Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 414
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
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.
Nat Commun ; 15(1): 4781, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38839766

ABSTRACT

Most vertebrates develop distinct females and males, where sex is determined by repeatedly evolved environmental or genetic triggers. Undifferentiated sex chromosomes and large genomes have caused major knowledge gaps in amphibians. Only a single master sex-determining gene, the dmrt1-paralogue (dm-w) of female-heterogametic clawed frogs (Xenopus; ZW♀/ZZ♂), is known across >8740 species of amphibians. In this study, by combining chromosome-scale female and male genomes of a non-model amphibian, the European green toad, Bufo(tes) viridis, with ddRAD- and whole genome pool-sequencing, we reveal a candidate master locus, governing a male-heterogametic system (XX♀/XY♂). Targeted sequencing across multiple taxa uncovered structural X/Y-variation in the 5'-regulatory region of the gene bod1l, where a Y-specific non-coding RNA (ncRNA-Y), only expressed in males, suggests that this locus initiates sex-specific differentiation. Developmental transcriptomes and RNA in-situ hybridization show timely and spatially relevant sex-specific ncRNA-Y and bod1l-gene expression in primordial gonads. This coincided with differential H3K4me-methylation in pre-granulosa/pre-Sertoli cells, pointing to a specific mechanism of amphibian sex determination.


Subject(s)
Sex Determination Processes , X Chromosome , Y Chromosome , Animals , Male , Female , Sex Determination Processes/genetics , Y Chromosome/genetics , X Chromosome/genetics , Amphibians/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , RNA, Untranslated/genetics , Genome , Evolution, Molecular
14.
Cell Death Dis ; 15(6): 396, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38839795

ABSTRACT

Klinefelter syndrome (47,XXY) causes infertility with a testicular histology comprising two types of Sertoli cell-only tubules, representing mature and immature-like Sertoli cells, and occasionally focal spermatogenesis. Here, we show that the immature-like Sertoli cells highly expressed XIST and had two X-chromosomes, while the mature Sertoli cells lacked XIST expression and had only one X-chromosome. Sertoli cells supporting focal spermatogenesis also lacked XIST expression and the additional X-chromosome, while the spermatogonia expressed XIST despite having only one X-chromosome. XIST was expressed in Sertoli cells until puberty, where a gradual loss was observed. Our results suggest that a micro-mosaic loss of the additional X-chromosome is needed for Sertoli cells to mature and to allow focal spermatogenesis.


Subject(s)
Klinefelter Syndrome , RNA, Long Noncoding , Sertoli Cells , Spermatogenesis , Klinefelter Syndrome/genetics , Klinefelter Syndrome/pathology , Klinefelter Syndrome/metabolism , Male , Sertoli Cells/metabolism , Sertoli Cells/pathology , Spermatogenesis/genetics , Animals , Humans , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Chromosomes, Human, X/genetics , X Chromosome/genetics
15.
Genes (Basel) ; 15(6)2024 May 23.
Article in English | MEDLINE | ID: mdl-38927599

ABSTRACT

The sex chromosomes of skinks are usually poorly differentiated and hardly distinguished by cytogenetic methods. Therefore, identifying sex chromosomes in species lacking easily recognizable heteromorphic sex chromosomes is necessary to fully understand sex chromosome diversity. In this paper, we employed cytogenetics, sex quantification of genes, and transcriptomic approaches to characterize the sex chromosomes in Plestiodon elegans. Cytogenetic examination of metaphases revealed a diploid number of 2n = 26, consisting of 12 macrochromosomes and 14 microchromosomes, with no significant heteromorphic chromosome pairs, speculating that the sex chromosomes may be homomorphic or poorly differentiated. The results of the sex quantification of genes showed that Calumenin (calu), COPI coat complex subunit γ 2 (copg2), and Smoothened (smo) were at half the dose in males as in females, suggesting that they are on the X chromosome. Transcriptomic data analysis from the gonads yielded the excess expression male-specific genes (n = 16), in which five PCR molecular markers were developed. Restricting the observed heterozygosity to males suggests the presence of homomorphic sex chromosomes in P. elegans, XX/XY. This is the first breakthrough in the study of the sex chromosomes of Plestiodon.


Subject(s)
Transcriptome , Animals , Male , Female , Transcriptome/genetics , Sex Chromosomes/genetics , X Chromosome/genetics , Gonads/metabolism , Cytogenetic Analysis/methods
16.
Nat Commun ; 15(1): 4983, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862555

ABSTRACT

Engineered sex ratio distorters (SRDs) have been proposed as a powerful component of genetic control strategies designed to suppress harmful insect pests. Two types of CRISPR-based SRD mechanisms have been proposed: X-shredding, which eliminates X-bearing sperm, and X-poisoning, which eliminates females inheriting disrupted X-chromosomes. These differences can have a profound impact on the population dynamics of SRDs when linked to the Y-chromosome: an X-shredder is invasive, constituting a classical meiotic Y-drive, whereas X-poisoning is self-limiting, unable to invade but also insulated from selection. Here, we establish X-poisoning strains in the malaria vector Anopheles gambiae targeting three X-linked genes during spermatogenesis, resulting in male bias. We find that sex distortion is primarily driven by a loss of X-bearing sperm, with limited evidence for postzygotic lethality of female progeny. By leveraging a Drosophila melanogaster model, we show unambiguously that engineered SRD traits can operate differently in these two insects. Unlike X-shredding, X-poisoning could theoretically operate at early stages of spermatogenesis. We therefore explore premeiotic Cas9 expression to target the mosquito X-chromosome. We find that, by pre-empting the onset of meiotic sex chromosome inactivation, this approach may enable the development of Y-linked SRDs if mutagenesis of spermatogenesis-essential genes is functionally balanced.


Subject(s)
Anopheles , Drosophila melanogaster , Gene Drive Technology , Sex Ratio , Spermatogenesis , X Chromosome , Animals , Male , Female , Anopheles/genetics , X Chromosome/genetics , Drosophila melanogaster/genetics , Gene Drive Technology/methods , Spermatogenesis/genetics , Mosquito Vectors/genetics , Genes, X-Linked , CRISPR-Cas Systems , Spermatozoa/metabolism , Animals, Genetically Modified
17.
Sci Rep ; 14(1): 13466, 2024 06 12.
Article in English | MEDLINE | ID: mdl-38866815

ABSTRACT

CRISPR-Cas9 technology has facilitated development of strategies that can potentially provide more humane and effective methods to control invasive vertebrate species, such as mice. One promising strategy is X chromosome shredding which aims to bias offspring towards males, resulting in a gradual and unsustainable decline of females. This method has been explored in insects with encouraging results. Here, we investigated this strategy in Mus musculus by targeting repeat DNA sequences on the X chromosome with the aim of inducing sufficient DNA damage to specifically eliminate X chromosome-bearing sperm during gametogenesis. We tested three different guide RNAs (gRNAs) targeting different repeats on the X chromosome, together with three male germline-specific promoters for inducing Cas9 expression at different stages of spermatogenesis. A modest bias towards mature Y-bearing sperm was detected in some transgenic males, although this did not translate into significant male-biasing of offspring. Instead, cleavage of the X chromosome during meiosis typically resulted in a spermatogenic block, manifest as small testes volume, empty tubules, low sperm concentration, and sub/infertility. Our study highlights the importance of controlling the timing of CRISPR-Cas9 activity during mammalian spermatogenesis and the sensitivity of spermatocytes to X chromosome disruption.


Subject(s)
CRISPR-Cas Systems , Spermatogenesis , X Chromosome , Animals , Male , Mice , Spermatogenesis/genetics , X Chromosome/genetics , Female , RNA, Guide, CRISPR-Cas Systems/genetics , Spermatozoa/metabolism , Mice, Transgenic , Meiosis/genetics
18.
Evol Anthropol ; 33(4): e22038, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38877873

ABSTRACT

Vertebrates exhibit sexual dimorphism in response to infectious diseases and in morbidity and mortality rates to various pathogens. Females are generally more immunocompetent than males, despite their increased reproductive burden and the immunosuppressive effects of gestation. In addition, females generally have lower incidences of cancer compared to males; however, they have higher rates of autoimmune disorders. These sex differences may be a result of life history differences, sexual selection, genetics, and/or the physiological effects of hormones. As highly social mammals with complex life histories, primates offer a unique opportunity to investigate the evolution of enhanced female immunocompetence. This review aims to examine the evidence of this immunity gap, understand current hypotheses for its evolution, and explore the potential role of X chromosome specific genes and heterozygosity within this framework.


Subject(s)
Primates , Animals , Female , Primates/immunology , Male , Sex Characteristics , Biological Evolution , Immunocompetence , X Chromosome , Anthropology, Physical
19.
Genetics ; 227(4)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38884610

ABSTRACT

53BP1 plays a crucial role in regulating DNA damage repair pathway choice and checkpoint signaling in somatic cells; however, its role in meiosis has remained enigmatic. In this study, we demonstrate that the Caenorhabditis elegans ortholog of 53BP1, HSR-9, associates with chromatin in both proliferating and meiotic germ cells. Notably, HSR-9 is enriched on the X chromosome pair in pachytene oogenic germ cells. HSR-9 is also present at kinetochores during both mitotic and meiotic divisions but does not appear to be essential for monitoring microtubule-kinetochore attachments or tension. Using cytological markers of different steps in recombinational repair, we found that HSR-9 influences the processing of a subset of meiotic double-stranded breaks into COSA-1-marked crossovers. Additionally, HSR-9 plays a role in meiotic X chromosome segregation under conditions where X chromosomes fail to pair, synapse, and recombine. Together, these results highlight that chromatin-associated HSR-9 has both conserved and unique functions in the regulation of meiotic chromosome behavior.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Chromatin , Chromosome Segregation , Germ Cells , Meiosis , X Chromosome , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , X Chromosome/genetics , Chromatin/metabolism , Chromatin/genetics , Germ Cells/metabolism , Recombinational DNA Repair , DNA Breaks, Double-Stranded
20.
Anim Biotechnol ; 35(1): 2323592, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38770771

ABSTRACT

Nucleic acid aptamers have been used in the past for the development of diagnostic methods against a number of targets such as bacteria, pesticides, cancer cells etc. In the present study, six rounds of Cell-SELEX were performed on a ssDNA aptamer library against X-enriched sperm cells from Sahiwal breed cattle. Sequencing was used to examine the aptamer sequences that shown affinity for sperm carrying the X chromosome in order to find any possible X-sperm-specific sequences. Out of 35 identified sequences, 14 were selected based on bioinformatics analysis like G-Score and Mfold structures. Further validation of their specificity was done via fluorescence microscopy. The interaction of biotinylated-aptamer with sperm was also determined by visualizing the binding of streptavidin coated magnetic beads on the head region of the sperm under bright field microscopy. Finally, a real-time experiment was designed for the validation of X-sperm enrichment by synthesized aptamer sequences. Among the studied sequences, aptamer 29a exhibited a higher affinity for X sperm compared to Y sperm in a mixed population of sperm cells. By using aptamer sequence 29a, we obtained an enrichment of 70% for X chromosome bearing sperm cells.


Subject(s)
Aptamers, Nucleotide , SELEX Aptamer Technique , Spermatozoa , X Chromosome , Male , Animals , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/genetics , Spermatozoa/chemistry , Cattle , X Chromosome/genetics , SELEX Aptamer Technique/methods
SELECTION OF CITATIONS
SEARCH DETAIL