Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 273
Filtrar
1.
Physiol Genomics ; 56(2): 179-193, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38047312

RESUMEN

Epigenetic alterations such as dysregulation of miRNAs have been reported to play important roles in interactions between genetic and environmental factors. In this study, we tested the hypothesis that induction of lung inflammation by inhaled allergens triggers a sex-specific miRNA regulation that is dependent on chromosome complement and hormonal milieu. We challenged the four core genotypes (FCGs) model through intranasal sensitization with a house dust mite (HDM) solution (or PBS as a control) for 5 wk. The FCG model allows four combinations of gonads and sex chromosomes: 1) XX mice with ovaries (XXF), 2) XY mice with testes (XYM), 3) XX mice with testes (XXM), and 4) XY mice with ovaries (XYF). Following the challenge (n = 5-7/group), we assessed the expression of 84 inflammatory miRNAs in lung tissue using a PCR array and cytokine levels in bronchoalveolar lavage fluid (BAL) by a multiplex protein assay (n = 4-7 animals/group). Our results showed higher levels of the chemokine KC (an Il-8 homolog) and IL-7 in BAL from XYF mice challenged with HDM. In addition, IL-17A was significantly higher in BAL from both XXF and XYF mice. A three-way interaction among treatment, gonads, and sex chromosome revealed 60 of 64 miRNAs that differed in expression depending on genotype; XXF, XXM, XYF, and XYM mice had 45, 32, 4, and 52 differentially expressed miRNAs, respectively. Regulatory networks of miRNAs identified in this study were implicated in pathways associated with asthma. Female gonadal hormonal effects may alter miRNA expression and contribute to the higher susceptibility of females to asthma.NEW & NOTEWORTHY miRNAs play important roles in regulating gene and environmental interactions. However, their role in mediating sex differences in allergic responses and lung diseases has not been elucidated. Our study used a targeted omics approach to characterize the contributions of gonadal hormones and chromosomal components to lung responses to an allergen challenge. Our results point to the influence of sex hormones in miRNA expression and proinflammatory markers in allergic airway inflammation.


Asunto(s)
Asma , MicroARNs , Femenino , Animales , Ratones , Masculino , Citocinas/genética , MicroARNs/genética , MicroARNs/metabolismo , Pulmón/metabolismo , Cromosomas Sexuales/genética , Cromosomas Sexuales/metabolismo , Asma/genética , Asma/metabolismo , Inflamación/genética , Inflamación/metabolismo , Líquido del Lavado Bronquioalveolar , Hormonas Gonadales/genética , Hormonas Gonadales/metabolismo , Modelos Animales de Enfermedad
2.
Nat Rev Nephrol ; 20(1): 37-55, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37993681

RESUMEN

Biological sex differences refer to differences between males and females caused by the sex chromosome complement (that is, XY or XX), reproductive tissues (that is, the presence of testes or ovaries), and concentrations of sex steroids (that is, testosterone or oestrogens and progesterone). Although these sex differences are binary for most human individuals and mice, transgender individuals receiving hormone therapy, individuals with genetic syndromes (for example, Klinefelter and Turner syndromes) and people with disorders of sexual development reflect the diversity in sex-based biology. The broad distribution of sex steroid hormone receptors across diverse cell types and the differential expression of X-linked and autosomal genes means that sex is a biological variable that can affect the function of all physiological systems, including the immune system. Sex differences in immune cell function and immune responses to foreign and self antigens affect the development and outcome of diverse diseases and immune responses.


Asunto(s)
Caracteres Sexuales , Cromosomas Sexuales , Animales , Femenino , Ratones , Humanos , Masculino , Cromosomas Sexuales/metabolismo , Ovario/metabolismo , Hormonas Esteroides Gonadales , Inmunidad
3.
Int J Mol Sci ; 24(24)2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38139211

RESUMEN

Gender-related methodology in biomedical sciences receives considerable attention, with numerous studies highlighting biological differences between cisgender males and females. These differences influence the clinical symptoms of various diseases and impact therapeutic approaches. In this in vitro study, we investigate the potential role of sex-chromosome-related dimorphism on steroidogenic enzymes, androgen receptor (AR) expression, and cellular translocation in primary human skeletal muscle cells before and after exposure to testosterone. We analyzed 46XY and 46XX cells for 17ß-hydroxysteroid dehydrogenase (17ß-HSD), 5α-reductase (5α-R2), aromatase (Cyp-19), and AR gene expression. We also compared AR expression and intracellular translocation after increasing exposure to testosterone. At baseline, we observed higher mRNA expression for 5α-R2 and AR in 46XY cells and higher Cyp-19 mRNA expression in 46XX cells. Following testosterone exposure, we observed an increase in AR expression and translocation in 46XX cells, even at the lowest dose of 0.5 nM, while significant changes in 46XY cells were observed only from 10 nM. Our in vitro results demonstrate that the diverse sex chromosome assets reflect important differences in muscle steroidogenesis. They support the concept that chromosomal disparities between males and females, even in vitro, lead to pivotal variations in cellular physiology and response. This understanding represents a crucial starting point in gender medicine, ensuring a precise approach in clinical practice, sports, and exercise settings and facilitating the translation of in vitro data to in vivo applicability.


Asunto(s)
Receptores Androgénicos , Testosterona , Masculino , Femenino , Humanos , Testosterona/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Caracteres Sexuales , Andrógenos/metabolismo , Oxidorreductasas/metabolismo , Colestenona 5 alfa-Reductasa/genética , Músculo Esquelético/metabolismo , Cromosomas Sexuales/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
Int J Mol Sci ; 23(20)2022 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-36293143

RESUMEN

For many decades to date, neuroendocrinologists have delved into the key contribution of gonadal hormones to the generation of sex differences in the developing brain and the expression of sex-specific physiological and behavioral phenotypes in adulthood. However, it was not until recent years that the role of sex chromosomes in the matter started to be seriously explored and unveiled beyond gonadal determination. Now we know that the divergent evolutionary process suffered by X and Y chromosomes has determined that they now encode mostly dissimilar genetic information and are subject to different epigenetic regulations, characteristics that together contribute to generate sex differences between XX and XY cells/individuals from the zygote throughout life. Here we will review and discuss relevant data showing how particular X- and Y-linked genes and epigenetic mechanisms controlling their expression and inheritance are involved, along with or independently of gonadal hormones, in the generation of sex differences in the brain.


Asunto(s)
Diferenciación Sexual , Cromosoma Y , Femenino , Masculino , Animales , Diferenciación Sexual/genética , Cromosomas Sexuales/genética , Cromosomas Sexuales/metabolismo , Caracteres Sexuales , Hormonas Gonadales/metabolismo , Encéfalo/metabolismo , Epigénesis Genética , Cromosoma X
5.
PLoS Genet ; 18(10): e1010469, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36251690

RESUMEN

Meiosis in males of higher dipterans is achiasmate. In their spermatocytes, pairing of homologs into bivalent chromosomes does not include synaptonemal complex and crossover formation. While crossovers preserve homolog conjunction until anaphase I during canonical meiosis, an alternative system is used in dipteran males. Mutant screening in Drosophila melanogaster has identified teflon (tef) as being required specifically for alternative homolog conjunction (AHC) of autosomal bivalents. The additional known AHC genes, snm, uno and mnm, are needed for the conjunction of autosomal homologs and of sex chromosomes. Here, we have analyzed the pattern of TEF protein expression. TEF is present in early spermatocytes but cannot be detected on bivalents at the onset of the first meiotic division, in contrast to SNM, UNO and MNM (SUM). TEF binds to polytene chromosomes in larval salivary glands, recruits MNM by direct interaction and thereby, indirectly, also SNM and UNO. However, chromosomal SUM association is not entirely dependent on TEF, and residual autosome conjunction occurs in tef null mutant spermatocytes. The higher tef requirement for autosomal conjunction is likely linked to the quantitative difference in the amount of SUM protein that provides conjunction of autosomes and sex chromosomes, respectively. During normal meiosis, SUM proteins are far more abundant on sex chromosomes compared to autosomes. Beyond promoting SUM recruitment, TEF has a stabilizing effect on SUM proteins. Increased SUM causes excess conjunction and consequential chromosome missegregation during meiosis I after co-overexpression. Similarly, expression of SUM without TEF, and even more potently with TEF, interferes with chromosome segregation during anaphase of mitotic divisions in somatic cells, suggesting that the known AHC proteins are sufficient for establishment of ectopic chromosome conjunction. Overall, our findings suggest that TEF promotes alternative homolog conjunction during male meiosis without being part of the final physical linkage between chromosomes.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Animales , Masculino , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Politetrafluoroetileno/metabolismo , Segregación Cromosómica/genética , Meiosis/genética , Cromosomas Sexuales/metabolismo , Emparejamiento Cromosómico
6.
Biol Lett ; 18(9): 20220116, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36069069

RESUMEN

The evolution of dosage compensation produces similar expression of sex-linked and autosomal genes in the heterogametic sex. The silkworm (Bombyx mori), a lepidopteran insect, has a female heterogametic WZ sex determination system. A Z-linked gene, Masculinizer (Masc), is the primary determinant of maleness and dosage compensation in B. mori. However, it remains unknown whether one of the two Z chromosomes is inactivated or both Z chromosomes are suppressed in B. mori males. Hence, we performed transcriptome analysis using hybrids between two B. mori strains and analysed allele-specific expression to distinguish these alternatives. Our analysis revealed that genes on both the maternal and paternal Z chromosomes are transcriptionally upregulated in Masc knocked down males. We therefore conclude that both Z chromosomes are transcriptionally downregulated in B. mori males, similar to the system in Caenorhabditis elegans.


Asunto(s)
Bombyx , Animales , Bombyx/genética , Bombyx/metabolismo , Compensación de Dosificación (Genética) , Regulación hacia Abajo , Femenino , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Masculino , Cromosomas Sexuales/genética , Cromosomas Sexuales/metabolismo
7.
Nat Rev Endocrinol ; 18(9): 574-583, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35705742

RESUMEN

Understanding sex differences in physiology and disease requires the identification of the molecular agents that cause phenotypic sex differences. Two groups of such agents are genes located on the sex chromosomes, and gonadal hormones. The former have coherent linkage to chromosomes that form differently in the two sexes under the influence of genomic forces that are not related to reproductive function, whereas the latter have a direct or indirect relationship to reproduction. Evidence published in the past 5 years supports the identification of several agents of sexual differentiation encoded by the X chromosome in mice, including Kdm5c, Kdm6a, Ogt and Xist. These X chromosome agents have wide pleiotropic effects, potentially influencing sex differences in many different tissues, a characteristic shared with the gonadal hormones. The identification of X chromosome agents of sexual differentiation will facilitate understanding of complex intersecting gene pathways underlying sex differences in disease.


Asunto(s)
Diferenciación Sexual , Cromosoma X , Animales , Femenino , Hormonas Gonadales/metabolismo , Humanos , Masculino , Ratones , Caracteres Sexuales , Cromosomas Sexuales/genética , Cromosomas Sexuales/metabolismo , Diferenciación Sexual/genética , Cromosoma X/metabolismo
8.
Circ Res ; 130(12): 1747-1762, 2022 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-35679362

RESUMEN

Sex is a key risk factor for many types of cardiovascular disease. It is imperative to understand the mechanisms underlying sex differences to devise optimal preventive and therapeutic approaches for all individuals. Both biological sex (determined by sex chromosomes and gonadal hormones) and gender (social and cultural behaviors associated with femininity or masculinity) influence differences between men and women in disease susceptibility and pathology. Here, we focus on the application of experimental mouse models that elucidate the influence of 2 components of biological sex-sex chromosome complement (XX or XY) and gonad type (ovaries or testes). These models have revealed that in addition to well-known effects of gonadal hormones, sex chromosome complement influences cardiovascular risk factors, such as plasma cholesterol levels and adiposity, as well as the development of atherosclerosis and pulmonary hypertension. One mechanism by which sex chromosome dosage influences cardiometabolic traits is through sex-biased expression of X chromosome genes that escape X inactivation. These include chromatin-modifying enzymes that regulate gene expression throughout the genome. The identification of factors that determine sex-biased gene expression and cardiometabolic traits will expand our mechanistic understanding of cardiovascular disease processes and provide insight into sex differences that remain throughout the lifespan as gonadal hormone levels alter with age.


Asunto(s)
Enfermedades Cardiovasculares , Caracteres Sexuales , Adiposidad , Animales , Enfermedades Cardiovasculares/genética , Femenino , Hormonas Gonadales/metabolismo , Humanos , Masculino , Ratones , Cromosomas Sexuales/genética , Cromosomas Sexuales/metabolismo , Cromosoma X/genética , Cromosoma X/metabolismo
9.
Nucleic Acids Res ; 50(13): 7350-7366, 2022 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-35766398

RESUMEN

The histone variant H3.3 is encoded by two distinct genes, H3f3a and H3f3b, exhibiting identical amino-acid sequence. H3.3 is required for spermatogenesis, but the molecular mechanism of its spermatogenic function remains obscure. Here, we have studied the role of each one of H3.3A and H3.3B proteins in spermatogenesis. We have generated transgenic conditional knock-out/knock-in (cKO/KI) epitope-tagged FLAG-FLAG-HA-H3.3B (H3.3BHA) and FLAG-FLAG-HA-H3.3A (H3.3AHA) mouse lines. We show that H3.3B, but not H3.3A, is required for spermatogenesis and male fertility. Analysis of the molecular mechanism unveils that the absence of H3.3B led to alterations in the meiotic/post-meiotic transition. Genome-wide RNA-seq reveals that the depletion of H3.3B in meiotic cells is associated with increased expression of the whole sex X and Y chromosomes as well as of both RLTR10B and RLTR10B2 retrotransposons. In contrast, the absence of H3.3B resulted in down-regulation of the expression of piRNA clusters. ChIP-seq experiments uncover that RLTR10B and RLTR10B2 retrotransposons, the whole sex chromosomes and the piRNA clusters are markedly enriched of H3.3. Taken together, our data dissect the molecular mechanism of H3.3B functions during spermatogenesis and demonstrate that H3.3B, depending on its chromatin localization, is involved in either up-regulation or down-regulation of expression of defined large chromatin regions.


Asunto(s)
Histonas , ARN Interferente Pequeño/metabolismo , Retroelementos , Espermatogénesis , Animales , Cromatina/genética , Histonas/genética , Histonas/metabolismo , Masculino , Ratones , Cromosomas Sexuales/metabolismo
10.
Cell Rep ; 39(2): 110636, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35417719

RESUMEN

Genetic networks are characterized by extensive buffering. During tumor evolution, disruption of functional redundancies can create de novo vulnerabilities that are specific to cancer cells. Here, we systematically search for cancer-relevant paralog interactions using CRISPR screens and publicly available loss-of-function datasets. Our analysis reveals >2,000 candidate dependencies, several of which we validate experimentally, including CSTF2-CSTF2T, DNAJC15-DNAJC19, FAM50A-FAM50B, and RPP25-RPP25L. We provide evidence that RPP25L can physically and functionally compensate for the absence of RPP25 as a member of the RNase P/MRP complexes in tRNA processing. Our analysis also reveals unexpected redundancies between sex chromosome genes. We show that chrX- and chrY-encoded paralogs, such as ZFX-ZFY, DDX3X-DDX3Y, and EIF1AX-EIF1AY, are functionally linked. Tumor cell lines from male patients with loss of chromosome Y become dependent on the chrX-encoded gene. We propose targeting of chrX-encoded paralogs as a general therapeutic strategy for human tumors that have lost the Y chromosome.


Asunto(s)
Neoplasias , Oncogenes , ARN Helicasas DEAD-box/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Masculino , Antígenos de Histocompatibilidad Menor/metabolismo , Neoplasias/genética , Proteínas de Unión al ARN/genética , Cromosomas Sexuales/metabolismo , Cromosoma X , Cromosoma Y
11.
J Nephrol ; 35(4): 1069-1078, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34292531

RESUMEN

BACKGROUND: There are arguments for individualized treatments and the necessity of non-invasive biomarkers for patients with IgA nephropathy (IgAN) according to gender, but the rationale remains unclear. We aimed to investigate the relationship between urine exosomal circular RNA (circRNA) levels, related genes, clinical features, and renal pathological features in IgA nephropathy patients of different genders. METHODS: Clinicopathological data from patients of different genders from a multicenter cohort were retrospectively analyzed. We used the Oxford classification to examine the severity of pathological damage in these patients. We compared clinical features and renal pathologies between IgAN patients of different genders. Using findings of urine exosomal circRNAs from male IgAN patients, we analyzed the relationship between this factor, the regulated genes located on the sex chromosomes, and renal pathologies. RESULTS: A total of 502 IgAN patients were included. The proportion of male patients with crescent formation was higher than that of females (p = 0.019). Multivariate logistic regression analysis showed that proteinuria was an independent marker for crescent formation in male and female patients with IgAN, while smoking and higher low-density lipoprotein cholesterol (LDL-C) levels were independent risk factors for crescent formation in males alone. Urine exosomal circRNA chrY:15478147-15481229- located on the Y chromosome in male patients was negatively correlated with the expressions of UTY in specific regions of the Y chromosome. CONCLUSION: Compared with female patients, males with IgAN had more severe renal dysfunction and a higher probability of glomerular crescent formation. Urine exosomal circRNA chrY:15478147-15481229- might participate in the pathogenesis of IgAN in male patients by altering UTY expressions.


Asunto(s)
Glomerulonefritis por IGA , Biomarcadores/metabolismo , Femenino , Glomerulonefritis por IGA/diagnóstico , Glomerulonefritis por IGA/genética , Glomerulonefritis por IGA/metabolismo , Humanos , Masculino , Proteinuria , ARN Circular , Estudios Retrospectivos , Cromosomas Sexuales/metabolismo , Cromosomas Sexuales/patología
12.
Cell Mol Life Sci ; 79(1): 18, 2021 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-34971404

RESUMEN

In mammalian male meiosis, the heterologous X and Y chromosomes remain unsynapsed and, as a result, are subject to meiotic sex chromosome inactivation (MSCI). MSCI is required for the successful completion of spermatogenesis. Following the initiation of MSCI, the X and Y chromosomes undergo various epigenetic modifications and are transformed into a nuclear body termed the XY body. Here, we review the mechanisms underlying the initiation of two essential, sequential processes in meiotic prophase I: MSCI and XY-body formation. The initiation of MSCI is directed by the action of DNA damage response (DDR) pathways; downstream of the DDR, unique epigenetic states are established, leading to the formation of the XY body. Accumulating evidence suggests that MSCI and subsequent XY-body formation may be driven by phase separation, a physical process that governs the formation of membraneless organelles and other biomolecular condensates. Thus, here we gather literature-based evidence to explore a phase separation hypothesis for the initiation of MSCI and the formation of the XY body.


Asunto(s)
Compensación de Dosificación (Genética) , Meiosis , Modelos Biológicos , Cromosomas Sexuales/metabolismo , Animales , Daño del ADN/genética , Reparación del ADN/genética , Humanos , Meiosis/genética
13.
PLoS One ; 16(12): e0261918, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34968397

RESUMEN

Yun7Ge is a giant egg mutant found in the silkworm variety Yun7. In comparison with the giant mutant Ge, the eggs of Yun7Ge are larger. The number of laid eggs and hatching rate of Yun7Ge are reduced, which is not conducive to reproduction. In this work, the target gene controlling giant egg trait is located on the Z chromosome and was determined through genetic analysis. Transcriptome results showed that phytanoyl-CoA dioxygenase domain-containing protein 1 (PHYHD1) on the Z chromosome was silenced, and the 25 chorion genes on chromosome 2 were remarkably downregulated. Sequence analysis showed that the 73.5 kb sequence including the PHYHD1 was replaced by a ~3.0 kb sequence. After knocking out the PHYHD1 by using CRISPR/Cas9, the chorion genes were significantly downregulated. Hence, the silencing of PHYHD1 leads to the downregulation of many chorion protein genes, thus directly causing giant eggs.


Asunto(s)
Bombyx/fisiología , Cáscara de Huevo/fisiología , Oxigenasas/química , Animales , Sistemas CRISPR-Cas , Corion/química , Cromosomas , Coenzima A/química , Regulación hacia Abajo , Femenino , Silenciador del Gen , Proteínas de Insectos/genética , Larva/genética , Masculino , Modelos Genéticos , Mutación , Fenotipo , Ácido Fitánico/análogos & derivados , Ácido Fitánico/química , Reacción en Cadena de la Polimerasa , Dominios Proteicos , RNA-Seq , Reproducción , Cromosomas Sexuales/metabolismo
14.
Dev Cell ; 56(21): 3019-3034.e7, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34655525

RESUMEN

Sex disparities in cardiac homeostasis and heart disease are well documented, with differences attributed to actions of sex hormones. However, studies have indicated sex chromosomes act outside of the gonads to function without mediation by gonadal hormones. Here, we performed transcriptional and proteomics profiling to define differences between male and female mouse hearts. We demonstrate, contrary to current dogma, cardiac sex disparities are controlled not only by sex hormones but also through a sex-chromosome mechanism. Using Turner syndrome (XO) and Klinefelter (XXY) models, we find the sex-chromosome pathway is established by X-linked gene dosage. We demonstrate cardiac sex disparities occur at the earliest stages of heart formation, a period before gonad formation. Using these datasets, we identify and define a role for alpha-1B-glycoprotein (A1BG), showing loss of A1BG leads to cardiac defects in females, but not males. These studies provide resources for studying sex-biased cardiac disease states.


Asunto(s)
Gónadas/crecimiento & desarrollo , Gónadas/metabolismo , Proteómica , Caracteres Sexuales , Cromosomas Sexuales/metabolismo , Animales , Femenino , Genes Ligados a X/genética , Masculino , Ratones , Proteómica/métodos
15.
Cell Death Dis ; 12(8): 780, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34373449

RESUMEN

FANCI is an essential component of Fanconi anemia pathway, which is responsible for the repair of DNA interstrand cross-links (ICLs). As an evolutionarily related partner of FANCD2, FANCI functions together with FANCD2 downstream of FA core complex. Currently, growing evidences showed that the essential role of FA pathway in male fertility. However, the underlying mechanisms for FANCI in regulating spermatogenesis remain unclear. In the present study, we found that the male Fanci-/- mice were sterile and exhibited abnormal spermatogenesis, including massive germ cell apoptosis in seminiferous tubules and dramatically decreased number of sperms in epididymis. Besides, FANCI deletion impaired maintenance of undifferentiated spermatogonia. Further investigation indicated that FANCI was essential for FANCD2 foci formation and regulated H3K4 and H3K9 methylation on meiotic sex chromosomes. These findings elucidate the role and mechanism of FANCI during spermatogenesis in mice and provide new insights into the etiology and molecular basis of nonobstructive azoospermia.


Asunto(s)
Anemia de Fanconi/metabolismo , Histonas/metabolismo , Meiosis , Espermatogénesis , Animales , Apoptosis , Diferenciación Celular , Epigénesis Genética , Eliminación de Gen , Infertilidad Masculina/genética , Lisina/metabolismo , Masculino , Metilación , Ratones , Especificidad de Órganos/genética , Profase , ARN Mensajero/genética , ARN Mensajero/metabolismo , Recombinación Genética/genética , Cromosomas Sexuales/metabolismo , Espermatogonias/metabolismo , Espermatozoides/metabolismo , Testículo/metabolismo
16.
Biol Reprod ; 105(5): 1154-1159, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34225362

RESUMEN

While an E3 ubiquitin ligase, RNF8, was initially reported to be required for histone-to-protamine exchange in spermiogenesis, we subsequently demonstrated that RNF8 is not involved in this process. Nevertheless, reflecting a lingering misunderstanding in the field, a growing number of studies have continued to postulate a requirement for RNF8 in the histone-to-protamine exchange. For example, a recent study claimed that a mouse PIWI protein, MIWI, controls RNF8-mediated histone-to-protamine exchange. Here, confirming our earlier conclusions, we show that RNF8 is required neither for the establishment of histone H4K16 acetylation, which is an initial step in histone removal during spermiogenesis, nor for the incorporation of two protamine proteins, PRM1 and PRM2. Thus, whereas RNF8 mediates ubiquitination of H2A on the sex chromosomes in meiosis, during the prior stage of spermatogenesis, our genetic evidence underscores that RNF8 is not involved in histone-to-protamine exchange.


Asunto(s)
Histonas/metabolismo , Protaminas/metabolismo , Espermatogénesis , Ubiquitina-Proteína Ligasas/genética , Acetilación , Animales , Transporte Biológico , Ensamble y Desensamble de Cromatina , Ratones , Ratones Noqueados , Cromosomas Sexuales/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
17.
Genes Dev ; 35(13-14): 1055-1070, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34140353

RESUMEN

The dosage compensation complex (DCC) of Drosophila identifies its X-chromosomal binding sites with exquisite selectivity. The principles that assure this vital targeting are known from the D. melanogaster model: DCC-intrinsic specificity of DNA binding, cooperativity with the CLAMP protein, and noncoding roX2 RNA transcribed from the X chromosome. We found that in D. virilis, a species separated from melanogaster by 40 million years of evolution, all principles are active but contribute differently to X specificity. In melanogaster, the DCC subunit MSL2 evolved intrinsic DNA-binding selectivity for rare PionX sites, which mark the X chromosome. In virilis, PionX motifs are abundant and not X-enriched. Accordingly, MSL2 lacks specific recognition. Here, roX2 RNA plays a more instructive role, counteracting a nonproductive interaction of CLAMP and modulating DCC binding selectivity. Remarkably, roX2 triggers a stable chromatin binding mode characteristic of DCC. Evidently, X-specific regulation is achieved by divergent evolution of protein, DNA, and RNA components.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Animales , Compensación de Dosificación (Genética) , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Cromosomas Sexuales/metabolismo , Factores de Transcripción/metabolismo , Cromosoma X/genética , Cromosoma X/metabolismo
18.
Int J Mol Sci ; 22(4)2021 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-33671248

RESUMEN

Cyclin-dependent kinases (CDKs) are crucial regulators of the eukaryotic cell cycle. The critical role of CDK2 in the progression of meiosis was demonstrated in a single mammalian species, the mouse. We used immunocytochemistry to study the localization of CDK2 during meiosis in seven rodent species that possess hetero- and homomorphic male sex chromosomes. To compare the distribution of CDK2 in XY and XX male sex chromosomes, we performed multi-round immunostaining of a number of marker proteins in meiotic chromosomes of the rat and subterranean mole voles. Antibodies to the following proteins were used: RAD51, a member of the double-stranded DNA break repair machinery; MLH1, a component of the DNA mismatch repair system; and SUN1, which is involved in the connection between the meiotic telomeres and nuclear envelope, alongside the synaptic protein SYCP3 and kinetochore marker CREST. Using an enhanced protocol, we were able to assess the distribution of as many as four separate proteins in the same meiotic cell. We showed that during prophase I, CDK2 localizes to telomeric and interstitial regions of autosomes in all species investigated (rat, vole, hamster, subterranean mole voles, and mole rats). In sex bivalents following synaptic specificity, the CDK2 signals were distributed in three different modes. In the XY bivalent in the rat and mole rat, we detected numerous CDK2 signals in asynaptic regions and a single CDK2 focus on synaptic segments, similar to the mouse sex chromosomes. In the mole voles, which have unique XX sex chromosomes in males, CDK2 signals were nevertheless distributed similarly to the rat XY sex chromosomes. In the vole, sex chromosomes did not synapse, but demonstrated CDK2 signals of varying intensity, similar to the rat X and Y chromosomes. In female mole voles, the XX bivalent had CDK2 pattern similar to autosomes of all species. In the hamster, CDK2 signals were revealed in telomeric regions in the short synaptic segment of the sex bivalent. We found that CDK2 signals colocalize with SUN1 and MLH1 signals in meiotic chromosomes in rats and mole voles, similar to the mouse. The difference in CDK2 manifestation at the prophase I sex chromosomes can be considered an example of the rapid chromosome evolution in mammals.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/metabolismo , Mamíferos/metabolismo , Profase Meiótica I , Cromosomas Sexuales/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Femenino , Masculino , Modelos Biológicos , Fase Paquiteno , Ratas , Espermatocitos/metabolismo
19.
Cells ; 10(2)2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33535518

RESUMEN

Sex determination directs development as male or female in sexually reproducing organisms. Evolutionary transitions in sex determination have occurred frequently, suggesting simple mechanisms behind the transitions, yet their detail remains elusive. Here we explore the links between mechanisms of transitions in sex determination and sex chromosome evolution at both recent and deeper temporal scales (<1 Myr; ~79 Myr). We studied a rare example of a species with intraspecific variation in sex determination, Carinascincus ocellatus, and a relative, Liopholis whitii, using c-banding and mapping of repeat motifs and a custom Y chromosome probe set to identify the sex chromosomes. We identified both unique and conserved regions of the Y chromosome among C. ocellatus populations differing in sex determination. There was no evidence for homology of sex chromosomes between C. ocellatus and L. whitii, suggesting independent evolutionary origins. We discuss sex chromosome homology between members of the subfamily Lygosominae and propose links between sex chromosome evolution, sex determination transitions, and karyotype evolution.


Asunto(s)
Cromosomas Sexuales/metabolismo , Análisis para Determinación del Sexo/métodos , Animales , Evolución Biológica , Femenino , Cariotipo , Masculino
20.
Nat Commun ; 12(1): 892, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33563972

RESUMEN

Given their copy number differences and unique modes of inheritance, the evolved gene content and expression of sex chromosomes is unusual. In many organisms the X and Y chromosomes are inactivated in spermatocytes, possibly as a defense mechanism against insertions into unpaired chromatin. In addition to current sex chromosomes, Drosophila has a small gene-poor X-chromosome relic (4th) that re-acquired autosomal status. Here we use single cell RNA-Seq on fly larvae to demonstrate that the single X and pair of 4th chromosomes are specifically inactivated in primary spermatocytes, based on measuring all genes or a set of broadly expressed genes in testis we identified. In contrast, genes on the single Y chromosome become maximally active in primary spermatocytes. Reduced X transcript levels are due to failed activation of RNA-Polymerase-II by phosphorylation of Serine 2 and 5.


Asunto(s)
Drosophila/genética , Cromosomas Sexuales/genética , Espermatocitos/metabolismo , Animales , Drosophila/crecimiento & desarrollo , Regulación de la Expresión Génica , Genes Ligados a X/genética , Genes Ligados a Y/genética , Larva/genética , Larva/crecimiento & desarrollo , Masculino , Especificidad de Órganos , ARN Polimerasa II/metabolismo , Cromosomas Sexuales/metabolismo , Espermatogénesis/genética , Testículo/citología , Testículo/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...