Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
EMBO Rep ; 24(6): e56316, 2023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-37099396

RESUMEN

Spermatozoa have a unique genome organization. Their chromatin is almost completely devoid of histones and is formed instead of protamines, which confer a high level of compaction and preserve paternal genome integrity until fertilization. Histone-to-protamine transition takes place in spermatids and is indispensable for the production of functional sperm. Here, we show that the H3K79-methyltransferase DOT1L controls spermatid chromatin remodeling and subsequent reorganization and compaction of the spermatozoon genome. Using a mouse model in which Dot1l is knocked-out (KO) in postnatal male germ cells, we found that Dot1l-KO sperm chromatin is less compact and has an abnormal content, characterized by the presence of transition proteins, immature protamine 2 forms and a higher level of histones. Proteomic and transcriptomic analyses performed on spermatids reveal that Dot1l-KO modifies the chromatin prior to histone removal and leads to the deregulation of genes involved in flagellum formation and apoptosis during spermatid differentiation. As a consequence of these chromatin and gene expression defects, Dot1l-KO spermatozoa have less compact heads and are less motile, which results in impaired fertility.


Asunto(s)
Cromatina , Histonas , Animales , Masculino , Diferenciación Celular/genética , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Expresión Génica , Histonas/metabolismo , Proteómica , Semen/metabolismo , Espermatogénesis/genética , Espermatozoides/metabolismo , Ratones
2.
Cell Mol Biol Lett ; 29(1): 74, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38750428

RESUMEN

By analyzing a mouse Interspecific Recombinant Congenic Strain (IRCS), we previously identified a quantitative trait locus (QTL), called Mafq1 on mouse chromosome 1, that is associated with male hypofertility and ultrastructural sperm abnormalities. Within this locus, we identified a new candidate gene that could be implicated in a reproductive phenotype: Tex44 (Testis-expressed protein 44). We thus performed a CRISPR/Cas9-mediated complete deletion of this gene in mice in order to study its function. Tex44-KO males were severely hypofertile in vivo and in vitro due to a drastic reduction of sperm motility which itself resulted from important morphological sperm abnormalities. Namely, Tex44-KO sperm showed a disorganized junction between the midpiece and the principal piece of the flagellum, leading to a 180° flagellar bending in this region. In addition, the loss of some axonemal microtubule doublets and outer dense fibers in the flagellum's principal piece has been observed. Our results suggest that, in mice, TEX44 is implicated in the correct set-up of the sperm flagellum during spermiogenesis and its absence leads to flagellar abnormalities and consequently to severe male hypofertility.


Asunto(s)
Infertilidad Masculina , Ratones Noqueados , Motilidad Espermática , Cola del Espermatozoide , Animales , Masculino , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Motilidad Espermática/genética , Cola del Espermatozoide/patología , Cola del Espermatozoide/metabolismo , Ratones , Espermatozoides/metabolismo , Espermatogénesis/genética , Flagelos/genética , Flagelos/metabolismo , Ratones Endogámicos C57BL , Sistemas CRISPR-Cas/genética
3.
Am J Hum Genet ; 105(6): 1148-1167, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31735292

RESUMEN

In humans, structural or functional defects of the sperm flagellum induce asthenozoospermia, which accounts for the main sperm defect encountered in infertile men. Herein we focused on morphological abnormalities of the sperm flagellum (MMAF), a phenotype also termed "short tails," which constitutes one of the most severe sperm morphological defects resulting in asthenozoospermia. In previous work based on whole-exome sequencing of a cohort of 167 MMAF-affected individuals, we identified bi-allelic loss-of-function mutations in more than 30% of the tested subjects. In this study, we further analyzed this cohort and identified five individuals with homozygous truncating variants in TTC29, a gene preferentially and highly expressed in the testis, and encoding a tetratricopeptide repeat-containing protein related to the intraflagellar transport (IFT). One individual carried a frameshift variant, another one carried a homozygous stop-gain variant, and three carried the same splicing variant affecting a consensus donor site. The deleterious effect of this last variant was confirmed on the corresponding transcript and protein product. In addition, we produced and analyzed TTC29 loss-of-function models in the flagellated protist T. brucei and in M. musculus. Both models confirmed the importance of TTC29 for flagellar beating. We showed that in T. brucei the TPR structural motifs, highly conserved between the studied orthologs, are critical for TTC29 axonemal localization and flagellar beating. Overall our work demonstrates that TTC29 is a conserved axonemal protein required for flagellar structure and beating and that TTC29 mutations are a cause of male sterility due to MMAF.


Asunto(s)
Astenozoospermia/etiología , Axonema/patología , Flagelos/patología , Infertilidad Masculina/etiología , Proteínas Asociadas a Microtúbulos/genética , Mutación , Animales , Astenozoospermia/metabolismo , Astenozoospermia/patología , Axonema/genética , Axonema/metabolismo , Evolución Molecular , Femenino , Fertilización In Vitro , Flagelos/genética , Flagelos/metabolismo , Humanos , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Masculino , Ratones Endogámicos C57BL , Trypanosoma brucei brucei/fisiología , Tripanosomiasis
4.
Nucleic Acids Res ; 48(8): 4115-4138, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32182340

RESUMEN

Epigenetic regulation of gene expression is tightly controlled by the dynamic modification of histones by chemical groups, the diversity of which has largely expanded over the past decade with the discovery of lysine acylations, catalyzed from acyl-coenzymes A. We investigated the dynamics of lysine acetylation and crotonylation on histones H3 and H4 during mouse spermatogenesis. Lysine crotonylation appeared to be of significant abundance compared to acetylation, particularly on Lys27 of histone H3 (H3K27cr) that accumulates in sperm in a cleaved form of H3. We identified the genomic localization of H3K27cr and studied its effects on transcription compared to the classical active mark H3K27ac at promoters and distal enhancers. The presence of both marks was strongly associated with highest gene expression. Assessment of their co-localization with transcription regulators (SLY, SOX30) and chromatin-binding proteins (BRD4, BRDT, BORIS and CTCF) indicated systematic highest binding when both active marks were present and different selective binding when present alone at chromatin. H3K27cr and H3K27ac finally mark the building of some sperm super-enhancers. This integrated analysis of omics data provides an unprecedented level of understanding of gene expression regulation by H3K27cr in comparison to H3K27ac, and reveals both synergistic and specific actions of each histone modification.


Asunto(s)
Elementos de Facilitación Genéticos , Epigénesis Genética , Código de Histonas , Regiones Promotoras Genéticas , Espermatogénesis/genética , Acetilcoenzima A/metabolismo , Acetilación , Acilcoenzima A/metabolismo , Animales , Evolución Biológica , Crotonatos/metabolismo , Genómica , Histonas/química , Histonas/metabolismo , Lisina/metabolismo , Masculino , Metabolómica , Ratones Endogámicos C57BL , Proteómica , Transcripción Genética , Levaduras/metabolismo , Levaduras/fisiología
5.
Int J Mol Sci ; 23(22)2022 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-36430291

RESUMEN

Fertilization is a complex process that requires successive stages and culminates in the adhesion/fusion of gamete membranes. If the question of the involvement of oocyte integrins has been swept away by deletion experiments, that of the involvement of sperm integrins remains to be further characterized. In the present study, we addressed the question of the feasibility of sperm-oocyte adhesion/fusion and early implantation in the absence of sperm ß1 integrin. Males and females with ß1 integrin-depleted sperm and oocytes were mated, and fertilization outcome was monitored by a gestational ultrasound analysis. Results suggest that although the sperm ß1 integrin participates in gamete adhesion/fusion, it is dispensable for fertilization in mice. However, sperm- and/or oocyte-originated integrin ß1 is essential for post-implantation development. Redundancy phenomena could be at the origin of a compensatory expression or alternative dimerization pattern.


Asunto(s)
Integrina beta1 , Interacciones Espermatozoide-Óvulo , Femenino , Ratones , Masculino , Animales , Integrina beta1/genética , Integrina beta1/metabolismo , Semen/metabolismo , Oocitos/metabolismo , Espermatozoides/metabolismo , Fertilización , Integrinas/metabolismo
6.
Mol Biol Evol ; 37(12): 3453-3468, 2020 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-32658962

RESUMEN

Transmission distorters (TDs) are genetic elements that favor their own transmission to the detriments of others. Slx/Slxl1 (Sycp3-like-X-linked and Slx-like1) and Sly (Sycp3-like-Y-linked) are TDs, which have been coamplified on the X and Y chromosomes of Mus species. They are involved in an intragenomic conflict in which each favors its own transmission, resulting in sex ratio distortion of the progeny when Slx/Slxl1 versus Sly copy number is unbalanced. They are specifically expressed in male postmeiotic gametes (spermatids) and have opposite effects on gene expression: Sly knockdown leads to the upregulation of hundreds of spermatid-expressed genes, whereas Slx/Slxl1-deficiency downregulates them. When both Slx/Slxl1 and Sly are knocked down, sex ratio distortion and gene deregulation are corrected. Slx/Slxl1 and Sly are, therefore, in competition but the molecular mechanism remains unknown. By comparing their chromatin-binding profiles and protein partners, we show that SLX/SLXL1 and SLY proteins compete for interaction with H3K4me3-reader SSTY1 (Spermiogenesis-specific-transcript-on-the-Y1) at the promoter of thousands of genes to drive their expression, and that the opposite effect they have on gene expression is mediated by different abilities to recruit SMRT/N-Cor transcriptional complex. Their target genes are predominantly spermatid-specific multicopy genes encoded by the sex chromosomes and the autosomal Speer/Takusan. Many of them have coamplified with not only Slx/Slxl1/Sly but also Ssty during muroid rodent evolution. Overall, we identify Ssty as a key element of the X versus Y intragenomic conflict, which may have influenced gene content and hybrid sterility beyond Mus lineage since Ssty amplification on the Y predated that of Slx/Slxl1/Sly.


Asunto(s)
Evolución Biológica , Proteínas Nucleares/genética , Proteínas/genética , Cromosoma X/genética , Cromosoma Y/genética , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/genética , Masculino , Ratones Endogámicos C57BL , Proteínas Nucleares/metabolismo , Proteínas Quinasas/genética , Proteínas/metabolismo , Espermátides/metabolismo , Sitio de Iniciación de la Transcripción
7.
Int J Mol Sci ; 22(4)2021 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-33669425

RESUMEN

Thanks to the analysis of an Interspecific Recombinant Congenic Strain (IRCS), we previously defined the Mafq1 quantitative trait locus as an interval on mouse Chromosome 1 associated with male hypofertility and ultrastructural abnormalities. We identified the Spermatogenesis associated protein 3 gene (Spata3 or Tsarg1) as a pertinent candidate within the Mafq1 locus and performed the CRISPR-Cas9 mediated complete deletion of the gene to investigate its function. Male mice deleted for Spata3 were normally fertile in vivo but exhibited a drastic reduction of efficiency in in vitro fertilization assays. Mobility parameters were normal but ultrastructural analyses revealed acrosome defects and an overabundance of lipids droplets in cytoplasmic remnants. The deletion of the Spata3 gene reproduces therefore partially the phenotype of the hypofertile IRCS strain.


Asunto(s)
Acrosoma/patología , Fertilización In Vitro/métodos , Eliminación de Gen , Infertilidad Masculina/genética , Proteínas/genética , Acrosoma/metabolismo , Acrosoma/ultraestructura , Animales , Sistemas CRISPR-Cas , Modelos Animales de Enfermedad , Femenino , Infertilidad Masculina/metabolismo , Gotas Lipídicas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Embarazo , Proteínas/metabolismo , Motilidad Espermática/genética , Espermatogénesis/genética , Testículo/metabolismo
8.
Int J Mol Sci ; 21(22)2020 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-33187358

RESUMEN

We have previously shown, using antibodies, that the sperm alpha6beta1 integrin is involved in mouse gamete fusion in vitro. Here we report the conditional knockdown of the sperm Itgb1 gene. It induced a drastic failure of sperm fusogenic ability with sperm accumulation in the perivitelline space of in vitro inseminated oocytes deleted or not for the Itgb1 gene. These data demonstrate that sperm, but not oocyte, beta1 integrin subunit is involved in gamete adhesion/fusion. Curiously, knockdown males were fertile in vivo probably because of the incomplete Cre-mediated deletion of the sperm Itgb1 floxed gene. Indeed, this was shown by Western blot analysis and confirmed by both the viability and litter size of pups obtained by mating partially sperm Itgb1 deleted males with females producing completely deleted Itgb1 oocytes. Because of the total peri-implantation lethality of Itgb1 deletion in mice, we assume that sperm that escaped the Itgb1 excision seemed to be preferentially used to fertilize in vivo. Here, we showed for the first time that the deletion, even partial, of the sperm Itgb1 gene makes the sperm unable to normally fertilize oocytes. However, to elucidate the question of the essentiality of its role during fertilization, further investigations using a mouse expressing a recombinase more effective in male germ cells are necessary.


Asunto(s)
Adhesión Celular/genética , Células Germinativas/fisiología , Integrina beta1/genética , Subunidades de Proteína/genética , Animales , Adhesión Celular/fisiología , Fusión Celular/métodos , Femenino , Fertilización/genética , Fertilización/fisiología , Masculino , Ratones , Ratones Noqueados , Oocitos/fisiología , Interacciones Espermatozoide-Óvulo/genética , Interacciones Espermatozoide-Óvulo/fisiología , Espermatozoides/fisiología
9.
Int J Mol Sci ; 21(22)2020 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-33198087

RESUMEN

Male fertility disorders often have their origin in disturbed spermatogenesis, which can be induced by genetic factors. In this study, we used interspecific recombinant congenic mouse strains (IRCS) to identify genes responsible for male infertility. Using ultrasonography, in vivo and in vitro fertilization (IVF) and electron microscopy, the phenotyping of several IRCS carrying mouse chromosome 1 segments of Mus spretus origin revealed a decrease in the ability of sperm to fertilize. This teratozoospermia included the abnormal anchoring of the acrosome to the nucleus and a persistence of residual bodies at the level of epididymal sperm midpiece. We identified a quantitative trait locus (QTL) responsible for these phenotypes and we have proposed a short list of candidate genes specifically expressed in spermatids. The future functional validation of candidate genes should allow the identification of new genes and mechanisms involved in male infertility.


Asunto(s)
Cromosomas Humanos Par 1/genética , Infertilidad Masculina/genética , Sitios de Carácter Cuantitativo/genética , Acrosoma/fisiología , Animales , Núcleo Celular/genética , Núcleo Celular/fisiología , Epidídimo/fisiología , Femenino , Humanos , Masculino , Ratones , Fenotipo , Espermátides/fisiología , Espermatogénesis/genética , Espermatozoides/fisiología , Teratozoospermia/genética
10.
Genes (Basel) ; 14(5)2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-37239343

RESUMEN

Spermiogenesis is the step during which post-meiotic cells, called spermatids, undergo numerous morphological changes and differentiate into spermatozoa. Thousands of genes have been described to be expressed at this stage and could contribute to spermatid differentiation. Genetically-engineered mouse models using Cre/LoxP or CrispR/Cas9 are the favored approaches to characterize gene function and better understand the genetic basis of male infertility. In the present study, we produced a new spermatid-specific Cre transgenic mouse line, in which the improved iCre recombinase is expressed under the control of the acrosomal vesicle protein 1 gene promoter (Acrv1-iCre). We show that Cre protein expression is restricted to the testis and only detected in round spermatids of stage V to VIII seminiferous tubules. The Acrv1-iCre line can conditionally knockout a gene during spermiogenesis with a > 95% efficiency. Therefore, it could be useful to unravel the function of genes during the late stage of spermatogenesis, but it can also be used to produce an embryo with a paternally deleted allele without causing early spermatogenesis defects.


Asunto(s)
Espermátides , Espermatozoides , Ratones , Animales , Masculino , Espermátides/metabolismo , Ratones Transgénicos , Espermatozoides/metabolismo , Integrasas/genética , Integrasas/metabolismo , Proteínas de la Membrana/metabolismo
11.
Science ; 371(6525)2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33414192

RESUMEN

Posttranslational modifications of the microtubule cytoskeleton have emerged as key regulators of cellular functions, and their perturbations have been linked to a growing number of human pathologies. Tubulin glycylation modifies microtubules specifically in cilia and flagella, but its functional and mechanistic roles remain unclear. In this study, we generated a mouse model entirely lacking tubulin glycylation. Male mice were subfertile owing to aberrant beat patterns of their sperm flagella, which impeded the straight swimming of sperm cells. Using cryo-electron tomography, we showed that lack of glycylation caused abnormal conformations of the dynein arms within sperm axonemes, providing the structural basis for the observed dysfunction. Our findings reveal the importance of microtubule glycylation for controlled flagellar beating, directional sperm swimming, and male fertility.


Asunto(s)
Dineínas Axonemales/metabolismo , Fertilidad/genética , Infertilidad Masculina/enzimología , Procesamiento Proteico-Postraduccional , Motilidad Espermática/genética , Cola del Espermatozoide/enzimología , Tubulina (Proteína)/metabolismo , Animales , Dineínas Axonemales/química , Cilios/enzimología , Microscopía por Crioelectrón , Modelos Animales de Enfermedad , Tomografía con Microscopio Electrónico , Infertilidad Masculina/genética , Masculino , Ratones , Ratones Noqueados , Tubulina (Proteína)/química
12.
Commun Biol ; 4(1): 691, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099857

RESUMEN

Chromatin of male and female gametes undergoes a number of reprogramming events during the transition from germ cell to embryonic developmental programs. Although the rearrangement of DNA methylation patterns occurring in the zygote has been extensively characterized, little is known about the dynamics of DNA modifications during spermatid maturation. Here, we demonstrate that the dynamics of 5-carboxylcytosine (5caC) correlate with active transcription of LINE-1 retroelements during murine spermiogenesis. We show that the open reading frames of active and evolutionary young LINE-1s are 5caC-enriched in round spermatids and 5caC is eliminated from LINE-1s and spermiogenesis-specific genes during spermatid maturation, being simultaneously retained at promoters and introns of developmental genes. Our results reveal an association of 5caC with activity of LINE-1 retrotransposons suggesting a potential direct role for this DNA modification in fine regulation of their transcription.


Asunto(s)
Citosina/análogos & derivados , Elementos de Nucleótido Esparcido Largo , Sistemas de Lectura Abierta , Espermátides/metabolismo , Animales , Citosina/metabolismo , Masculino , Ratones , Espermátides/citología , Espermatogénesis , Transcripción Genética
13.
Sci Rep ; 10(1): 5335, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32210282

RESUMEN

Three genes are known to be essential for gamete adhesion/fusion (Cd9, Izumo1 and Juno). Here, we confirmed that Spaca6 null males are infertile and showed that their sperm accumulate in the perivitelline space but are unable to fuse with oocyte. Like IZUMO1, SPACA6 which is expressed by human sperm, is remained on the equatorial segment after acrosomal reaction and is involved in human fertilization since an anti-SPACA6 antibody inhibited it. Despite the similarity of the phenotypes caused by Spaca6 and Izumo1 knockouts, these are not redundant and the essential relocation of IZUMO1 is not affected by the lack of SPACA6. We propose a model in which IZUMO1 and SPACA6 would be part of a molecular complex necessary for gamete fusion and that their concomitant presence would be required for the recruitment of another essential molecular actor, such as a fusogen, for the fusion to take place.


Asunto(s)
Proteínas de Plasma Seminal/metabolismo , Interacciones Espermatozoide-Óvulo/fisiología , Espermatozoides/fisiología , Reacción Acrosómica , Animales , Células COS , Chlorocebus aethiops , Femenino , Fertilización In Vitro , Humanos , Inmunoglobulinas/genética , Inmunoglobulinas/metabolismo , Infertilidad Masculina/genética , Infertilidad Masculina/terapia , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Proteínas de Plasma Seminal/genética , Cabeza del Espermatozoide/fisiología , Inyecciones de Esperma Intracitoplasmáticas
14.
Curr Biol ; 29(21): 3692-3698.e4, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31630954

RESUMEN

The mouse sex chromosomes exhibit an extraordinary level of copy number amplification of postmeiotically expressed genes [1, 2], driven by an "arms race" (genomic conflict) between the X and Y chromosomes over the control of offspring sex ratio. The sex-linked ampliconic transcriptional regulators Slx and Sly [3-7] have opposing effects on global transcription levels of the sex chromosomes in haploid spermatids via regulation of postmeiotic sex chromatin (PMSC) [8-11] and opposing effects on offspring sex ratio. Partial deletions of the Y chromosome (Yq) that reduce Sly copy number lead to global overexpression of sex-linked genes in spermatids and either a distorted sex ratio in favor of females (smaller deletions) or sterility (larger deletions) [12-16]. Despite a large body of work studying the role of the sex chromosomes in regulating spermatogenesis (recent reviews [17-20]), most studies do not address differential fertility effects on X- and Y-bearing cells. Hence, in this study, we concentrate on identifying physiological differences between X- and Y-bearing sperm from Yq-deleted males that affect their relative fertilizing ability and consequently lead to sex ratio skewing. We show that X- and Y-bearing sperm in these males have differential motility and morphology but are equally able to penetrate the cumulus and fertilize the egg once at the site of fertilization. The altered motility is thus deduced to be the proximate cause of the skew. This represents the first demonstration of a specific difference in sperm function associated with sex ratio skewing.


Asunto(s)
Evolución Biológica , Cromosomas Sexuales/fisiología , Motilidad Espermática , Espermatozoides/fisiología , Cromosoma Y/genética , Animales , Masculino , Ratones , Razón de Masculinidad
15.
Epigenetics Chromatin ; 11(1): 2, 2018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-29329550

RESUMEN

BACKGROUND: Histones organize DNA into chromatin through a variety of processes. Among them, a vast diversity of histone variants can be incorporated into chromatin and finely modulate its organization and functionality. Classically, the study of histone variants has largely relied on antibody-based assays. However, antibodies have a limited efficiency to discriminate between highly similar histone variants. RESULTS: In this study, we established a mass spectrometry-based analysis to address this challenge. We developed a targeted proteomics method, using selected reaction monitoring or parallel reaction monitoring, to quantify a maximum number of histone variants in a single multiplexed assay, even when histones are present in a crude extract. This strategy was developed on H2A and H2B variants, using 55 peptides corresponding to 25 different histone sequences, among which a few differ by a single amino acid. The methodology was then applied to mouse testis extracts in which almost all histone variants are expressed. It confirmed the abundance profiles of several testis-specific histones during successive stages of spermatogenesis and the existence of predicted H2A.L.1 isoforms. This methodology was also used to explore the over-expression pattern of H2A.L.1 isoforms in a mouse model of male infertility. CONCLUSIONS: Our results demonstrate that targeted proteomics is a powerful method to quantify highly similar histone variants and isoforms. The developed method can be easily transposed to the study of human histone variants, whose abundance can be deregulated in various diseases.


Asunto(s)
Histonas/metabolismo , Espectrometría de Masas/métodos , Proteómica/métodos , Testículo/crecimiento & desarrollo , Secuencia de Aminoácidos , Animales , Epigénesis Genética , Histonas/análisis , Histonas/química , Humanos , Masculino , Ratones , Especificidad de Órganos , Péptidos/análisis , Espermatogénesis , Testículo/metabolismo
16.
Cell Death Differ ; 24(6): 1029-1044, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28475176

RESUMEN

Sperm differentiation requires unique transcriptional regulation and chromatin remodeling after meiosis to ensure proper compaction and protection of the paternal genome. Abnormal sperm chromatin remodeling can induce sperm DNA damage, embryo lethality and male infertility, yet, little is known about the factors which regulate this process. Deficiency in Sly, a mouse Y chromosome-encoded gene expressed only in postmeiotic male germ cells, has been shown to result in the deregulation of hundreds of sex chromosome-encoded genes associated with multiple sperm differentiation defects and subsequent male infertility. The underlying mechanism remained, to date, unknown. Here, we show that SLY binds to the promoter of sex chromosome-encoded and autosomal genes highly expressed postmeiotically and involved in chromatin regulation. Specifically, we demonstrate that Sly knockdown directly induces the deregulation of sex chromosome-encoded H2A variants and of the H3K79 methyltransferase DOT1L. The modifications prompted by loss of Sly alter the postmeiotic chromatin structure and ultimately result in abnormal sperm chromatin remodeling with negative consequences on the sperm genome integrity. Altogether our results show that SLY is a regulator of sperm chromatin remodeling. Finally we identified that SMRT/N-CoR repressor complex is involved in gene regulation during sperm differentiation since members of this complex, in particular TBL1XR1, interact with SLY in postmeiotic male germ cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ensamble y Desensamble de Cromatina , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Represoras/metabolismo , Cromosomas Sexuales/metabolismo , Espermatozoides/metabolismo , Proteínas Adaptadoras del Transporte Vesicular , Animales , Cromosomas de los Mamíferos/genética , Cromosomas de los Mamíferos/metabolismo , ADN/metabolismo , Regulación de la Expresión Génica , Masculino , Ratones , Cromosomas Sexuales/genética , Espermatogénesis , Espermatozoides/fisiología
17.
FEBS J ; 281(6): 1571-84, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24456183

RESUMEN

In mammals, X- and Y-encoded genes are transcriptionally shut down during male meiosis, but expression of many of them is (re)activated in spermatids after meiosis. Post-meiotic XY gene expression is regulated by active epigenetic marks, which are de novo incorporated in the sex chromatin of spermatids, and by repressive epigenetic marks inherited during meiosis; alterations in this process lead to male infertility. In the mouse, post-meiotic XY gene expression is known to depend on genetic information carried by the male-specific region of the Y chromosome long arm (MSYq). The MSYq gene Sly has been shown to be a key regulator of post-meiotic sex chromosome gene expression and is necessary for the maintenance/recruitment of repressive epigenetic marks on the sex chromatin, but studies suggest that another MSYq gene may also be required. The best candidate to date is Ssty, an MSYq multi-copy gene of unknown function. Here, we show that SSTY proteins are specifically expressed in round and elongating spermatids, and co-localize with post-meiotic sex chromatin. Moreover, SSTY proteins interact with SLY protein and its X-linked homolog SLX/SLXL1, and may be required for localization of SLX/SLY proteins in the spermatid nucleus and sex chromatin. Our data suggest that SSTY is a second MSYq factor involved in the control of XY gene expression during sperm differentiation. As Slx/Slxl1 and Sly genes have been shown to be involved in the XY intra-genomic conflict, which affects the offspring sex ratio, Ssty may constitute another player in this conflict.


Asunto(s)
Proteínas/genética , Proteínas/metabolismo , Cromatina Sexual/genética , Cromatina Sexual/metabolismo , Secuencia de Aminoácidos , Animales , Núcleo Celular/metabolismo , Epigénesis Genética , Femenino , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Homología de Secuencia de Aminoácido , Espermátides/metabolismo , Espermatogénesis/genética , Cromosoma X/genética , Cromosoma Y/genética
18.
PLoS One ; 8(4): e62919, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23638166

RESUMEN

Drastic membrane reorganization occurs when mammalian sperm binds to and fuses with the oocyte membrane. Two oocyte protein families are essential for fertilization, tetraspanins and glycosylphosphatidylinositol-anchored proteins. The firsts are associated to tetraspanin-enriched microdomains and the seconds to lipid rafts. Here we report membrane raft involvement in mouse fertilization assessed by cholesterol modulation using methyl-ß-cyclodextrin. Cholesterol removal induced: (1) a decrease of the fertilization rate and index; and (2) a delay in the extrusion of the second polar body. Cholesterol repletion recovered the fertilization ability of cholesterol-depleted oocytes, indicating reversibility of these effects. In vivo time-lapse analyses using fluorescent cholesterol permitted to identify the time-point at which the probe is mainly located at the plasma membrane enabling the estimation of the extent of the cholesterol depletion. We confirmed that the mouse oocyte is rich in rafts according to the presence of the raft marker lipid, ganglioside GM1 on the membrane of living oocytes and we identified the coexistence of two types of microdomains, planar rafts and caveolae-like structures, by terms of two differential rafts markers, flotillin-2 and caveolin-1, respectively. Moreover, this is the first report that shows characteristic caveolae-like invaginations in the mouse oocyte identified by electron microscopy. Raft disruption by cholesterol depletion disturbed the subcellular localization of the signal molecule c-Src and the inhibition of Src kinase proteins prevented second polar body extrusion, consistent with a role of Src-related kinases in fertilization via signaling complexes. Our data highlight the functional importance of intact membrane rafts for mouse fertilization and its dependence on cholesterol.


Asunto(s)
Colesterol/deficiencia , Fertilización , Microdominios de Membrana/metabolismo , Oocitos/citología , Animales , Transporte Biológico/efectos de los fármacos , Compuestos de Boro/metabolismo , Caveolina 1/metabolismo , Supervivencia Celular/efectos de los fármacos , Femenino , Fertilización/efectos de los fármacos , Gangliósido G(M1)/metabolismo , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Masculino , Microdominios de Membrana/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Ovulación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Tetraspaninas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA