylation is low at TSSs and flanking regions. Here, we examined the DNA methylation status of the tammar wallaby X chromosome during spermatogenesis to determine the DNA methylation profile of the paternal X prior to and at fertilization. Whole genome enzymatic methylation sequencing was carried out on enriched flow-sorted populations of premeiotic, meiotic, and postmeiotic cells. We observed that the X displayed a pattern of DNA methylation from spermatogonia to mature sperm that reflected the inactive X in female somatic tissue. Therefore, the paternal X chromosome arrives at the egg with a DNA methylation profile that reflects the transcriptionally silent X in adult female somatic tissue. We present this epigenetic signature as a candidate for the long sought-after imprint for paternal XCI in marsupials.


Subject(s)
DNA Methylation , X Chromosome Inactivation , X Chromosome , Animals , X Chromosome Inactivation/genetics , Male , Female , X Chromosome/genetics , Genomic Imprinting , Spermatogenesis/genetics , Macropodidae/genetics , Ovum/metabolism , Marsupialia/genetics , Spermatozoa/metabolism , Epigenesis, Genetic
12.
Commun Biol ; 7(1): 906, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39068254

ABSTRACT

Sexual dimorphism arises because of divergent fitness optima between the sexes. Phenotypic divergence between sexes can range from mild to extreme. Fireflies, bioluminescent beetles, present various degrees of sexual dimorphism, with species showing very mild sexual dimorphism to species presenting female-specific neoteny, posing a unique framework to investigate the evolution of sexually dimorphic traits across species. In this work, we present novel assembled genomes of two firefly species, Lamprohiza splendidula and Luciola italica, species with different degrees of sexual dimorphism. We uncover high synteny conservation of the X-chromosome across ~ 180 Mya and find full X-chromosome dosage compensation in our two fireflies, hinting at common mechanism upregulating the single male X-chromosome. Different degrees of sex-biased expressed genes were found across two body parts showing different proportions of expression conservation between species. Interestingly, we do not find X-chromosome enrichment of sex-biased genes, but retrieve autosomal enrichment of sex-biased genes. We further uncover higher nucleotide diversity in the intronic regions of sex-biased genes, hinting at a maintenance of heterozygosity through sexual selection. We identify different levels of sex-biased gene expression divergence including a set of genes showing conserved sex-biased gene expression between species. Divergent and conserved sex-biased genes are good candidates to test their role in the maintenance of sexually dimorphic traits.


Subject(s)
Dosage Compensation, Genetic , Fireflies , Sex Characteristics , Animals , Female , Male , Fireflies/genetics , Genome, Insect , X Chromosome/genetics , Gene Expression Regulation
13.
Sci Adv ; 10(30): eado5716, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39058769

ABSTRACT

The three-dimensional (3D) organization of chromatin within the nucleus is crucial for gene regulation. However, the 3D architectural features that coordinate the activation of an entire chromosome remain largely unknown. We introduce an omics method, RNA-associated chromatin DNA-DNA interactions, that integrates RNA polymerase II (RNAPII)-mediated regulome with stochastic optical reconstruction microscopy to investigate the landscape of noncoding RNA roX2-associated chromatin topology for gene equalization to achieve dosage compensation. Our findings reveal that roX2 anchors to the target gene transcription end sites (TESs) and spreads in a distinctive boot-shaped configuration, promoting a more open chromatin state for hyperactivation. Furthermore, roX2 arches TES to transcription start sites to enhance transcriptional loops, potentially facilitating RNAPII convoying and connecting proximal promoter-promoter transcriptional hubs for synergistic gene regulation. These TESs cluster as roX2 compartments, surrounded by inactive domains for coactivation of multiple genes within the roX2 territory. In addition, roX2 structures gradually form and scaffold for stepwise coactivation in dosage compensation.


Subject(s)
Chromatin , RNA Polymerase II , X Chromosome , Chromatin/metabolism , Chromatin/genetics , X Chromosome/genetics , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Animals , RNA, Untranslated/genetics , Gene Expression Regulation , Dosage Compensation, Genetic , Promoter Regions, Genetic , Transcription Initiation Site
14.
Nat Commun ; 15(1): 5571, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956041

ABSTRACT

Statin drugs lower blood cholesterol levels for cardiovascular disease prevention. Women are more likely than men to experience adverse statin effects, particularly new-onset diabetes (NOD) and muscle weakness. Here we find that impaired glucose homeostasis and muscle weakness in statin-treated female mice are associated with reduced levels of the omega-3 fatty acid, docosahexaenoic acid (DHA), impaired redox tone, and reduced mitochondrial respiration. Statin adverse effects are prevented in females by administering fish oil as a source of DHA, by reducing dosage of the X chromosome or the Kdm5c gene, which escapes X chromosome inactivation and is normally expressed at higher levels in females than males. As seen in female mice, we find that women experience more severe reductions than men in DHA levels after statin administration, and that DHA levels are inversely correlated with glucose levels. Furthermore, induced pluripotent stem cells from women who developed NOD exhibit impaired mitochondrial function when treated with statin, whereas cells from men do not. These studies identify X chromosome dosage as a genetic risk factor for statin adverse effects and suggest DHA supplementation as a preventive co-therapy.


Subject(s)
Docosahexaenoic Acids , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Mitochondria , X Chromosome , Animals , Female , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Humans , X Chromosome/genetics , Docosahexaenoic Acids/pharmacology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Gene Dosage , Mice, Inbred C57BL , Blood Glucose/metabolism , Blood Glucose/drug effects , Glucose/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/chemically induced , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism
15.
Cell ; 187(14): 3541-3562.e51, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38996487

ABSTRACT

Analyses of ancient DNA typically involve sequencing the surviving short oligonucleotides and aligning to genome assemblies from related, modern species. Here, we report that skin from a female woolly mammoth (†Mammuthus primigenius) that died 52,000 years ago retained its ancient genome architecture. We use PaleoHi-C to map chromatin contacts and assemble its genome, yielding 28 chromosome-length scaffolds. Chromosome territories, compartments, loops, Barr bodies, and inactive X chromosome (Xi) superdomains persist. The active and inactive genome compartments in mammoth skin more closely resemble Asian elephant skin than other elephant tissues. Our analyses uncover new biology. Differences in compartmentalization reveal genes whose transcription was potentially altered in mammoths vs. elephants. Mammoth Xi has a tetradic architecture, not bipartite like human and mouse. We hypothesize that, shortly after this mammoth's death, the sample spontaneously freeze-dried in the Siberian cold, leading to a glass transition that preserved subfossils of ancient chromosomes at nanometer scale.


Subject(s)
Genome , Mammoths , Skin , Animals , Mammoths/genetics , Genome/genetics , Female , Elephants/genetics , Chromatin/genetics , Fossils , DNA, Ancient/analysis , Mice , Humans , X Chromosome/genetics
16.
G3 (Bethesda) ; 14(9)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39001870

ABSTRACT

In organisms with the XY sex-determination system, there is an imbalance in the inheritance and transmission of the X chromosome between males and females. Unlike an autosomal allele, an X-linked recessive allele in a female will have phenotypic effects on its male counterpart. Thus, genes located on the X chromosome are of particular interest to researchers in molecular evolution and genetics. Here we present a model for selection with two alleles of X-linkage to understand fitness components associated with genes on the X chromosome. We apply this model to the fitness analysis of an X-linked gene, OdsH (16D), in the fruit fly Drosophila melanogaster. The function of OdsH is involved in sperm production and the gene is rapidly evolving under positive selection. Using site-directed gene targeting, we generated functional and defective OdsH variants tagged with the eye-color marker gene white. We compare the allele frequency changes of the two OdsH variants, each directly competing against a wild-type OdsH allele in concurrent but separate experimental populations. After 20 generations, the two genetically modified OdsH variants displayed a 40% difference in allele frequencies, with the functional OdsH variant demonstrating an advantage over the defective variant. Using maximum likelihood estimation, we determined the fitness components associated with the OdsH alleles in males and females. Our analysis revealed functional aspects of the fitness determinants associated with OdsH, and that sex-specific fertility and viability consequences both contribute to selection on an X-linked gene.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Homeodomain Proteins , Animals , Female , Male , Alleles , Drosophila melanogaster/genetics , Drosophila Proteins/genetics , Gene Frequency , Genes, X-Linked , Genetic Fitness , Genetic Linkage , Models, Genetic , Selection, Genetic , X Chromosome/genetics , Homeodomain Proteins/genetics
17.
Cell Genom ; 4(8): 100607, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-38996479

ABSTRACT

Chondrichthyes is an important lineage to reconstruct the evolutionary history of vertebrates. Here, we analyzed genome synteny for six chondrichthyan chromosome-level genomes. Our comparative analysis reveals a slow evolutionary rate of chromosomal changes, with infrequent but independent fusions observed in sharks, skates, and chimaeras. The chondrichthyan common ancestor had a proto-vertebrate-like karyotype, including the presence of 18 microchromosome pairs. The X chromosome is a conversed microchromosome shared by all sharks, suggesting a likely common origin of the sex chromosome at least 181 million years ago. We characterized the Y chromosomes of two sharks that are highly differentiated from the X except for a small young evolutionary stratum and a small pseudoautosomal region. We found that shark sex chromosomes lack global dosage compensation but that dosage-sensitive genes are locally compensated. Our study on shark chromosome evolution enhances our understanding of shark sex chromosomes and vertebrate chromosome evolution.


Subject(s)
Evolution, Molecular , Genomics , Karyotype , Sex Chromosomes , Sharks , Animals , Sharks/genetics , Genomics/methods , Sex Chromosomes/genetics , Male , Female , Synteny/genetics , Phylogeny , Dosage Compensation, Genetic , X Chromosome/genetics , Genome/genetics
18.
Nat Genet ; 56(8): 1737-1749, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39039278

ABSTRACT

The structural maintenance of chromosome (SMC) complexes-cohesin and condensins-are crucial for chromosome separation and compaction during cell division. During the interphase, mammalian cohesins additionally fold the genome into loops and domains. Here we show that, in Caenorhabditis elegans, a species with holocentric chromosomes, condensin I is the primary, long-range loop extruder. The loss of condensin I and its X-specific variant, condensin IDC, leads to genome-wide decompaction, chromosome mixing and disappearance of X-specific topologically associating domains, while reinforcing fine-scale epigenomic compartments. In addition, condensin I/IDC inactivation led to the upregulation of X-linked genes and unveiled nuclear bodies grouping together binding sites for the X-targeting loading complex of condensin IDC. C. elegans condensin I/IDC thus uniquely organizes holocentric interphase chromosomes, akin to cohesin in mammals, as well as regulates X-chromosome gene expression.


Subject(s)
Adenosine Triphosphatases , Caenorhabditis elegans Proteins , Caenorhabditis elegans , DNA-Binding Proteins , Multiprotein Complexes , X Chromosome , Animals , Caenorhabditis elegans/genetics , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , X Chromosome/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Cohesins , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Interphase/genetics , Genome, Helminth , Genes, X-Linked , Chromosomes/genetics
19.
Curr Opin Genet Dev ; 87: 102235, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39053028

ABSTRACT

To regulate gene expression, the macromolecular components of the mammalian interphase nucleus are spatially organized into a myriad of functional compartments. Over the past decade, increasingly sophisticated genomics, microscopy, and functional approaches have probed this organization in unprecedented detail. These investigations have linked chromatin-associated noncoding RNAs to specific nuclear compartments and uncovered mechanisms by which these RNAs establish such domains. In this review, we focus on the long non-coding RNA Xist and summarize new evidence demonstrating the significance of chromatin reconfiguration in creating the inactive X-chromosome compartment. Differences in chromatin compaction correlate with distinct levels of gene repression on the X-chromosome, potentially explaining how human XIST can induce chromosome-wide dampening and silencing of gene expression at different stages of human development.


Subject(s)
Dosage Compensation, Genetic , RNA, Long Noncoding , X Chromosome , Humans , Animals , Dosage Compensation, Genetic/genetics , X Chromosome/genetics , RNA, Long Noncoding/genetics , Chromatin/genetics , X Chromosome Inactivation/genetics , RNA, Untranslated/genetics , Mammals/genetics
20.
Nat Genet ; 56(8): 1678-1688, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39060501

ABSTRACT

X chromosome inactivation (XCI) generates clonal heterogeneity within XX individuals. Combined with sequence variation between human X chromosomes, XCI gives rise to intra-individual clonal diversity, whereby two sets of clones express mutually exclusive sequence variants present on one or the other X chromosome. Here we ask whether such clones merely co-exist or potentially interact with each other to modulate the contribution of X-linked diversity to organismal development. Focusing on X-linked coding variation in the human STAG2 gene, we show that Stag2variant clones contribute to most tissues at the expected frequencies but fail to form lymphocytes in Stag2WT Stag2variant mouse models. Unexpectedly, the absence of Stag2variant clones from the lymphoid compartment is due not solely to cell-intrinsic defects but requires continuous competition by Stag2WT clones. These findings show that interactions between epigenetically diverse clones can operate in an XX individual to shape the contribution of X-linked genetic diversity in a cell-type-specific manner.


Subject(s)
Chromosomes, Human, X , Genes, X-Linked , Genetic Variation , X Chromosome Inactivation , Humans , Animals , X Chromosome Inactivation/genetics , Mice , Chromosomes, Human, X/genetics , Female , Cell Cycle Proteins/genetics , Antigens, Nuclear/genetics , Lymphocytes/metabolism , X Chromosome/genetics , Cohesins
SELECTION OF CITATIONS
SEARCH DETAIL