Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Mol Genet Genomics ; 297(4): 1151-1167, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35704117

RESUMEN

Supernumerary B chromosomes (Bs) are dispensable genetic elements widespread in eukaryotes and are poorly understood mainly in relation to mechanisms of maintenance and transmission. The cichlid Astatotilapia latifasciata can harbor Bs in a range of 0 (named B -) and 1-2 (named B +). The B in A. latifasciata is rich in several classes of repetitive DNA sequences, contains protein coding genes, and affects hosts in diverse ways, including sex-biased effects. To advance in the knowledge about the mechanisms of maintenance and transmission of B chromosomes in A. latifasciata, here, we studied the meiotic behavior in males and transmission rates of A. latifasciata B chromosome. We also analyzed structurally and functionally the predicted B chromosome copies of the cell cycle genes separin-like, tubb1-like and kif11-like. We identified in the meiotic structure relative to the B chromosome the presence of proteins associated with Synaptonemal Complex organization (SMC3, SYCP1 and SYCP3) and found that the B performs self-pairing. These data suggest that isochromosome formation was a step during B chromosome evolution and this element is in a stage of diversification of the two arms keeping the self-pairing behavior to protect the A chromosome complement of negative effects of recombination. Moreover, we observed no occurrence of B-drive and confirmed the presence of cell cycle genes copies in the B chromosome and their transcription in encephalon, muscle and gonads, which can indicates beneficial effects to hosts and contribute to B maintenance.


Asunto(s)
Cíclidos , Animales , Cromosomas/genética , Cíclidos/genética , Masculino , Meiosis/genética , Secuencias Repetitivas de Ácidos Nucleicos
2.
BMC Evol Biol ; 20(1): 55, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32408858

RESUMEN

BACKGROUND: Meiosis is essential for sexual reproduction and generates genetically diverse haploid gametes from a diploid germ cell. Reduction of ploidy depends on active chromosome movements during early meiotic prophase I. Chromosome movements require telomere attachment to the nuclear envelope. This attachment is mediated by telomere adaptor proteins. Telomere adaptor proteins have to date been identified in fission yeast and mice. In the mouse, they form a complex composed of the meiotic proteins TERB1, TERB2, and MAJIN. No sequence similarity was observed between these three mouse proteins and the adaptor proteins of fission yeast, raising the question of the evolutionary history and significance of this specific protein complex. RESULT: Here, we show the TERB1, TERB2, and MAJIN proteins are found throughout the Metazoa and even in early-branching non-bilateral phyla such as Cnidaria, Placozoa and Porifera. Metazoan TERB1, TERB2, and MAJIN showed comparable domain architecture across all clades. Furthermore, the protein domains involved in the formation of the complex as well as those involved for the interaction with the telomere shelterin protein and the LINC complexes revealed high sequence similarity. Finally, gene expression in the cnidarian Hydra vulgaris provided evidence that the TERB1-TERB2-MAJIN complex is selectively expressed in the germ line. CONCLUSION: Our results indicate that the TERB1-TERB2-MAJIN complex has an ancient origin in metazoans, suggesting conservation of meiotic functions.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Meiosis/genética , Proteínas de la Membrana/genética , Filogenia , Proteínas de Unión a Telómeros/genética , Telómero/genética , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/química , Proteínas de Ciclo Celular/química , Femenino , Regulación de la Expresión Génica , Células Germinativas/metabolismo , Gónadas/metabolismo , Masculino , Proteínas de la Membrana/química , Ratones , Dominios Proteicos , Proteínas de Unión a Telómeros/química , Proteínas de Unión a Telómeros/metabolismo
3.
Chromosoma ; 128(3): 443-451, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30793238

RESUMEN

The synaptonemal complex is an evolutionarily conserved, supramolecular structure that holds the homologous chromosomes together during the pachytene stage of the first meiotic prophase. Among vertebrates, synaptonemal complex dynamics has been analyzed in mouse spermatocytes following the assembly of its components from leptotene to pachytene stages. With few exceptions, a detailed study of the disassembly of SCs and the behavior of SC components at recombination sites at the onset of diplotene has not been accomplished. Here, we describe for the first time the progressive disassembly of the SC in chicken oocytes during the initial steps of desynapsis using immunolocalization of specific SC proteins and super-resolution microscopy. We found that transverse filament protein SYCP1 and central element component SYCE3 remain associated with the lateral elements at the beginning of chromosomal axis separation. As the separation between lateral elements widens, these proteins eventually disappear, without any evidence of subsequent association. Our observations support the idea that post-translational modifications of the central region components have a role at the initial phases of the SC disassembly. At the crossover sites, signaled by persistent MLH1 foci, the central region proteins are no longer detected when the SYCP3-positive lateral elements are widely separated. These findings are indicative that SC disassembly follows a general pattern along the desynaptic bivalents. The present work shows that the use of avian oocytes at prophase I provides a valuable model to explore the time course and chromosomal localization of SC proteins and its relationship with local changes along meiotic bivalents.


Asunto(s)
Pollos/genética , Microscopía Confocal , Oocitos/metabolismo , Complejo Sinaptonémico/metabolismo , Animales , Biomarcadores , Segregación Cromosómica , Femenino , Técnica del Anticuerpo Fluorescente , Sitios Genéticos , Meiosis
4.
Mol Hum Reprod ; 26(7): 485-497, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32402064

RESUMEN

More than 50% of cases of primary ovarian insufficiency (POI) and nonobstructive azoospermia in humans are classified as idiopathic infertility. Meiotic defects may relate to at least some of these cases. Mutations in genes coding for synaptonemal complex (SC) components have been identified in humans, and hypothesized to be causative for the observed infertile phenotype. Mutation SYCE1 c.721C>T (former c.613C>T)-a familial mutation reported in two sisters with primary amenorrhea-was the first such mutation found in an SC central element component-coding gene. Most fundamental mammalian oogenesis events occur during the embryonic phase, and eventual defects are identified many years later, thus leaving few possibilities to study the condition's etiology and pathogenesis. Aiming to validate an approach to circumvent this difficulty, we have used the CRISPR/Cas9 technology to generate a mouse model with an SYCE1 c.721C>T equivalent genome alteration. We hereby present the characterization of the homozygous mutant mice phenotype, compared to their wild type and heterozygous littermates. Our results strongly support a causative role of this mutation for the POI phenotype in human patients, and the mechanisms involved would relate to defects in homologous chromosome synapsis. No SYCE1 protein was detected in homozygous mutants and Syce1 transcript level was highly diminished, suggesting transcript degradation as the basis of the infertility mechanism. This is the first report on the generation of a humanized mouse model line for the study of an infertility-related human mutation in an SC component-coding gene, thus representing a proof of principle.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/genética , Mutación Puntual/genética , Insuficiencia Ovárica Primaria/genética , Animales , Emparejamiento Cromosómico/genética , Emparejamiento Cromosómico/fisiología , Proteínas de Unión al ADN/genética , Femenino , Citometría de Flujo , Predisposición Genética a la Enfermedad/genética , Homocigoto , Humanos , Inmunohistoquímica , Meiosis/genética , Meiosis/fisiología , Ratones , Mutación/genética
5.
RNA Biol ; 17(3): 350-365, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31869276

RESUMEN

The discovery of a large number of long noncoding RNAs (lncRNAs), and the finding that they may play key roles in different biological processes, have started to provide a new perspective in the understanding of gene regulation. It has been shown that the testes express the highest amount of lncRNAs among different vertebrate tissues. However, although some studies have addressed the characterization of lncRNAs along spermatogenesis, an exhaustive analysis of the differential expression of lncRNAs at its different stages is still lacking. Here, we present the results for lncRNA transcriptome profiling along mouse spermatogenesis, employing highly pure flow sorted spermatogenic stage-specific cell populations, strand-specific RNAseq, and a combination of up-to-date bioinformatic pipelines for analysis. We found that the vast majority of testicular lncRNA genes are expressed at post-meiotic stages (i.e. spermiogenesis), which are characterized by extensive post-transcriptional regulation. LncRNAs at different spermatogenic stages shared common traits in terms of transcript length, exon number, and biotypes. Most lncRNAs were lincRNAs, followed by a high representation of antisense (AS) lncRNAs. Co-expression analyses showed a high correlation along the different spermatogenic stage transitions between the expression patterns of AS lncRNAs and their overlapping protein-coding genes, raising possible clues about lncRNA-related regulatory mechanisms. Interestingly, we observed the co-localization of an AS lncRNA and its host sense mRNA in the chromatoid body, a round spermatids-specific organelle that has been proposed as a reservoir of RNA-related regulatory machinery. An additional, intriguing observation is the almost complete lack of detectable expression for Y-linked testicular lncRNAs, despite that a high number of lncRNA genes are annotated for this chromosome.


Asunto(s)
ARN Largo no Codificante/genética , Espermatogénesis/fisiología , Animales , Regulación de la Expresión Génica , Masculino , Ratones , ARN sin Sentido , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Espermátides/citología , Espermátides/fisiología , Espermatogénesis/genética , Testículo/citología , Testículo/fisiología
6.
Proc Natl Acad Sci U S A ; 112(7): 2029-33, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25646409

RESUMEN

Synaptonemal complexes (SCs) are meiosis-specific multiprotein complexes that are essential for synapsis, recombination, and segregation of homologous chromosomes, but the molecular organization of SCs remains unclear. We used immunofluorescence labeling in combination with super-resolution imaging and average position determination to investigate the molecular architecture of SCs. Combination of 2D super-resolution images recorded from different areas of the helical ladder-like structure allowed us to reconstruct the 3D molecular organization of the mammalian SC with isotropic resolution. The central element is composed of two parallel cables at a distance of ∼ 100 nm, which are oriented perpendicular to two parallel cables of the lateral element arranged at a distance of ∼ 220 nm. The two parallel cable elements form twisted helical structures that are connected by transversal filaments by their N and C termini. A single-cell preparation generates sufficient localizations to compile a 3D model of the SC with nanometer precision.


Asunto(s)
Complejo Sinaptonémico/ultraestructura , Animales , Masculino , Ratones
7.
Chromosoma ; 125(3): 355-60, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26968413

RESUMEN

The synaptonemal complex (SC), a key structure of meiosis that assembles during prophase I, has been initially described 60 years ago. Since then, the structure has been described in many sexually reproducing organisms. However, the SC protein components were characterized in only few model organisms. Surprisingly, they lacked an apparent evolutionary relationship despite the conserved structural organization of the SC. For better understanding of this obvious discrepancy, the evolutionary history of the SC and its individual components has been investigated in Metazoa in detail. The results are consistent with the notion of a single origin of the metazoan SC and provide evidence for a dynamic evolutionary history of the SC components. In this mini review, we recapitulate and discuss new insights into metazoan SC evolution.


Asunto(s)
Evolución Molecular , Complejo Sinaptonémico/genética , Complejo Sinaptonémico/metabolismo , Animales , Humanos
8.
J Cell Sci ; 128(1): 88-99, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25380821

RESUMEN

In most organisms, telomeres attach to the nuclear envelope at the onset of meiosis to promote the crucial processes of pairing, recombination and synapsis during prophase I. This attachment of meiotic telomeres is mediated by the specific distribution of several nuclear envelope components that interact with the attachment plates of the synaptonemal complex. We have determined by immunofluorescence and electron microscopy that the ablation of the kinase CDK2 alters the nuclear envelope in mouse spermatocytes, and that the proteins SUN1, KASH5 (also known as CCDC155) and lamin C2 show an abnormal cap-like distribution facing the centrosome. Strikingly, some telomeres are not attached to the nuclear envelope but remain at the nuclear interior where they are associated with SUN1 and with nuclear-envelope-detached vesicles. We also demonstrate that mouse testis CDK2 phosphorylates SUN1 in vitro. We propose that during mammalian prophase I the kinase CDK2 is a key factor governing the structure of the nuclear envelope and the telomere-led chromosome movements essential for homolog pairing.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/metabolismo , Profase Meiótica I/fisiología , Membrana Nuclear/metabolismo , Espermatocitos/metabolismo , Telómero/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Quinasa 2 Dependiente de la Ciclina/genética , Proteínas del Citoesqueleto , Laminina/genética , Laminina/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Membrana Nuclear/genética , Proteínas Nucleares/genética , Fosforilación/fisiología , Espermatocitos/citología , Telómero/genética
9.
PLoS Genet ; 10(2): e1004099, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24586178

RESUMEN

LINC complexes are evolutionarily conserved nuclear envelope bridges, composed of SUN (Sad-1/UNC-84) and KASH (Klarsicht/ANC-1/Syne/homology) domain proteins. They are crucial for nuclear positioning and nuclear shape determination, and also mediate nuclear envelope (NE) attachment of meiotic telomeres, essential for driving homolog synapsis and recombination. In mice, SUN1 and SUN2 are the only SUN domain proteins expressed during meiosis, sharing their localization with meiosis-specific KASH5. Recent studies have shown that loss of SUN1 severely interferes with meiotic processes. Absence of SUN1 provokes defective telomere attachment and causes infertility. Here, we report that meiotic telomere attachment is not entirely lost in mice deficient for SUN1, but numerous telomeres are still attached to the NE through SUN2/KASH5-LINC complexes. In Sun1(-/-) meiocytes attached telomeres retained the capacity to form bouquet-like clusters. Furthermore, we could detect significant numbers of late meiotic recombination events in Sun1(-/-) mice. Together, this indicates that even in the absence of SUN1 telomere attachment and their movement within the nuclear envelope per se can be functional.


Asunto(s)
Meiosis/genética , Proteínas de la Membrana/genética , Proteínas Asociadas a Microtúbulos/genética , ARN Largo no Codificante/genética , Proteínas de Unión a Telómeros/genética , Animales , Proteínas de Ciclo Celular/genética , Proteínas del Citoesqueleto , Ratones , Complejos Multiproteicos/genética , Membrana Nuclear/genética , Proteínas Nucleares/genética , Telómero/genética
10.
BMC Genomics ; 17: 294, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27094866

RESUMEN

BACKGROUND: Spermatogenesis is a complex differentiation process that involves the successive and simultaneous execution of three different gene expression programs: mitotic proliferation of spermatogonia, meiosis, and spermiogenesis. Testicular cell heterogeneity has hindered its molecular analyses. Moreover, the characterization of short, poorly represented cell stages such as initial meiotic prophase ones (leptotene and zygotene) has remained elusive, despite their crucial importance for understanding the fundamentals of meiosis. RESULTS: We have developed a flow cytometry-based approach for obtaining highly pure stage-specific spermatogenic cell populations, including early meiotic prophase. Here we combined this methodology with next generation sequencing, which enabled the analysis of meiotic and postmeiotic gene expression signatures in mouse with unprecedented reliability. Interestingly, we found that a considerable number of genes involved in early as well as late meiotic processes are already on at early meiotic prophase, with a high proportion of them being expressed only for the short time lapse of lepto-zygotene stages. Besides, we observed a massive change in gene expression patterns during medium meiotic prophase (pachytene) when mostly genes related to spermiogenesis and sperm function are already turned on. This indicates that the transcriptional switch from meiosis to post-meiosis takes place very early, during meiotic prophase, thus disclosing a higher incidence of post-transcriptional regulation in spermatogenesis than previously reported. Moreover, we found that a good proportion of the differential gene expression in spermiogenesis corresponds to up-regulation of genes whose expression starts earlier, at pachytene stage; this includes transition protein-and protamine-coding genes, which have long been claimed to switch on during spermiogenesis. In addition, our results afford new insights concerning X chromosome meiotic inactivation and reactivation. CONCLUSIONS: This work provides for the first time an overview of the time course for the massive onset and turning off of the meiotic and spermiogenic genetic programs. Importantly, our data represent a highly reliable information set about gene expression in pure testicular cell populations including early meiotic prophase, for further data mining towards the elucidation of the molecular bases of male reproduction in mammals.


Asunto(s)
Fase Paquiteno/genética , Espermatogénesis/genética , Transcriptoma , Animales , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Masculino , Profase Meiótica I/genética , Ratones , Reproducibilidad de los Resultados , Análisis de Secuencia de ARN , Espermatogonias/citología , Cromosoma X/genética
11.
Cytogenet Genome Res ; 150(2): 77-85, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27997882

RESUMEN

Human infertility is often classified as idiopathic in both males and females. Meiotic errors may account for at least part of these cases. As the synaptonemal complex (SC, a meiosis-specific protein scaffold) is essential for successful meiosis progression, in this paper, we analyzed the mutations in genes coding for SC components described in infertile patients to assess to what extent alterations in the SC can be related to human infertility. So far, mutations in SYCP3 and SYCE1 genes have been reported. While most SYCP3 mutations are heterozygous mutations with dominant-negative effect on the region encoding the C-terminal coiled coil of the protein, SYCE1 mutations are homozygous, which is consistent with a recessive inheritance. Similarities and differences between males and females as well as between mice and humans have been found and are discussed herein. The results suggest that a low percentage of human infertility cases may be explained by mutations in genes coding for SC components. The characterization of these mutations, together with available information from the study of knockout mice, will enable a deeper understanding of the underlying molecular bases for some of the cases of idiopathic infertility.


Asunto(s)
Fertilidad/genética , Mutación , Complejo Sinaptonémico/genética , Animales , Proteínas de Ciclo Celular , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Complejo Sinaptonémico/ultraestructura
12.
PLoS Genet ; 9(1): e1003261, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23382700

RESUMEN

The nuclear lamina is the structural scaffold of the nuclear envelope and is well known for its central role in nuclear organization and maintaining nuclear stability and shape. In the past, a number of severe human disorders have been identified to be associated with mutations in lamins. Extensive research on this topic has provided novel important clues about nuclear lamina function. These studies have contributed to the knowledge that the lamina constitutes a complex multifunctional platform combining both structural and regulatory functions. Here, we report that, in addition to the previously demonstrated significance for somatic cell differentiation and maintenance, the nuclear lamina is also an essential determinant for germ cell development. Both male and female mice lacking the short meiosis-specific A-type lamin C2 have a severely defective meiosis, which at least in the male results in infertility. Detailed analysis revealed that lamin C2 is required for telomere-driven dynamic repositioning of meiotic chromosomes. Loss of lamin C2 affects precise synapsis of the homologs and interferes with meiotic double-strand break repair. Taken together, our data explain how the nuclear lamina contributes to meiotic chromosome behaviour and accurate genome haploidization on a mechanistic level.


Asunto(s)
Células Germinativas , Recombinación Homóloga/genética , Laminina , Meiosis/genética , Lámina Nuclear , Animales , Diferenciación Celular , Cromosomas/genética , Femenino , Células Germinativas/crecimiento & desarrollo , Células Germinativas/metabolismo , Humanos , Infertilidad Masculina/etiología , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Laminina/genética , Laminina/metabolismo , Masculino , Ratones , Mutación , Lámina Nuclear/genética , Lámina Nuclear/metabolismo
13.
Proc Natl Acad Sci U S A ; 109(41): 16588-93, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-23012415

RESUMEN

The synaptonemal complex (SC) is a key structure of meiosis, mediating the stable pairing (synapsis) of homologous chromosomes during prophase I. Its remarkable tripartite structure is evolutionarily well conserved and can be found in almost all sexually reproducing organisms. However, comparison of the different SC protein components in the common meiosis model organisms Saccharomyces cerevisiae, Arabidopsis thaliana, Caenorhabditis elegans, Drosophila melanogaster, and Mus musculus revealed no sequence homology. This discrepancy challenged the hypothesis that the SC arose only once in evolution. To pursue this matter we focused on the evolution of SYCP1 and SYCP3, the two major structural SC proteins of mammals. Remarkably, our comparative bioinformatic and expression studies revealed that SYCP1 and SYCP3 are also components of the SC in the basal metazoan Hydra. In contrast to previous assumptions, we therefore conclude that SYCP1 and SYCP3 form monophyletic groups of orthologous proteins across metazoans.


Asunto(s)
Hydra/genética , Meiosis/genética , Proteínas Nucleares/genética , Complejo Sinaptonémico/genética , Secuencia de Aminoácidos , Animales , Western Blotting , Clonación Molecular , ADN Complementario/química , ADN Complementario/genética , Proteínas de Unión al ADN , Expresión Génica , Hydra/metabolismo , Inmunohistoquímica , Hibridación in Situ , Microscopía Electrónica , Datos de Secuencia Molecular , Proteínas Nucleares/clasificación , Proteínas Nucleares/metabolismo , Filogenia , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Complejo Sinaptonémico/metabolismo , Complejo Sinaptonémico/ultraestructura
14.
Cytogenet Genome Res ; 144(4): 299-305, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25831978

RESUMEN

During the first meiotic prophase, chromosome synapsis is mediated by the synaptonemal complex (SC), an evolutionarily conserved meiosis-specific structure. In mammals, 7 SC protein components have been identified so far. Despite some controversy in the past, we have shown that SC proteins are ancient in metazoans and very likely formed an ancestral SC structure in the ancestor of metazoans. Protein components SYCP1, SYCP3, SYCE2, and TEX12 were identified in basal-branching metazoans, while other components (SYCE1 and SYCE3) are more recent elements. However, the evolutionary history of mammalian SYCP2 is not known. Here, we investigated this aspect with the aid of bioinformatic tools as well as with RNA and protein expression analysis. We conclude that SYCP2 belongs to the group of ancient SC proteins that was already present in the common ancestor of metazoans more than 500 million years ago.


Asunto(s)
Hydra/metabolismo , Mamíferos/metabolismo , Complejo Sinaptonémico/genética , Complejo Sinaptonémico/metabolismo , Testículo/metabolismo , Animales , Biología Computacional/métodos , Evolución Molecular , Hydra/anatomía & histología , Hydra/clasificación , Hydra/genética , Masculino , Mamíferos/anatomía & histología , Mamíferos/genética , Especificidad de Órganos , Filogenia , Homología de Secuencia de Aminoácido
15.
PLoS Genet ; 7(5): e1002088, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21637789

RESUMEN

The synaptonemal complex (SC) is a proteinaceous, meiosis-specific structure that is highly conserved in evolution. During meiosis, the SC mediates synapsis of homologous chromosomes. It is essential for proper recombination and segregation of homologous chromosomes, and therefore for genome haploidization. Mutations in human SC genes can cause infertility. In order to gain a better understanding of the process of SC assembly in a model system that would be relevant for humans, we are investigating meiosis in mice. Here, we report on a newly identified component of the murine SC, which we named SYCE3. SYCE3 is strongly conserved among mammals and localizes to the central element (CE) of the SC. By generating a Syce3 knockout mouse, we found that SYCE3 is required for fertility in both sexes. Loss of SYCE3 blocks synapsis initiation and results in meiotic arrest. In the absence of SYCE3, initiation of meiotic recombination appears to be normal, but its progression is severely impaired resulting in complete absence of MLH1 foci, which are presumed markers of crossovers in wild-type meiocytes. In the process of SC assembly, SYCE3 is required downstream of transverse filament protein SYCP1, but upstream of the other previously described CE-specific proteins. We conclude that SYCE3 enables chromosome loading of the other CE-specific proteins, which in turn would promote synapsis between homologous chromosomes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Fertilidad , Meiosis , Proteínas Nucleares/metabolismo , Complejo Sinaptonémico/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Animales , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Clonación Molecular , Intercambio Genético , Proteínas de Unión al ADN , Femenino , Genotipo , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Homólogo 1 de la Proteína MutL , Proteínas Nucleares/genética , Ovario/metabolismo , Ovario/ultraestructura , Unión Proteica , Alineación de Secuencia , Espermatocitos/metabolismo , Testículo/metabolismo , Testículo/ultraestructura , Transfección
16.
Exp Cell Res ; 318(12): 1340-6, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22394509

RESUMEN

The synaptonemal complex (SC) is a proteinaceous structure of chromosome bivalents whose assembly is indispensable for the successful progression of the first meiotic division of sexually reproducing organisms. In this mini-review we will focus on recent progress dealing with the composition and assembly of the mammalian SC. These advances mainly resulted from the systematic use of knockout mice for all known mammalian SC proteins as well as from protein polymerization studies performed in heterologous systems.


Asunto(s)
Mamíferos/fisiología , Meiosis/fisiología , Multimerización de Proteína/fisiología , Recombinación Genética/fisiología , Complejo Sinaptonémico/fisiología , Animales , Humanos , Mamíferos/genética , Mamíferos/metabolismo , Meiosis/genética , Ratones , Ratones Noqueados , Modelos Biológicos , Multimerización de Proteína/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Subunidades de Proteína/fisiología , Recombinación Genética/genética , Complejo Sinaptonémico/genética , Complejo Sinaptonémico/metabolismo
17.
J Cell Biol ; 176(6): 741-7, 2007 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-17339376

RESUMEN

Synapsis is the process by which paired chromosome homologues closely associate in meiosis before crossover. In the synaptonemal complex (SC), axial elements of each homologue connect through molecules of SYCP1 to the central element, which contains the proteins SYCE1 and -2. We have derived mice lacking SYCE2 protein, producing males and females in which meiotic chromosomes align and axes form but do not synapse. Sex chromosomes are unaligned, not forming a sex body. Additionally, markers of DNA breakage and repair are retained on the axes, and crossover is impaired, culminating in both males and females failing to produce gametes. We show that SC formation can initiate at sites of SYCE1/SYCP1 localization but that these points of initiation cannot be extended in the absence of SYCE2. SC assembly is thus dependent on SYCP1, SYCE1, and SYCE2. We provide a model to explain this based on protein-protein interactions.


Asunto(s)
Reparación del ADN , Proteínas Nucleares/fisiología , Recombinación Genética , Complejo Sinaptonémico/metabolismo , Animales , Roturas del ADN de Doble Cadena , Femenino , Masculino , Ratones , Ratones Noqueados , Modelos Genéticos , Mutación , Proteínas Nucleares/genética , Oocitos/citología , Cromosomas Sexuales/fisiología , Espermatocitos/citología
18.
PLoS Genet ; 5(2): e1000393, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19247432

RESUMEN

In mammals, the synaptonemal complex is a structure required to complete crossover recombination. Although suggested by cytological work, in vivo links between the structural proteins of the synaptonemal complex and the proteins of the recombination process have not previously been made. The central element of the synaptonemal complex is traversed by DNA at sites of recombination and presents a logical place to look for interactions between these components. There are four known central element proteins, three of which have previously been mutated. Here, we complete the set by creating a null mutation in the Syce1 gene in mouse. The resulting disruption of synapsis in these animals has allowed us to demonstrate a biochemical interaction between the structural protein SYCE2 and the repair protein RAD51. In normal meiosis, this interaction may be responsible for promoting homologous synapsis from sites of recombination.


Asunto(s)
Emparejamiento Cromosómico , Reparación del ADN , Proteínas Nucleares/genética , Complejo Sinaptonémico/metabolismo , Animales , Femenino , Gametogénesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/metabolismo , Unión Proteica , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Recombinación Genética , Espermatocitos/citología , Espermatocitos/metabolismo , Complejo Sinaptonémico/genética
19.
Front Cell Dev Biol ; 9: 626020, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33748111

RESUMEN

Molecular studies of meiosis in mammals have been long relegated due to some intrinsic obstacles, namely the impossibility to reproduce the process in vitro, and the difficulty to obtain highly pure isolated cells of the different meiotic stages. In the recent years, some technical advances, from the improvement of flow cytometry sorting protocols to single-cell RNAseq, are enabling to profile the transcriptome and its fluctuations along the meiotic process. In this mini-review we will outline the diverse methodological approaches that have been employed, and some of the main findings that have started to arise from these studies. As for practical reasons most studies have been carried out in males, and mostly using mouse as a model, our focus will be on murine male meiosis, although also including specific comments about humans. Particularly, we will center on the controversy about gene expression during early meiotic prophase; the widespread existing gap between transcription and translation in meiotic cells; the expression patterns and potential roles of meiotic long non-coding RNAs; and the visualization of meiotic sex chromosome inactivation from the RNAseq perspective.

20.
PLoS One ; 16(5): e0251028, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33945571

RESUMEN

SPATS1 (spermatogenesis-associated, serine-rich 1) is an evolutionarily conserved, testis-specific protein that is differentially expressed during rat male meiotic prophase. Some reports have suggested a link between SPATS1 underexpression/mutation and human pathologies such as male infertility and testicular cancer. Given the absence of functional studies, we generated a Spats1 loss-of-function mouse model using CRISPR/Cas9 technology. The phenotypic analysis showed no overt phenotype in Spats1-/- mice, with both males and females being fertile. Flow cytometry and histological analyses did not show differences in the testicular content and histology between WT and knockout mice. Moreover, no significant differences in sperm concentration, motility, and morphology, were observed between WT and KO mice. These results were obtained both for young adults and for aged animals. Besides, although an involvement of SPATS1 in the Wnt signaling pathway has been suggested, we did not detect changes in the expression levels of typical Wnt pathway-target genes in mutant individuals. Thus, albeit Spats1 alteration might be a risk factor for male testicular health, we hereby show that this gene is not individually essential for male fertility and spermatogenesis in mouse.


Asunto(s)
Fertilidad/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Espermatogénesis/fisiología , Secuencia de Aminoácidos , Animales , Femenino , Infertilidad Masculina/metabolismo , Masculino , Meiosis/fisiología , Ratones , Ratones Noqueados , Neoplasias de Células Germinales y Embrionarias/metabolismo , Serina/metabolismo , Recuento de Espermatozoides/métodos , Motilidad Espermática/fisiología , Espermatozoides/metabolismo , Neoplasias Testiculares/metabolismo , Testículo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA