Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Genet ; 56: 145-164, 2022 11 30.
Article in English | MEDLINE | ID: mdl-35977408

ABSTRACT

Various stem cells in the body are tasked with maintaining tissue homeostasis throughout the life of an organism and thus must be resilient to intrinsic and extrinsic challenges such as infection and injury. Crucial to these challenges is genome maintenance because a high mutational load and persistent DNA lesions impact the production of essential gene products at proper levels and compromise optimal stem cell renewal and differentiation. Genome maintenance requires a robust and well-regulated DNA damage response suited to maintaining specific niches and tissues. In this review, we explore the similarities and differences between diverse stem cell types derived from (or preceding) all germ layers, including extraembryonic tissues. These cells utilize different strategies, including implementation of robust repair mechanisms, modulation of cell cycle checkpoints best suited to eliminating compromised cells, minimization of cell divisions, and differentiation in response to excessive damage.


Subject(s)
Mammals , Stem Cells , Animals , Cell Differentiation/genetics , Germ Layers , Mutation
2.
Cell ; 157(6): 1257-1261, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24906145

ABSTRACT

Germ cells are the ultimate stem cells, and reports of their in vitro derivation generate excitement due to potential applications in reproductive medicine. To date, there is no firm evidence that meiosis, the hallmark of gametogenesis, can be faithfully replicated outside of the gonad. We propose benchmarks for evaluating in vitro derivation of germ cells, facilitating realization of their potential.


Subject(s)
Cytological Techniques/standards , Gametogenesis , Germ Cells/cytology , Meiosis , Stem Cells/cytology , Female , Humans , Male
3.
Genes Dev ; 34(23-24): 1637-1649, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33184219

ABSTRACT

Germ cells specified during fetal development form the foundation of the mammalian germline. These primordial germ cells (PGCs) undergo rapid proliferation, yet the germline is highly refractory to mutation accumulation compared with somatic cells. Importantly, while the presence of endogenous or exogenous DNA damage has the potential to impact PGCs, there is little known about how these cells respond to stressors. To better understand the DNA damage response (DDR) in these cells, we exposed pregnant mice to ionizing radiation (IR) at specific gestational time points and assessed the DDR in PGCs. Our results show that PGCs prior to sex determination lack a G1 cell cycle checkpoint. Additionally, the response to IR-induced DNA damage differs between female and male PGCs post-sex determination. IR of female PGCs caused uncoupling of germ cell differentiation and meiotic initiation, while male PGCs exhibited repression of piRNA metabolism and transposon derepression. We also used whole-genome single-cell DNA sequencing to reveal that genetic rescue of DNA repair-deficient germ cells (Fancm-/- ) leads to increased mutation incidence and biases. Importantly, our work uncovers novel insights into how PGCs exposed to DNA damage can become developmentally defective, leaving only those genetically fit cells to establish the adult germline.


Subject(s)
DNA Damage , DNA/radiation effects , Embryonic Germ Cells/radiation effects , Germ Cells/radiation effects , Mutation/genetics , Radiation, Ionizing , Animals , Cell Cycle Checkpoints/genetics , Cell Differentiation/genetics , Cell Differentiation/radiation effects , DNA Transposable Elements/radiation effects , Embryonic Germ Cells/cytology , Female , Male , Meiosis/genetics , Meiosis/radiation effects , Mice , Oocytes/cytology , Oocytes/radiation effects , Pregnancy , RNA, Small Interfering/metabolism , Sex Factors
4.
Trends Genet ; 40(4): 326-336, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38177041

ABSTRACT

Meiosis is essential for gamete production in all sexually reproducing organisms. It entails two successive cell divisions without DNA replication, producing haploid cells from diploid ones. This process involves complex morphological and molecular differentiation that varies across species and between sexes. Specialized genomic events like meiotic recombination and chromosome segregation are tightly regulated, including preparation for post-meiotic development. Research in model organisms, notably yeast, has shed light on the genetic and molecular aspects of meiosis and its regulation. Although mammalian meiosis research faces challenges, particularly in replicating gametogenesis in vitro, advances in genetic and genomic technologies are providing mechanistic insights. Here we review the genetics and molecular biology of meiotic gene expression control, focusing on mammals.


Subject(s)
Meiosis , Saccharomyces cerevisiae , Animals , Meiosis/genetics , Saccharomyces cerevisiae/genetics , Gametogenesis/genetics , Chromosome Segregation/genetics , DNA Replication , Mammals
5.
Proc Natl Acad Sci U S A ; 120(30): e2219925120, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37459509

ABSTRACT

Infertility is a heterogeneous condition, with genetic causes thought to underlie a substantial fraction of cases. Genome sequencing is becoming increasingly important for genetic diagnosis of diseases including idiopathic infertility; however, most rare or minor alleles identified in patients are variants of uncertain significance (VUS). Interpreting the functional impacts of VUS is challenging but profoundly important for clinical management and genetic counseling. To determine the consequences of these variants in key fertility genes, we functionally evaluated 11 missense variants in the genes ANKRD31, BRDT, DMC1, EXO1, FKBP6, MCM9, M1AP, MEI1, MSH4 and SEPT12 by generating genome-edited mouse models. Nine variants were classified as deleterious by most functional prediction algorithms, and two disrupted a protein-protein interaction (PPI) in the yeast two hybrid (Y2H) assay. Though these genes are essential for normal meiosis or spermiogenesis in mice, only one variant, observed in the MCM9 gene of a male infertility patient, compromised fertility or gametogenesis in the mouse models. To explore the disconnect between predictions and outcomes, we compared pathogenicity calls of missense variants made by ten widely used algorithms to 1) those annotated in ClinVar and 2) those evaluated in mice. All the algorithms performed poorly in terms of predicting the effects of human missense variants modeled in mice. These studies emphasize caution in the genetic diagnoses of infertile patients based primarily on pathogenicity prediction algorithms and emphasize the need for alternative and efficient in vitro or in vivo functional validation models for more effective and accurate VUS description to either pathogenic or benign categories.


Subject(s)
Infertility, Male , Mutation, Missense , Humans , Male , Mice , Animals , Reproduction , Alleles , Infertility, Male/genetics , Disease Models, Animal , Septins/genetics
6.
Nature ; 567(7746): 105-108, 2019 03.
Article in English | MEDLINE | ID: mdl-30787433

ABSTRACT

Genomic instability can trigger cellular responses that include checkpoint activation, senescence and inflammation1,2. Although genomic instability has been extensively studied in cell culture and cancer paradigms, little is known about its effect during embryonic development, a period of rapid cellular proliferation. Here we report that mutations in the heterohexameric minichromosome maintenance complex-the DNA replicative helicase comprising MCM2 to MCM73,4-that cause genomic instability render female mouse embryos markedly more susceptible than males to embryonic lethality. This bias was not attributable to X chromosome-inactivation defects, differential replication licensing or X versus Y chromosome size, but rather to 'maleness'-XX embryos could be rescued by transgene-mediated sex reversal or testosterone administration. The ability of exogenous or endogenous testosterone to protect embryos was related to its anti-inflammatory properties5. Ibuprofen, a non-steroidal anti-inflammatory drug, rescued female embryos that contained mutations in not only the Mcm genes but also the Fancm gene; similar to MCM mutants, Fancm mutant embryos have increased levels of genomic instability (measured as the number of cells with micronuclei) from compromised replication fork repair6. In addition, deficiency in the anti-inflammatory IL10 receptor was synthetically lethal with the Mcm4Chaos3 helicase mutant. Our experiments indicate that, during development, DNA damage associated with DNA replication induces inflammation that is preferentially lethal to female embryos, because male embryos are protected by high levels of intrinsic testosterone.


Subject(s)
Embryo Loss/genetics , Genomic Instability/genetics , Inflammation/genetics , Minichromosome Maintenance Proteins/genetics , Mutation , Sex Characteristics , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Proliferation , DNA Damage , DNA Helicases/genetics , DNA Replication , Embryo Loss/pathology , Embryo Loss/prevention & control , Embryonic Development/drug effects , Embryonic Development/genetics , Female , Ibuprofen/pharmacology , Inflammation/pathology , Inflammation/prevention & control , Male , Mice , Minichromosome Maintenance Complex Component 4/genetics , Minichromosome Maintenance Proteins/deficiency , Placenta/metabolism , Placenta/pathology , Pregnancy , Receptors, Interleukin-10/deficiency , Receptors, Interleukin-10/genetics , Synthetic Lethal Mutations , Testosterone/pharmacology
7.
Mol Cell ; 67(6): 1026-1036.e2, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28844861

ABSTRACT

Pairing and synapsis of homologous chromosomes during meiosis is crucial for producing genetically normal gametes and is dependent upon repair of SPO11-induced double-strand breaks (DSBs) by homologous recombination. To prevent transmission of genetic defects, diverse organisms have evolved mechanisms to eliminate meiocytes containing unrepaired DSBs or unsynapsed chromosomes. Here we show that the CHK2 (CHEK2)-dependent DNA damage checkpoint culls not only recombination-defective mouse oocytes but also SPO11-deficient oocytes that are severely defective in homolog synapsis. The checkpoint is triggered in oocytes that accumulate a threshold level of spontaneous DSBs (∼10) in late prophase I, the repair of which is inhibited by the presence of HORMAD1/2 on unsynapsed chromosome axes. Furthermore, Hormad2 deletion rescued the fertility of oocytes containing a synapsis-proficient, DSB repair-defective mutation in a gene (Trip13) required for removal of HORMADs from synapsed chromosomes, suggesting that many meiotic DSBs are normally repaired by intersister recombination in mice.


Subject(s)
Checkpoint Kinase 2/metabolism , Chromosome Pairing , DNA Damage , Meiosis , Oocytes/enzymology , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Death , Checkpoint Kinase 2/genetics , Endodeoxyribonucleases/deficiency , Endodeoxyribonucleases/genetics , Female , Fertility , Genotype , Infertility, Female/enzymology , Infertility, Female/genetics , Infertility, Female/pathology , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Oocytes/pathology , Pachytene Stage , Phenotype , Recombinational DNA Repair , Time Factors , Tissue Culture Techniques
8.
Cell ; 159(1): 216, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-28934603
9.
BMC Genomics ; 24(1): 641, 2023 Oct 26.
Article in English | MEDLINE | ID: mdl-37884859

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are important post-transcriptional gene regulators controlling cellular lineage specification and differentiation during embryonic development, including the gastrointestinal system. However, miRNA-mediated regulatory mechanisms involved in early embryonic development of human small intestine (SI) remains underexplored. To explore candidate roles for miRNAs in prenatal SI lineage specification in humans, we used a multi-omic analysis strategy in a directed differentiation model that programs human pluripotent stem cells toward the SI lineage. RESULTS: We leveraged small RNA-seq to define the changing miRNA landscape, and integrated chromatin run-on sequencing (ChRO-seq) and RNA-seq to define genes subject to significant post-transcriptional regulation across the different stages of differentiation. Small RNA-seq profiling revealed temporal dynamics of miRNA signatures across different developmental events of the model, including definitive endoderm formation, SI lineage specification and SI regional patterning. Our multi-omic, integrative analyses showed further that the elevation of miR-182 and reduction of miR-375 are key events during SI lineage specification. We demonstrated that loss of miR-182 leads to an increase in the foregut master marker SOX2. We also used single-cell analyses in murine adult intestinal crypts to support a life-long role for miR-375 in the regulation of Zfp36l2. Finally, we uncovered opposing roles of SMAD4 and WNT signaling in regulating miR-375 expression during SI lineage specification. Beyond the mechanisms highlighted in this study, we also present a web-based application for exploration of post-transcriptional regulation and miRNA-mediated control in the context of early human SI development. CONCLUSION: The present study uncovers a novel facet of miRNAs in regulating prenatal SI development. We leveraged multi-omic, systems biology approaches to discover candidate miRNA regulators associated with early SI developmental events in a human organoid model. In this study, we highlighted miRNA-mediated post-transcriptional regulation relevant to the event of SI lineage specification. The candidate miRNA regulators that we identified for the other stages of SI development also warrant detailed characterization in the future.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs , Humans , Animals , Mice , Cell Differentiation/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Intestine, Small/metabolism , Organoids/metabolism
10.
PLoS Biol ; 18(10): e3000903, 2020 10.
Article in English | MEDLINE | ID: mdl-33075054

ABSTRACT

Genetic diversity in offspring is induced by meiotic recombination, which is initiated between homologs at >200 sites originating from meiotic double-strand breaks (DSBs). Of this initial pool, only 1-2 DSBs per homolog pair will be designated to form meiotic crossovers (COs), where reciprocal genetic exchange occurs between parental chromosomes. Cyclin-dependent kinase 2 (CDK2) is known to localize to so-called "late recombination nodules" (LRNs) marking incipient CO sites. However, the role of CDK2 kinase activity in the process of CO formation remains uncertain. Here, we describe the phenotype of 2 Cdk2 point mutants with elevated or decreased activity, respectively. Elevated CDK2 activity was associated with increased numbers of LRN-associated proteins, including CDK2 itself and the MutL homolog 1 (MLH1) component of the MutLγ complex, but did not lead to increased numbers of COs. In contrast, reduced CDK2 activity leads to the complete absence of CO formation during meiotic prophase I. Our data suggest an important role for CDK2 in regulating MLH1 focus numbers and that the activity of this kinase is a key regulatory factor in the formation of meiotic COs.


Subject(s)
Crossing Over, Genetic , Cyclin-Dependent Kinase 2/metabolism , Meiosis , Animals , Apoptosis , Cell Cycle Checkpoints , Cell Cycle Proteins/metabolism , Chromosome Pairing , DNA Repair , Histones/metabolism , Ligases/metabolism , Male , Meiotic Prophase I , Metaphase , Mice, Inbred C57BL , Mice, Transgenic , MutL Protein Homolog 1/metabolism , Pachytene Stage , Sex Chromosomes/genetics , Spermatozoa/cytology , Spermatozoa/metabolism , Synaptonemal Complex/metabolism , Telomere/metabolism
11.
J Microsc ; 291(3): 237-247, 2023 09.
Article in English | MEDLINE | ID: mdl-37413663

ABSTRACT

Lightsheet microscopy offers an ideal method for imaging of large (mm-cm scale) biological tissues rendered transparent via optical clearing protocols. However the diversity of clearing technologies and tissue types, and how these are adapted to the microscope can make tissue mounting complicated and somewhat irreproducible. Tissue preparation for imaging can involve glues and or equilibration in a variety of expensive and/or proprietary formulations. Here we present practical advice for mounting and capping cleared tissues in optical cuvettes for macroscopic imaging, providing a standardised 3D cell that can be imaged routinely and relatively inexpensively. We show that acrylic cuvettes cause minimal spherical aberration with objective numerical apertures less than 0.65. Furthermore, we describe methods for aligning and assessing the light sheets, discriminating fluorescence from autofluorescence, identifying chromatic artefacts due to differential scattering and removing streak artefacts such that they do not confound downstream 3D object segmentation analyses, with mouse embryo, liver and heart imaging as demonstrated examples.


Subject(s)
Histological Techniques , Microscopy , Mice , Animals , Imaging, Three-Dimensional/methods
12.
Proc Natl Acad Sci U S A ; 117(24): 13680-13688, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32493750

ABSTRACT

Sex determination in mammals is governed by antagonistic interactions of two genetic pathways, imbalance in which may lead to disorders/differences of sex development (DSD) in human. Among 46,XX individuals with testicular DSD (TDSD) or ovotesticular DSD (OTDSD), testicular tissue is present in the gonad. Although the testis-determining gene SRY is present in many cases, the etiology is unknown in most SRY-negative patients. We performed exome sequencing on 78 individuals with 46,XX TDSD/OTDSD of unknown genetic etiology and identified seven (8.97%) with heterozygous variants affecting the fourth zinc finger (ZF4) of Wilms' tumor 1 (WT1) (p.Ser478Thrfs*17, p.Pro481Leufs*15, p.Lys491Glu, p.Arg495Gln [x3], p.Arg495Gly). The variants were de novo in six families (P = 4.4 × 10-6), and the incidence of WT1 variants in 46,XX DSD is enriched compared to control populations (P < 1.8 × 10-4). The introduction of ZF4 mutants into a human granulosa cell line resulted in up-regulation of endogenous Sertoli cell transcripts and Wt1Arg495Gly/Arg495Gly XX mice display masculinization of the fetal gonads. The phenotype could be explained by the ability of the mutated proteins to physically interact with and sequester a key pro-ovary factor ß-CATENIN, which may lead to up-regulation of testis-specific pathway. Our data show that unlike previous association of WT1 and 46,XY DSD, ZF4 variants of WT1 are a relatively common cause of 46,XX TDSD/OTDSD. This expands the spectrum of phenotypes associated with WT1 variants and shows that the WT1 protein affecting ZF4 can function as a protestis factor in an XX chromosomal context.


Subject(s)
46, XX Testicular Disorders of Sex Development/metabolism , Testis/metabolism , WT1 Proteins/metabolism , 46, XX Testicular Disorders of Sex Development/genetics , 46, XX Testicular Disorders of Sex Development/pathology , Animals , Child, Preschool , Female , Humans , Infant , Male , Mice , Ovary/growth & development , Ovary/metabolism , Testis/growth & development , Testis/pathology , WT1 Proteins/chemistry , WT1 Proteins/genetics , Zinc Fingers , beta Catenin/genetics , beta Catenin/metabolism
13.
Hum Mol Genet ; 29(20): 3402-3411, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33075816

ABSTRACT

Approximately 7% of men worldwide suffer from infertility, with sperm abnormalities being the most common defect. Though genetic causes are thought to underlie a substantial fraction of idiopathic cases, the actual molecular bases are usually undetermined. Because the consequences of most genetic variants in populations are unknown, this complicates genetic diagnosis even after genome sequencing of patients. Some patients with ciliopathies, including primary ciliary dyskinesia and Bardet-Biedl syndrome, also suffer from infertility because cilia and sperm flagella share several characteristics. Here, we identified two deleterious alleles of RABL2A, a gene essential for normal function of cilia and flagella. Our in silico predictions and in vitro assays suggest that both alleles destabilize the protein. We constructed and analyzed mice homozygous for these two single-nucleotide polymorphisms, Rabl2L119F (rs80006029) and Rabl2V158F (rs200121688), and found that they exhibit ciliopathy-associated disorders including male infertility, early growth retardation, excessive weight gain in adulthood, heterotaxia, pre-axial polydactyly, neural tube defects and hydrocephalus. Our study provides a paradigm for triaging candidate infertility variants in the population for in vivo functional validation, using computational, in vitro and in vivo approaches.


Subject(s)
Ciliopathies/etiology , Infertility, Male/etiology , Polymorphism, Single Nucleotide , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/physiology , Animals , Ciliopathies/pathology , Female , Humans , Infertility, Male/pathology , Male , Mice , Phenotype
14.
Development ; 146(21)2019 11 06.
Article in English | MEDLINE | ID: mdl-31582414

ABSTRACT

The ability of men to remain fertile throughout their lives depends upon establishment of a spermatogonial stem cell (SSC) pool from gonocyte progenitors, and thereafter balancing SSC renewal versus terminal differentiation. Here, we report that precise regulation of the cell cycle is crucial for this balance. Whereas cyclin-dependent kinase 2 (Cdk2) is not necessary for mouse viability or gametogenesis stages prior to meiotic prophase I, mice bearing a deregulated allele (Cdk2Y15S ) are severely deficient in spermatogonial differentiation. This allele disrupts an inhibitory phosphorylation site (Tyr15) for the kinase WEE1. Remarkably, Cdk2Y15S/Y15S mice possess abnormal clusters of mitotically active SSC-like cells, but these are eventually removed by apoptosis after failing to differentiate properly. Analyses of lineage markers, germ cell proliferation over time, and single cell RNA-seq data revealed delayed and defective differentiation of gonocytes into SSCs. Biochemical and genetic data demonstrated that Cdk2Y15S is a gain-of-function allele causing elevated kinase activity, which underlies these differentiation defects. Our results demonstrate that precise regulation of CDK2 kinase activity in male germ cell development is crucial for the gonocyte-to-spermatogonia transition and long-term spermatogenic homeostasis.


Subject(s)
Cell Differentiation , Cell Lineage , Cyclin-Dependent Kinase 2/metabolism , Germ Cells/enzymology , Spermatogonia/cytology , Alleles , Animals , Apoptosis , CRISPR-Cas Systems , Cell Proliferation , Cluster Analysis , Crosses, Genetic , Germ Cells/cytology , Heterozygote , Homeostasis , Male , Mass Spectrometry , Meiosis , Mice , Mutagenesis, Site-Directed , Phenotype , Phosphorylation , RNA, Small Cytoplasmic/metabolism , Seminiferous Tubules/metabolism , Spermatogenesis , Spermatogonia/metabolism , Testis/metabolism , Transcriptome
15.
Chromosoma ; 129(1): 69-82, 2020 03.
Article in English | MEDLINE | ID: mdl-31940063

ABSTRACT

Long transgenes are often used in mammalian genetics, e.g., to rescue mutations in large genes. In the course of experiments addressing the genetic basis of hybrid sterility caused by meiotic defects in mice bearing different alleles of Prdm9, we discovered that introduction of copy-number variation (CNV) via two independent insertions of long transgenes containing incomplete Prdm9 decreased testicular weight and epididymal sperm count. Transgenic animals displayed increased occurrence of seminiferous tubules with apoptotic cells at 18 days postpartum (dpp) corresponding to late meiotic prophase I, but not at 21 dpp. We hypothesized that long transgene insertions could cause asynapsis, but the immunocytochemical data revealed that the adult transgenic testes carried a similar percentage of asynaptic pachytene spermatocytes as the controls. These transgenic spermatocytes displayed less crossovers but similar numbers of unrepaired meiotic breaks. Despite slightly increased frequency of metaphase I spermatocytes with univalent chromosome(s) and reduced numbers of metaphase II spermatocytes, cytological studies did not reveal increased apoptosis in tubules containing the metaphase spermatocytes, but found an increased percentage of tubules carrying apoptotic spermatids. Sperm counts of subfertile animals inversely correlated with the transcription levels of the Psmb1 gene encoded within these two transgenes. The effect of the transgenes was dependent on sex and genetic background. Our results imply that the fertility of transgenic hybrid animals is not compromised by the impaired meiotic synapsis of homologous chromosomes, but can be negatively influenced by the increased expression of the introduced genes.


Subject(s)
Cell Cycle Checkpoints/genetics , DNA Copy Number Variations , Fertility/genetics , Pachytene Stage/genetics , Transgenes , Animals , Apoptosis/genetics , DNA Breaks, Double-Stranded , Female , Genetic Background , Male , Mice , Organ Size , Sperm Count , Spermatocytes/metabolism , Testis/anatomy & histology , Testis/metabolism
16.
Biol Reprod ; 104(1): 8-10, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33057575

ABSTRACT

The laboratory mouse is the most widely used animal model for studying the genetics and biology of mammalian development and reproduction. Embryonic stem cell (ESC) gene targeting technology, and the sophisticated genomic manipulations it allowed, was unique to this organism for a long period of time; this was a major factor in the mouse's rise to pre-eminence as a model system over the past three decades or so. The recent advent of CRISPR/Cas9 technology has democratized the application of genome editing to essentially all organisms. Nevertheless, the scientific infrastructure behind the mouse still makes it the organism of choice for studying molecular mechanisms of mammalian development, and for modeling human development and disease.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Gene Targeting , Genes, Lethal , Animals , Mice , Reproduction
17.
Mol Cell ; 50(1): 67-81, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23523368

ABSTRACT

Animal germ cells produce PIWI-interacting RNAs (piRNAs), small silencing RNAs that suppress transposons and enable gamete maturation. Mammalian transposon-silencing piRNAs accumulate early in spermatogenesis, whereas pachytene piRNAs are produced later during postnatal spermatogenesis and account for >95% of all piRNAs in the adult mouse testis. Mutants defective for pachytene piRNA pathway proteins fail to produce mature sperm, but neither the piRNA precursor transcripts nor the trigger for pachytene piRNA production is known. Here, we show that the transcription factor A-MYB initiates pachytene piRNA production. A-MYB drives transcription of both pachytene piRNA precursor RNAs and the mRNAs for core piRNA biogenesis factors including MIWI, the protein through which pachytene piRNAs function. A-MYB regulation of piRNA pathway proteins and piRNA genes creates a coherent feedforward loop that ensures the robust accumulation of pachytene piRNAs. This regulatory circuit, which can be detected in rooster testes, likely predates the divergence of birds and mammals.


Subject(s)
Meiosis , Proto-Oncogene Proteins c-myb/metabolism , RNA, Small Interfering/biosynthesis , Spermatogenesis , Testis/metabolism , Trans-Activators/metabolism , Animals , Argonaute Proteins/deficiency , Argonaute Proteins/genetics , Biological Evolution , Chickens , Endodeoxyribonucleases/deficiency , Endodeoxyribonucleases/genetics , Feedback, Physiological , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genotype , High-Throughput Nucleotide Sequencing , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Pachytene Stage , Phenotype , Proto-Oncogene Proteins c-myb/deficiency , Proto-Oncogene Proteins c-myb/genetics , Testis/growth & development , Trans-Activators/deficiency , Trans-Activators/genetics , Transcription, Genetic , Transcriptional Activation
18.
Genesis ; 58(8): e23368, 2020 08.
Article in English | MEDLINE | ID: mdl-32343484

ABSTRACT

Maintaining genome integrity in the germline is essential for survival and propagation of a species. In both mouse and human, germ cells originate during fetal development and are hypersensitive to both endogenous and exogenous DNA damaging agents. Currently, mechanistic understanding of how primordial germ cells respond to DNA damage is limited in part by the tools available to study these cells. We developed a mouse transgenic reporter strain expressing a 53BP1-mCherry fusion protein under the control of the Oct4ΔPE embryonic germ cell-specific promoter. This reporter binds sites of DNA double strand breaks (DSBs) on chromatin, forming foci. Using ionizing radiation as a DNA DSB-inducing agent, we show that the transgenic reporter expresses specifically in the embryonic germ cells of both sexes and forms DNA damage induced foci in both a dose- and time-dependent manner. The dynamic time-sensitive and dose-sensitive DNA damage detection ability of this transgenic reporter, in combination with its specific expression in embryonic germ cells, makes it a versatile and valuable tool for increasing our understanding of DNA damage responses in these unique cells.


Subject(s)
DNA Damage , Embryonic Germ Cells/metabolism , Genes, Reporter , Genetic Engineering/methods , Animals , Chromatin/metabolism , DNA Breaks, Double-Stranded , Female , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Protein Binding , Red Fluorescent Protein
19.
Hum Mol Genet ; 27(22): 3911-3918, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30085085

ABSTRACT

Whole-exome or whole-genome sequencing is becoming routine in clinical situations for identifying mutations underlying presumed genetic causes of disease including infertility. While this is a powerful approach for implicating polymorphisms or de novo mutations in genes plausibly related to the phenotype, a greater challenge is to definitively prove causality. This is a crucial requisite for treatment, especially for infertility, in which validation options are limited. In this study, we created a mouse model of a putative infertility allele, DMC1M200V. DMC1 encodes a RecA homolog essential for meiotic recombination and fertility in mice. This allele was originally implicated as being responsible for the sterility of a homozygous African woman, a conclusion supported by subsequent biochemical analyses of the mutant protein and by studies of yeast with the orthologous amino acid change. Here, we found that Dmc1M200V/M200V male and female mice are fully fertile and do not exhibit any gonadal abnormalities. Detailed immunocytological analysis of meiosis revealed no defects suggestive of compromised fertility. This study serves as a cautionary tale for making conclusions about consequences of genetic variants, especially with respect to infertility, and emphasizes the importance of conducting relevant biological assays for making accurate diagnoses in the era of genomic medicine.


Subject(s)
Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Infertility/genetics , Meiosis/genetics , Nuclear Proteins/genetics , Alleles , Animals , Disease Models, Animal , Female , Gonads/growth & development , Gonads/pathology , Humans , Infertility/physiopathology , Male , Mice , Mutation , Phosphate-Binding Proteins , Recombinases , Recombination, Genetic
20.
Reproduction ; 160(1): 53-64, 2020 07.
Article in English | MEDLINE | ID: mdl-32272448

ABSTRACT

Reduced fertility of male mouse hybrids relative to their parents, or hybrid sterility, is governed by the hybrid sterility 1 (Hst1) locus. Rescue experiments with transgenes carrying sequences within or near Hst1 manifested that Hst1 contains the gene encoding meiosis-specific histone methyltransferase PRDM9. The Prdm9 gene is responsible for partial meiotic arrest, testicular atrophy, and low sperm count in (C57BL/6J x PWD)F1 mouse hybrids. Here we report that these male hybrids suffer an additional reproductive disadvantage, decreased sperm quality, which is (i) further exacerbated by the introduction of long transgenes carrying sequences from Hst1 with incomplete Prdm9 into their genome and (ii) controlled by the Prdm9 dosage. These transgenic male hybrids displayed the features of severe oligoasthenoteratozoospermia (OAT), a human infertility syndrome characterized by a low number of spermatozoa with poor motility and morphological abnormalities. Analysis of spermiogenesis in these mice revealed acrosome detachment, aberrant elongation and condensation of the nucleus. As a result, the transgenic sperm had acrosome malformations, abnormal chromatin packaging, and fragmented DNA with elevated base oxidation, revealed by using multiple methods. Heterozygosity for one null Prdm9 allele improved meiotic progression and sperm quality of both non- and transgenic hybrids. Our results indicate that genomic analysis of OAT patients should include consideration of allelic variants in PRDM9, and our transgenic models can serve as tools to understand the diverse molecular processes that, when perturbed, can cause this disease.


Subject(s)
Gene Expression Regulation , Histone-Lysine N-Methyltransferase/physiology , Infertility, Male/pathology , Meiosis , Oligospermia/pathology , Sperm Motility , Animals , Infertility, Male/etiology , Infertility, Male/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligospermia/etiology , Oligospermia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL