Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2770: 53-62, 2024.
Article in English | MEDLINE | ID: mdl-38351446

ABSTRACT

In the adult mouse testis, germ cells of various developmental cell states co-exist. FACS isolation of cells stained with the DNA dye Hoechst 33342 has been used for many years to sub-divide these cells based on their DNA content. This approach provides an efficient way to obtain broad categories of male germ cells: pre-meiotic spermatogonia, meiotic spermatocytes and post-meiotic spermatids. The addition of a red filter for Hoechst staining enables further sub-division of spermatocytes depending on sub-stages of meiotic prophase. However, separation of different stage spermatids using Hoechst staining alone is not possible. We recently reported a methodology, combining Hoechst staining with a second DNA dye (SYTO16) that enables the further separation of these cells into three sub-populations: round, early elongating, and late elongating spermatids (Gill et al., Cytometry A 101:529-536, 2022). This method makes it possible to obtain rapidly and simply pure fractions of male germ cells from multiple developental stages from the same animal.


Subject(s)
Benzimidazoles , Spermatogenesis , Testis , Mice , Animals , Male , Meiosis , Spermatids , Spermatocytes , Germ Cells , Staining and Labeling , DNA
2.
Genome Res ; 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38129075

ABSTRACT

In mammals, the adult testis is the tissue with the highest diversity in gene expression. Much of that diversity is attributed to germ cells, primarily meiotic spermatocytes and postmeiotic haploid spermatids. Exploiting a newly developed cell purification method, we profiled the transcriptomes of such postmitotic germ cells of mice. We used a de novo transcriptome assembly approach and identified thousands of novel expressed transcripts characterized by features distinct from those of known genes. Novel loci tend to be short in length, monoexonic, and lowly expressed. Most novel genes have arisen recently in evolutionary time and possess low coding potential. Nonetheless, we identify several novel protein-coding genes harboring open reading frames that encode proteins containing matches to conserved protein domains. Analysis of mass-spectrometry data from adult mouse testes confirms protein production from several of these novel genes. We also examine overlap between transcripts and repetitive elements. We find that although distinct families of repeats are expressed with differing temporal dynamics during spermatogenesis, we do not observe a general mode of regulation wherein repeats drive expression of nonrepetitive sequences in a cell type-specific manner. Finally, we observe many fairly long antisense transcripts originating from canonical gene promoters, pointing to pervasive bidirectional promoter activity during spermatogenesis that is distinct and more frequent compared with somatic cells.

3.
J Assist Reprod Genet ; 40(2): 235-239, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36534231

ABSTRACT

Microscopy has long played a pivotal role in the field of assisted reproductive technology (ART). The advent of artificial intelligence (AI) has opened the door for new approaches to sperm and oocyte assessment and selection, with the potential for improved ART outcomes.


Subject(s)
Artificial Intelligence , Reproductive Medicine , Male , Animals , Microscopy , Semen , Reproductive Techniques, Assisted
4.
Clin Epigenetics ; 14(1): 185, 2022 12 26.
Article in English | MEDLINE | ID: mdl-36572941

ABSTRACT

BACKGROUND: It has been suggested that antenatal exposure to environmental endocrine disruptors is responsible for adverse trends in male reproductive health, including male infertility, impaired semen quality, cryptorchidism and testicular cancer, a condition known as testicular dysgenesis syndrome. Anogenital distance (AGD) is an anthropomorphic measure of antenatal exposure to endocrine disruptors, with higher exposure levels leading to shortened AGD. We hypothesized that exposure to endocrine disruptors could lead to changes in DNA methylation during early embryonic development, which could then persist in the sperm of infertile men with shortened AGD. RESULTS: Using fluorescence activated cell sorting based on staining with either YO-PRO-1 (YOPRO) or chromomycin-3 (CMA3), we isolated four sperm fractions from eleven infertile men with short AGD and ten healthy semen donors. We examined DNA methylation in these sorted spermatozoa using reduced representation bisulfite sequencing. We found that fractions of spermatozoa from infertile men stained with CMA3 or YOPRO were more likely to contain transposable elements harboring an estrogen receptor response element (ERE). Abnormal sperm (as judged by high CMA3 or YOPRO staining) from infertile men shows substantial hypomethylation in estrogenic Alu sequences. Conversely, normal sperm fractions (as judged by low CMA3 or YO-PRO-1 staining) of either healthy donors or infertile patients were more likely to contain hypermethylated Alu sequences with ERE. CONCLUSIONS: Shortened AGD, as related to previous exposure to endocrine disruptors, and male infertility are accompanied by increased presence of hormonal response elements in the differentially methylated regulatory sequences of the genome of sperm fractions characterized by chromatin decondensation and apoptosis.


Subject(s)
Endocrine Disruptors , Infertility, Male , Testicular Neoplasms , Humans , Male , Female , Pregnancy , Semen Analysis , Testicular Neoplasms/genetics , DNA Methylation , Endocrine Disruptors/metabolism , Semen , Spermatozoa/metabolism , Infertility, Male/chemically induced , Infertility, Male/genetics , Estrogens/metabolism , Repetitive Sequences, Nucleic Acid
5.
Cytometry A ; 101(6): 529-536, 2022 06.
Article in English | MEDLINE | ID: mdl-35128792

ABSTRACT

During spermatogenesis, mammalian male germ cells undergo multiple developmental processes, including meiosis and post-meiotic differentiation (spermiogenesis). To understand the transitions between different cellular states it is essential to isolate pure populations of cells at different stages of development. Previous approaches enabled the isolation of cells from different stages of meiotic prophase I, but techniques to sub-fractionate unfixed, post-meiotic spermatids have been lacking. Here we report the development of a protocol enabling simultaneous isolation of cells at different stages of meiotic prophase and post-meiotic differentiation from testes of adult mice. This approach builds on existing fluorescence activated cell sorting protocols designed to purify cells in different stages of meiotic prophase I. By utilizing the specific spectral properties that two different DNA dyes (Hoechst 33342 and SYTO 16) exhibit when bound to chromatin of different stage male germ cells, we obtain highly pure populations of cells in relatively large numbers. This FACS protocol will enable immunocytological and molecular characterization studies of fractionated meiotic and haploid germ cells from both wild type and genetically mutant animals.


Subject(s)
Meiosis , Spermatids , Animals , DNA/metabolism , Germ Cells/metabolism , Male , Mammals/genetics , Mice , Spermatogenesis/genetics , Staining and Labeling , Testis
6.
Nature ; 588(7839): 642-647, 2020 12.
Article in English | MEDLINE | ID: mdl-33177713

ABSTRACT

Gene-expression programs define shared and species-specific phenotypes, but their evolution remains largely uncharacterized beyond the transcriptome layer1. Here we report an analysis of the co-evolution of translatomes and transcriptomes using ribosome-profiling and matched RNA-sequencing data for three organs (brain, liver and testis) in five mammals (human, macaque, mouse, opossum and platypus) and a bird (chicken). Our within-species analyses reveal that translational regulation is widespread in the different organs, in particular across the spermatogenic cell types of the testis. The between-species divergence in gene expression is around 20% lower at the translatome layer than at the transcriptome layer owing to extensive buffering between the expression layers, which especially preserved old, essential and housekeeping genes. Translational upregulation specifically counterbalanced global dosage reductions during the evolution of sex chromosomes and the effects of meiotic sex-chromosome inactivation during spermatogenesis. Despite the overall prevalence of buffering, some genes evolved faster at the translatome layer-potentially indicating adaptive changes in expression; testis tissue shows the highest fraction of such genes. Further analyses incorporating mass spectrometry proteomics data establish that the co-evolution of transcriptomes and translatomes is reflected at the proteome layer. Together, our work uncovers co-evolutionary patterns and associated selective forces across the expression layers, and provides a resource for understanding their interplay in mammalian organs.


Subject(s)
Evolution, Molecular , Mammals/genetics , Protein Biosynthesis , Transcriptome/genetics , Animals , Brain/metabolism , Chickens/genetics , Female , Genes, X-Linked/genetics , Humans , Liver/metabolism , Macaca/genetics , Male , Mice , Opossums/genetics , Organ Specificity/genetics , Platypus/genetics , Protein Biosynthesis/genetics , RNA-Seq , Ribosomes/metabolism , Sex Chromosomes/genetics , Species Specificity , Spermatogenesis/genetics , Testis/metabolism , Up-Regulation
7.
EMBO J ; 39(13): e103697, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32395866

ABSTRACT

Chromatin integrity is essential for cellular homeostasis. Polycomb group proteins modulate chromatin states and transcriptionally repress developmental genes to maintain cell identity. They also repress repetitive sequences such as major satellites and constitute an alternative state of pericentromeric constitutive heterochromatin at paternal chromosomes (pat-PCH) in mouse pre-implantation embryos. Remarkably, pat-PCH contains the histone H3.3 variant, which is absent from canonical PCH at maternal chromosomes, which is marked by histone H3 lysine 9 trimethylation (H3K9me3), HP1, and ATRX proteins. Here, we show that SUMO2-modified CBX2-containing Polycomb Repressive Complex 1 (PRC1) recruits the H3.3-specific chaperone DAXX to pat-PCH, enabling H3.3 incorporation at these loci. Deficiency of Daxx or PRC1 components Ring1 and Rnf2 abrogates H3.3 incorporation, induces chromatin decompaction and breakage at PCH of exclusively paternal chromosomes, and causes their mis-segregation. Complementation assays show that DAXX-mediated H3.3 deposition is required for chromosome stability in early embryos. DAXX also regulates repression of PRC1 target genes during oogenesis and early embryogenesis. The study identifies a novel critical role for Polycomb in ensuring heterochromatin integrity and chromosome stability in mouse early development.


Subject(s)
Embryo, Mammalian/embryology , Genomic Instability , Heterochromatin/metabolism , Histones/metabolism , Polycomb Repressive Complex 1/metabolism , Sumoylation , Animals , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Female , Heterochromatin/genetics , Histones/genetics , Male , Mice , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Polycomb Repressive Complex 1/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism
8.
Mol Cell ; 75(3): 483-497.e9, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31253574

ABSTRACT

In mammals, ∼100 deubiquitinases act on ∼20,000 intracellular ubiquitination sites. Deubiquitinases are commonly regarded as constitutively active, with limited regulatory and targeting capacity. The BRCA1-A and BRISC complexes serve in DNA double-strand break repair and immune signaling and contain the lysine-63 linkage-specific BRCC36 subunit that is functionalized by scaffold subunits ABRAXAS and ABRO1, respectively. The molecular basis underlying BRCA1-A and BRISC function is currently unknown. Here we show that in the BRCA1-A complex structure, ABRAXAS integrates the DNA repair protein RAP80 and provides a high-affinity binding site that sequesters the tumor suppressor BRCA1 away from the break site. In the BRISC structure, ABRO1 binds SHMT2α, a metabolic enzyme enabling cancer growth in hypoxic environments, which we find prevents BRCC36 from binding and cleaving ubiquitin chains. Our work explains modularity in the BRCC36 DUB family, with different adaptor subunits conferring diversified targeting and regulatory functions.


Subject(s)
BRCA1 Protein/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Deubiquitinating Enzymes/genetics , Histone Chaperones/genetics , Neoplasms/genetics , Binding Sites/genetics , Carrier Proteins/genetics , Cell Nucleus/genetics , Cell Nucleus/immunology , Cytoplasm/genetics , Cytoplasm/immunology , DNA Breaks, Double-Stranded , DNA Repair/immunology , Deubiquitinating Enzymes/immunology , HeLa Cells , Humans , Immunity, Cellular/genetics , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Neoplasms/immunology , Nuclear Matrix-Associated Proteins/genetics , Protein Binding/genetics , Ubiquitin/genetics , Ubiquitin-Specific Proteases/genetics , Ubiquitination/genetics
9.
Science ; 362(6412): 291-292, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30337398

Subject(s)
Ovum , Humans
10.
EMBO J ; 36(21): 3097-3099, 2017 11 02.
Article in English | MEDLINE | ID: mdl-28978668

Subject(s)
Meiosis , Humans
11.
PLoS Genet ; 11(9): e1005531, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26378784

ABSTRACT

The chromosomal program of meiotic prophase, comprising events such as laying down of meiotic cohesins, synapsis between homologs, and homologous recombination, must be preceded and enabled by the regulated induction of meiotic prophase genes. This gene regulatory program is poorly understood, particularly in organisms with a segregated germline. We characterized the gene regulatory program of meiotic prophase as it occurs in the mouse fetal ovary. By profiling gene expression in the mouse fetal ovary in mutants with whole tissue and single-cell techniques, we identified 104 genes expressed specifically in pre-meiotic to pachytene germ cells. We characterized the regulation of these genes by 1) retinoic acid (RA), which induces meiosis, 2) Dazl, which is required for germ cell competence to respond to RA, and 3) Stra8, a downstream target of RA required for the chromosomal program of meiotic prophase. Initial induction of practically all identified meiotic prophase genes requires Dazl. In the presence of Dazl, RA induces at least two pathways: one Stra8-independent, and one Stra8-dependent. Genes vary in their induction by Stra8, spanning fully Stra8-independent, partially Stra8-independent, and fully Stra8-dependent. Thus, Stra8 regulates the entirety of the chromosomal program but plays a more nuanced role in governing the gene expression program. We propose that Stra8-independent gene expression enables the stockpiling of selected meiotic structural proteins prior to the commencement of the chromosomal program. Unexpectedly, we discovered that Stra8 is required for prompt down-regulation of itself and Rec8. Germ cells that have expressed and down-regulated Stra8 are refractory to further Stra8 expression. Negative feedback of Stra8, and subsequent resistance to further Stra8 expression, may ensure a single, restricted pulse of Stra8 expression. Collectively, our findings reveal a gene regulatory logic by which germ cells prepare for the chromosomal program of meiotic prophase, and ensure that it is induced only once.


Subject(s)
Gene Regulatory Networks , Meiotic Prophase I , Ovary/embryology , Sheep/embryology , Adaptor Proteins, Signal Transducing/genetics , Animals , Down-Regulation , Female , Gene Expression Regulation, Developmental , Ovary/cytology
12.
PLoS Genet ; 10(8): e1004541, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25102060

ABSTRACT

In all sexually reproducing organisms, cells of the germ line must transition from mitosis to meiosis. In mice, retinoic acid (RA), the extrinsic signal for meiotic initiation, activates transcription of Stra8, which is required for meiotic DNA replication and the subsequent processes of meiotic prophase. Here we report that RA also activates transcription of Rec8, which encodes a component of the cohesin complex that accumulates during meiotic S phase, and which is essential for chromosome synapsis and segregation. This RA induction of Rec8 occurs in parallel with the induction of Stra8, and independently of Stra8 function, and it is conserved between the sexes. Further, RA induction of Rec8, like that of Stra8, requires the germ-cell-intrinsic competence factor Dazl. Our findings strengthen the importance of RA and Dazl in the meiotic transition, provide important details about the Stra8 pathway, and open avenues to investigate early meiosis through analysis of Rec8 induction and function.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Meiosis/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , RNA-Binding Proteins/genetics , Tretinoin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , DNA Replication/genetics , Female , Gene Expression Regulation, Developmental/drug effects , Germ Cells/growth & development , Male , Mice , Mitosis/genetics , Nuclear Proteins/biosynthesis , Ovary/drug effects , Ovary/growth & development , Phosphoproteins/biosynthesis , RNA-Binding Proteins/biosynthesis , Signal Transduction/drug effects , Testis/drug effects , Testis/growth & development , Transcription, Genetic/drug effects , Tretinoin/administration & dosage
13.
Curr Opin Cell Biol ; 24(3): 387-96, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22445736

ABSTRACT

At fertilization, fusion of two differentiated gametes forms the zygote that is capable of forming all of the varied cell lineages of an organism. It is widely thought that the acquisition of totipotency involves extensive epigenetic reprogramming of the germline state into an embryonic state. However, recent data argue that this reprogramming is incomplete and that substantial epigenetic information passes from one generation to the next. In this review we summarize the changes in chromatin states that take place during mammalian gametogenesis and examine the evidence that early mammalian embryogenesis may be affected by inheritance of epigenetic information from the parental generation.


Subject(s)
Embryonic Development/physiology , Epigenomics , Animals , Cell Lineage/genetics , Chromatin/metabolism , Chromatin/physiology , DNA Methylation , Embryonic Development/genetics , Fertilization/genetics , Fertilization/physiology , Gametogenesis/genetics , Gametogenesis/physiology , Germ Cells/physiology , Humans , Inheritance Patterns , Mammals/genetics , Zygote/metabolism , Zygote/physiology
14.
Proc Natl Acad Sci U S A ; 108(34): 14163-8, 2011 Aug 23.
Article in English | MEDLINE | ID: mdl-21844366

ABSTRACT

Mir-290 through mir-295 (mir-290-295) is a mammalian-specific microRNA (miRNA) cluster that, in mice, is expressed specifically in early embryos and embryonic germ cells. Here, we show that mir-290-295 plays important roles in embryonic development as indicated by the partially penetrant lethality of mutant embryos. In addition, we show that in surviving mir-290-295-deficient embryos, female but not male fertility is compromised. This impairment in fertility arises from a defect in migrating primordial germ cells and occurs equally in male and female mutant animals. Male mir-290-295(-/-) mice, due to the extended proliferative lifespan of their germ cells, are able to recover from this initial germ cell loss and are fertile. Female mir-290-295(-/-) mice are unable to recover and are sterile, due to premature ovarian failure.


Subject(s)
Embryo Loss/genetics , Embryo Loss/pathology , Germ Cells/metabolism , Germ Cells/pathology , MicroRNAs/metabolism , Penetrance , Aging/pathology , Animals , Animals, Newborn , Apoptosis , Cell Count , Cell Cycle , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Female , Fertility/genetics , Gene Expression Regulation, Developmental , Gonads/growth & development , Gonads/pathology , Infertility, Female/genetics , Infertility, Female/pathology , Male , Mice , Mice, Mutant Strains , MicroRNAs/genetics
15.
Proc Natl Acad Sci U S A ; 108(18): 7443-8, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21504946

ABSTRACT

Mammalian oocytes and spermatozoa derive from fetal cells shared by the sexes. These primordial germ cells (PGCs) migrate to the developing somatic gonad, giving rise to oocytes or spermatozoa. These opposing sexual fates are determined not by the PGCs' own sex chromosome constitution (XX or XY), but by the sexual identity of the fetal gonad that they enter. We asked whether PGCs undergo a developmental transition that enables them to respond to feminizing or masculinizing cues from fetal ovary or testis. We conducted in vivo genetic studies of DAZL, an RNA-binding protein expressed in both ovarian and testicular germ cells. We found that germ cells in C57BL/6 Dazl--deficient fetuses-whether XX or XY--migrate to the gonad but do not develop either male or female features. Instead, they remain in a sexually undifferentiated state similar to that of migrating PGCs. Thus, germ cells in C57BL/6 Dazl-deficient fetuses do not respond to sexual cues from ovary or testis, whereas the earlier processes of germ cell specification and migration are unaffected. We propose that PGCs of both XX and XY fetuses undergo licensing, an active developmental transition that enables the resultant gametogenesis-competent cells to respond to feminizing or masculinizing cues produced by the fetal ovary or testis and hence to embark on oogenesis or spermatogenesis. In C57BL/6 mice, Dazl is required for licensing. Licensing serves as a gateway from the embryonic processes shared between the sexes--germ cell specification and migration--to the sex-specific pathways of oogenesis and spermatogenesis.


Subject(s)
Cell Differentiation/physiology , Gametogenesis/physiology , Germ Cells/cytology , Germ Cells/physiology , RNA-Binding Proteins/metabolism , Animals , Bromodeoxyuridine , Cell Movement/physiology , DNA Primers/genetics , Female , Fetus , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction
16.
Science ; 322(5908): 1685-7, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-19074348

ABSTRACT

Retinoic acid (RA) is an essential extrinsic inducer of meiotic initiation in mammalian germ cells. However, RA acts too widely in mammalian development to account, by itself, for the cell-type and temporal specificity of meiotic initiation. We considered parallels to yeast, in which extrinsic and intrinsic factors combine to restrict meiotic initiation. We demonstrate that, in mouse embryos, extrinsic and intrinsic factors together regulate meiotic initiation. The mouse RNA-binding protein DAZL, which is expressed by postmigratory germ cells, is a key intrinsic factor, enabling those cells to initiate meiosis in response to RA. Within a brief developmental window, Dazl-expressing germ cells in both XX and XY embryos actively acquire the ability to interpret RA as a meiosis-inducing signal.


Subject(s)
Embryo, Mammalian/cytology , Germ Cells/cytology , Meiosis , RNA-Binding Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Cell Cycle Proteins/metabolism , Cell Nucleus/ultrastructure , DNA Breaks , DNA Repair , DNA-Binding Proteins , Embryo, Mammalian/physiology , Endodeoxyribonucleases , Esterases/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ovary/embryology , Ovary/physiology , Phosphate-Binding Proteins , Phosphoproteins/genetics , Phosphoproteins/metabolism , Proteins/metabolism , RNA-Binding Proteins/genetics , Testis/embryology , Testis/physiology , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...