Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Genes Dev ; 36(11-12): 752-763, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35738678

ABSTRACT

Self-renewal of spermatogonial stem cells is vital to lifelong production of male gametes and thus fertility. However, the underlying mechanisms remain enigmatic. Here, we show that DOT1L, the sole H3K79 methyltransferase, is required for spermatogonial stem cell self-renewal. Mice lacking DOT1L fail to maintain spermatogonial stem cells, characterized by a sequential loss of germ cells from spermatogonia to spermatids and ultimately a Sertoli cell only syndrome. Inhibition of DOT1L reduces the stem cell activity after transplantation. DOT1L promotes expression of the fate-determining HoxC transcription factors in spermatogonial stem cells. Furthermore, H3K79me2 accumulates at HoxC9 and HoxC10 genes. Our findings identify an essential function for DOT1L in adult stem cells and provide an epigenetic paradigm for regulation of spermatogonial stem cells.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Spermatogonia , Stem Cells , Animals , Cell Differentiation , Male , Mice , Spermatogonia/cytology , Spermatogonia/metabolism , Stem Cells/cytology , Stem Cells/metabolism
2.
Genes Dev ; 34(11-12): 745-750, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32381626

ABSTRACT

DNA methylation is a major silencing mechanism of transposable elements (TEs). Here we report that TEX15, a testis-specific protein, is required for TE silencing. TEX15 is expressed in embryonic germ cells and functions during genome-wide epigenetic reprogramming. The Tex15 mutant exhibits DNA hypomethylation in TEs at a level similar to Mili and Dnmt3c but not Miwi2 mutants. TEX15 is associated with MILI in testis. As loss of Tex15 causes TE desilencing with intact piRNA production, our results identify TEX15 as a new essential epigenetic regulator that may function as a nuclear effector of MILI to silence TEs by DNA methylation.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA Transposable Elements/genetics , Gene Silencing/physiology , Germ Cells/metabolism , Animals , DNA Methylation , Embryonic Germ Cells/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Developmental/genetics , Male , Mice , Mutation
3.
Development ; 150(9)2023 05 01.
Article in English | MEDLINE | ID: mdl-37082953

ABSTRACT

Histone modifications regulate chromatin remodeling and gene expression in development and diseases. DOT1L, the sole histone H3K79 methyltransferase, is essential for embryonic development. Here, we report that DOT1L regulates male fertility in mouse. DOT1L associates with MLLT10 in testis. DOT1L and MLLT10 localize to the sex chromatin in meiotic and post-meiotic germ cells in an inter-dependent manner. Loss of either DOT1L or MLLT10 leads to reduced testis weight, decreased sperm count and male subfertility. H3K79me2 is abundant in elongating spermatids, which undergo the dramatic histone-to-protamine transition. Both DOT1L and MLLT10 are essential for H3K79me2 modification in germ cells. Strikingly, histones are substantially retained in epididymal sperm from either DOT1L- or MLLT10-deficient mice. These results demonstrate that H3K79 methylation promotes histone replacement during spermiogenesis.


Subject(s)
Histones , Semen , Animals , Male , Mice , Fertility , Histone Methyltransferases/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Methylation , Methyltransferases/genetics , Semen/metabolism , Spermatogenesis/genetics , Transcription Factors/metabolism
4.
PLoS Genet ; 19(5): e1010566, 2023 05.
Article in English | MEDLINE | ID: mdl-37126510

ABSTRACT

Transposable elements constitute nearly half of the mammalian genome and play important roles in genome evolution. While a multitude of both transcriptional and post-transcriptional mechanisms exist to silence transposable elements, control of transposition in vivo remains poorly understood. MOV10, an RNA helicase, is an inhibitor of mobilization of retrotransposons and retroviruses in cell culture assays. Here we report that MOV10 restricts LINE1 retrotransposition in mice. Although MOV10 is broadly expressed, its loss causes only incomplete penetrance of embryonic lethality, and the surviving MOV10-deficient mice are healthy and fertile. Biochemically, MOV10 forms a complex with UPF1, a key component of the nonsense-mediated mRNA decay pathway, and primarily binds to the 3' UTR of somatically expressed transcripts in testis. Consequently, loss of MOV10 results in an altered transcriptome in testis. Analyses using a LINE1 reporter transgene reveal that loss of MOV10 leads to increased LINE1 retrotransposition in somatic and reproductive tissues from both embryos and adult mice. Moreover, the degree of LINE1 retrotransposition inhibition is dependent on the Mov10 gene dosage. Furthermore, MOV10 deficiency reduces reproductive fitness over successive generations. Our findings demonstrate that MOV10 attenuates LINE1 retrotransposition in a dosage-dependent manner in mice.


Subject(s)
DNA Transposable Elements , RNA Helicases , Animals , Male , Mice , Nonsense Mediated mRNA Decay , Retroelements/genetics , RNA Helicases/genetics , RNA Helicases/metabolism
5.
Dev Biol ; 508: 46-63, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38242343

ABSTRACT

Male germ cells are connected by intercellular bridges (ICBs) in a syncytium due to incomplete cytokinesis. Syncytium is thought to be important for synchronized germ cell development by interchange of cytoplasmic factors via ICBs. Mammalian ADP-ribosylation factor 6 (ARF6) is a small GTPase that is involved in many cellular mechanisms including but not limited to regulating cellular structure, motility, vesicle trafficking and cytokinesis. ARF6 localizes to ICBs in spermatogonia and spermatocytes in mice. Here we report that mice with global depletion of ARF6 in adulthood using Ubc-CreERT2 display no observable phenotypes but are male sterile. ARF6-deficient males display a progressive loss of germ cells, including LIN28A-expressing spermatogonia, and ultimately develop Sertoli-cell-only syndrome. Specifically, intercellular bridges are lost in ARF6-deficient testis. Furthermore, germ cell-specific inactivation using the Ddx4-CreERT2 results in the same testicular morphological phenotype, showing the germ cell-intrinsic requirement of ARF6. Therefore, ARF6 is essential for spermatogenesis in mice and this function is conserved from Drosophila to mammals.


Subject(s)
ADP-Ribosylation Factor 6 , Spermatogenesis , Animals , Female , Male , Mice , Drosophila , Mammals , Spermatocytes , Spermatogenesis/genetics , Spermatogonia , Testis
6.
Development ; 149(2)2022 01 15.
Article in English | MEDLINE | ID: mdl-34919125

ABSTRACT

The CatSper cation channel is essential for sperm capacitation and male fertility. The multi-subunit CatSper complexes form highly organized calcium signaling nanodomains on flagellar membranes. Here, we report identification of an uncharacterized protein, C2CD6, as a subunit of the mouse CatSper complex. C2CD6 contains a calcium-dependent, membrane-targeting C2 domain. C2CD6 associates with the CatSper calcium-selective, core-forming subunits. Deficiency of C2CD6 depletes the CatSper nanodomains from the flagellum and results in male sterility. C2CD6-deficient sperm are defective in hyperactivation and fail to fertilize oocytes both in vitro and in vivo. CatSper currents are present but at a significantly lower level in C2CD6-deficient sperm. Transient treatments with either Ca2+ ionophore, starvation, or a combination of both restore the fertilization capacity of C2CD6-deficient sperm. C2CD6 interacts with EFCAB9, a pH-dependent calcium sensor in the CatSper complex. We postulate that C2CD6 facilitates incorporation of the CatSper complex into the flagellar plasma membrane and may function as a calcium sensor. The identification of C2CD6 may enable the long-sought reconstitution of the CatSper ion channel complex in a heterologous system for male contraceptive development.


Subject(s)
Calcium Channels , Infertility, Male , Sperm Tail , Animals , Female , Male , Mice , Action Potentials , Calcium/metabolism , Calcium Channels/metabolism , Calcium-Binding Proteins/metabolism , Infertility, Male/genetics , Mice, Inbred C57BL , Protein Multimerization , Protein Transport , Sperm Motility , Sperm Tail/metabolism , Sperm Tail/physiology
7.
PLoS Genet ; 18(9): e1010416, 2022 09.
Article in English | MEDLINE | ID: mdl-36129965

ABSTRACT

Control over gene expression is exerted, in multiple stages of spermatogenesis, at the post-transcriptional level by RNA binding proteins (RBPs). We identify here an essential role in mammalian spermatogenesis and male fertility for 'RNA binding protein 46' (RBM46). A highly evolutionarily conserved gene, Rbm46 is also essential for fertility in both flies and fish. We found Rbm46 expression was restricted to the mouse germline, detectable in males in the cytoplasm of premeiotic spermatogonia and meiotic spermatocytes. To define its requirement for spermatogenesis, we generated Rbm46 knockout (KO, Rbm46-/-) mice; although male Rbm46-/- mice were viable and appeared grossly normal, they were infertile. Testes from adult Rbm46-/- mice were small, with seminiferous tubules containing only Sertoli cells and few undifferentiated spermatogonia. Using genome-wide unbiased high throughput assays RNA-seq and 'enhanced crosslinking immunoprecipitation' coupled with RNA-seq (eCLIP-seq), we discovered RBM46 could bind, via a U-rich conserved consensus sequence, to a cohort of mRNAs encoding proteins required for completion of differentiation and subsequent meiotic initiation. In summary, our studies support an essential role for RBM46 in regulating target mRNAs during spermatogonia differentiation prior to the commitment to meiosis in mice.


Subject(s)
RNA-Binding Proteins/metabolism , Spermatogenesis , Spermatogonia , Animals , Cell Differentiation/genetics , Male , Mammals/genetics , Meiosis/genetics , Mice , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Spermatocytes/metabolism , Spermatogenesis/genetics , Spermatogonia/metabolism , Testis
8.
Reproduction ; 167(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38401263

ABSTRACT

In brief: The dissociation of HORMA domain protein 2 (HORMAD2) from the synaptonemal complex is tightly regulated. This study reveals that the N-terminal region of HORMAD2 is critical for its dissociation from synapsed meiotic chromosomes. Abstract: During meiosis, homologous chromosomes undergo synapsis and recombination. HORMA domain proteins regulate key processes in meiosis. Mammalian HORMAD1 and HORMAD2 localize to unsynapsed chromosome axes but are removed upon synapsis by the TRIP13 AAA+ ATPase. TRIP13 engages the N-terminal region of HORMA domain proteins to induce an open conformation, resulting in the disassembly of protein complexes. Here, we report introduction of a 3×FLAG-HA tag to the N-terminus of HORMAD2 in mice. Coimmunoprecipitation coupled with mass spectrometry identified HORMAD1 and SYCP2 as HORMAD2-associated proteins in the testis. Unexpectedly, the N-terminal tagging of HORMAD2 resulted in its abnormal persistence along synapsed regions in pachynema and ectopic localization to telomeres in diplonema. Super-resolution microscopy revealed that 3×FLAG-HA-HORMAD2 was distributed along the central region of the synaptonemal complex, whereas wild-type HORMAD1 persisted along the lateral elements in 3×FLAG-HA-HORMAD2 meiocytes. Although homozygous mice completed meiosis and were fertile, homozygous males exhibited a significant reduction in sperm count. Collectively, these results suggest that the N-terminus of HORMAD2 is important for its timely removal from meiotic chromosome axes.


Subject(s)
Cell Cycle Proteins , Semen , Animals , Male , Mice , Cell Cycle Proteins/metabolism , Chromosome Pairing , Mammals/genetics , Meiosis , Meiotic Prophase I , Semen/metabolism , Synaptonemal Complex/metabolism
9.
Nucleic Acids Res ; 50(9): 5129-5144, 2022 05 20.
Article in English | MEDLINE | ID: mdl-35489071

ABSTRACT

Homeostasis of meiotic DNA double strand breaks (DSB) is critical for germline genome integrity and homologous recombination. Here we demonstrate an essential role for SKP1, a constitutive subunit of the SCF (SKP1-Cullin-F-box) ubiquitin E3 ligase, in early meiotic processes. SKP1 restrains accumulation of HORMAD1 and the pre-DSB complex (IHO1-REC114-MEI4) on the chromosome axis in meiotic germ cells. Loss of SKP1 prior to meiosis leads to aberrant localization of DSB repair proteins and a failure in synapsis initiation in meiosis of both males and females. Furthermore, SKP1 is crucial for sister chromatid cohesion during the pre-meiotic S-phase. Mechanistically, FBXO47, a meiosis-specific F-box protein, interacts with SKP1 and HORMAD1 and targets HORMAD1 for polyubiquitination and degradation in HEK293T cells. Our results support a model wherein the SCF ubiquitin E3 ligase prevents hyperactive DSB formation through proteasome-mediated degradation of HORMAD1 and subsequent modulation of the pre-DSB complex during meiosis.


Subject(s)
DNA Breaks, Double-Stranded , SKP Cullin F-Box Protein Ligases , Cell Cycle Proteins/metabolism , DNA , Female , HEK293 Cells , Homologous Recombination , Humans , Male , Meiosis/genetics , SKP Cullin F-Box Protein Ligases/genetics , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/genetics
10.
PLoS Genet ; 17(2): e1009265, 2021 02.
Article in English | MEDLINE | ID: mdl-33635934

ABSTRACT

Piwi-interacting RNAs (piRNAs) play critical roles in protecting germline genome integrity and promoting normal spermiogenic differentiation. In mammals, there are two populations of piRNAs: pre-pachytene and pachytene. Transposon-rich pre-pachytene piRNAs are expressed in fetal and perinatal germ cells and are required for retrotransposon silencing, whereas transposon-poor pachytene piRNAs are expressed in spermatocytes and round spermatids and regulate mRNA transcript levels. MOV10L1, a germ cell-specific RNA helicase, is essential for the production of both populations of piRNAs. Although the requirement of the RNA helicase domain located in the MOV10L1 C-terminal region for piRNA biogenesis is well known, its large N-terminal region remains mysterious. Here we report a novel Mov10l1 mutation, named yama, in the Mov10l1 N-terminal region. The yama mutation results in a single amino acid substitution V229E. The yama mutation causes meiotic arrest, de-repression of transposable elements, and male sterility because of defects in pre-pachytene piRNA biogenesis. Moreover, restricting the Mov10l1 mutation effects to later stages in germ cell development by combining with a postnatal conditional deletion of a complementing wild-type allele causes absence of pachytene piRNAs, accumulation of piRNA precursors, polar conglomeration of piRNA pathway proteins in spermatocytes, and spermiogenic arrest. Mechanistically, the V229E substitution in MOV10L1 reduces its interaction with PLD6, an endonuclease that generates the 5' ends of piRNA intermediates. Our results uncover an important role for the MOV10L1-PLD6 interaction in piRNA biogenesis throughout male germ cell development.


Subject(s)
Infertility, Male/genetics , Meiosis/genetics , Mitochondrial Proteins/metabolism , Phospholipase D/metabolism , RNA Helicases/metabolism , RNA, Small Interfering/metabolism , Retroelements/genetics , Spermatogenesis/genetics , Alleles , Animals , Gene Silencing , Germ Cells/metabolism , Germ Cells/pathology , HEK293 Cells , Humans , Male , Mice , Mitochondrial Proteins/genetics , Mutation , Pachytene Stage/genetics , Phospholipase D/genetics , RNA Helicases/genetics , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Spermatids/metabolism , Spermatocytes/metabolism , Testis/metabolism
11.
Development ; 147(8)2020 04 12.
Article in English | MEDLINE | ID: mdl-32188631

ABSTRACT

Continual spermatogenesis relies on the actions of an undifferentiated spermatogonial population that is composed of stem cells and progenitors. Here, using mouse models, we explored the role of RNA-binding proteins (RBPs) in regulation of the biological activities of this population. Proteins bound to polyadenylated RNAs in primary cultures of undifferentiated spermatogonia were captured with oligo (dT)-conjugated beads after UV-crosslinking and profiled by proteomics (termed mRBPome capture), yielding a putative repertoire of 473 RBPs. From this database, the RBP TRIM71 was identified and found to be expressed by stem and progenitor spermatogonia in prepubertal and adult mouse testes. Tissue-specific deletion of TRIM71 in the male germline led to reduction of the undifferentiated spermatogonial population and a block in transition to the differentiating state. Collectively, these findings demonstrate a key role of the RBP system in regulation of the spermatogenic lineage and may provide clues about the influence of RBPs on the biology of progenitor cell populations in other lineages.


Subject(s)
Proteome/metabolism , RNA-Binding Proteins/metabolism , Spermatogonia/cytology , Transcription Factors/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Gene Expression Regulation, Developmental , Male , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Testis/cytology , Up-Regulation/genetics
12.
Nucleic Acids Res ; 49(9): 5106-5123, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33939832

ABSTRACT

The transition from meiotic spermatocytes to postmeiotic haploid germ cells constitutes an essential step in spermatogenesis. The epigenomic regulatory mechanisms underlying this transition remain unclear. Here, we find a prominent transcriptomic switch from the late spermatocytes to the early round spermatids during the meiotic-to-postmeiotic transition, which is associated with robust histone acetylation changes across the genome. Among histone deacetylases (HDACs) and acetyltransferases, we find that HDAC3 is selectively expressed in the late meiotic and early haploid stages. Three independent mouse lines with the testis-specific knockout of HDAC3 show infertility and defects in meiotic exit with an arrest at the late stage of meiosis or early stage of round spermatids. Stage-specific RNA-seq and histone acetylation ChIP-seq analyses reveal that HDAC3 represses meiotic/spermatogonial genes and activates postmeiotic haploid gene programs during meiotic exit, with associated histone acetylation alterations. Unexpectedly, abolishing HDAC3 catalytic activity by missense mutations in the nuclear receptor corepressor (NCOR or SMRT) does not cause infertility, despite causing histone hyperacetylation as HDAC3 knockout, demonstrating that HDAC3 enzyme activity is not required for spermatogenesis. Motif analysis of the HDAC3 cistrome in the testes identified SOX30, which has a similar spatiotemporal expression pattern as HDAC3 during spermatogenesis. Depletion of SOX30 in the testes abolishes the genomic recruitment of the HDAC3 to the binding sites. Collectively, these results establish the SOX30/HDAC3 signaling as a key regulator of the transcriptional program in a deacetylase-independent manner during the meiotic-to-postmeiotic transition in spermatogenesis.


Subject(s)
Fertility/genetics , Gene Expression Regulation , Histone Deacetylases/physiology , Meiosis/genetics , Spermatogenesis/genetics , Transcriptional Activation , Acetylation , Animals , Cellular Reprogramming/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , SOX Transcription Factors/metabolism , Spermatids/cytology , Spermatids/metabolism , Testis/metabolism
13.
Genes Dev ; 29(6): 617-29, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25762440

ABSTRACT

Piwi-piRNA (Piwi-interacting RNA) ribonucleoproteins (piRNPs) enforce retrotransposon silencing, a function critical for preserving the genome integrity of germ cells. The molecular functions of most of the factors that have been genetically implicated in primary piRNA biogenesis are still elusive. Here we show that MOV10L1 exhibits 5'-to-3' directional RNA-unwinding activity in vitro and that a point mutation that abolishes this activity causes a failure in primary piRNA biogenesis in vivo. We demonstrate that MOV10L1 selectively binds piRNA precursor transcripts and is essential for the generation of intermediate piRNA processing fragments that are subsequently loaded to Piwi proteins. Multiple analyses suggest an intimate coupling of piRNA precursor processing with elements of local secondary structures such as G quadruplexes. Our results support a model in which MOV10L1 RNA helicase activity promotes unwinding and funneling of the single-stranded piRNA precursor transcripts to the endonuclease that catalyzes the first cleavage step of piRNA processing.


Subject(s)
RNA Helicases/metabolism , RNA, Small Interfering/metabolism , Animals , Female , G-Quadruplexes , Male , Mice , Protein Binding , Protein Structure, Secondary , RNA Helicases/chemistry , RNA Helicases/genetics , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA, Small Interfering/biosynthesis , Ribonucleoproteins/metabolism
14.
Biol Reprod ; 107(1): 157-167, 2022 07 25.
Article in English | MEDLINE | ID: mdl-35554494

ABSTRACT

Although hundreds of knockout mice show infertility as a major phenotype, the causative genic mutations of male infertility in humans remain rather limited. Here, we report the identification of a missense mutation (D136G) in the X-linked TAF7L gene as a potential cause of oligozoospermia in men. The human aspartate (D136) is evolutionally conserved across species, and its change to glycine (G) is predicted to be detrimental. Genetic complementation experiments in budding yeast demonstrate that the conserved aspartate or its analogous asparagine (N) residue in yeast TAF7 is essential for cell viability and thus its mutation to G is lethal. Although the corresponding D144G substitution in the mouse Taf7l gene does not affect male fertility, RNA-seq analyses reveal alterations in transcriptomic profiles in the Taf7l (D144G) mutant testes. These results support TAF7L mutation as a risk factor for oligozoospermia in humans.


Subject(s)
Infertility, Male , Oligospermia , TATA-Binding Protein Associated Factors , Transcription Factor TFIID , Animals , Aspartic Acid , Genes, X-Linked/genetics , Humans , Infertility, Male/genetics , Male , Mice , Mutation , Mutation, Missense , Oligospermia/genetics , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics
15.
Nucleic Acids Res ; 48(21): 12219-12233, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33166385

ABSTRACT

Meiotic recombination enables reciprocal exchange of genetic information between parental chromosomes and is essential for fertility. MEIOB, a meiosis-specific ssDNA-binding protein, regulates early meiotic recombination. Here we report that the human infertility-associated missense mutation (N64I) in MEIOB causes protein degradation and reduced crossover formation in mouse testes. Although the MEIOB N64I substitution is associated with human infertility, the point mutant mice are fertile despite meiotic defects. Meiob mutagenesis identifies serine 67 as a critical residue for MEIOB. Biochemically, these two mutations (N64I and S67 deletion) cause self-aggregation of MEIOB and sharply reduced protein half-life. Molecular genetic analyses of both point mutants reveal an important role for MEIOB in crossover formation in late meiotic recombination. Furthermore, we find that the MEIOB protein levels directly correlate with the severity of meiotic defects. Our results demonstrate that MEIOB regulates meiotic recombination in a dosage-dependent manner.


Subject(s)
DNA, Single-Stranded/genetics , DNA-Binding Proteins/genetics , Mutation, Missense , Testis/metabolism , Animals , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems , Chromosome Pairing , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Female , Gene Dosage , Gene Editing , HEK293 Cells , Homologous Recombination , Humans , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , Male , Mice , Mice, Transgenic , Ovary/metabolism , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Species Specificity , Testis/pathology
16.
PLoS Genet ; 15(2): e1007952, 2019 02.
Article in English | MEDLINE | ID: mdl-30716097

ABSTRACT

Meiotic recombination permits exchange of genetic material between homologous chromosomes. The replication protein A (RPA) complex, the predominant ssDNA-binding complex, is required for nearly all aspects of DNA metabolism, but its role in mammalian meiotic recombination remains unknown due to the embryonic lethality of RPA mutant mice. RPA is a heterotrimer of RPA1, RPA2, and RPA3. We find that loss of RPA1, the largest subunit, leads to disappearance of RPA2 and RPA3, resulting in the absence of the RPA complex. Using an inducible germline-specific inactivation strategy, we find that loss of RPA completely abrogates loading of RAD51/DMC1 recombinases to programmed meiotic DNA double strand breaks, thus blocking strand invasion required for chromosome pairing and synapsis. Surprisingly, loading of MEIOB, SPATA22, and ATR to DNA double strand breaks is RPA-independent and does not promote RAD51/DMC1 recruitment in the absence of RPA. Finally, inactivation of RPA reduces crossover formation. Our results demonstrate that RPA plays two distinct roles in meiotic recombination: an essential role in recombinase recruitment at early stages and an important role in promoting crossover formation at later stages.


Subject(s)
Homologous Recombination , Meiosis/genetics , Replication Protein A/metabolism , Animals , Cell Cycle Proteins/metabolism , Chromosome Pairing , Crossing Over, Genetic , DNA Breaks, Double-Stranded , DNA Replication , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Nuclear Proteins/metabolism , Phosphate-Binding Proteins , Protein Stability , Rad51 Recombinase/deficiency , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Replication Protein A/deficiency , Replication Protein A/genetics , Spermatocytes/cytology , Spermatocytes/metabolism
17.
PLoS Genet ; 14(5): e1007412, 2018 05.
Article in English | MEDLINE | ID: mdl-29799838

ABSTRACT

The N6-methyladenosine (m6A) modification is the most prevalent internal RNA modification in eukaryotes. The majority of m6A sites are found in the last exon and 3' UTRs. Here we show that the nuclear m6A reader YTHDC1 is essential for embryo viability and germline development in mouse. Specifically, YTHDC1 is required for spermatogonial development in males and for oocyte growth and maturation in females; Ythdc1-deficient oocytes are blocked at the primary follicle stage. Strikingly, loss of YTHDC1 leads to extensive alternative polyadenylation in oocytes, altering 3' UTR length. Furthermore, YTHDC1 deficiency causes massive alternative splicing defects in oocytes. The majority of splicing defects in mutant oocytes are rescued by introducing wild-type, but not m6A-binding-deficient, YTHDC1. YTHDC1 is associated with the pre-mRNA 3' end processing factors CPSF6, SRSF3, and SRSF7. Thus, YTHDC1 plays a critical role in processing of pre-mRNA transcripts in the oocyte nucleus and may have similar non-redundant roles throughout fetal development.


Subject(s)
Alternative Splicing/genetics , Mice/growth & development , Nerve Tissue Proteins/genetics , Oocytes/growth & development , Polyadenylation/genetics , RNA Splicing Factors/genetics , 3' Untranslated Regions/genetics , Adenosine/analogs & derivatives , Adenosine/metabolism , Animals , Cell Nucleus/metabolism , Cleavage And Polyadenylation Specificity Factor/metabolism , Embryonic Development/genetics , Exons/genetics , Female , Male , Mice/genetics , Mice, Transgenic , Mutation , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/metabolism , Oocytes/metabolism , RNA Precursors/genetics , RNA Splicing Factors/deficiency , RNA Splicing Factors/metabolism , RNA, Messenger/genetics , Serine-Arginine Splicing Factors/metabolism , Spermatogonia/growth & development , Spermatogonia/metabolism
18.
PLoS Genet ; 14(1): e1007175, 2018 01.
Article in English | MEDLINE | ID: mdl-29329290

ABSTRACT

The microrchidia (MORC) family proteins are chromatin-remodelling factors and function in diverse biological processes such as DNA damage response and transposon silencing. Here, we report that mouse Morc2b encodes a functional germ cell-specific member of the MORC protein family. Morc2b arose specifically in the rodent lineage through retrotransposition of Morc2a during evolution. Inactivation of Morc2b leads to meiotic arrest and sterility in both sexes. Morc2b-deficient spermatocytes and oocytes exhibit failures in chromosomal synapsis, blockades in meiotic recombination, and increased apoptosis. Loss of MORC2B causes mis-regulated expression of meiosis-specific genes. Furthermore, we find that MORC2B interacts with MORC2A, its sequence paralogue. Our results demonstrate that Morc2b, a relatively recent gene, has evolved an essential role in meiosis and fertility.


Subject(s)
Fertility/genetics , Meiosis/genetics , Transcription Factors/physiology , Animals , Chromosome Pairing/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/metabolism , Sequence Homology , Spermatocytes/metabolism , Transcription Factors/genetics
19.
Genesis ; 58(5): e23355, 2020 05.
Article in English | MEDLINE | ID: mdl-31990142

ABSTRACT

H1 histones bind to linker DNA. H1t (H1f6), a testis-specific linker histone variant, is present in pachytene spermatocytes and spermatids. The expression of H1t histone coincides with the acquisition of metaphase I competence in pachytene spermatocytes. Here we report the generation of H1t-GFP transgenic mice. The H1t-GFP (H1 histone testis-green fluorescence protein) fusion protein expression recapitulates the endogenous H1t expression pattern. This protein appears first in mid pachytene spermatocytes in stage V seminiferous tubules, persists in round spermatids and elongating spermatids, but is absent in elongated spermatids. The strong green fluorescence signal, due to the high abundance of H1t-GFP, is maintained in spermatocytes after induction towards metaphase I through treatment with okadaic acid. Therefore, H1t-GFP can be used as a visual marker for monitoring the progression of meiosis in vitro and in vivo, as well as fluorescence-activated cell sorting (FACS) sorting of germ cells.


Subject(s)
Histones/genetics , Spermatids/metabolism , Transgenes , Animals , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histones/metabolism , Male , Mice , Mice, Transgenic , Okadaic Acid/pharmacology , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Testis/drug effects , Testis/metabolism
20.
Biol Reprod ; 103(2): 333-342, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32463099

ABSTRACT

MEIOB and SPATA22 are meiosis-specific proteins, interact with each other, and are essential for meiotic recombination and fertility. Aspartic acid 383 (D383) in MEIOB is critical for its interaction with SPATA22 in biochemical studies. Here we report that genetic studies validate the requirement of D383 for the function of MEIOB in mice. The MeiobD383A/D383A mice display meiotic arrest due to depletion of both MEIOB and SPATA22 proteins in the testes. We developed a cell-based bimolecular fluorescence complementation (BiFC) assay, in which MEIOB and SPATA22 are fused to split YFP moieties and their co-expression in cultured cells leads to the MEIOB-SPATA22 dimerization and reconstitution of the fluorophore. As expected, the interaction-disrupting D383A substitution results in the absence of YFP fluorescence in the BiFC assay. A high-throughput screen of small molecule libraries identified candidate hit compounds at a rate of 0.7%. Isocotoin, a hit compound from the natural product library, inhibits the MEIOB-SPATA22 interaction and promotes their degradation in HEK293 cells in a dose-dependent manner. Therefore, the BiFC assay can be employed to screen for small molecule inhibitors that disrupt protein-protein interactions or promote degradation of meiosis-specific proteins.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Meiosis/physiology , Testis/metabolism , Animals , Fertility/physiology , HEK293 Cells , Humans , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL