Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
2.
Mol Genet Genomics ; 297(4): 1151-1167, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35704117

ABSTRACT

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.


Subject(s)
Cichlids , Animals , Chromosomes/genetics , Cichlids/genetics , Male , Meiosis/genetics , Repetitive Sequences, Nucleic Acid
3.
PLoS One ; 16(5): e0251028, 2021.
Article in English | MEDLINE | ID: mdl-33945571

ABSTRACT

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.


Subject(s)
Fertility/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Spermatogenesis/physiology , Amino Acid Sequence , Animals , Female , Infertility, Male/metabolism , Male , Meiosis/physiology , Mice , Mice, Knockout , Neoplasms, Germ Cell and Embryonal/metabolism , Serine/metabolism , Sperm Count/methods , Sperm Motility/physiology , Spermatozoa/metabolism , Testicular Neoplasms/metabolism , Testis/metabolism
4.
Front Cell Dev Biol ; 9: 626020, 2021.
Article in English | MEDLINE | ID: mdl-33748111

ABSTRACT

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.

5.
Nat Commun ; 11(1): 3222, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32591508

ABSTRACT

The synaptonemal complex (SC) is a meiosis-specific nuclear multiprotein complex that is essential for proper synapsis, recombination and segregation of homologous chromosomes. We combined structured illumination microscopy (SIM) with different expansion microscopy (ExM) protocols including U-ExM, proExM, and magnified analysis of the proteome (MAP) to investigate the molecular organization of the SC. Comparison with structural data obtained by single-molecule localization microscopy of unexpanded SCs allowed us to investigate ultrastructure preservation of expanded SCs. For image analysis, we developed an automatic image processing software that enabled unbiased comparison of structural properties pre- and post-expansion. Here, MAP-SIM provided the best results and enabled reliable three-color super-resolution microscopy of the SCs of a whole set of chromosomes in a spermatocyte with 20-30 nm spatial resolution. Our data demonstrate that post-expansion labeling by MAP-SIM improves immunolabeling efficiency and allowed us thus to unravel previously hidden details of the molecular organization of SCs.


Subject(s)
Microscopy/methods , Synaptonemal Complex/metabolism , Animals , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Imaging, Three-Dimensional , Mice, Inbred C57BL , Proteome/metabolism , Signal Processing, Computer-Assisted
6.
Mol Hum Reprod ; 26(7): 485-497, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32402064

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/genetics , Point Mutation/genetics , Primary Ovarian Insufficiency/genetics , Animals , Chromosome Pairing/genetics , Chromosome Pairing/physiology , DNA-Binding Proteins/genetics , Female , Flow Cytometry , Genetic Predisposition to Disease/genetics , Homozygote , Humans , Immunohistochemistry , Meiosis/genetics , Meiosis/physiology , Mice , Mutation/genetics
7.
BMC Evol Biol ; 20(1): 55, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32408858

ABSTRACT

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.


Subject(s)
Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Meiosis/genetics , Membrane Proteins/genetics , Phylogeny , Telomere-Binding Proteins/genetics , Telomere/genetics , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Cell Cycle Proteins/chemistry , Female , Gene Expression Regulation , Germ Cells/metabolism , Gonads/metabolism , Male , Membrane Proteins/chemistry , Mice , Protein Domains , Telomere-Binding Proteins/chemistry , Telomere-Binding Proteins/metabolism
8.
RNA Biol ; 17(3): 350-365, 2020 03.
Article in English | MEDLINE | ID: mdl-31869276

ABSTRACT

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.


Subject(s)
RNA, Long Noncoding/genetics , Spermatogenesis/physiology , Animals , Gene Expression Regulation , Male , Mice , RNA, Antisense , RNA, Messenger/metabolism , Reproducibility of Results , Spermatids/cytology , Spermatids/physiology , Spermatogenesis/genetics , Testis/cytology , Testis/physiology
9.
Sci Rep ; 9(1): 16102, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31695079

ABSTRACT

The synaptonemal complex is a multiprotein complex, which mediates the synapsis and recombination between homologous chromosomes during meiosis. The complex is comprised of two lateral elements and a central element connected by perpendicular transverse filaments (TFs). A 3D model based on actual morphological data of the SC is missing. Here, we applied electron tomography (ET) and manual feature extraction to generate a quantitative 3D model of the murine SC. We quantified the length (90 nm) and width (2 nm) of the TFs. Interestingly, the 80 TFs/µm are distributed asymmetrically in the central region of the SC challenging available models of SC organization. Furthermore, our detailed 3D topological analysis does not support a bilayered organization of the central region as proposed earlier. Overall, our quantitative analysis is relevant to understand the functions and dynamics of the SC and provides the basis for analyzing multiprotein complexes in their morphological context using ET.


Subject(s)
Chromosome Pairing , Chromosomes/genetics , Meiosis , Animals , Chromosomes/ultrastructure , Electron Microscope Tomography , Male , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Synaptonemal Complex/genetics , Synaptonemal Complex/ultrastructure , Testis/cytology
10.
Commun Biol ; 2: 376, 2019.
Article in English | MEDLINE | ID: mdl-31633067

ABSTRACT

Telomere movements during meiotic prophase I facilitate synapsis and recombination of homologous chromosomes. Hereby, chromosome movements depend on the dynamic attachment of meiotic telomeres to the nuclear envelope and generation of forces that actively move the telomeres. In most eukaryotes, forces that move telomeres are generated in the cytoplasm by microtubule-associated motor proteins and transduced into the nucleus through the LINC complexes of the nuclear envelope. Meiotic LINC complexes, in mouse comprised of SUN1/2 and KASH5, selectively localize to the attachment sites of meiotic telomeres. For a better understanding of meiotic telomere dynamics, here we provide quantitative information of telomere attachment sites that we have generated with the aid of electron microscope tomography (EM tomography). Our data on the number, length, width, distribution and relation with microtubules of the reconstructed structures indicate that an average number of 76 LINC complexes would be required to move a telomere attachment site.


Subject(s)
Microtubules/ultrastructure , Telomere/ultrastructure , Animals , Binding Sites , Cell Cycle Proteins/metabolism , Chromosome Pairing , Cytoskeletal Proteins/metabolism , Electron Microscope Tomography , Male , Meiosis , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Nuclear Proteins/metabolism , Telomere/metabolism , Telomere-Binding Proteins/metabolism , Testis/metabolism , Testis/ultrastructure
11.
Chromosoma ; 128(3): 443-451, 2019 09.
Article in English | MEDLINE | ID: mdl-30793238

ABSTRACT

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.


Subject(s)
Chickens/genetics , Microscopy, Confocal , Oocytes/metabolism , Synaptonemal Complex/metabolism , Animals , Biomarkers , Chromosome Segregation , Female , Fluorescent Antibody Technique , Genetic Loci , Meiosis
12.
Nat Commun ; 9(1): 5355, 2018 12 17.
Article in English | MEDLINE | ID: mdl-30559341

ABSTRACT

Meiotic chromosomes undergo rapid prophase movements, which are thought to facilitate the formation of inter-homologue recombination intermediates that underlie synapsis, crossing over and segregation. The meiotic telomere complex (MAJIN, TERB1, TERB2) tethers telomere ends to the nuclear envelope and transmits cytoskeletal forces via the LINC complex to drive these rapid movements. Here, we report the molecular architecture of the meiotic telomere complex through the crystal structure of MAJIN-TERB2, together with light and X-ray scattering studies of wider complexes. The MAJIN-TERB2 2:2 hetero-tetramer binds strongly to DNA and is tethered through long flexible linkers to the inner nuclear membrane and two TRF1-binding 1:1 TERB2-TERB1 complexes. Our complementary structured illumination microscopy studies and biochemical findings reveal a telomere attachment mechanism in which MAJIN-TERB2-TERB1 recruits telomere-bound TRF1, which is then displaced during pachytene, allowing MAJIN-TERB2-TERB1 to bind telomeric DNA and form a mature attachment plate.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/genetics , Telomere-Binding Proteins/genetics , Telomere/genetics , Telomeric Repeat Binding Protein 1/genetics , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Line , Crystallography, X-Ray , DNA-Binding Proteins , Humans , Meiosis/genetics , Multiprotein Complexes/metabolism , Protein Folding , Telomere/metabolism
13.
Methods Mol Biol ; 1840: 3-15, 2018.
Article in English | MEDLINE | ID: mdl-30141033

ABSTRACT

Electron microscope (EM) tomography is a powerful technique that enables the three-dimensional analysis of subcellular structures at high resolution. We have applied this method to the quantitative analysis of LINC complex distribution and interaction with the cytoskeleton in meiotic cells from male mice. In this chapter, we describe methods to generate and analyze the tomograms.


Subject(s)
Cytoskeleton/ultrastructure , Electron Microscope Tomography , Meiosis , Nuclear Envelope/ultrastructure , Telomere/ultrastructure , Animals , Electron Microscope Tomography/methods , Male , Mice
14.
Methods Cell Biol ; 145: 335-346, 2018.
Article in English | MEDLINE | ID: mdl-29957213

ABSTRACT

This chapter describes how two different superresolution microscopy techniques, namely, structured illumination microscopy and direct stochastic optical reconstruction microscopy, can be used to analyze the molecular architecture of the synaptonemal complex. The experimental protocols provided allow the construction of precise localization maps for different synaptonemal complex proteins.


Subject(s)
Microscopy, Fluorescence/methods , Synaptonemal Complex/physiology , Animals , Mice
15.
Aging (Albany NY) ; 10(5): 951-972, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29754147

ABSTRACT

The cnidarian Hydra is known for its unlimited lifespan and non-senescence, due to the indefinite self-renewal capacity of its stem cells. While proteins of the Lamin family are recognized as critical factors affecting senescence and longevity in human and mice, their putative role in the extreme longevity and non-senescence in long-living animals remains unknown. Here we analyze the role of a single lamin protein in non-senescence of Hydra. We demonstrate that proliferation of stem cells in Hydra is robust against the disturbance of Lamin expression and localization. While Lamin is indispensable for Hydra, the stem cells tolerate overexpression, downregulation and mislocalization of Lamin, and disturbances in the nuclear envelope structure. This extraordinary robustness may underlie the indefinite self-renewal capacity of stem cells and the non-senescence of Hydra. A relatively low complexity of the nuclear envelope architecture in basal Metazoa might allow for their extreme lifespans, while an increasing complexity of the nuclear architecture in bilaterians resulted in restricted lifespans.


Subject(s)
Cellular Senescence/physiology , Hydra/physiology , Lamins/metabolism , Nuclear Lamina/metabolism , Stem Cells/metabolism , Aging/metabolism , Animals , Longevity/physiology
16.
Cytogenet Genome Res ; 150(2): 77-85, 2016.
Article in English | MEDLINE | ID: mdl-27997882

ABSTRACT

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.


Subject(s)
Fertility/genetics , Mutation , Synaptonemal Complex/genetics , Animals , Cell Cycle Proteins , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins , Female , Humans , Male , Mice , Mice, Knockout , Nuclear Proteins/genetics , Synaptonemal Complex/ultrastructure
17.
Nat Commun ; 7: 13298, 2016 10 31.
Article in English | MEDLINE | ID: mdl-27796301

ABSTRACT

Meiotic recombination generates crossovers between homologous chromosomes that are essential for genome haploidization. The synaptonemal complex is a 'zipper'-like protein assembly that synapses homologue pairs together and provides the structural framework for processing recombination sites into crossovers. Humans show individual differences in the number of crossovers generated across the genome. Recently, an anonymous gene variant in C14ORF39/SIX6OS1 was identified that influences the recombination rate in humans. Here we show that C14ORF39/SIX6OS1 encodes a component of the central element of the synaptonemal complex. Yeast two-hybrid analysis reveals that SIX6OS1 interacts with the well-established protein synaptonemal complex central element 1 (SYCE1). Mice lacking SIX6OS1 are defective in chromosome synapsis at meiotic prophase I, which provokes an arrest at the pachytene-like stage and results in infertility. In accordance with its role as a modifier of the human recombination rate, SIX6OS1 is essential for the appropriate processing of intermediate recombination nodules before crossover formation.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Fertility , Synaptonemal Complex/metabolism , Animals , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosome Pairing , Crossing Over, Genetic , DNA-Binding Proteins , Electroporation , Female , Genetic Variation , Genome , HEK293 Cells , Haploidy , Humans , Male , Meiosis , Mice , Nuclear Proteins/metabolism , Recombination, Genetic , Testis/pathology , Transcription, Genetic , Two-Hybrid System Techniques
18.
Methods Mol Biol ; 1411: 195-208, 2016.
Article in English | MEDLINE | ID: mdl-27147043

ABSTRACT

Linear eukaryotic chromosomes are capped by the telomeres, which consist of highly repetitive nucleotide sequences bound by several telomere-specific proteins. While the general role of telomeres is to protect chromosomes from degradation and end-to-end fusion, during meiosis they are assigned with a distinct and without doubt highly fascinating function. During meiosis, telomeres attach to the nuclear envelope and mediate characteristic chromosome movements, essential for correct haploidization of the genome. Here, we provide elaborate tools to study telomeres in mammalian meiotic germ cells, which include (co-)immunofluorescence staining procedures on cell spreads and paraffin-embedded tissues. We provide detailed procedures for fluorescence labeling of telomeric DNA (Telo-FISH) to visualize telomeres at the light microscopic level, which we often use in combination with immunofluorescence staining of meiotic proteins. We also present a protocol for detection of telomeric DNA at the electron microscopic level (EM-ISH). We finally describe how meiotic telomeres can be visualized by common electron microscopic methods and how they can be analyzed at the ultrastructural level by immunogold labeling of telomere components or associated structures.


Subject(s)
Meiosis/genetics , Telomere Homeostasis , Telomere/genetics , Animals , Chromosomes, Mammalian , Female , In Situ Hybridization, Fluorescence , Male , Mice , Microscopy, Electron , Nuclear Envelope/metabolism , Oocytes/metabolism , Ovary/metabolism , Spermatocytes/metabolism , Testis/metabolism
19.
BMC Genomics ; 17: 294, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27094866

ABSTRACT

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.


Subject(s)
Pachytene Stage/genetics , Spermatogenesis/genetics , Transcriptome , Animals , Gene Expression Profiling , Gene Expression Regulation, Developmental , High-Throughput Nucleotide Sequencing , Male , Meiotic Prophase I/genetics , Mice , Reproducibility of Results , Sequence Analysis, RNA , Spermatogonia/cytology , X Chromosome/genetics
20.
Chromosoma ; 125(3): 355-60, 2016 06.
Article in English | MEDLINE | ID: mdl-26968413

ABSTRACT

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.


Subject(s)
Evolution, Molecular , Synaptonemal Complex/genetics , Synaptonemal Complex/metabolism , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...