Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 200
Filtrar
1.
Genome Res ; 32(11-12): 1993-2002, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36418059

RESUMEN

Human sex differences arise from gonadal hormones and sex chromosomes. Studying the direct effects of sex chromosomes in humans is still challenging. Here we studied how the sex chromosomes can modulate gene expression and the outcome of mutations across the genome by exploiting the tendency of cancer cell lines to lose or gain sex chromosomes. We inferred the dosage of the sex chromosomes in 355 female and 408 male cancer cell lines and used it to dissect the contributions of the Y and X Chromosomes to sex-biased gene expression. Furthermore, based on genome-wide CRISPR screens, we identified genes whose essentiality is different between male and female cells depending on the sex chromosomes. The most significant genes were X-linked genes compensated by Y-linked paralogs. Our sex-based analysis identifies genes that, when mutated, can affect male and female cells differently and reinforces the roles of the X and Y Chromosomes in sex-specific cell function.


Asunto(s)
Neoplasias , Cromosomas Sexuales , Femenino , Masculino , Humanos , Cromosomas Sexuales/genética , Cromosoma Y , Cromosoma X , Genes Ligados a X , Genes Ligados a Y , Caracteres Sexuales , Neoplasias/genética
2.
Hum Mol Genet ; 30(R2): R296-R300, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34328177

RESUMEN

The Y chromosome is the most gene-deficient chromosome in the human genome (though not the smallest chromosome) and has largely been sequestered away from large-scale studies of the effects of genetics on human health. Here I review the literature, focusing on the last 2 years, for recent evidence of the role of the Y chromosome in protecting from or contributing to disease. Although many studies have focused on Y chromosome gene copy number and variants in fertility, the role of the Y chromosome in human health is now known to extend too many other conditions including the development of multiple cancers and Alzheimer's disease. I further include the discussion of current technology and methods for analyzing Y chromosome variation. The true role of the Y chromosome and associated genetic variants in human disease will only become clear when the Y chromosome is integrated into larger studies of human genetic variation, rather than being analyzed in isolation.


Asunto(s)
Cromosomas Humanos Y , Susceptibilidad a Enfermedades , Genoma Humano , Homeostasis , Evolución Molecular , Regulación de la Expresión Génica , Genes Ligados a Y , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Humanos , Masculino
3.
Genome Res ; 30(6): 860-873, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32461223

RESUMEN

Little is known about how human Y-Chromosome gene expression directly contributes to differences between XX (female) and XY (male) individuals in nonreproductive tissues. Here, we analyzed quantitative profiles of Y-Chromosome gene expression across 36 human tissues from hundreds of individuals. Although it is often said that Y-Chromosome genes are lowly expressed outside the testis, we report many instances of elevated Y-Chromosome gene expression in a nonreproductive tissue. A notable example is EIF1AY, which encodes eukaryotic translation initiation factor 1A Y-linked, together with its X-linked homolog EIF1AX Evolutionary loss of a Y-linked microRNA target site enabled up-regulation of EIF1AY, but not of EIF1AX, in the heart. Consequently, this essential translation initiation factor is nearly twice as abundant in male as in female heart tissue at the protein level. Divergence between the X and Y Chromosomes in regulatory sequence can therefore lead to tissue-specific Y-Chromosome-driven sex biases in expression of critical, dosage-sensitive regulatory genes.


Asunto(s)
Cromosomas Humanos Y , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genes Ligados a Y , Transcriptoma , Cromosomas Humanos X/genética , Biología Computacional/métodos , Evolución Molecular , Femenino , Perfilación de la Expresión Génica/métodos , Genes Ligados a X , Humanos , Masculino , MicroARNs/genética , Especificidad de Órganos/genética
4.
Heredity (Edinb) ; 130(5): 312-319, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36914794

RESUMEN

Although containing genes important for sex determination, genetic variation within the Y chromosome was traditionally predicted to contribute little to the expression of sexually dimorphic traits. This prediction was shaped by the assumption that the chromosome harbours few protein-coding genes, and that capacity for Y-linked variation to shape adaptation would be hindered by the chromosome's lack of recombination and holandric inheritance. Consequently, most studies exploring the genotypic contributions to sexually dimorphic traits have focused on the autosomes and X chromosome. Yet, several studies have now demonstrated that the Y chromosome harbours variation affecting male fitness, moderating the expression of hundreds of genes across the nuclear genome. Furthermore, emerging results have shown that expression of this Y-linked variation may be sensitive to environmental heterogeneity, leading to the prediction that Y-mediated gene-by-environment interactions will shape the expression of sexually dimorphic phenotypes. We tested this prediction, investigating whether genetic variation across six distinct Y chromosome haplotypes affects the expression of locomotor activity, at each of two temperatures (20 and 28 °C) in male fruit flies (Drosophila melanogaster). Locomotor activity is a sexually dimorphic trait in this species, previously demonstrated to be under intralocus sexual conflict. We demonstrate Y haplotype effects on male locomotor activity, but the rank order and magnitude of these effects were unaltered by differences in temperature. Our study contributes to a growing number of studies demonstrating Y-linked effects moderating expression of traits evolving under sexually antagonistic selection, suggesting a role for the Y chromosome in shaping outcomes of sexual conflict.


Asunto(s)
Drosophila melanogaster , Genes Ligados a Y , Animales , Masculino , Drosophila melanogaster/genética , Cromosoma Y/genética , Cromosoma X/genética , Locomoción
5.
Am J Respir Crit Care Med ; 206(2): 186-196, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35504005

RESUMEN

Rationale: Idiopathic pulmonary arterial hypertension (PAH) is a terminal pulmonary vascular disease characterized by increased pressure, right ventricular failure, and death. PAH exhibits a striking sex bias and is up to four times more prevalent in females. Understanding the molecular basis behind sex differences could help uncover novel therapies. Objectives: We previously discovered that the Y chromosome is protective against hypoxia-induced experimental pulmonary hypertension (PH), which may contribute to sex differences in PAH. Here, we identify the gene responsible for Y-chromosome protection, investigate key downstream autosomal genes, and demonstrate a novel preclinical therapy. Methods: To test the effect of Y-chromosome genes on PH development, we knocked down each Y-chromosome gene expressed in the lung by means of intratracheal instillation of siRNA in gonadectomized male mice exposed to hypoxia and monitored changes in right ventricular and pulmonary artery hemodynamics. We compared the lung transcriptome of Uty knockdown mouse lungs to those of male and female PAH patient lungs to identify common downstream pathogenic chemokines and tested the effects of these chemokines on human pulmonary artery endothelial cells. We further inhibited the activity of these chemokines in two preclinical pulmonary hypertension models to test the therapeutic efficacy. Measurements and Main Results: Knockdown of the Y-chromosome gene Uty resulted in more severe PH measured by increased right ventricular pressure and decreased pulmonary artery acceleration time. RNA sequencing revealed an increase in proinflammatory chemokines Cxcl9 and Cxcl10 as a result of Uty knockdown. We found CXCL9 and CXCL10 significantly upregulated in human PAH lungs, with more robust upregulation in females with PAH. Treatment of human pulmonary artery endothelial cells with CXCL9 and CXCL10 triggered apoptosis. Inhibition of Cxcl9 and Cxcl10 expression in male Uty knockout mice and CXCL9 and CXCL10 activity in female rats significantly reduced PH severity. Conclusions:Uty is protective against PH. Reduction of Uty expression results in increased expression of proinflammatory chemokines Cxcl9 and Cxcl10, which trigger endothelial cell death and PH. Inhibition of CLXC9 and CXLC10 rescues PH development in multiple experimental models.


Asunto(s)
Quimiocinas , Hipertensión Pulmonar , Antígenos de Histocompatibilidad Menor , Proteínas Nucleares , Animales , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Hipertensión Pulmonar Primaria Familiar/genética , Femenino , Genes Ligados a Y , Humanos , Hipertensión Pulmonar/genética , Hipoxia , Masculino , Ratones , Antígenos de Histocompatibilidad Menor/genética , Proteínas Nucleares/genética , Arteria Pulmonar , Ratas
6.
Biol Reprod ; 106(6): 1312-1326, 2022 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-35293998

RESUMEN

Using mice with Y chromosome deficiencies and supplementing Zfy transgenes, we, and others, have previously shown that the loss of Y chromosome Zfy1 and Zfy2 genes is associated with infertility and spermiogenic defects and that the addition of Zfy transgenes rescues these defects. In these past studies, the absence of Zfy was linked to the loss of other Y chromosome genes, which might have contributed to spermiogenic phenotypes. Here, we used CRISPR/Cas9 to specifically remove open reading frame of Zfy1, Zfy2, or both Zfy1 and Zfy2, and generated Zfy knockout (KO) and double knockout (DKO) mice. Zfy1 KO and Zfy2 KO mice were both fertile, but the latter had decreased litters size and sperm number, and sperm headshape abnormalities. Zfy DKO males were infertile and displayed severe spermatogenesis defects. Postmeiotic arrest largely prevented production of sperm and the few sperm that were produced all displayed gross headshape abnormalities and structural defects within head and tail. Infertility of Zfy DKO mice could be overcome by injection of spermatids or sperm directly to oocytes, and the resulting male offspring had the same spermiogenic phenotype as their fathers. The study is the first describing detailed phenotypic characterization of mice with the complete Zfy gene loss. It provides evidence supporting that the presence of at least one Zfy homolog is essential for male fertility and development of normal sperm functional in unassisted fertilization. The data also show that while the loss of Zfy1 is benign, the loss of Zfy2 is mildly detrimental for spermatogenesis.


Asunto(s)
Proteínas de Unión al ADN , Genes Ligados a Y , Infertilidad , Factores de Transcripción , Animales , Proteínas de Unión al ADN/genética , Infertilidad/genética , Masculino , Ratones , Espermatogénesis/genética , Espermatozoides , Factores de Transcripción/genética , Cromosoma Y/genética
7.
PLoS Genet ; 15(9): e1008369, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31525193

RESUMEN

The Y chromosome harbors nine multi-copy ampliconic gene families expressed exclusively in testis. The gene copies within each family are >99% identical to each other, which poses a major challenge in evaluating their copy number. Recent studies demonstrated high variation in Y ampliconic gene copy number among humans. However, how this variation affects expression levels in human testis remains understudied. Here we developed a novel computational tool Ampliconic Copy Number Estimator (AmpliCoNE) that utilizes read sequencing depth information to estimate Y ampliconic gene copy number per family. We applied this tool to whole-genome sequencing data of 149 men with matched testis expression data whose samples are part of the Genotype-Tissue Expression (GTEx) project. We found that the Y ampliconic gene families with low copy number in humans were deleted or pseudogenized in non-human great apes, suggesting relaxation of functional constraints. Among the Y ampliconic gene families, higher copy number leads to higher expression. Within the Y ampliconic gene families, copy number does not influence gene expression, rather a high tolerance for variation in gene expression was observed in testis of presumably healthy men. No differences in gene expression levels were found among major Y haplogroups. Age positively correlated with expression levels of the HSFY and PRY gene families in the African subhaplogroup E1b, but not in the European subhaplogroups R1b and I1. We also found that expression of five Y ampliconic gene families is coordinated with that of their non-Y (i.e. X or autosomal) homologs. Indeed, five ampliconic gene families had consistently lower expression levels when compared to their non-Y homologs suggesting dosage regulation, while the HSFY family had higher expression levels than its X homolog and thus lacked dosage regulation.


Asunto(s)
Cromosomas Humanos Y/genética , Genes Ligados a Y/genética , Análisis de Secuencia de ADN/métodos , Animales , Cromosomas Humanos Y/fisiología , Variaciones en el Número de Copia de ADN/genética , Bases de Datos Genéticas , Compensación de Dosificación (Genética)/genética , Compensación de Dosificación (Genética)/fisiología , Epigénesis Genética/genética , Dosificación de Gen/genética , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Genes Ligados a Y/fisiología , Factores de Transcripción del Choque Térmico/genética , Factores de Transcripción del Choque Térmico/metabolismo , Humanos , Masculino , Familia de Multigenes/genética , Testículo/metabolismo
8.
Proc Natl Acad Sci U S A ; 116(33): 16577-16582, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31371505

RESUMEN

Parkinson's disease (PD) is a debilitating neurodegenerative disorder caused by the loss of midbrain dopamine (DA) neurons. While the cause of DA cell loss in PD is unknown, male sex is a strong risk factor. Aside from the protective actions of sex hormones in females, emerging evidence suggests that sex-chromosome genes contribute to the male bias in PD. We previously showed that the Y-chromosome gene, SRY, directly regulates adult brain function in males independent of gonadal hormone influence. SRY protein colocalizes with DA neurons in the male substantia nigra, where it regulates DA biosynthesis and voluntary movement. Here we demonstrate that nigral SRY expression is highly and persistently up-regulated in animal and human cell culture models of PD. Remarkably, lowering nigral SRY expression with antisense oligonucleotides in male rats diminished motor deficits and nigral DA cell loss in 6-hydroxydopamine (6-OHDA)-induced and rotenone-induced rat models of PD. The protective effect of the SRY antisense oligonucleotides was associated with male-specific attenuation of DNA damage, mitochondrial degradation, and neuroinflammation in the toxin-induced rat models of PD. Moreover, reducing nigral SRY expression diminished or removed the male bias in nigrostriatal degeneration, mitochondrial degradation, DNA damage, and neuroinflammation in the 6-OHDA rat model of PD, suggesting that SRY directly contributes to the sex differences in PD. These findings demonstrate that SRY directs a previously unrecognized male-specific mechanism of DA cell death and suggests that suppressing nigral Sry synthesis represents a sex-specific strategy to slow or prevent DA cell loss in PD.


Asunto(s)
Genes Ligados a Y , Neuroprotección/genética , Enfermedad de Parkinson/genética , Animales , Daño del ADN , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación/patología , Masculino , Mitofagia/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Oligonucleótidos Antisentido/farmacología , Oxidopamina , Enfermedad de Parkinson/fisiopatología , Ratas , Proteína de la Región Y Determinante del Sexo/genética , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
9.
Genome Res ; 28(12): 1841-1851, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30381290

RESUMEN

Despite claims that the mammalian Y Chromosome is on a path to extinction, comparative sequence analysis of primate Y Chromosomes has shown the decay of the ancestral single-copy genes has all but ceased in this eutherian lineage. The suite of single-copy Y-linked genes is highly conserved among the majority of eutherian Y Chromosomes due to strong purifying selection to retain dosage-sensitive genes. In contrast, the ampliconic regions of the Y Chromosome, which contain testis-specific genes that encode the majority of the transcripts on eutherian Y Chromosomes, are rapidly evolving and are thought to undergo species-specific turnover. However, ampliconic genes are known from only a handful of species, limiting insights into their long-term evolutionary dynamics. We used a clone-based sequencing approach employing both long- and short-read sequencing technologies to assemble ∼2.4 Mb of representative ampliconic sequence dispersed across the domestic cat Y Chromosome, and identified the major ampliconic gene families and repeat units. We analyzed fluorescence in situ hybridization, qPCR, and whole-genome sequence data from 20 cat species and revealed that ampliconic gene families are conserved across the cat family Felidae but show high transcript diversity, copy number variation, and structural rearrangement. Our analysis of ampliconic gene evolution unveils a complex pattern of long-term gene content stability despite extensive structural variation on a nonrecombining background.


Asunto(s)
Variaciones en el Número de Copia de ADN , Evolución Molecular , Amplificación de Genes , Genes Ligados a Y , Familia de Multigenes , Cromosoma Y , Animales , Gatos , Cromosomas Artificiales Bacterianos , Biología Computacional/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Hibridación Fluorescente in Situ , Masculino , Filogenia , Transcripción Genética , Secuenciación Completa del Genoma
10.
Proc Biol Sci ; 287(1937): 20200469, 2020 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-33081607

RESUMEN

The coordination between mitochondrial and nuclear genes is crucial to eukaryotic organisms. Predicting the nature of these epistatic interactions can be difficult because of the transmission asymmetry of the genes involved. While autosomes and X-linked genes are transmitted through both sexes, genes on the Y chromosome and in the mitochondrial genome are uniparentally transmitted through males and females, respectively. Here, we generate 36 otherwise isogenic Drosophila melanogaster strains differing only in the geographical origin of their mitochondrial genome and Y chromosome, to experimentally examine the effects of the uniparentally inherited parts of the genome, as well as their interaction, in males. We assay longevity and gene expression through RNA-sequencing. We detect an important role for both mitochondrial and Y-linked genes, as well as extensive mitochondrial-Y chromosome epistasis. In particular, genes involved in male reproduction appear to be especially sensitive to such interactions, and variation on the Y chromosome is associated with differences in longevity. Despite these interactions, we find no evidence that the mitochondrial genome and Y chromosome are co-adapted within a geographical region. Overall, our study demonstrates a key role for the uniparentally inherited parts of the genome for male biology, but also that mito-nuclear interactions are complex and not easily predicted from simple transmission asymmetries.


Asunto(s)
Drosophila melanogaster , Epistasis Genética/fisiología , Cromosoma Y/genética , Animales , Núcleo Celular , ADN Mitocondrial , Femenino , Genes Ligados a Y , Genoma Mitocondrial , Longevidad , Masculino , Mitocondrias
11.
Sex Transm Dis ; 47(5): 314-320, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32187172

RESUMEN

BACKGROUND: Preexposure prophylaxis (PrEP) can reduce HIV acquisition among female sex workers (FSWs). However, changes in condomless sex frequency after PrEP initiation could reduce PrEP effectiveness when PrEP adherence is suboptimal as well as increase the risk of acquiring other sexually transmitted infections. Objective measures of condomless sex may be more accurate for determining changes in sexual behavior than self-reported measures. METHODS: We longitudinally measured self-reported condom use, number of clients, and presence of Y-chromosomal DNA (Yc-DNA) in vaginal swabs among 267 FSWs accessing PrEP at 4 clinics in Senegal between 2015 and 2016. We assessed trends in sexual behavior over time since PrEP initiation using generalized estimating equations and evaluated predictors of Yc-DNA detection. RESULTS: We found no increase in self-reported condomless sex with clients (odds ratio [OR], 0.94; 95% confidence interval [CI], 0.89-1.00), main partners (OR, 0.99; 95% CI, 0.96-1.02), or Yc-DNA detection (OR, 0.99; 95% CI, 0.90-1.08) over time since initiation. Y-chromosomal DNA was detected in 34 (22%) of 154 swabs tested and in 15 (26%) of 58 swabs from FSW reporting consistent condom use among both clients and main partners. Self-reported condom use with clients or main partners did not predict Yc-DNA detection. CONCLUSIONS: In a FSW PrEP demonstration project in Senegal, we found no evidence of risk compensation among FSWs on PrEP as measured by self-reported behavior or through Yc-DNA detection. Y-chromosomal DNA detection was frequently detected among FSWs reporting consistent condom use, highlighting limitations of self-reported sexual behavioral measures.


Asunto(s)
Fármacos Anti-VIH/administración & dosificación , Condones , Genes Ligados a Y , Profilaxis Pre-Exposición/estadística & datos numéricos , Trabajadores Sexuales , Conducta Sexual/estadística & datos numéricos , Sexo Inseguro/estadística & datos numéricos , Adulto , ADN , Femenino , Infecciones por VIH/diagnóstico , Infecciones por VIH/epidemiología , Infecciones por VIH/prevención & control , Humanos , Persona de Mediana Edad , Evaluación de Programas y Proyectos de Salud , Senegal/epidemiología , Parejas Sexuales
12.
Transgenic Res ; 29(4): 395-407, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32607872

RESUMEN

Advances in genome editing tools have reduced barriers to the creation of animal models. Due to their anatomical and physiological similarities to humans, there has been a growing need for pig models to study human diseases, for xenotransplantation and translational research. The ability to determine the sex of genetically modified embryos, cells or fetuses is beneficial for every project involving the production of transgenic animals. This strategy can improve the time-efficiency and lower the production costs. Additionally, sex assessment is very useful for wildlife studies to understand population behavior and structure. Thus, we developed a simple and fast PCR-based protocol for sex determination in pigs by using a unique primer set to amplify either the DDX3X or DDX3Y gene. The sex was 100% correctly assigned when tail genomic DNA, Day-35 fetus and hair samples from pigs were used. For both blastocysts and oocytes (84.6% and 96.5% of efficacy, respectively) the unidentified samples were potentially due to a limitation in sample size. Our assay also worked for domestic sheep (Ovis aries), American bison (Bison bison) and European cattle (Bos taurus) samples and by in silico analysis we confirmed X-Y amplicon length polymorphisms for the DDX3 gene in 12 other mammalian species. This PCR protocol for determining sex in pig tissues and cells showed to be simple, specific, highly reproducible and less time consuming as well as an important tool for other livestock species and wildlife studies.


Asunto(s)
ARN Helicasas DEAD-box/genética , Genes Ligados a X , Genes Ligados a Y , Variación Genética , Análisis de Secuencia de ADN/métodos , Análisis para Determinación del Sexo/métodos , Animales , Bison , Bovinos , Femenino , Masculino , Reacción en Cadena de la Polimerasa , Oveja Doméstica , Porcinos
13.
Proc Natl Acad Sci U S A ; 114(44): 11721-11726, 2017 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-29078298

RESUMEN

The Y chromosome is a unique genetic environment defined by a lack of recombination and male-limited inheritance. The Drosophila Y chromosome has been gradually acquiring genes from the rest of the genome, with only seven Y-linked genes being gained over the past 63 million years (0.12 gene gains per million years). Using a next-generation sequencing (NGS)-powered genomic scan, we show that gene transfers to the Y chromosome are much more common than previously suspected: at least 25 have arisen across three Drosophila species over the past 5.4 million years (1.67 per million years for each lineage). The gene transfer rate is significantly lower in Drosophila melanogaster than in the Drosophila simulans clade, primarily due to Y-linked retrotranspositions being significantly more common in the latter. Despite all Y-linked gene transfers being evolutionarily recent (<1 million years old), only three showed evidence for purifying selection (ω ≤ 0.14). Thus, although the resulting Y-linked functional gene acquisition rate (0.25 new genes per million years) is double the longer-term estimate, the fate of most new Y-linked genes is defined by rapid degeneration and pseudogenization. Our results show that Y-linked gene traffic, and the molecular mechanisms governing these transfers, can diverge rapidly between species, revealing the Drosophila Y chromosome to be more dynamic than previously appreciated. Our analytical method provides a powerful means to identify Y-linked gene transfers and will help illuminate the evolutionary dynamics of the Y chromosome in Drosophila and other species.


Asunto(s)
Drosophila/genética , Genes Ligados a Y , Cromosoma Y/genética , Animales , Cromosomas de Insectos , Proteínas de Drosophila/genética , Evolución Molecular , Femenino , Genes de Insecto , Masculino , Filogenia , Especificidad de la Especie , Translocación Genética
14.
Biol Reprod ; 100(3): 697-710, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30289439

RESUMEN

The fertility of sex-reversed XY female mice is severely impaired by a massive loss of oocytes and failure of meiotic progression. This phenomenon remains an outstanding mystery. We sought to determine the molecular etiology of XY oocyte dysfunction by generating sex-reversed females that bear genetic ablation of Sry, a vital sex determination gene, on an inbred C57BL/6 background. These mutant mice, termed XYsry- mutants, showed severe attrition of germ cells during fetal development, resulting in the depletion of ovarian germ cells prior to sexual maturation. Comprehensive transcriptome analyses of primordial germ cells (PGCs) and postnatal oocytes demonstrated that XYsry- females had deviated significantly from normal developmental processes during the stages of mitotic proliferation. The impaired proliferation of XYsry- PGCs was associated with aberrant ß-catenin signaling and the excessive expression of transposable elements. Upon entry to the meiotic stage, XYsry- oocytes demonstrated extensive defects, including the impairment of crossover formation, the failure of primordial follicle maintenance, and no capacity for embryo development. Together, these results suggest potential molecular causes for germ cell disruption in sex-reversed female mice, thereby providing insights into disorders of sex differentiation in humans, such as "Swyer syndrome," in which patients with an XY karyotype present as typical females and are infertile.


Asunto(s)
Disgenesia Gonadal 46 XY/fisiopatología , Oocitos/crecimiento & desarrollo , Proteína de la Región Y Determinante del Sexo/genética , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Ligados a Y , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mitosis , Mutación , Transcriptoma
15.
Hum Reprod ; 34(6): 1117-1125, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31111890

RESUMEN

STUDY QUESTION: Is there an increased prevalence of male microchimerism in women with Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome, as evidence of fetal exposure to blood and anti-Müllerian hormone (AMH) from a (vanished) male co-twin resulting in regression of the Müllerian duct derivatives? SUMMARY ANSWER: Predominant absence of male microchimerism in adult women with MRKH syndrome does not support our hypothesis that intrauterine blood exchange with a (vanished) male co-twin is the pathophysiological mechanism. WHAT IS KNOWN ALREADY: The etiology of MRKH is unclear. Research on the phenotype analogous condition in cattle (freemartinism) has yielded the hypothesis that Müllerian duct development is inhibited by exposure to AMH in utero. In cattle, the male co-twin has been identified as the source for AMH, which is transferred via placental blood exchange. In human twins, a similar exchange of cellular material has been documented by detection of chimerism, but it is unknown whether this has clinical consequences. STUDY DESIGN, SIZE, DURATION: An observational case-control study was performed to compare the presence of male microchimerism in women with MRKH syndrome and control women. Through recruitment via the Dutch patients' association of women with MRKH (comprising 300 members who were informed by email or regular mail), we enrolled 96 patients between January 2017 and July 2017. The control group consisted of 100 women who reported never having been pregnant. PARTICIPANTS/MATERIALS, SETTING, METHODS: After written informed consent, peripheral blood samples were obtained by venipuncture, and genomic DNA was extracted. Male microchimerism was detected by Y-chromosome-specific real-time quantitative PCR, with use of DYS14 marker. Possible other sources for microchimerism, for example older brothers, were evaluated using questionnaire data. MAIN RESULTS AND THE ROLE OF CHANCE: The final analysis included 194 women: 95 women with MRKH syndrome with a mean age of 40.9 years and 99 control women with a mean age of 30.2 years. In total, 54 women (56.8%) were identified as having typical MRKH syndrome, and 41 women (43.2%) were identified as having atypical MRKH syndrome (when extra-genital malformations were present). The prevalence of male microchimerism was significantly higher in the control group than in the MRKH group (17.2% versus 5.3%, P = 0.009). After correcting for age, women in the control group were 5.8 times more likely to have male microchimerism (odds ratio 5.84 (CI 1.59-21.47), P = 0.008). The mean concentration of male microchimerism in the positive samples was 56.0 male genome equivalent per 1 000 000 cells. The prevalence of male microchimerism was similar in women with typical MRKH syndrome and atypical MRKH syndrome (5.6% versus 4.9%, P = 0.884). There were no differences between women with or without microchimerism in occurrence of alternative sources of XY cells, such as older brothers, previous blood transfusion, or history of sexual intercourse. LIMITATIONS, REASON FOR CAUTION: We are not able to draw definitive conclusions regarding the occurrence of AMH exchange during embryologic development in women with MRKH syndrome. Our subject population includes all adult women and therefore is reliant on long-term prevalence of microchimerism. Moreover, we have only tested blood, and, theoretically, the cells may have grafted anywhere in the body during development. It must also be considered that the exchange of AMH may occur without the transfusion of XY cells and therefore cannot be discovered by chimerism detection. WIDER IMPLICATIONS OF THE FINDINGS: This is the first study to test the theory that freemartinism causes the MRKH syndrome in humans. The study aimed to test the presence of male microchimerism in women with MRKH syndrome as a reflection of early fetal exposure to blood and AMH from a male (vanished) co-twin. We found that male microchimerism was only present in 5.3% of the women with MRKH syndrome, a significantly lower percentage than in the control group (17.2%). Our results do not provide evidence for an increased male microchimerism in adult women with MRKH as a product of intrauterine blood exchange. However, the significant difference in favor of the control group is of interest to the ongoing discussion on microchimeric cell transfer and the possible sources of XY cells. STUDY FUNDING/COMPETING INTEREST(S): None. TRIAL REGISTRATION NUMBER: Dutch trial register, NTR5961.


Asunto(s)
Trastornos del Desarrollo Sexual 46, XX/genética , Quimerismo , Anomalías Congénitas/genética , Genes Ligados a Y/genética , Conductos Paramesonéfricos/anomalías , Conductos Paramesonéfricos/crecimiento & desarrollo , Trastornos del Desarrollo Sexual 46, XX/sangre , Trastornos del Desarrollo Sexual 46, XX/diagnóstico , Adulto , Biomarcadores/análisis , Estudios de Casos y Controles , Anomalías Congénitas/sangre , Anomalías Congénitas/diagnóstico , Femenino , Humanos , Persona de Mediana Edad , Prevalencia , Reacción en Cadena en Tiempo Real de la Polimerasa , Adulto Joven
16.
Int J Legal Med ; 133(4): 1033-1035, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30032460

RESUMEN

In the present study, blood samples of 984 unrelated Han individuals were collected from Dongfang, Southern China, after informed consent. A total of 29 Y-chromosomal short tandem repeat (Y-STR) were analyzed, including DYF387S1, DYS19, DYS385ab, DYS389I, DYS389II, DYS390, DYS391, DYS392, DYS393, DYS437, DYS438, DYS439, DYS444, DYS447, DYS448, DYS449, DYS456, DYS458, DYS460, DYS481, DYS508, DYS518, DYS533, DYS576, DYS635, DYS643 and GATAH4. A total of 749 different haplotypes were found among 984 individuals, of which 645 were unique. The haplotype diversity was 0.9988 and the discrimination capacity was 0.7612, while the match probability was 0.0025. The smallest genetic distance (RST = 0.0155) was found between the Dongfang Han population and Guizhou Han population, while the largest genetic distance (RST = 0.1284) was observed with Gansu Tibetan.


Asunto(s)
Pueblo Asiatico/genética , Cromosomas Humanos Y/genética , Genes Ligados a Y/genética , Repeticiones de Microsatélite/genética , China , ADN/genética , Dermatoglifia del ADN/métodos , Haplotipos , Humanos , Masculino
17.
J Dairy Sci ; 102(8): 7226-7236, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31202648

RESUMEN

The mammalian Y chromosome gene families in the ampliconic region are expressed predominantly or exclusively in the testis, and their copy number variations (CNV) are significantly associated with male reproductive traits, suggesting they have important roles in spermatogenesis and testicular development. ZNF280AY (zinc finger protein 280A, Y-linked) is a member of the zinc finger protein family and has been identified as a bovid-specific Y-chromosome gene. The current study applied a reliable quantitative real-time PCR method to estimate the CNV of ZNF280AY in 715 bulls across 21 cattle breeds and to further investigate the association of the CNV of ZNF280AY with bull reproductive traits and ZNF280AY mRNA expression levels in adult testis. The results revealed that the median copy number of ZNF280AY was 47, and the copy number varied from 11 to 154, showing significant CNV between and within the investigated cattle breeds. In addition, all 715 bulls were classified into Y1, Y2, and Y3 lineage groups based on a rapid genotyping method described previously. Pairwise comparisons indicated that bulls belonging to the Y1 lineage had a significantly lower median copy number (40) than bulls belonging to the Y2 (52) and Y3 lineages (57). Association analysis revealed that the CNV of ZNF280AY was correlated negatively with the percentage of normal sperm and sperm concentration in Holstein bulls, whereas no significant correlation was observed with ejaculation volume, total sperm count, sperm motility, postthaw motility (PTM), and scrotal circumference in Holstein and Simmental bulls. Furthermore, no correlation was observed between ZNF280AY copy number and ZNF280AY mRNA expression levels in the testis. The current study suggests that the CNV of the ZNF280AY gene family is associated with male reproductive traits and may serve as a valuable marker for early bull fertility selection in Holstein breeding programs.


Asunto(s)
Bovinos/genética , Variaciones en el Número de Copia de ADN , Fertilidad/genética , Regulación de la Expresión Génica , Genes Ligados a Y/genética , Reproducción/genética , Cromosoma Y/genética , Animales , Cruzamiento , Bovinos/fisiología , Marcadores Genéticos/genética , Genotipo , Masculino , Especificidad de Órganos , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa/veterinaria , Especificidad de la Especie , Recuento de Espermatozoides/veterinaria , Motilidad Espermática/genética , Espermatogénesis/genética , Testículo/fisiología , Dedos de Zinc/genética
18.
Reprod Domest Anim ; 54(3): 580-584, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30597663

RESUMEN

Chromosomal abnormalities are a major cause of infertility and reproductive problems in equids. Nowadays, their detection is rising due to the use of new diagnostic tools based on molecular markers instead of karyotyping. Reports of this kind of genetic aberrations in domestic donkeys (Equus asinus) are extremely scarce, despite their importance in human activities. In the present study, we analysed the implementation of a short-tandem-repeat (STR)-based molecular method initially developed for horses, as a diagnostic tool to detect chromosomal abnormalities in donkeys. The frequency of five X-linked (LEX003, LEX026, TKY38, TKY270 and UCEDQ502) and one Y-linked (ECAYM2) molecular markers and one Y-linked gene (sex-determining region Y, SRY) was characterized in 121 donkeys from two diverse breeds, the Spanish Andalusian and the African Moroccan breeds. The molecular panel showed 100% sensitivity and 99.67% specificity in detecting 10 different chromosomal abnormalities in the species. In conclusion, this methodology is a valid, rapid and low-cost tool for the detection and characterization of chromosomal abnormalities in domestic donkeys.


Asunto(s)
Equidae/genética , Pruebas Genéticas/veterinaria , Infertilidad/veterinaria , Aberraciones Cromosómicas Sexuales , Animales , Cruzamiento , Femenino , Genes Ligados a X , Genes Ligados a Y , Pruebas Genéticas/métodos , Infertilidad/diagnóstico , Infertilidad/genética , Cariotipificación , Masculino , Repeticiones de Microsatélite , Marruecos , España
19.
Int J Mol Sci ; 21(1)2019 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-31905822

RESUMEN

Sex-related growth differences between male and female embryos remain an attractive subject for reproductive biologists. This study aimed to investigate the endogenous factors that play a crucial role in the pace of early development between male and female bovine embryos. Using sex pre-selected semen by Y-specific monoclonal antibodies for the production of bovine embryos, we characterized the critical endogenous factors that are responsible for creating the development differences, especially during the pre-implantation period between male and female embryos. Our results showed that at day seven, (57.8%) Y-sperm sorted in vitro cultured embryos reached the expanded blastocyst (BL) stage, whereas the X-sperm sorted group were only 25%. Y-BLs showed higher mRNA abundance of pluripotency and developmental competency regulators, such as Oct4 and IGF1-R. Interestingly, Y-sperm sorted BLs had a homogeneous mitochondrial distribution pattern, higher mitochondrial membrane potential (∆Ñ°m), efficient OXPHOS (oxidative phosphorylation) system and well-encountered production of ROS (reactive oxygen species) level. Moreover, Y-blastocysts (BLs) showed less utilization of glucose metabolism relative to the X-BLs group. Importantly, both sexes showed differences in the timing of epigenetic events. All these factors directly or indirectly orchestrate the whole embryonic progression and may help in the faster and better quality yield of BL in the Y-sperm sorted group compared to the X counterpart group.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Blastocisto/metabolismo , Desarrollo Embrionario/inmunología , Cromosoma Y , Animales , Bovinos/embriología , Embrión de Mamíferos , Desarrollo Embrionario/genética , Femenino , Genes Ligados a X , Genes Ligados a Y , Glucosa/metabolismo , Cinética , Masculino , Potencial de la Membrana Mitocondrial , Mitocondrias , Fosforilación , Factores Sexuales , Espermatozoides , Cromosoma X
20.
Am J Med Genet B Neuropsychiatr Genet ; 180(7): 471-482, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31161682

RESUMEN

We describe a unique male with a dicentric Y chromosome whose phenotype was compared to that of males with 47,XYY (XYY). The male Y-chromosome aneuploidy XYY is associated with physical, behavioral/cognitive phenotypes, and autism spectrum disorders. We hypothesize that increased risk for these phenotypes is caused by increased copy number/overexpression of Y-encoded genes. Specifically, an extra copy of the neuroligin gene NLGN4Y might elevate the risk of autism in boys with XYY. We present a unique male with the karyotype 46,X,idic(Y)(q11.22), which includes duplication of the Y short arm and proximal long arm and deletion of the distal long arm, evaluated his physical, behavioral/cognitive, and neuroimaging/magnetoencephalography (MEG) phenotypes, and measured blood RNA expression of Y genes. The proband had tall stature and cognitive function within the typical range, without autism features. His blood RNA showed twofold increase in expression of Yp genes versus XY controls, and absent expression of deleted Yq genes, including NLGN4Y. The M100 latencies were similar to findings in typically developing males. In summary, the proband had overexpression of a subset of Yp genes, absent NLGN4Y expression, without ASD findings or XYY-MEG latency findings. These results are consistent with a role for NLGN4Y overexpression in the etiology of behavioral phenotypes associated with XYY. Further investigation of NLGN4Y as an ASD risk gene in XYY is warranted. The genotype and phenotype(s) of this subject may also provide insight into how Y chromosome genes contribute to normal male development and the male predominance in ASD.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/genética , Cromosomas Humanos Y/genética , Cariotipo XYY/fisiopatología , Adolescente , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/fisiopatología , Trastorno Autístico/genética , Trastorno Autístico/fisiopatología , Moléculas de Adhesión Celular Neuronal/metabolismo , Niño , Aberraciones Cromosómicas , Variaciones en el Número de Copia de ADN/genética , Dosificación de Gen/genética , Genes Ligados a Y/genética , Humanos , Cariotipificación , Masculino , Pruebas Neuropsicológicas , Fenotipo , Cariotipo XYY/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA