Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.873
Filtrar
1.
Science ; 385(6715): 1347-1354, 2024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39298575

RESUMO

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.


Assuntos
Mecanismo Genético de Compensação de Dose , Lagartos , RNA Longo não Codificante , Cromossomo X , Animais , Feminino , Masculino , Acetilação , Epigênese Genética , Evolução Molecular , Histonas/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Cromossomo X/genética , Lagartos/genética
2.
Development ; 151(15)2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39140247

RESUMO

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.


Assuntos
Mecanismo Genético de Compensação de Dose , Mamíferos , Inativação do Cromossomo X , Cromossomo X , Animais , Humanos , Cromossomo X/genética , Mamíferos/genética , Inativação do Cromossomo X/genética , Dosagem de Genes , Evolução Molecular
3.
PLoS Genet ; 20(8): e1011238, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39146391

RESUMO

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.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Processos de Determinação Sexual , Cromossomo X , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/crescimento & desenvolvimento , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Masculino , Feminino , Desenvolvimento Embrionário/genética , Cromossomo X/genética , Processos de Determinação Sexual/genética , Histonas/metabolismo , Histonas/genética , Mecanismo Genético de Compensação de Dose , Embrião não Mamífero/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
Genome Res ; 34(7): 997-1007, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39103228

RESUMO

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.


Assuntos
Evolução Molecular , Genoma de Inseto , Gafanhotos , Cromossomo X , Animais , Cromossomo X/genética , Masculino , Feminino , Gafanhotos/genética , Genes Ligados ao Cromossomo X , Cromossomos de Insetos/genética , Locusta migratoria/genética , Mecanismo Genético de Compensação de Dose
5.
Cell Genom ; 4(8): 100607, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-38996479

RESUMO

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.


Assuntos
Evolução Molecular , Genômica , Cariótipo , Cromossomos Sexuais , Tubarões , Animais , Tubarões/genética , Genômica/métodos , Cromossomos Sexuais/genética , Masculino , Feminino , Sintenia/genética , Filogenia , Mecanismo Genético de Compensação de Dose , Cromossomo X/genética , Genoma/genética
6.
Proc Natl Acad Sci U S A ; 121(32): e2322360121, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-39074288

RESUMO

Heteromorphic sex chromosomes (XY or ZW) present problems of gene dosage imbalance between sexes and with autosomes. A need for dosage compensation has long been thought to be critical in vertebrates. However, this was questioned by findings of unequal mRNA abundance measurements in monotreme mammals and birds. Here, we demonstrate unbalanced mRNA levels of X genes in platypus males and females and a correlation with differential loading of histone modifications. We also observed unbalanced transcripts of Z genes in chicken. Surprisingly, however, we found that protein abundance ratios were 1:1 between the sexes in both species, indicating a post-transcriptional layer of dosage compensation. We conclude that sex chromosome output is maintained in chicken and platypus (and perhaps many other non therian vertebrates) via a combination of transcriptional and post-transcriptional control, consistent with a critical importance of sex chromosome dosage compensation.


Assuntos
Galinhas , Mecanismo Genético de Compensação de Dose , Ornitorrinco , Cromossomos Sexuais , Animais , Galinhas/genética , Cromossomos Sexuais/genética , Masculino , Feminino , Ornitorrinco/genética , Transcrição Gênica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
7.
Commun Biol ; 7(1): 906, 2024 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-39068254

RESUMO

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.


Assuntos
Mecanismo Genético de Compensação de Dose , Vaga-Lumes , Caracteres Sexuais , Animais , Feminino , Masculino , Vaga-Lumes/genética , Genoma de Inseto , Cromossomo X/genética , Regulação da Expressão Gênica
8.
Sci Adv ; 10(30): eado5716, 2024 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-39058769

RESUMO

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.


Assuntos
Cromatina , RNA Polimerase II , Cromossomo X , Cromatina/metabolismo , Cromatina/genética , Cromossomo X/genética , RNA Polimerase II/metabolismo , RNA Polimerase II/genética , Animais , RNA não Traduzido/genética , Regulação da Expressão Gênica , Mecanismo Genético de Compensação de Dose , Regiões Promotoras Genéticas , Sítio de Iniciação de Transcrição
9.
Curr Opin Genet Dev ; 87: 102235, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39053028

RESUMO

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.


Assuntos
Mecanismo Genético de Compensação de Dose , RNA Longo não Codificante , Cromossomo X , Humanos , Animais , Mecanismo Genético de Compensação de Dose/genética , Cromossomo X/genética , RNA Longo não Codificante/genética , Cromatina/genética , Inativação do Cromossomo X/genética , RNA não Traduzido/genética , Mamíferos/genética
10.
Nat Struct Mol Biol ; 31(10): 1589-1600, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38834912

RESUMO

XIST (X-inactive specific transcript) long noncoding RNA (lncRNA) is responsible for X chromosome inactivation (XCI) in placental mammals, yet it accumulates on both X chromosomes in human female preimplantation embryos without triggering X chromosome silencing. The XACT (X-active coating transcript) lncRNA coaccumulates with XIST on active X chromosomes and may antagonize XIST function. Here, we used human embryonic stem cells in a naive state of pluripotency to assess the function of XIST and XACT in shaping the X chromosome chromatin and transcriptional landscapes during preimplantation development. We show that XIST triggers the deposition of polycomb-mediated repressive histone modifications and dampens the transcription of most X-linked genes in a SPEN-dependent manner, while XACT deficiency does not significantly affect XIST activity or X-linked gene expression. Our study demonstrates that XIST is functional before XCI, confirms the existence of a transient process of X chromosome dosage compensation and reveals that XCI and dampening rely on the same set of factors.


Assuntos
Cromossomos Humanos X , RNA Longo não Codificante , Inativação do Cromossomo X , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Cromossomos Humanos X/genética , Cromossomos Humanos X/metabolismo , Feminino , Células-Tronco Embrionárias Humanas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Desenvolvimento Embrionário/genética , Cromatina/metabolismo , Mecanismo Genético de Compensação de Dose , Genes Ligados ao Cromossomo X
11.
Genetics ; 227(3)2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38718207

RESUMO

Organisms with differentiated sex chromosomes must accommodate unequal gene dosage in males and females. Male fruit flies increase X-linked gene expression to compensate for hemizygosity of their single X chromosome. Full compensation requires localization of the Male-Specific Lethal (MSL) complex to active genes on the male X, where it modulates chromatin to elevate expression. The mechanisms that identify X chromatin are poorly understood. The euchromatic X is enriched for AT-rich, ∼359 bp satellites termed the 1.688X repeats. Autosomal insertions of 1.688X DNA enable MSL recruitment to nearby genes. Ectopic expression of dsRNA from one of these repeats produces siRNA and partially restores X-localization of MSLs in males with defective X recognition. Surprisingly, expression of double-stranded RNA from three other 1.688X repeats failed to rescue males. We reconstructed dsRNA-expressing transgenes with sequence from two of these repeats and identified phasing of repeat DNA, rather than sequence or orientation, as the factor that determines rescue of males with defective X recognition. Small RNA sequencing revealed that siRNA was produced in flies with a transgene that rescues, but not in those carrying a transgene with the same repeat but different phasing. We demonstrate that pericentromeric X heterochromatin promotes X recognition through a maternal effect, potentially mediated by small RNA from closely related heterochromatic repeats. This suggests that the sources of siRNAs promoting X recognition are highly redundant. We propose that enrichment of satellite repeats on Drosophilid X chromosomes facilitates the rapid evolution of differentiated sex chromosomes by marking the X for compensation.


Assuntos
Mecanismo Genético de Compensação de Dose , RNA Interferente Pequeno , Cromossomo X , Animais , Masculino , Cromossomo X/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Feminino , Sequências Repetitivas de Ácido Nucleico , Drosophila melanogaster/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
12.
Nature ; 630(8015): 149-157, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38778096

RESUMO

Accessing the natural genetic diversity of species unveils hidden genetic traits, clarifies gene functions and allows the generalizability of laboratory findings to be assessed. One notable discovery made in natural isolates of Saccharomyces cerevisiae is that aneuploidy-an imbalance in chromosome copy numbers-is frequent1,2 (around 20%), which seems to contradict the substantial fitness costs and transient nature of aneuploidy when it is engineered in the laboratory3-5. Here we generate a proteomic resource and merge it with genomic1 and transcriptomic6 data for 796 euploid and aneuploid natural isolates. We find that natural and lab-generated aneuploids differ specifically at the proteome. In lab-generated aneuploids, some proteins-especially subunits of protein complexes-show reduced expression, but the overall protein levels correspond to the aneuploid gene dosage. By contrast, in natural isolates, more than 70% of proteins encoded on aneuploid chromosomes are dosage compensated, and average protein levels are shifted towards the euploid state chromosome-wide. At the molecular level, we detect an induction of structural components of the proteasome, increased levels of ubiquitination, and reveal an interdependency of protein turnover rates and attenuation. Our study thus highlights the role of protein turnover in mediating aneuploidy tolerance, and shows the utility of exploiting the natural diversity of species to attain generalizable molecular insights into complex biological processes.


Assuntos
Aneuploidia , Complexo de Endopeptidases do Proteassoma , Proteólise , Proteoma , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Mecanismo Genético de Compensação de Dose , Variação Genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Proteoma/metabolismo , Proteoma/genética , Proteômica , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Ubiquitinação , Perfilação da Expressão Gênica , Genômica
13.
Genes (Basel) ; 15(5)2024 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-38790266

RESUMO

Genomic imbalance in aneuploidy is often detrimental to organisms. To gain insight into the molecular basis of aneuploidies in humans, we analyzed transcriptome data from several autosomal and sex chromosome aneuploidies. The results showed that in human aneuploid cells, genes located on unvaried chromosomes are inversely or proportionally trans-modulated, while a subset of genes on the varied chromosomes are compensated. Less genome-wide modulation is found for sex chromosome aneuploidy compared with autosomal aneuploidy due to X inactivation and the retention of dosage sensitive regulators on both sex chromosomes to limit the effective dosage change. We also found that lncRNA and mRNA can have different responses to aneuploidy. Furthermore, we analyzed the relationship between dosage-sensitive transcription factors and their targets, which illustrated the modulations and indicates genomic imbalance is related to stoichiometric changes in components of gene regulatory complexes.In summary, this study demonstrates the existence of trans-acting effects and compensation mechanisms in human aneuploidies and contributes to our understanding of gene expression regulation in unbalanced genomes and disease states.


Assuntos
Aneuploidia , Humanos , Regulação da Expressão Gênica , RNA Longo não Codificante/genética , Mecanismo Genético de Compensação de Dose , Transcriptoma/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Genoma Humano
14.
Trends Genet ; 40(7): 564-579, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38677904

RESUMO

Progressive recombination loss is a common feature of sex chromosomes. Yet, the evolutionary drivers of this phenomenon remain a mystery. For decades, differences in trait optima between sexes (sexual antagonism) have been the favoured hypothesis, but convincing evidence is lacking. Recent years have seen a surge of alternative hypotheses to explain progressive extensions and maintenance of recombination suppression: neutral accumulation of sequence divergence, selection of nonrecombining fragments with fewer deleterious mutations than average, sheltering of recessive deleterious mutations by linkage to heterozygous alleles, early evolution of dosage compensation, and constraints on recombination restoration. Here, we explain these recent hypotheses and dissect their assumptions, mechanisms, and predictions. We also review empirical studies that have brought support to the various hypotheses.


Assuntos
Recombinação Genética , Cromossomos Sexuais , Cromossomos Sexuais/genética , Animais , Humanos , Evolução Molecular , Masculino , Feminino , Seleção Genética/genética , Mutação , Mecanismo Genético de Compensação de Dose , Modelos Genéticos
15.
PLoS Biol ; 22(4): e3002605, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38687805

RESUMO

Although sex chromosomes have evolved from autosomes, they often have unusual regulatory regimes that are sex- and cell-type-specific such as dosage compensation (DC) and meiotic sex chromosome inactivation (MSCI). The molecular mechanisms and evolutionary forces driving these unique transcriptional programs are critical for genome evolution but have been, in the case of MSCI in Drosophila, subject to continuous debate. Here, we take advantage of the younger sex chromosomes in D. miranda (XR and the neo-X) to infer how former autosomes acquire sex-chromosome-specific regulatory programs using single-cell and bulk RNA sequencing and ribosome profiling, in a comparative evolutionary context. We show that contrary to mammals and worms, the X down-regulation through germline progression is most consistent with the shutdown of DC instead of MSCI, resulting in half gene dosage at the end of meiosis for all 3 X's. Moreover, lowly expressed germline and meiotic genes on the neo-X are ancestrally lowly expressed, instead of acquired suppression after sex linkage. For the young neo-X, DC is incomplete across all tissue and cell types and this dosage imbalance is rescued by contributions from Y-linked gametologs which produce transcripts that are translated to compensate both gene and protein dosage. We find an excess of previously autosomal testis genes becoming Y-specific, showing that the neo-Y and its masculinization likely resolve sexual antagonism. Multicopy neo-sex genes are predominantly expressed during meiotic stages of spermatogenesis, consistent with their amplification being driven to interfere with mendelian segregation. Altogether, this study reveals germline regulation of evolving sex chromosomes and elucidates the consequences these unique regulatory mechanisms have on the evolution of sex chromosome architecture.


Assuntos
Drosophila , Células Germinativas , Meiose , RNA-Seq , Cromossomos Sexuais , Análise de Célula Única , Testículo , Animais , Masculino , Testículo/metabolismo , Cromossomos Sexuais/genética , Análise de Célula Única/métodos , Células Germinativas/metabolismo , Drosophila/genética , Drosophila/metabolismo , RNA-Seq/métodos , Meiose/genética , Mecanismo Genético de Compensação de Dose , Evolução Molecular , Feminino , Cromossomo X/genética , Análise da Expressão Gênica de Célula Única
16.
G3 (Bethesda) ; 14(6)2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38491905

RESUMO

Drosophila melanogaster males have one X chromosome while females have two. This creates an imbalance in X:A gene dosage between the sexes. This imbalance is corrected by increasing transcription from male X-linked genes approximately 2-fold. This process involves the Male-Specific Lethal (MSL) complex, which is recruited to Chromatin Entry Sites (CES) and transcribed X-linked genes, where it modifies chromatin to increase expression. Repetitive sequences strikingly enriched in X euchromatin, the 1.688X satellite repeats, also promote recruitment of the MSL complex to nearby genes. Unlike CES, the 1.688X repeats do not recruit the MSL complex directly. The genetic architecture of recruitment by these DNA elements remains speculative. To facilitate dissection of the mechanism of recruitment, we developed a luciferase reporter system for recruitment of compensation to an autosome. The system was validated by knock down of genes known to participate in compensation. Knock down of factors genetically linked to X recognition reveals that 1.688X repeats recruit through a different mechanism than the CES. Our findings suggest that 1.688X repeats play a larger role during embryogenesis, whereas the contribution of 1.688X repeats and CES is equivalent later in development. Our studies also reveal unexpected complexity and potential interdependence of recruiting elements.


Assuntos
Cromatina , Proteínas de Drosophila , Drosophila melanogaster , Cromossomo X , Animais , Drosophila melanogaster/genética , Cromossomo X/genética , Cromatina/metabolismo , Cromatina/genética , Masculino , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Feminino , Genes Ligados ao Cromossomo X , Mecanismo Genético de Compensação de Dose
17.
Open Biol ; 14(3): 230270, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38471568

RESUMO

The Drosophila male-specific lethal (MSL) complex binds to the male X chromosome to activate transcription. It comprises five proteins (MSL1, MSL2, MSL3, male absent on the first (MOF), and maleless (MLE)) and two long noncoding RNAs (lncRNAs; roX1 and roX2). The MLE helicase remodels the roX lncRNAs, enabling the lncRNA-mediated assembly of the Drosophila dosage compensation complex. MSL2 is expressed only in males and interacts with the N-terminal zinc finger of the transcription factor chromatin-linked adapter for MSL proteins (CLAMP), which is important for the specific recruitment of the MSL complex to the male X chromosome. Here, we found that MLE's unstructured C-terminal region interacts with the sixth zinc-finger domain of CLAMP. In vitro, 4-5 zinc fingers are critical for the specific DNA-binding of CLAMP with GA repeats, which constitute the core motif at the high affinity binding sites for MSL proteins. Deleting the CLAMP binding region in MLE decreases the association of MSL proteins with the male X chromosome and increases male lethality. These results suggest that interactions of unstructured regions in MSL2 and MLE with CLAMP zinc finger domains are important for the specific recruitment of the MSL complex to the male X chromosome.


Assuntos
Proteínas de Drosophila , RNA Longo não Codificante , Animais , Masculino , Drosophila/metabolismo , Proteínas de Ligação a DNA/metabolismo , Drosophila melanogaster/genética , Proteínas de Drosophila/metabolismo , RNA Longo não Codificante/metabolismo , Proteínas Nucleares/metabolismo , Mecanismo Genético de Compensação de Dose , Dedos de Zinco , Cromossomo X/metabolismo
18.
PLoS Genet ; 20(3): e1010719, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38457441

RESUMO

DNA methylation is a key regulator of eukaryote genomes, and is of particular relevance in the regulation of gene expression on the sex chromosomes, with a key role in dosage compensation in mammalian XY systems. In the case of birds, dosage compensation is largely absent, with it being restricted to two small Male Hyper-Methylated (MHM) regions on the Z chromosome. To investigate how variation in DNA methylation is regulated on the Z chromosome we utilised a wild x domestic advanced intercross in the chicken, with both hypothalamic methylomes and transcriptomes assayed in 124 individuals. The relatively large numbers of individuals allowed us to identify additional genomic MHM regions on the Z chromosome that were significantly differentially methylated between the sexes. These regions appear to down-regulate local gene expression in males, but not remove it entirely (unlike the lncRNAs identified in the initial MHM regions). These MHM regions were further tested and the most balanced genes appear to show decreased expression in males, whilst methylation appeared to be far more correlated with gene expression in the less balanced, as compared to the most balanced genes. In addition, quantitative trait loci (QTL) that regulate variation in methylation on the Z chromosome, and those loci that regulate methylation on the autosomes that derive from the Z chromosome were mapped. Trans-effect hotspots were also identified that were based on the autosomes but affected the Z, and also one that was based on the Z chromosome but that affected both autosomal and sex chromosome DNA methylation regulation. We show that both cis and trans loci that originate from the Z chromosome never exhibit an interaction with sex, whereas trans loci originating from the autosomes but affecting the Z chromosome always display such an interaction. Our results highlight how additional MHM regions are actually present on the Z chromosome, and they appear to have smaller-scale effects on gene expression in males. Quantitative variation in methylation is also regulated both from the autosomes to the Z chromosome, and from the Z chromosome to the autosomes.


Assuntos
Galinhas , Cromossomos Sexuais , Animais , Masculino , Galinhas/genética , Metilação de DNA/genética , Mecanismo Genético de Compensação de Dose , Genoma , Mamíferos/genética , Cromossomos Sexuais/genética
19.
Genome Biol Evol ; 16(3)2024 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-38319079

RESUMO

Reptiles exhibit a variety of modes of sex determination, including both temperature-dependent and genetic mechanisms. Among those species with genetic sex determination, sex chromosomes of varying heterogamety (XX/XY and ZZ/ZW) have been observed with different degrees of differentiation. Karyotype studies have demonstrated that Gila monsters (Heloderma suspectum) have ZZ/ZW sex determination and this system is likely homologous to the ZZ/ZW system in the Komodo dragon (Varanus komodoensis), but little else is known about their sex chromosomes. Here, we report the assembly and analysis of the Gila monster genome. We generated a de novo draft genome assembly for a male using 10X Genomics technology. We further generated and analyzed short-read whole genome sequencing and whole transcriptome sequencing data for three males and three females. By comparing female and male genomic data, we identified four putative Z chromosome scaffolds. These putative Z chromosome scaffolds are homologous to Z-linked scaffolds identified in the Komodo dragon. Further, by analyzing RNAseq data, we observed evidence of incomplete dosage compensation between the Gila monster Z chromosome and autosomes and a lack of balance in Z-linked expression between the sexes. In particular, we observe lower expression of the Z in females (ZW) than males (ZZ) on a global basis, though we find evidence suggesting local gene-by-gene compensation. This pattern has been observed in most other ZZ/ZW systems studied to date and may represent a general pattern for female heterogamety in vertebrates.


Assuntos
Animais Peçonhentos , Heloderma suspectum , Lagartos , Animais , Masculino , Feminino , Lagartos/genética , Cromossomos Sexuais/genética , Cariótipo , Mecanismo Genético de Compensação de Dose
20.
Cancer Med ; 12(24): 22130-22155, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37987212

RESUMO

The gene dosage compensation hypothesis presents a mechanism through which the expression of certain genes is modulated to compensate for differences in the dose of genes when additional chromosomes are present. It is one of the means through which cancer cells actively cope with the potential damaging effects of aneuploidy, a hallmark of most cancers. Dosage compensation arises through several processes, including downregulation or overexpression of specific genes and the relocation of dosage-sensitive genes. In cancer, a majority of compensated genes are generally thought to be regulated at the translational or post-translational level, and include the basic components of a compensation loop, including sensors of gene dosage and modulators of gene expression. Post-translational regulation is mostly undertaken by a general degradation or aggregation of remaining protein subunits of macromolecular complexes. An increasingly important role has also been observed for transcriptional level regulation. This article reviews the process of targeted gene dosage compensation in cancer and other biological conditions, along with the mechanisms by which cells regulate specific genes to restore cellular homeostasis. These mechanisms represent potential targets for the inhibition of dosage compensation of specific genes in aneuploid cancers. This article critically examines the process of targeted gene dosage compensation in cancer and other biological contexts, alongside the criteria for identifying genes subject to dosage compensation and the intricate mechanisms by which cells orchestrate the regulation of specific genes to reinstate cellular homeostasis. Ultimately, our aim is to gain a comprehensive understanding of the intricate nature of a systems-level property. This property hinges upon the kinetic parameters of regulatory motifs, which we have termed "gene dosage sensor loops." These loops have the potential to operate at both the transcriptional and translational levels, thus emerging as promising candidates for the inhibition of dosage compensation in specific genes. Additionally, they represent novel and highly specific therapeutic targets in the context of aneuploid cancer.


Assuntos
Mecanismo Genético de Compensação de Dose , Neoplasias , Humanos , Dosagem de Genes , Regulação da Expressão Gênica , Aneuploidia , Regulação para Baixo , Neoplasias/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA