Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.184
1.
Nat Commun ; 15(1): 3330, 2024 Apr 29.
Article En | MEDLINE | ID: mdl-38684656

Meiotic prophase progression is differently regulated in males and females. In males, pachytene transition during meiotic prophase is accompanied by robust alteration in gene expression. However, how gene expression is regulated differently to ensure meiotic prophase completion in males remains elusive. Herein, we identify HSF5 as a male germ cell-specific heat shock transcription factor (HSF) for meiotic prophase progression. Genetic analyzes and single-cell RNA-sequencing demonstrate that HSF5 is essential for progression beyond the pachytene stage under non-stress conditions rather than heat stress. Chromatin binding analysis in vivo and DNA-binding assays in vitro suggest that HSF5 binds to promoters in a subset of genes associated with chromatin organization. HSF5 recognizes a DNA motif different from typical heat shock elements recognized by other canonical HSFs. This study suggests that HSF5 is an atypical HSF that is required for the gene expression program for pachytene transition during meiotic prophase in males.


Heat Shock Transcription Factors , Male , Animals , Heat Shock Transcription Factors/metabolism , Heat Shock Transcription Factors/genetics , Mice , Pachytene Stage/genetics , Chromatin/metabolism , Chromatin/genetics , Spermatocytes/metabolism , Spermatocytes/cytology , Promoter Regions, Genetic/genetics , Heat-Shock Response/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Spermatogenesis/genetics , Meiotic Prophase I , Mice, Knockout
2.
J Cell Physiol ; 239(4): e31201, 2024 Apr.
Article En | MEDLINE | ID: mdl-38284481

Dynamic nuclear architecture and chromatin organizations are the key features of the mid-prophase I in mammalian meiosis. The chromatin undergoes major changes, including meiosis-specific spatiotemporal arrangements and remodeling, the establishment of chromatin loop-axis structure, pairing, and crossing over between homologous chromosomes, any deficiencies in these events may induce genome instability, subsequently leading to failure to produce gametes and infertility. Despite the significance of chromatin structure, little is known about the location of chromatin marks and the necessity of their balance during meiosis prophase I. Here, we show a thorough cytological study of the surface-spread meiotic chromosomes of mouse spermatocytes for H3K9,14,18,23,27,36, H4K12,16 acetylation, and H3K4,9,27,36 methylation. Active acetylation and methylation marks on H3 and H4, such as H3K9ac, H3K14ac, H3K18ac, H3K36ac, H3K56ac, H4K12ac, H4K16ac, and H3K36me3 exhibited pan-nuclear localization away from heterochromatin. In comparison, repressive marks like H3K9me3 and H3K27me3 are localized to heterochromatin. Further, taking advantage of the delivery of small-molecule chemical inhibitors methotrexate (heterochromatin enhancer), heterochromatin inhibitor, anacardic acid (histone acetyltransferase inhibitor), trichostatin A (histone deacetylase inhibitor), IOX1 (JmjC demethylases inhibitor), and AZ505 (methyltransferase inhibitor) in seminiferous tubules through the rete testis route, revealed that alteration in histone modifications enhanced the centromere mislocalization, chromosome breakage, altered meiotic recombination and reduced sperm count. Specifically, IOX1 and AZ505 treatment shows severe meiotic phenotypes, including altering chromosome axis length and chromatin loop size via transcriptional regulation of meiosis-specific genes. Our findings highlight the importance of balanced chromatin modifications in meiotic prophase I chromosome organization and instability.


Histones , Meiotic Prophase I , Protein Processing, Post-Translational , Spermatocytes , Animals , Male , Mice , Chromatin/genetics , Heterochromatin , Histones/metabolism , Meiosis , Spermatocytes/cytology , Spermatocytes/metabolism
3.
Cell Rep ; 37(11): 110110, 2021 12 14.
Article En | MEDLINE | ID: mdl-34910909

Mechanisms driving the prolonged meiotic prophase I in mammals are poorly understood. RNA helicase YTHDC2 is critical for mitosis to meiosis transition. However, YTHDC2 is highly expressed in pachytene cells. Here we identify an essential role for YTHDC2 in meiotic progression. Specifically, YTHDC2 deficiency causes microtubule-dependent telomere clustering and apoptosis at the pachytene stage of prophase I. Depletion of YTHDC2 results in a massively dysregulated transcriptome in pachytene cells, with a tendency toward upregulation of genes normally expressed in mitotic germ cells and downregulation of meiotic transcripts. Dysregulation does not correlate with m6A status, and YTHDC2-bound mRNAs are enriched in genes upregulated in mutant germ cells, revealing that YTHDC2 primarily targets mRNAs for degradation. Furthermore, altered transcripts in mutant pachytene cells encode microtubule network proteins. Our results demonstrate that YTHDC2 regulates the pachytene stage by perpetuating a meiotic transcriptome and preventing microtubule network changes that could lead to telomere clustering.


Meiosis , Microtubules/physiology , Pachytene Stage , RNA Helicases/physiology , Spermatocytes/cytology , Telomere , Transcriptome , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spermatocytes/metabolism
4.
Int J Mol Sci ; 22(24)2021 Dec 18.
Article En | MEDLINE | ID: mdl-34948376

Long noncoding RNAs (lncRNAs) are composed of nucleotides located in the nucleus and cytoplasm; these are transcribed by RNA polymerase II and are greater than 200 nt in length. LncRNAs fulfill important functions in a variety of biological processes, including genome imprinting, cell differentiation, apoptosis, stem cell pluripotency, X chromosome inactivation and nuclear transport. As high throughput sequencing technology develops, a substantial number of lncRNAs have been found to be related to a variety of biological processes, such as development of the testes, maintaining the self-renewal and differentiation of spermatogonial stem cells, and regulating spermatocyte meiosis. These indicate that lncRNAs can be used as biomarkers and potential therapeutic targets for male infertility. However, only a few comprehensive reviews have described the role of lncRNAs in male reproduction. In this paper, we summarize recent findings relating to the role of lncRNAs in spermatogenesis, their potential as biomarkers for male infertility and the relationship between reproductive arrest and transgenerational effects. Finally, we suggest specific targets for the treatment of male infertility from the perspective of lncRNAs.


Infertility, Male/genetics , RNA, Long Noncoding/genetics , Spermatogenesis , Animals , Cell Proliferation , Humans , Infertility, Male/pathology , Infertility, Male/therapy , Male , Meiosis , RNA, Long Noncoding/analysis , Spermatocytes/cytology , Spermatocytes/metabolism , Spermatocytes/pathology
5.
Int J Mol Sci ; 22(16)2021 Aug 18.
Article En | MEDLINE | ID: mdl-34445597

Spermatogenesis is a complicated process involving mitotically proliferating spermatogonial cells, meiotically dividing spermatocytes, and spermatid going through maturation into spermatozoa. The post-translational modifications of proteins play important roles in this biological process. S-palmitoylation is one type of protein modifications catalyzed by zinc finger Asp-His-His-Cys (ZDHHC)-family palmitoyl S-acyltransferases. There are 23 mammalian ZDHHCs that have been identified in mouse. Among them, Zdhhc19 is highly expressed in adult testis. However, the in vivo function of Zdhhc19 in mouse spermatogenesis and fertility remains unknown. In this study, we knocked out the Zdhhc19 gene by generating a 2609 bp deletion from exon 3 to exon 6 in mice. No differences were found in testis morphology and testis/body weight ratios upon Zdhhc19 deletion. Spermatogenesis was not disrupted in Zdhhc19 knockout mice, in which properly developed TRA98+ germ cells, SYCP3+ spermatocytes, and TNP1+ spermatids/spermatozoa were detected in seminiferous tubules. Nevertheless, Zdhhc19 knockout mice were male infertile. Zdhhc19 deficient spermatozoa exhibited multiple defects including abnormal morphology of sperm tails and heads, decreased motility, and disturbed acrosome reaction. All of these led to the inability of Zdhhc19 mutant sperm to fertilize oocytes in IVF assays. Taken together, our results support the fact that Zdhhc19 is a testis enriched gene dispensable for spermatogenesis, but is essential for sperm functions in mice.


Acyltransferases/physiology , Fertilization , Sperm Motility , Spermatocytes/cytology , Spermatogenesis , Spermatozoa/physiology , Acrosome Reaction , Animals , Female , Male , Mice , Mice, Knockout , Spermatocytes/physiology
6.
PLoS Genet ; 17(6): e1009655, 2021 06.
Article En | MEDLINE | ID: mdl-34181646

During spermatogenesis, the process in which sperm for fertilization are produced from germline cells, gene expression is spatiotemporally highly regulated. In Drosophila, successful expression of extremely large male fertility factor genes on Y-chromosome spanning some megabases due to their gigantic intron sizes is crucial for spermatogenesis. Expression of such extremely large genes must be challenging, but the molecular mechanism that allows it remains unknown. Here we report that a novel RNA-binding protein Maca, which contains two RNA-recognition motifs, is crucial for this process. maca null mutant male flies exhibited a failure in the spermatid individualization process during spermatogenesis, lacked mature sperm, and were completely sterile, while maca mutant female flies were fully fertile. Proteomics and transcriptome analyses revealed that both protein and mRNA abundance of the gigantic male fertility factor genes kl-2, kl-3, and kl-5 (kl genes) are significantly decreased, where the decreases of kl-2 are particularly dramatic, in maca mutant testes. Splicing of the kl-3 transcripts was also dysregulated in maca mutant testes. All these physiological and molecular phenotypes were rescued by a maca transgene in the maca mutant background. Furthermore, we found that in the control genetic background, Maca is exclusively expressed in spermatocytes in testes and enriched at Y-loop A/C in the nucleus, where the kl-5 primary transcripts are localized. Our data suggest that Maca increases transcription processivity, promotes successful splicing of gigantic introns, and/or protects transcripts from premature degradation, of the kl genes. Our study identified a novel RNA-binding protein Maca that is crucial for successful expression of the gigantic male fertility factor genes, spermatogenesis, and male fertility.


Drosophila melanogaster/genetics , RNA-Binding Proteins/genetics , Spermatids/metabolism , Spermatocytes/metabolism , Spermatogenesis/genetics , Transcriptome , Animals , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Female , Fertility/genetics , Gene Expression Regulation , Gene Ontology , Genes, Reporter , Genetic Complementation Test , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Molecular Sequence Annotation , Mutation , RNA-Binding Proteins/metabolism , Spermatids/cytology , Spermatids/growth & development , Spermatocytes/cytology , Spermatocytes/growth & development , Testis/cytology , Testis/metabolism , Y Chromosome/chemistry
7.
PLoS Genet ; 17(5): e1009412, 2021 05.
Article En | MEDLINE | ID: mdl-33961623

Meiosis is a cell division process with complex chromosome events where various molecules must work in tandem. To find meiosis-related genes, we screened evolutionarily conserved and reproductive tract-enriched genes using the CRISPR/Cas9 system and identified potassium channel tetramerization domain containing 19 (Kctd19) as an essential factor for meiosis. In prophase I, Kctd19 deficiency did not affect synapsis or the DNA damage response, and chiasma structures were also observed in metaphase I spermatocytes of Kctd19 KO mice. However, spermatocytes underwent apoptotic elimination during the metaphase-anaphase transition. We were able to rescue the Kctd19 KO phenotype with an epitope-tagged Kctd19 transgene. By immunoprecipitation-mass spectrometry, we confirmed the association of KCTD19 with zinc finger protein 541 (ZFP541) and histone deacetylase 1 (HDAC1). Phenotyping of Zfp541 KO spermatocytes demonstrated XY chromosome asynapsis and recurrent DNA damage in the late pachytene stage, leading to apoptosis. In summary, our study reveals that KCTD19 associates with ZFP541 and HDAC1, and that both KCTD19 and ZFP541 are essential for meiosis in male mice.


Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Genes, Essential , Meiosis , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Anaphase , Animals , CRISPR-Cas Systems/genetics , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Nucleus/metabolism , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/genetics , Chromosome Pairing , Conserved Sequence , DNA Damage , Evolution, Molecular , Fertility/genetics , Histone Deacetylase 1/metabolism , Male , Meiotic Prophase I , Metaphase , Mice , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Pachytene Stage , Phenotype , Spermatids/cytology , Spermatocytes/cytology , Spermatocytes/metabolism , Testis/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Transgenes
8.
Nat Commun ; 12(1): 2981, 2021 05 20.
Article En | MEDLINE | ID: mdl-34016985

The spatial folding of chromosomes inside the nucleus has regulatory effects on gene expression, yet the impact of genome reshuffling on this organization remains unclear. Here, we take advantage of chromosome conformation capture in combination with single-nucleotide polymorphism (SNP) genotyping and analysis of crossover events to study how the higher-order chromatin organization and recombination landscapes are affected by chromosomal fusions in the mammalian germ line. We demonstrate that chromosomal fusions alter the nuclear architecture during meiosis, including an increased rate of heterologous interactions in primary spermatocytes, and alterations in both chromosome synapsis and axis length. These disturbances in topology were associated with changes in genomic landscapes of recombination, resulting in detectable genomic footprints. Overall, we show that chromosomal fusions impact the dynamic genome topology of germ cells in two ways: (i) altering chromosomal nuclear occupancy and synapsis, and (ii) reshaping landscapes of recombination.


Chromatin/metabolism , Chromosomes/metabolism , Recombination, Genetic , Spermatocytes/metabolism , Animals , Biological Evolution , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , Chromatin/genetics , Chromosome Pairing/genetics , Chromosome Segregation , Chromosomes/genetics , Europe , Fertility/genetics , Genotyping Techniques/methods , Male , Mice , Polymorphism, Single Nucleotide , Primary Cell Culture , Semen Analysis , Spermatocytes/cytology
9.
Food Chem Toxicol ; 154: 112245, 2021 Aug.
Article En | MEDLINE | ID: mdl-33940107

Dietary pattern and cooking methods are important factors to determine the nutrients supplementation for male reproduction. Methionine and choline are two methyl donors in daily diet, which could mediate the lipid metabolism, but their effects on the sperms are not clear. In this study, we fed the mice with methionine-choline deficient (MCD) diet or the baked MCD diet for 6 weeks to evaluate this dietary pattern and the appended high temperature cooking on the spermatogenesis. The results have shown that MCD diet induced testis degradation and the damage of spermatocytes, reduced sperm vitality, motility, but elevated sperm deformity. Additionally, baking of MCD diet aggravated the testis injury, further reduced sperm density, sperm motility, and decreased normal sperm morphology dramatically. These changes were not related to the blood-testis barrier nor the Leydig cells dysfunction, but related to spermatocytes lost and apoptosis. The spermatocyte apoptosis was mediated by reticulum stress, including GRP78, XBP-1 and CHOP gene expression. Our study has shown the importance of methionine and choline in diet, and emphasized the crucial role of cooking condition, which are dietary factors to influence the quality of sperms.


Choline Deficiency/metabolism , Cooking , Diet , Methionine/deficiency , Testis/cytology , Animals , Apoptosis , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Male , Mice , Mice, Inbred C57BL , Sperm Count , Sperm Motility , Spermatocytes/cytology
10.
Cytogenet Genome Res ; 161(1-2): 14-22, 2021.
Article En | MEDLINE | ID: mdl-33725692

Amplified sequences constitute a large part of mammalian genomes. A chromosome 1 containing 2 large (up to 50 Mb) homogeneously staining regions (HSRs) separated by a small inverted euchromatic region is present in many natural populations of the house mouse (Mus musculus musculus). The HSRs are composed of a long-range repeat cluster, Sp100-rs, with a repeat length of 100 kb. In order to understand the organization and function of HSRs in meiotic chromosomes, we examined synapsis and recombination in male mice hetero- and homozygous for the HSR-carrying chromosome using FISH with an HSR-specific DNA probe and immunolocalization of the key meiotic proteins. In all homozygous and heterozygous pachytene nuclei, we observed fully synapsed linear homomorphic bivalents 1 marked by the HSR FISH probe. The synaptic adjustment in the heterozygotes was bilateral: the HSR-carrying homolog was shortened and the wild-type homolog was elongated. The adjustment was reversible: desynapsis at diplotene was accompanied by elongation of the HSRs. Immunolocalization of H3K9me2/3 indicated that the HSRs in the meiotic chromosome retained the epigenetic modification typical for C-heterochromatin in somatic cells. MLH1 foci, marking mature recombination nodules, were detected in the proximal HSR band in heterozygotes and in both HSR bands of homozygotes. Unequal crossing over within the long-range repeat cluster can cause variation in size of the HSRs, which has been detected in the natural populations of the house mouse.


Chromosome Mapping , Meiosis , Recombination, Genetic , Animals , Cell Nucleus/metabolism , Chromosome Aberrations , Chromosome Banding , DNA/genetics , Epigenesis, Genetic , Female , Heterozygote , Histones/genetics , Homozygote , In Situ Hybridization, Fluorescence , Karyotyping , Male , Mice , Mice, Inbred C57BL , Multigene Family , Spermatocytes/cytology
11.
STAR Protoc ; 2(1): 100294, 2021 03 19.
Article En | MEDLINE | ID: mdl-33532739

Here, we describe a detailed protocol for the isolation of purified populations of viable spermatogenic cells derived from the non-human primate model organism Macaca fascicularis (cynomolgus). Using fluorescence-activated cell sorting (FACS), we describe methods to isolate spermatogonia and primary spermatocytes ranging across the sub-stages of meiosis prophase I. These cell populations can be used with a variety of downstream assays, including single-cell approaches such as RNA sequencing, chromatin immunoprecipitation, quantitative RT-PCR, and immunocytochemistry. For complete details on the use and execution of this protocol, please refer to Lau et al. (2020).


Cell Separation , Spermatocytes/cytology , Testis/cytology , Animals , Macaca fascicularis , Male
12.
J Vis Exp ; (167)2021 01 14.
Article En | MEDLINE | ID: mdl-33522502

Isolation of meiotic spermatocytes is essential to investigate molecular mechanisms underlying meiosis and spermatogenesis. Although there are established cell isolation protocols using Hoechst 33342 staining in combination with fluorescence-activated cell sorting, it requires cell sorters equipped with an ultraviolet laser. Here we describe a cell isolation protocol using the DyeCycle Violet (DCV) stain, a low cytotoxicity DNA binding dye structurally similar to Hoechst 33342. DCV can be excited by both ultraviolet and violet lasers, which improves the flexibility of equipment choice, including a cell sorter not equipped with an ultraviolet laser. Using this protocol, one can isolate three live-cell subpopulations in meiotic prophase I, including leptotene/zygotene, pachytene, and diplotene spermatocytes, as well as post-meiotic round spermatids. We also describe a protocol to prepare single-cell suspension from mouse testes. Overall, the procedure requires a short time to complete (4-5 hours depending on the number of needed cells), which facilitates many downstream applications.


Cell Membrane Permeability , Cell Separation/methods , DNA/metabolism , Spermatocytes/cytology , Spermatogenesis , Animals , Benzimidazoles/metabolism , Cell Survival , Dissection , Flow Cytometry , Fluorescence , Male , Meiosis , Mice , Pachytene Stage , Scattering, Radiation , Spermatids/cytology , Staining and Labeling , Testis/cytology
13.
Development ; 148(5)2021 03 05.
Article En | MEDLINE | ID: mdl-33558389

Many specialized cells use unconventional strategies of cytoskeletal control. Nematode spermatocytes discard their actin and tubulin following meiosis, and instead employ the regulated assembly/disassembly of the Major Sperm Protein (MSP) to drive sperm motility. However, prior to the meiotic divisions, MSP is sequestered through its assembly into paracrystalline structures called fibrous bodies (FBs). The accessory proteins that direct this sequestration process have remained mysterious. This study reveals SPE-18 as an intrinsically disordered protein that is essential for MSP assembly within FBs. In spe-18 mutant spermatocytes, MSP forms disorganized cortical fibers, and the cells arrest in meiosis without forming haploid sperm. In wild-type spermatocytes, SPE-18 localizes to pre-FB complexes and functions with the kinase SPE-6 to localize MSP assembly. Changing patterns of SPE-18 localization uncover previously unappreciated complexities in FB maturation. Later, within newly individualized spermatids, SPE-18 is rapidly lost, yet SPE-18 loss alone is insufficient for MSP disassembly. Our findings reveal an alternative strategy for sequestering cytoskeletal elements, not as monomers but in localized, bundled polymers. Additionally, these studies provide an important example of disordered proteins promoting ordered cellular structures.


Caenorhabditis elegans Proteins/metabolism , Intrinsically Disordered Proteins/metabolism , Spermatocytes/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Cell Cycle Checkpoints , Cytoskeleton/metabolism , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Male , Meiosis , Mutagenesis , Sequence Alignment , Spermatids/metabolism , Spermatocytes/cytology , Spermatocytes/growth & development , Spermatogenesis
14.
Methods Mol Biol ; 2153: 267-286, 2021.
Article En | MEDLINE | ID: mdl-32840786

Crossing-over between homologous chromosomes is essential for accurate chromosome segregation at anaphase-I of meiosis. Defective crossing-over is associated with infertility, pregnancy miscarriage, and congenital disease. This chapter presents optimized protocols for the analysis of meiotic crossovers at the cytological level in spermatocytes and oocytes from mouse. The first approach employs immunocytology to detect MLH1, a DNA mismatch-repair protein that specifically marks crossover sites in the pachytene stage of meiotic prophase-I. These immunocytological methods have general utility for the analysis of other recombination steps, such as initiation and DNA strand exchange. The second approach visualizes chiasmata, the points of physical exchange between homologous chromosomes that are present during the diakinesis and metaphase-I stages. Both approaches are readily adaptable to the analysis of crossing over in other vertebrate species.


Crossing Over, Genetic , MutL Protein Homolog 1/metabolism , Oocytes/cytology , Spermatocytes/cytology , Aneuploidy , Animals , Cells, Cultured , Chromosomes, Mammalian/metabolism , Female , Immunohistochemistry , Male , Mice , Oocytes/metabolism , Pachytene Stage , Spermatocytes/metabolism
15.
J Biol Chem ; 296: 100130, 2021.
Article En | MEDLINE | ID: mdl-33262216

Meiosis, which produces haploid progeny, is critical to ensuring both faithful genome transmission and genetic diversity. Proteasomes play critical roles at various stages of spermatogenesis, including meiosis, but the underlying mechanisms remain unclear. The atypical proteasomes, which contain the activator PA200, catalyze the acetylation-dependent degradation of the core histones in elongated spermatids and DNA repair in somatic cells. We show here that the testis-specific proteasome subunit α4s/PSMA8 is essential for male fertility by promoting proper formation of spermatoproteasomes, which harbor both PA200 and constitutive catalytic subunits. Immunostaining of a spermatocyte marker, SYCP3, indicated that meiosis was halted at the stage of spermatocytes in the α4s-deficient testes. α4s stimulated the in vitro degradation of the acetylated core histones, instead of nonacetylated histones, by the PA200-proteasome. Deletion of α4s blocked degradation of the core histones at DNA damage loci in spermatocytes, leading to meiotic arrest at metaphase I. Thus, α4s is required for histone degradation at meiotic DNA damage loci, proper progression of meiosis, and fertility in males by promoting proper formation of spermatoproteasomes. These results are important for understanding male infertility and might provide potential targets for male contraception or treatment of male infertility.


DNA Repair , Histones/metabolism , Infertility, Male/pathology , Meiosis , Proteasome Endopeptidase Complex/metabolism , Spermatocytes/cytology , Spermatogenesis , Animals , DNA Damage , Infertility, Male/etiology , Infertility, Male/metabolism , Male , Mice , Mice, Inbred C57BL , Proteasome Endopeptidase Complex/genetics , Spermatids , Spermatocytes/metabolism
16.
DNA Cell Biol ; 40(2): 209-218, 2021 Feb.
Article En | MEDLINE | ID: mdl-33337266

Poly (ADP-ribose) polymerase-1 (Parp1) is a member of nuclear enzymes family involved in to the response to genotoxic stresses, DNA repair, and is critical for the maintenance of genome stability. During gametogenesis, genome stability is essential for inheritance and formation of healthy gametes. The latter involves DNA double-strand break (DSB)-driven pairing of homologous chromosomes in first meiotic prophase. By analysis of DSB repair kinetics in male meiotic prophase cells of homologous recombination (HR) and nonhomologous end joining (NHEJ)-deficient mouse models, we previously demonstrated an interplay between HR and the conventional NHEJ repair pathway. In the current work, we evaluate the relative contribution of Parp1-dependent NHEJ to the repair of ectopic ionizing radiation (IR)-induced DSBs in control and Parp1-inhibited mouse pachytene spermatocytes before and after the completion of meiotic recombination in stages VI-XI. The disappearance of large, exogenous DSB-related γ-H2AX foci was quantified 1 and 8 h after 1 Gy γ-irradiation of control and 3,4-dihydro-5-[4-(1-piperidinyl)butoxy]-1(2H)quinolinone (DPQ) Parp1-inhibited mice. Late pachytene control spermatocytes obtained 8 h after IR had repaired >80% of DSBs observed at 1 h after IR. However, only 64% of DSBs were repaired in late spermatocytes of DPQ-treated (Parp1-inhibited) mice. Thus, it appears that Parp1 contributes to the repair of a fraction of DSBs in late prophase I, providing further insights in DNA repair pathway choreography during spermatogenic differentiation.


DNA Breaks, Double-Stranded , DNA Repair , Pachytene Stage/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Spermatocytes/cytology , Spermatocytes/radiation effects , Animals , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/radiation effects , Male , Mice , Mice, Inbred C57BL , Pachytene Stage/radiation effects , Spermatocytes/metabolism
17.
J Genet Genomics ; 47(8): 451-465, 2020 08.
Article En | MEDLINE | ID: mdl-33250349

Meiosis is a specialized cell division for producing haploid gametes in sexually reproducing organisms. In this study, we have independently identified a novel meiosis protein male meiosis recombination regulator (MAMERR)/4930432K21Rik and showed that it is indispensable for meiosis prophase I progression in male mice. Using super-resolution structured illumination microscopy, we found that MAMERR functions at the same double-strand breaks as the replication protein A and meiosis-specific with OB domains/spermatogenesis associated 22 complex. We generated a Mamerr-deficient mouse model by deleting exons 3-6 and found that most of Mamerr-/- spermatocytes were arrested at pachynema and failed to progress to diplonema, although they exhibited almost intact synapsis and progression to the pachytene stage along with XY body formation. Further mechanistic studies revealed that the recruitment of DMC1/RAD51 and heat shock factor 2-binding protein in Mamerr-/- spermatocytes was only mildly impaired with a partial reduction in double-strand break repair, whereas a substantial reduction in ubiquitination on the autosomal axes and on the XY body appeared as a major phenotype in Mamerr-/- spermatocytes. We propose that MAMERR may participate in meiotic prophase I progression by regulating the ubiquitination of key meiotic proteins on autosomes and XY chromosomes, and in the absence of MAMERR, the repressed ubiquitination of key meiotic proteins leads to pachytene arrest and cell death.


Cell Cycle Proteins/genetics , Chromosomes/genetics , Meiosis/genetics , Meiotic Prophase I/genetics , Animals , Chromosome Pairing/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , Male , Mice , Recombination, Genetic/genetics , Spermatocytes/cytology , Spermatogenesis/genetics
18.
Anim Reprod Sci ; 222: 106608, 2020 Nov.
Article En | MEDLINE | ID: mdl-33039822

Spermatogenesis is important for male fertility, but has not been well-studied in Opsariichthys bidens, an economically important freshwater fish in China. In this study, there was investigation of the cytological features of spermatogenesis in O. bidens using light microscopy, transmission electron microscopy, and immunofluorescence detection of microtubules. O. bidens has tubular testis. Spermatogenesis in O. bidens is of the cystic type, in which the spermatogenic cells develop into spermatozoa in cysts. There was asynchronous development of primary spermatocytes within a single cyst. Spermiogenesis was classified as Type I, which develops into a Type I aquasperm with an oval nucleus, a small and simple midpiece, a flagellum and no acrosome. There was a nuage in spermatogonia, spermatocytes, and spermatids in different developmental stages of spermatids which may have important functions in fish spermatogenesis. Furthermore, microtubule dynamics may be involved in spermatid reshaping, material transport, and polar distribution of organelles during spermiogenesis.


Cyprinidae/physiology , Spermatogenesis/physiology , Spermatozoa/cytology , Testis/cytology , Animals , Aquaculture , China , Cytoskeleton/physiology , Fresh Water , Male , Meiosis , Microscopy, Electron , Microtubules/ultrastructure , Sertoli Cells/cytology , Sertoli Cells/ultrastructure , Spermatids/cytology , Spermatids/ultrastructure , Spermatocytes/cytology , Spermatocytes/ultrastructure , Spermatogonia/cytology , Spermatogonia/ultrastructure , Spermatozoa/ultrastructure , Testis/ultrastructure
19.
Elife ; 92020 10 13.
Article En | MEDLINE | ID: mdl-33047671

Meiotic recombination starts with the formation of DNA double-strand breaks (DSBs) at specific genomic locations that correspond to PRDM9-binding sites. The molecular steps occurring from PRDM9 binding to DSB formation are unknown. Using proteomic approaches to find PRDM9 partners, we identified HELLS, a member of the SNF2-like family of chromatin remodelers. Upon functional analyses during mouse male meiosis, we demonstrated that HELLS is required for PRDM9 binding and DSB activity at PRDM9 sites. However, HELLS is not required for DSB activity at PRDM9-independent sites. HELLS is also essential for 5-hydroxymethylcytosine (5hmC) enrichment at PRDM9 sites. Analyses of 5hmC in mice deficient for SPO11, which catalyzes DSB formation, and in PRDM9 methyltransferase deficient mice reveal that 5hmC is triggered at DSB-prone sites upon PRDM9 binding and histone modification, but independent of DSB activity. These findings highlight the complex regulation of the chromatin and epigenetic environments at PRDM9-specified hotspots.


5-Methylcytosine/analogs & derivatives , DNA Breaks, Double-Stranded , DNA Helicases/metabolism , Histone-Lysine N-Methyltransferase/genetics , 5-Methylcytosine/metabolism , Animals , Binding Sites , Endodeoxyribonucleases/metabolism , HeLa Cells , Histone-Lysine N-Methyltransferase/metabolism , Homologous Recombination , Humans , Male , Mice , Mice, Knockout , Proteomics , Spermatocytes/cytology , Testis/metabolism
20.
Development ; 147(22)2020 11 27.
Article En | MEDLINE | ID: mdl-33060131

In sexually reproducing metazoans, spermatogenesis is the process by which uncommitted germ cells give rise to haploid sperm. Work in model systems has revealed mechanisms controlling commitment to the sperm fate, but how this fate is subsequently executed remains less clear. While studying the well-established role of the conserved nuclear hormone receptor transcription factor, NHR-23/NR1F1, in regulating C. elegans molting, we discovered that NHR-23/NR1F1 is also constitutively expressed in developing primary spermatocytes and is a critical regulator of spermatogenesis. In this novel role, NHR-23/NR1F1 functions downstream of the canonical sex-determination pathway. Degron-mediated depletion of NHR-23/NR1F1 within hermaphrodite or male germlines causes sterility due to an absence of functional sperm, as depleted animals produce arrested primary spermatocytes rather than haploid sperm. These spermatocytes arrest in prometaphase I and fail to either progress to anaphase or attempt spermatid-residual body partitioning. They make sperm-specific membranous organelles but fail to assemble their major sperm protein into fibrous bodies. NHR-23/NR1F1 appears to function independently of the known SPE-44 gene regulatory network, revealing the existence of an NHR-23/NR1F1-mediated module that regulates the spermatogenesis program.


Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Spermatids/metabolism , Spermatocytes/metabolism , Spermatogenesis/physiology , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Male , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Spermatids/cytology , Spermatocytes/cytology
...