Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nature ; 620(7976): 1047-1053, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37459895

ABSTRACT

Zygotic genome activation (ZGA) activates the quiescent genome to enable the maternal-to-zygotic transition1,2. However, the identity of transcription factors that underlie mammalian ZGA in vivo remains elusive. Here we show that OBOX, a PRD-like homeobox domain transcription factor family (OBOX1-OBOX8)3-5, are key regulators of mouse ZGA. Mice deficient for maternally transcribed Obox1/2/5/7 and zygotically expressed Obox3/4 had a two-cell to four-cell arrest, accompanied by impaired ZGA. The Obox knockout defects could be rescued by restoring either maternal and zygotic OBOX, which suggests that maternal and zygotic OBOX redundantly support embryonic development. Chromatin-binding analysis showed that Obox knockout preferentially affected OBOX-binding targets. Mechanistically, OBOX facilitated the 'preconfiguration' of RNA polymerase II, as the polymerase relocated from the initial one-cell binding targets to ZGA gene promoters and distal enhancers. Impaired polymerase II preconfiguration in Obox mutants was accompanied by defective ZGA and chromatin accessibility transition, as well as aberrant activation of one-cell polymerase II targets. Finally, ectopic expression of OBOX activated ZGA genes and MERVL repeats in mouse embryonic stem cells. These data thus demonstrate that OBOX regulates mouse ZGA and early embryogenesis.


Subject(s)
Embryonic Development , Gene Expression Regulation, Developmental , Genome , Homeodomain Proteins , Transcription Factors , Zygote , Animals , Mice , Chromatin/genetics , Chromatin/metabolism , Embryonic Development/genetics , Enhancer Elements, Genetic/genetics , Genome/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mouse Embryonic Stem Cells/metabolism , Mutation , Promoter Regions, Genetic/genetics , RNA Polymerase II/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism , Zygote/metabolism
2.
Genome Biol ; 24(1): 166, 2023 Jul 13.
Article in English | MEDLINE | ID: mdl-37443062

ABSTRACT

BACKGROUND: The oocyte-to-embryo transition (OET) converts terminally differentiated gametes into a totipotent embryo and is critically controlled by maternal mRNAs and proteins, while the genome is silent until zygotic genome activation. How the transcriptome, translatome, and proteome are coordinated during this critical developmental window remains poorly understood. RESULTS: Utilizing a highly sensitive and quantitative mass spectrometry approach, we obtain high-quality proteome data spanning seven mouse stages, from full-grown oocyte (FGO) to blastocyst, using 100 oocytes/embryos at each stage. Integrative analyses reveal distinct proteome reprogramming compared to that of the transcriptome or translatome. FGO to 8-cell proteomes are dominated by FGO-stockpiled proteins, while the transcriptome and translatome are more dynamic. FGO-originated proteins frequently persist to blastocyst while corresponding transcripts are already downregulated or decayed. Improved concordance between protein and translation or transcription is observed for genes starting translation upon meiotic resumption, as well as those transcribed and translated only in embryos. Concordance between protein and transcription/translation is also observed for proteins with short half-lives. We built a kinetic model that predicts protein dynamics by incorporating both initial protein abundance in FGOs and translation kinetics across developmental stages. CONCLUSIONS: Through integrative analyses of datasets generated by ultrasensitive methods, our study reveals that the proteome shows distinct dynamics compared to the translatome and transcriptome during mouse OET. We propose that the remarkably stable oocyte-originated proteome may help save resources to accommodate the demanding needs of growing embryos. This study will advance our understanding of mammalian OET and the fundamental principles governing gene expression.


Subject(s)
Proteome , Transcriptome , Animals , Mice , Proteome/metabolism , Embryo, Mammalian/metabolism , Blastocyst/metabolism , Oocytes/metabolism , Gene Expression Regulation, Developmental , Mammals/metabolism
3.
Nat Cell Biol ; 24(6): 968-980, 2022 06.
Article in English | MEDLINE | ID: mdl-35697785

ABSTRACT

In mammals, translational control plays critical roles during oocyte-to-embryo transition (OET) when transcription ceases. However, the underlying regulatory mechanisms remain challenging to study. Here, using low-input Ribo-seq (Ribo-lite), we investigated translational landscapes during OET using 30-150 mouse oocytes or embryos per stage. Ribo-lite can also accommodate single oocytes. Combining PAIso-seq to interrogate poly(A) tail lengths, we found a global switch of translatome that closely parallels changes of poly(A) tails upon meiotic resumption. Translation activation correlates with polyadenylation and is supported by polyadenylation signal proximal cytoplasmic polyadenylation elements (papCPEs) in 3' untranslated regions. By contrast, translation repression parallels global de-adenylation. The latter includes transcripts containing no CPEs or non-papCPEs, which encode many transcription regulators that are preferentially re-activated before zygotic genome activation. CCR4-NOT, the major de-adenylation complex, and its key adaptor protein BTG4 regulate translation downregulation often independent of RNA decay. BTG4 is not essential for global de-adenylation but is required for selective gene de-adenylation and production of very short-tailed transcripts. In sum, our data reveal intimate interplays among translation, RNA stability and poly(A) tail length regulation underlying mammalian OET.


Subject(s)
Embryonic Development , Oocytes , 3' Untranslated Regions/genetics , Animals , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Mammals/genetics , Mammals/metabolism , Mice , Oocytes/metabolism , Polyadenylation , Protein Biosynthesis , RNA, Messenger/genetics
4.
Nucleic Acids Res ; 50(10): 5599-5616, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35640597

ABSTRACT

Maternal-to-zygotic transition (MZT) is the first and key step in the control of animal development and intimately related to changes in chromatin structure and histone modifications. H2AK119ub1, an important epigenetic modification in regulating chromatin configuration and function, is primarily catalyzed by PRC1 and contributes to resistance to transcriptional reprogramming in mouse embryos. In this study, the genome-wide dynamic distribution of H2AK119ub1 during MZT in mice was investigated using chromosome immunoprecipitation and sequencing. The results indicated that H2AK119ub1 accumulated in fully grown oocytes and was enriched at the TSSs of maternal genes, but was promptly declined after meiotic resumption at genome-wide including the TSSs of early zygotic genes, by a previously unidentified mechanism. Genetic evidences indicated that ubiquitin-specific peptidase 16 (USP16) is the major deubiquitinase for H2AK119ub1 in mouse oocytes. Conditional knockout of Usp16 in oocytes did not impair their survival, growth, or meiotic maturation. However, oocytes lacking USP16 have defects when undergoing zygotic genome activation or gaining developmental competence after fertilization, potentially associated with high levels of maternal H2AK119ub1 deposition on the zygotic genomes. Taken together, H2AK119ub1 level is declined during oocyte maturation by an USP16-dependent mechanism, which ensures zygotic genome reprogramming and transcriptional activation of essential early zygotic genes.


Subject(s)
Histones , Lysine , Animals , Chromatin/genetics , Gene Expression Regulation, Developmental , Histones/genetics , Histones/metabolism , Lysine/metabolism , Mice , Oocytes/metabolism , Oogenesis/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Zygote
5.
Nat Metab ; 3(10): 1372-1384, 2021 10.
Article in English | MEDLINE | ID: mdl-34650276

ABSTRACT

During early mammalian embryogenesis, changes in cell growth and proliferation depend on strict genetic and metabolic instructions. However, our understanding of metabolic reprogramming and its influence on epigenetic regulation in early embryo development remains elusive. Here we show a comprehensive metabolomics profiling of key stages in mouse early development and the two-cell and blastocyst embryos, and we reconstructed the metabolic landscape through the transition from totipotency to pluripotency. Our integrated metabolomics and transcriptomics analysis shows that while two-cell embryos favour methionine, polyamine and glutathione metabolism and stay in a more reductive state, blastocyst embryos have higher metabolites related to the mitochondrial tricarboxylic acid cycle, and present a more oxidative state. Moreover, we identify a reciprocal relationship between α-ketoglutarate (α-KG) and the competitive inhibitor of α-KG-dependent dioxygenases, L-2-hydroxyglutarate (L-2-HG), where two-cell embryos inherited from oocytes and one-cell zygotes display higher L-2-HG, whereas blastocysts show higher α-KG. Lastly, increasing 2-HG availability impedes erasure of global histone methylation markers after fertilization. Together, our data demonstrate dynamic and interconnected metabolic, transcriptional and epigenetic network remodelling during early mouse embryo development.


Subject(s)
Embryo, Mammalian/metabolism , Embryonic Development/genetics , Animals , Metabolomics , Mice , Transcriptome
6.
Nucleic Acids Res ; 47(21): 11387-11402, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31598710

ABSTRACT

Zar1 was one of the earliest mammalian maternal-effect genes to be identified. Embryos derived from Zar1-null female mice are blocked before zygotic genome activation; however, the underlying mechanism remains unclear. By knocking out Zar1 and its homolog Zar2 in mice, we revealed a novel function of these genes in oocyte meiotic maturation. Zar1/2-deleted oocytes displayed delayed meiotic resumption and polar body-1 emission and a higher incidence of abnormal meiotic spindle formation and chromosome aneuploidy. The grown oocytes of Zar1/2-null mice contained decreased levels of many maternal mRNAs and displayed a reduced level of protein synthesis. Key maturation-associated changes failed to occur in the Zar1/2-null oocytes, including the translational activation of maternal mRNAs encoding the cell-cycle proteins cyclin B1 and WEE2, as well as maternal-to-zygotic transition (MZT) licensing factor BTG4. Consequently, maternal mRNA decay was impaired and MZT was abolished. ZAR1/2 bound mRNAs to regulate the translational activity of their 3'-UTRs and interacted with other oocyte proteins, including mRNA-stabilizing protein MSY2 and cytoplasmic lattice components. These results countered the traditional view that ZAR1 only functions after fertilization and highlight a previously unrecognized role of ZAR1/2 in regulating the maternal transcriptome and translational activation in maturing oocytes.


Subject(s)
Egg Proteins/physiology , Meiosis/genetics , Oocytes/physiology , Oogenesis/genetics , Protein Biosynthesis/genetics , Proteins/physiology , RNA, Messenger, Stored/genetics , Animals , Cells, Cultured , Egg Proteins/genetics , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Pregnancy , Proteins/genetics , Transcription Factors , Transcriptome/genetics
7.
Nat Commun ; 10(1): 3387, 2019 07 29.
Article in English | MEDLINE | ID: mdl-31358751

ABSTRACT

Spermatogenesis is tightly regulated by ubiquitination and proteasomal degradation, especially during spermiogenesis, in which histones are replaced by protamine. However, the functions of proteasomal activity in meiosis I and II remain elusive. Here, we show that PSMA8-associated proteasomes are essential for the degradation of meiotic proteins and the progression of meiosis I during spermatogenesis. PSMA8 is expressed in spermatocytes from the pachytene stage, and assembles a type of testis-specific core proteasome. Deletion of PSMA8 decreases the abundance of proteasome in testes. Meiotic proteins that are normally degraded at late prophase I, such as RAD51 and RPA1, remain stable in PSMA8-deleted spermatocytes. Moreover, PSMA8-null spermatocytes exhibit delayed M-phase entry and are finally arrested at this stage, resulting in male infertility. However, PSMA8 is neither expressed nor required for female meiotic progression. Thus, meiosis I progression in spermatogenesis, particularly entry into and exit from M-phase, requires the proteasomal activity of PSMA8-associated proteasomes.


Subject(s)
Meiotic Prophase I/genetics , Proteasome Endopeptidase Complex/genetics , Spermatogenesis/genetics , Testis/enzymology , Animals , Cell Division/genetics , Female , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Pachytene Stage/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Spermatocytes/enzymology , Spermatocytes/metabolism
8.
Sci Adv ; 5(1): eaau9780, 2019 01.
Article in English | MEDLINE | ID: mdl-30746471

ABSTRACT

Segregation of homologous chromosomes in meiosis I is tightly regulated by their physical links, or crossovers (COs), generated from DNA double-strand breaks (DSBs) through meiotic homologous recombination. In budding yeast, three ZMM (Zip1/2/3/4, Mer3, Msh4/5) proteins, Zip2, Zip4, and Spo16, form a "ZZS" complex, functioning to promote meiotic recombination via a DSB repair pathway. Here, we identified the mammalian ortholog of Spo16, termed SPO16, which interacts with the mammalian ortholog of Zip2 (SHOC1/MZIP2), and whose functions are evolutionarily conserved to promote the formation of COs. SPO16 localizes to the recombination nodules, as SHOC1 and TEX11 do. SPO16 is required for stabilization of SHOC1 and proper localization of other ZMM proteins. The DSBs formed in SPO16-deleted meiocytes were repaired without COs formation, although synapsis is less affected. Therefore, formation of SPO16-SHOC1 complex-associated recombination intermediates is a key step facilitating meiotic recombination that produces COs from yeast to mammals.


Subject(s)
Crossing Over, Genetic , DNA-Binding Proteins/metabolism , Meiotic Prophase I/physiology , Microtubule-Associated Proteins/metabolism , Animals , Cation Transport Proteins/metabolism , Chromosome Segregation/physiology , Chromosomes, Mammalian/genetics , DNA Breaks, Double-Stranded , Female , HeLa Cells , Humans , Male , Mice
9.
Cell Death Differ ; 26(7): 1251-1266, 2019 07.
Article in English | MEDLINE | ID: mdl-30283081

ABSTRACT

During mammalian oocyte growth, chromatin configuration transition from the nonsurrounded nucleolus (NSN) to surrounded nucleolus (SN) type plays a key role in the regulation of gene expression and acquisition of meiotic and developmental competence by the oocyte. Nonetheless, the mechanism underlying chromatin configuration maturation in oocytes is poorly understood. Here we show that nucleolar protein DCAF13 is an important component of the ribosomal RNA (rRNA)-processing complex and is essential for oocyte NSN-SN transition in mice. A conditional knockout of Dcaf13 in oocytes led to the arrest of oocyte development in the NSN configuration, follicular atresia, premature ovarian failure, and female sterility. The DCAF13 deficiency resulted in pre-rRNA accumulation in oocytes, whereas the total mRNA level was not altered. Further exploration showed that DCAF13 participated in the 18S rRNA processing in growing oocytes. The lack of 18S rRNA because of DCAF13 deletion caused a ribosome assembly disorder and then reduced global protein synthesis. DCAF13 interacted with a protein of the core box C/D ribonucleoprotein, fibrillarin, i.e., a factor of early pre-rRNA processing. When fibrillarin was knocked down in the oocytes from primary follicles, follicle development was inhibited as well, indicating that an rRNA processing defect in the oocyte indeed stunts chromatin configuration transition and follicle development. Taken together, these results elucidated the in vivo function of novel nucleolar protein DCAF13 in maintaining mammalian oogenesis.


Subject(s)
Oocytes/metabolism , Ovarian Follicle/metabolism , RNA Processing, Post-Transcriptional , RNA, Ribosomal/metabolism , RNA-Binding Proteins/metabolism , Animals , Cells, Cultured , Female , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Oocytes/cytology , Oocytes/growth & development , Ovarian Follicle/cytology , Ovarian Follicle/growth & development
10.
EMBO J ; 37(24)2018 12 14.
Article in English | MEDLINE | ID: mdl-30478191

ABSTRACT

Meiotic resumption-coupled degradation of maternal transcripts occurs during oocyte maturation in the absence of mRNA transcription. The CCR4-NOT complex has been identified as the main eukaryotic mRNA deadenylase. In vivo functional and mechanistic information regarding its multiple subunits remains insufficient. Cnot6l, one of four genes encoding CCR4-NOT catalytic subunits, is preferentially expressed in mouse oocytes. Genetic deletion of Cnot6l impaired deadenylation and degradation of a subset of maternal mRNAs during oocyte maturation. Overtranslation of these undegraded mRNAs caused microtubule-chromosome organization defects, which led to activation of spindle assembly checkpoint and meiotic cell cycle arrest at prometaphase. Consequently, Cnot6l-/- female mice were severely subfertile. The function of CNOT6L in maturing oocytes is mediated by RNA-binding protein ZFP36L2, not maternal-to-zygotic transition licensing factor BTG4, which interacts with catalytic subunits CNOT7 and CNOT8 of CCR4-NOT Thus, recruitment of different adaptors by different catalytic subunits ensures stage-specific degradation of maternal mRNAs by CCR4-NOT This study provides the first direct genetic evidence that CCR4-NOT-dependent and particularly CNOT6L-dependent decay of selective maternal mRNAs is a prerequisite for meiotic maturation of oocytes.


Subject(s)
Meiosis , Oocytes/metabolism , RNA Stability , RNA, Messenger/metabolism , Ribonucleases/metabolism , Animals , Exoribonucleases , Female , Gene Deletion , Mice , Mice, Knockout , Oocytes/cytology , Proteins/genetics , Proteins/metabolism , RNA, Messenger/genetics , Repressor Proteins , Ribonucleases/genetics , Tristetraprolin/genetics , Tristetraprolin/metabolism
11.
Cell Death Dis ; 9(12): 1144, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30451825

ABSTRACT

Global transcriptional activity increases as oocytes grow and is silenced in fully grown oocytes. Thus, the chromatin configuration varies during oocyte growth, but the molecular mechanisms regulating these changes remain to be clarified. Here, we studied a susceptibility gene of polycystic ovary syndrome (PCOS), RPS26, which is a ribosomal protein-encoding gene that is highly expressed in the ovary, but the functions of which remain unknown. Specific knockout of Rps26 in mouse oocytes resulted in retarded follicle development from pre-antral follicles to antral follicles, while the chromatin configurations of the oocytes were arrested at the transition from the non-surrounded nucleolus (NSN) to surrounded nucleolus (SN)-type. As a consequence, all oocytes died by postnatal day 84 resulting in premature ovarian failure (POF). Loss of Rps26 in oocytes led to decreased mRNA transcription and low levels of histone trimethylation on H3K4/H3K9 and DNA methylation at 5-cytosine, high levels of which are required for oocytes to transform from NSN to SN-type. Low protein levels of oocyte-derived growth differentiation factor 9, bone morphogenetic protein 15, and the oocyte-granulosa cell gap junction protein connexin 37 inhibited oocyte growth and retarded follicle development. The disruption of the phosphoinositide 3-kinase/protein kinase B/Forkhead box O-3a pathway contributed to oocyte death and follicle atresia. These results provide genetic clues for the clinical diagnosis of POF, especially in PCOS patients without treatment.


Subject(s)
Oocytes/metabolism , Ovarian Follicle/growth & development , Primary Ovarian Insufficiency/genetics , Ribosomal Proteins/genetics , Animals , Cell Nucleolus/genetics , DNA Methylation/genetics , Female , Humans , Mice , Mice, Knockout , Oocytes/growth & development , Oocytes/pathology , Oogenesis/genetics , Ovarian Follicle/metabolism , Ovarian Follicle/pathology , Primary Ovarian Insufficiency/pathology , Transcription, Genetic
12.
EMBO J ; 37(18)2018 09 14.
Article in English | MEDLINE | ID: mdl-30111536

ABSTRACT

Mammalian oocytes and zygotes have the unique ability to reprogram a somatic cell nucleus into a totipotent state. SUV39H1/2-mediated histone H3 lysine-9 trimethylation (H3K9me3) is a major barrier to efficient reprogramming. How SUV39H1/2 activities are regulated in early embryos and during generation of induced pluripotent stem cells (iPSCs) remains unclear. Since expression of the CRL4 E3 ubiquitin ligase in oocytes is crucial for female fertility, we analyzed putative CRL4 adaptors (DCAFs) and identified DCAF13 as a novel CRL4 adaptor that is essential for preimplantation embryonic development. Dcaf13 is expressed from eight-cell to morula stages in both murine and human embryos, and Dcaf13 knockout in mice causes preimplantation-stage mortality. Dcaf13 knockout embryos are arrested at the eight- to sixteen-cell stage before compaction, and this arrest is accompanied by high levels of H3K9me3. Mechanistically, CRL4-DCAF13 targets SUV39H1 for polyubiquitination and proteasomal degradation and therefore facilitates H3K9me3 removal and zygotic gene expression. Taken together, CRL4-DCAF13-mediated SUV39H1 degradation is an essential step for progressive genome reprogramming during preimplantation embryonic development.


Subject(s)
Blastocyst/metabolism , Embryonic Development , Gene Expression Regulation, Developmental , Induced Pluripotent Stem Cells/metabolism , Methyltransferases/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Animals , Blastocyst/cytology , Enzyme Stability , Histones/genetics , Histones/metabolism , Induced Pluripotent Stem Cells/cytology , Methyltransferases/genetics , Mice , Mice, Inbred ICR , Mice, Knockout , Oocytes/cytology , Oocytes/metabolism , Proteolysis , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitination/genetics
13.
Endocrinology ; 158(11): 3988-3999, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28938432

ABSTRACT

The balances of mitochondrial dynamic changes, mitochondrial morphology, and mitochondrial number are critical in cell metabolism. Once they are disturbed, disorders in these processes generally cause diseases or even death in animals. We performed large-scale genetic screenings in fruit flies and discovered the mitoguardin gene (Miga) that encodes for a mitochondrial outer membrane protein. To examine the physiological functions of its mammalian homologs Miga1 and Miga2, we generated Miga1 and Miga2 single- and double-knockout mouse strains and found that the knockout mice were viable, but the females were subfertile. The ovarian phenotypes of these mice suggested that the MIGA1/2 proteins play an important role in ovulation and ovarian steroidogenesis. In vivo and in vitro analyses of Miga1/2-knockout granulosa cells showed severe defects in luteinization and steroidogenesis and disordered mitochondrial morphology and function in response to gonadotropins. This is a report of genes involved in mitochondrial fusion and morphology-regulating mitochondrial functions during ovulation and luteinization. These results suggest a mechanism of gonadotropin-regulated ovarian endocrine functions and provide clues for therapeutic treatments of infertile females.


Subject(s)
Gonadal Hormones/metabolism , Membrane Proteins/physiology , Mitochondria/physiology , Mitochondrial Proteins/physiology , Ovary/metabolism , Ovulation/genetics , Animals , Cells, Cultured , Female , Gonadotropins/metabolism , Granulosa Cells/metabolism , Luteinization/genetics , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Ovulation/metabolism
14.
Mol Hum Reprod ; 23(10): 698-707, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28961951

ABSTRACT

STUDY QUESTION: What is the physiological function of Yes-associated protein-1 (Yap1), a susceptibility gene for polycystic ovary syndrome (PCOS), in ovarian granulosa cells (GCs)? SUMMARY ANSWER: Physiologically, steroid sex hormones stimulate follicle growth by activating YAP1; however, the preovulatory inhibition of YAP1 activity in GCs is a prerequisite of LH actions. WHAT IS KNOWN ALREADY: PCOS is a common gynecologic and endocrine disease with multiple short and long-term consequences. Many PCOS patients suffer anovulation caused by hyperandrogenism, but its etiology remains unclear. STUDY DESIGN, SIZE, DURATION: To study the effect of acute hyperandrogenism on ovulation, we injected pregnant mare serum gonadotrophin (PMSG)-primed (44 h) pubertal mice with dihydrotestosterone (DHT), the major biologically active form of androgen, in a superovulation assay. We investigated if YAP1 is regulated by testosterone and if it is potentially involved in follicle development and ovulation. Cultured primary GCs were subjected to Yap1 depletion by RNA interference and Yap1 overexpression by adenoviral infections. PARTICIPANTS/MATERIALS, SETTING, METHODS: Female mice at postnatal day (PD)-21~23 were analyzed to avoid the complexity of ovarian functions associated with estrous cycles and endogenous surges of gonadotropins. Immature mice were injected intraperitoneally with five IU PMSG to stimulate preovulatory follicle development followed 44 h later with five IU hCG to stimulate ovulation. For DHT treatments, female mice at PD23 were injected intraperitoneally with five IU PMSG followed 44 h later with five IU hCG alone (as control) or five IU hCG plus 100 µg DHT, which was dissolved in 0.1 ml DMSO. Methods of gene expression detection used include immunohistochemistry, immunofluorescence, Western blotting and quantitative PCR. More than three biological and technical replicates were included in each experiments. MAIN RESULTS AND THE ROLE OF CHANCE: we provide novel evidence in a mouse model that YAP1 is required for proliferation of ovarian GCs, but is down-regulated by LH through the extracellular-regulated kinase-1/2 (ERK1/2) cascade. Acute hyperandrogenism blocks LH actions and causes oligo-ovulation by activating YAP1. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: Results shown were obtained only in mouse, and need to be further confirmed in human samples. WIDER IMPLICATIONS OF THE FINDINGS: These findings not only elucidated the role of YAP1 in maintaining normal ovarian functions, but also link the YAP1 deregulation to the pathogenesis of PCOS. STUDY FUNDING AND COMPETING INTEREST(S): This study is funded by the National Key Research and Development Program of China (2016YFC1000600 and 2017YFSF1001500) and National Natural Science Foundation of China (31528016, 31371449 and 31671558). The authors have no competing interests.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Chorionic Gonadotropin/pharmacology , Dihydrotestosterone/pharmacology , Granulosa Cells/drug effects , Hyperandrogenism/genetics , Phosphoproteins/genetics , Polycystic Ovary Syndrome/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Cell Proliferation/drug effects , Disease Models, Animal , Female , Gene Expression Regulation , Gonadotropins, Equine/pharmacology , Granulosa Cells/cytology , Granulosa Cells/metabolism , Horses , Humans , Hyperandrogenism/metabolism , Hyperandrogenism/pathology , Luteinizing Hormone/genetics , Luteinizing Hormone/metabolism , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Ovary/cytology , Ovary/drug effects , Ovary/metabolism , Ovulation/drug effects , Phosphoproteins/metabolism , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Primary Cell Culture , Signal Transduction , YAP-Signaling Proteins
15.
Nat Struct Mol Biol ; 23(5): 387-94, 2016 05.
Article in English | MEDLINE | ID: mdl-27065194

ABSTRACT

The mRNAs stored in oocytes undergo general decay during the maternal-zygotic transition (MZT), and their stability is tightly interconnected with meiotic cell-cycle progression. However, the factors that trigger decay of maternal mRNA and couple this event to oocyte meiotic maturation remain elusive. Here, we identified B-cell translocation gene-4 (BTG4) as an MZT licensing factor in mice. BTG4 bridged CNOT7, a catalytic subunit of the CCR4-NOT deadenylase, to eIF4E, a key translation initiation factor, and facilitated decay of maternal mRNA. Btg4-null females produced morphologically normal oocytes but were infertile, owing to early developmental arrest. The intrinsic MAP kinase cascade in oocytes triggered translation of Btg4 mRNA stored in fully grown oocytes by targeting the 3' untranslated region, thereby coupling CCR4-NOT deadenylase-mediated decay of maternal mRNA with oocyte maturation and fertilization. This is a key step in oocyte cytoplasmic maturation that determines the developmental potential of mammalian embryos.


Subject(s)
Cell Cycle Proteins/physiology , Oocytes/physiology , 3' Untranslated Regions , Animals , Base Sequence , Cells, Cultured , Eukaryotic Initiation Factor-4E/physiology , Exoribonucleases , Female , Gene Expression , Gene Expression Regulation, Developmental , Gene Knockout Techniques , MAP Kinase Signaling System , Male , Meiosis , Mice, Inbred ICR , Mice, Knockout , Proteins/metabolism , RNA Stability , Repressor Proteins , Ribonucleases
16.
Cell Res ; 26(3): 275-87, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26902285

ABSTRACT

In early mammalian embryos, the genome is transcriptionally quiescent until the zygotic genome activation (ZGA) which occurs 2-3 days after fertilization. Despite a long-standing effort, maternal transcription factors regulating this crucial developmental event remain largely elusive. Here, using maternal and paternal mouse models of Yap1 deletion, we show that maternally accumulated yes-associated protein (YAP) in oocyte is essential for ZGA. Maternal Yap1-knockout embryos exhibit a prolonged two-cell stage and develop into the four-cell stage at a much slower pace than the wild-type controls. Transcriptome analyses identify YAP target genes in early blastomeres; two of which, Rpl13 and Rrm2, are required to mediate maternal YAP's effect in conferring developmental competence on preimplantation embryos. Furthermore, the physiological YAP activator, lysophosphatidic acid, can substantially improve early development of wild-type, but not maternal Yap1-knockout embryos in both oviduct and culture. These observations provide insights into the mechanisms of ZGA, and suggest potentials of YAP activators in improving the developmental competence of cultured embryos in assisted human reproduction and animal biotechnology.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Oocytes/metabolism , Phosphoproteins/metabolism , Zygote/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Cells, Cultured , Embryo, Mammalian/metabolism , Embryonic Development/drug effects , Genome , HEK293 Cells , Humans , Lysophospholipids/pharmacology , Mice , Phosphoproteins/genetics , YAP-Signaling Proteins
17.
Oncotarget ; 7(2): 1155-67, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26716412

ABSTRACT

Mitochondrial dynamics change mitochondrial morphological features and numbers as a part of adaptive cellular metabolism, which is vital for most eukaryotic cells and organisms. A disease or even death of an animal can occur if these dynamics are disrupted. Using large-scale genetic screening in fruit flies, we previously found the gene mitoguardin (Miga), which encodes a mitochondrial outer-membrane protein and promotes mitochondrial fusion. Knockout mouse strains were generated for the mammalian Miga homologs Miga1 and Miga2. Miga1/2-/- females show greatly reduced quality of oocytes and early embryos and are subfertile. Mitochondria became clustered in the cytoplasm of oocytes from the germinal-vesicle stage to meiosis II; production of reactive oxygen species increased in mitochondria and caused damage to mitochondrial ultrastructures. Additionally, reduced ATP production, a decreased mitochondrial-DNA copy number, and lower mitochondrial membrane potential were detected in Miga1/2-/- oocytes during meiotic maturation. These changes resulted in low rates of polar-body extrusion during oocyte maturation, reduced developmental potential of the resulting early embryos, and consequently female subfertility. We provide direct evidence that MIGA1/2-regulated mitochondrial dynamics is crucial for mitochondrial functions, ensure oocyte maturation, and maintain the developmental potential.


Subject(s)
Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics , Mitochondrial Proteins/metabolism , Oocytes/metabolism , Adenosine Triphosphate/metabolism , Animals , Blastocyst/cytology , Blastocyst/drug effects , Blastocyst/metabolism , Blotting, Western , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Cells, Cultured , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Female , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/genetics , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron, Transmission , Mitochondria/genetics , Mitochondria/ultrastructure , Mitochondrial Proteins/genetics , Oocytes/drug effects , Oocytes/growth & development , Proton Ionophores/pharmacology , Reactive Oxygen Species/metabolism
18.
Hum Reprod ; 31(1): 169-82, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26621853

ABSTRACT

STUDY QUESTION: Does a novel long-acting recombinant human FSH, KN015, a heterodimer composed of FSHα and FSHß-Fc/Fc, offer a potential FSH alternative? SUMMARY ANSWER: KN015 had in vitro activity and superior in vivo bioactivity than recombinant human FSH (rhFSH), suggesting KN015 could serve as a potential FSH agonist for clinical therapy. WHAT IS KNOWN ALREADY: rhFSH has very short half-life so that repeat injections are needed, resulting in discomfort and inconvenience for patients. The longest-acting rhFSH available in clinics is corifollitropin alpha (FSH-CTP), but its half-life is not long enough to sustain the whole therapy period, and additional injections of rhFSH are needed. STUDY DESIGN, SIZE, DURATION: Plasmids containing FSHα, FSHß-Fc and Fc cDNA were transfected into Chinese hamster ovary (CHO) cells for KN015 production. The pharmacokinetics of KN015 was investigated in 6-week-old SD rats (n = 6/group) and healthy Cynomolgus monkeys in two different dose groups (n = 2/group). A series of experiments were designed for in vitro and in vivo characterization of the bioactivity of KN015 relative to rhFSH. PARTICIPANTS/MATERIALS, SETTING, METHODS: The purity and molecular weight of KN015 were determined by reducing and non-reducing SDS-PAGE. To measure KN015 half-life, sera were collected at increasing time points and the remaining FSH concentration was measured by enzyme-linked immunosorbent assay. To assess the bioactivity of KN015 versus rhFSH in vitro, firstly cAMP production was assessed in CHO cells expressing FSH receptor (FSHR) with the treatment of Fc/Fc, rhFSH or KN015 at eight different doses (0.03, 0.09, 0.28, 0.83, 2.5, 7.5, 22.5, 67.5 nM), and secondly cumulus oocyte complexes (COCs; n = 20/group) of ICR mice (primed-PMSG 44 h before sacrificed) were collected and cultured in medium containing 1.25 pM Fc/Fc, rhFSH or KN015 at 37°C and then germinal vesicle breakdown (GVBD) and COC expansion were observed at 4 and 16 h, respectively. The in vivo activity of KN015 was compared with rhFSH by ovary weight gain and ovulation assays. In the former, ovary weight gains in 21-day-old female SD rats, after a single subcutaneous injection of KN015, were compared with those after several injections of rhFSH over a range of doses (n = 8/group). Sera were harvested for estradiol (E2) analysis, and the ovaries were processed for hematoxylin and eosin (HE) staining, immunohistochemistry (IHC), TdT (terminal deoxynucleotidyl transferase)-mediated dUDP nick-end labeling (TUNEL), RT-PCR and western blot. In the latter, 26-day-old female SD rats (n = 8/group) were injected with different doses of KN015 or rhFSH, and were sacrificed at 24 h after an injection of hCG (20 IU/rat). Moreover, the molecular responses stimulated by KN015 or rhFSH in the ovary were also analyzed through detecting expression of the FSH target genes (Cyp19a1, Fshr and Lhcgr) and phosphatidylinositide 3-kinase (PI3K) pathway activation. MAIN RESULTS AND THE ROLE OF CHANCE: KN015 has a molecular weight of 82 kD and its half-life is 84 h in SD rats (10-fold longer than that of rhFSH) and 215 h in Cynomolgus monkeys. The EC50 value of the cAMP induction in CHO cells (KN015 versus rhFSH, 1.84 versus 0.87 nM), COC expansion and oocyte maturation assays showed KN015 had approximately half of rhFSH's activity in vitro. A single dose of KN015 (1.5 pmol/rat, 166.1 ± 19.7 mg, P < 0.01) stimulated significantly larger ovary weight gain than several injections of rhFSH (1.5 pmol/rat, 59.3 ± 28.1 mg, P < 0.01). The serum E2 level in the KN015 group was significantly higher than that in rhFSH group. The number of oocytes obtained by ovulation induction was comparable with or higher in the KN015 group than in the rhFSH group. KN015 was more effective than rhFSH in inducing FSH target genes (Cyp19a1, Fshr, Lhcgr) or activating the PI3K pathway in vivo. Moreover, a single injection of KN015 promoted granulosa cell proliferation and prevented follicle atresia to the same extent as several injections of rhFSH. LIMITATIONS, REASONS FOR CAUTION: All assays in this study were operated only in animals and clinical trials are needed to confirm they can be extrapolated to humans. WIDER IMPLICATIONS OF THE FINDINGS: KN015 is a valuable alternative to FSH and may have great potential for therapeutic applications. STUDY FUNDING/COMPETING INTERESTS: This study was supported by National Basic Research Program of China (2011|CB944504, 2012CB944403) and National Natural Science Foundation of China (81172473, 31371449). The authors have no conflicts of interest to declare.


Subject(s)
Follicle Stimulating Hormone/agonists , Ovary/drug effects , Ovulation/drug effects , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/pharmacokinetics , Animals , Cricetinae , Female , Follicle Stimulating Hormone, beta Subunit , Macaca fascicularis , Mice , Mice, Inbred ICR , Peptide Fragments , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/administration & dosage
19.
J Genet Genomics ; 42(9): 477-85, 2015 Sep 20.
Article in English | MEDLINE | ID: mdl-26408092

ABSTRACT

Previous studies revealed that extracellular regulated kinase-1 and -2 (ERK1/2) cascade plays pivotal roles in regulating oocyte meiotic cell cycle progression. However, most knowledge about the in vivo function of ERK1/2 in mammalian oocytes was indirectly obtained from analyzing the phenotypes of Mos knockout mice. In this study, we knocked out Erk1 and Erk2 in mouse oocytes as early as the primordial follicle stage using the well-characterized Gdf9-Cre mouse model, and for the first time directly investigated the in vivo function of ERK1/2 in mouse oocytes. In this novel mouse model, we observed that ERK1/2 activities in oocyte are dispensable for primordial follicle maintenance, activation and follicle growth. Different from the Mos null oocytes, the ERK1/2-deleted oocytes had well-assembled spindles at metaphase I (MI), extruded polar body-1 (PB1) with normal sizes, and did not undergo a full parthenogenetic activation characterized for pronuclear formation. However, the ovulated ERK1/2-deleted oocytes had poorly-assembled metaphase II (MII) spindles, spontaneously released polar body-2 (PB2), and were arrested at another metaphase called metaphase III (MIII). In addition, ERK1/2 deletion prevented male pronuclear formation after fertilization, and caused female infertility. In conclusion, these results indicate that ERK1/2 activities are required for not only MII-arrest maintenance, but also efficient pronuclear formation in mouse oocytes.


Subject(s)
Cell Nucleus/metabolism , Metaphase , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oocytes/cytology , Oocytes/enzymology , Animals , Chromatids/metabolism , Female , Gene Deletion , Male , Mice , Mitogen-Activated Protein Kinase 1/deficiency , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/deficiency , Mitogen-Activated Protein Kinase 3/genetics , Ovarian Follicle/cytology , Ovarian Follicle/growth & development , Polar Bodies/cytology , Spindle Apparatus/metabolism
20.
Nat Commun ; 6: 8017, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26281983

ABSTRACT

Oocyte meiosis is a specialized cell cycle that gives rise to fertilizable haploid gametes and is precisely controlled in various dimensions. We recently found that E3 ubiquitin ligase CRL4 is required for female fertility by regulating DNA hydroxymethylation to maintain oocyte survival and to promote zygotic genome reprogramming. However, not all phenotypes of CRL4-deleted oocytes could be explained by this mechanism. Here we show that CRL4 controls oocyte meiotic maturation by proteasomal degradation of protein phosphatase 2A scaffold subunit, PP2A-A. Oocyte-specific deletion of DDB1 or DCAF1 (also called VPRBP) results in delayed meiotic resumption and failure to complete meiosis I along with PP2A-A accumulation. DCAF1 directly binds to and results in the poly-ubiquitination of PP2A-A. Moreover, combined deletion of Ppp2r1a rescues the meiotic defects caused by DDB1/DCAF1 deficiency. These results provide in vivo evidence that CRL4-directed PP2A-A degradation is physiologically essential for regulating oocyte meiosis and female fertility.


Subject(s)
DNA-Binding Proteins/metabolism , Meiosis/physiology , Oocytes/enzymology , Oocytes/physiology , Protein Phosphatase 2/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , DNA-Binding Proteins/genetics , Female , Gene Deletion , Gene Expression Regulation, Enzymologic/physiology , HEK293 Cells , HeLa Cells , Humans , Mice , Protein Phosphatase 2/genetics , Signal Transduction , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...