Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Genet ; 56(4): 697-709, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38509386

RESUMO

In mice, exit from the totipotent two-cell (2C) stage embryo requires silencing of the 2C-associated transcriptional program. However, the molecular mechanisms involved in this process remain poorly understood. Here we demonstrate that the 2C-specific transcription factor double homeobox protein (DUX) mediates an essential negative feedback loop by inducing the expression of DUXBL to promote this silencing. We show that DUXBL gains accessibility to DUX-bound regions specifically upon DUX expression. Furthermore, we determine that DUXBL interacts with TRIM24 and TRIM33, members of the TRIM superfamily involved in gene silencing, and colocalizes with them in nuclear foci upon DUX expression. Importantly, DUXBL overexpression impairs 2C-associated transcription, whereas Duxbl inactivation in mouse embryonic stem cells increases DUX-dependent induction of the 2C-transcriptional program. Consequently, DUXBL deficiency in embryos results in sustained expression of 2C-associated transcripts leading to early developmental arrest. Our study identifies DUXBL as an essential regulator of totipotency exit enabling the first divergence of cell fates.


Assuntos
Genes Homeobox , Proteínas de Homeodomínio , Células-Tronco Embrionárias Murinas , Fatores de Transcrição , Animais , Camundongos , Diferenciação Celular , Regulação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo
2.
Cell Rep ; 42(10): 113232, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37824328

RESUMO

TRPM7 (transient receptor potential cation channel subfamily M member 7) is a chanzyme with channel and kinase domains essential for embryo development. Using gamete-specific Trpm7-null lines, we report that TRPM7-mediated Mg2+ influx is indispensable for reaching the blastocyst stage. TRPM7 is expressed dynamically from gametes to blastocysts; displays stage-specific localization on the plasma membrane, cytoplasm, and nucleus; and undergoes cleavage that produces C-terminal kinase fragments. TRPM7 underpins Mg2+ homeostasis, and excess Mg2+ but not Zn2+ or Ca2+ overcomes the arrest of Trpm7-null embryos; expressing Trpm7 mRNA restores development, but mutant versions fail or are partially rescued. Transcriptomic analyses of Trpm7-null embryos reveal an abundance of oxidative stress-pathway genes, confirmed by mitochondrial dysfunction, and a reduction in transcription factor networks essential for proliferation; Mg2+ supplementation corrects these defects. Hence, TRPM7 underpins Mg2+ homeostasis in preimplantation embryos, prevents oxidative stress, and promotes gene expression patterns necessary for developmental progression and cell-lineage specification.


Assuntos
Desenvolvimento Embrionário , Magnésio , Canais de Cátion TRPM , Animais , Camundongos , Citoplasma/metabolismo , Regulação da Expressão Gênica , Células Germinativas/metabolismo , Canais de Cátion TRPM/metabolismo , Magnésio/metabolismo
3.
Nature ; 620(7976): 1047-1053, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37459895

RESUMO

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.


Assuntos
Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Genoma , Proteínas de Homeodomínio , Fatores de Transcrição , Zigoto , Animais , Camundongos , Cromatina/genética , Cromatina/metabolismo , Desenvolvimento Embrionário/genética , Elementos Facilitadores Genéticos/genética , Genoma/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Mutação , Regiões Promotoras Genéticas/genética , RNA Polimerase II/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Zigoto/metabolismo
4.
Biol Reprod ; 107(6): 1439-1451, 2022 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-36130203

RESUMO

Egg activation in mammals is triggered by oscillations in egg intracellular calcium (Ca2+) level. Ca2+ oscillation patterns can be modified in vitro by changing the ionic composition of culture media or in vivo by conditions affecting mitochondrial function, such as obesity and inflammation. In mice, disruption of Ca2+ oscillations in vitro impacts embryo development and offspring growth. Here we tested the hypothesis that, even without in vitro manipulation, abnormal Ca2+ signaling following fertilization impacts offspring growth. Plasma membrane Ca2+ ATPases (PMCA) extrude cytosolic Ca2+ to restore Ca2+ homeostasis. To disrupt Ca2+ signaling in vivo, we conditionally deleted PMCA1 (cKO) in oocytes. As anticipated, in vitro fertilized cKO eggs had increased Ca2+ exposure relative to controls. To assess the impact on offspring growth, cKO females were mated to wild type males to generate pups that had high Ca2+ exposure at fertilization. Because these offspring would be heterozygous, we also tested the impact of global PMCA1 heterozygosity on offspring growth. Control heterozygous pups that had normal Ca2+ at fertilization were generated by mating wild type females to heterozygous males; these control offspring weighed significantly less than their wild type siblings. However, heterozygous offspring from cKO eggs (and high Ca2+ exposure) were larger than heterozygous controls at 12 week-of-age and males had altered body composition. Our results show that global PMCA1 haploinsufficiency impacts growth and support that abnormal Ca2+ signaling after fertilization in vivo has a long-term impact on offspring weight. These findings are relevant for environmental and medical conditions affecting Ca2+ handling and for design of culture conditions and procedures for domestic animal and human assisted reproduction.


Assuntos
Sinalização do Cálcio , Cálcio , Masculino , Feminino , Camundongos , Humanos , Animais , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Fertilização/fisiologia , Zigoto/metabolismo , Oócitos/metabolismo , Mamíferos/metabolismo
5.
Front Cell Dev Biol ; 9: 762057, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34805168

RESUMO

Superovulation is a common approach to maximize the number of eggs available for either clinical assisted reproductive technologies or experimental animal studies. This procedure provides supraphysiological amounts of gonadotropins to promote continued growth and maturation of ovarian follicles that otherwise would undergo atresia. There is evidence in mice, cows, sheep, and humans that superovulation has a detrimental impact on the quality of the resulting ovulated eggs or embryos. Here we tested the hypothesis that eggs derived from superovulation have a reduced capacity to support calcium oscillations, which are a critical factor in the success of embryo development. Eggs were obtained from mice that were either naturally cycling or underwent a standard superovulation protocol. The eggs were either parthenogenetically activated using strontium or fertilized in vitro while undergoing monitoring of calcium oscillatory patterns. Following parthenogenetic activation, superovulated eggs had a slightly delayed onset and longer duration of the first calcium transient, but no differences in oscillation persistence, frequency, or total calcium signal. However, in vitro fertilized superovulated eggs had no differences in any of these measures of calcium oscillatory behavior relative to spontaneously ovulated eggs. These findings indicate that although subtle differences in calcium signaling can be detected following parthenogenetic activation, superovulation does not disrupt physiological calcium signaling at fertilization, supporting the use of this method for both clinical and experimental purposes.

6.
Nat Genet ; 53(8): 1207-1220, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34267371

RESUMO

In mammalian embryos, proper zygotic genome activation (ZGA) underlies totipotent development. Double homeobox (DUX)-family factors participate in ZGA, and mouse Dux is required for forming cultured two-cell (2C)-like cells. Remarkably, in mouse embryonic stem cells, Dux is activated by the tumor suppressor p53, and Dux expression promotes differentiation into expanded-fate cell types. Long-read sequencing and assembly of the mouse Dux locus reveals its complex chromatin regulation including putative positive and negative feedback loops. We show that the p53-DUX/DUX4 regulatory axis is conserved in humans. Furthermore, we demonstrate that cells derived from patients with facioscapulohumeral muscular dystrophy (FSHD) activate human DUX4 during p53 signaling via a p53-binding site in a primate-specific subtelomeric long terminal repeat (LTR)10C element. In summary, our work shows that p53 activation convergently evolved to couple p53 to Dux/DUX4 activation in embryonic stem cells, embryos and cells from patients with FSHD, potentially uniting the developmental and disease regulation of DUX-family factors and identifying evidence-based therapeutic opportunities for FSHD.


Assuntos
Proteínas de Homeodomínio/genética , Células-Tronco Embrionárias Murinas/fisiologia , Distrofia Muscular Facioescapuloumeral/patologia , Proteína Supressora de Tumor p53/genética , Animais , Diferenciação Celular/genética , Reprogramação Celular , Dano ao DNA , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Camundongos Knockout , Células-Tronco Embrionárias Murinas/citologia , Distrofia Muscular Facioescapuloumeral/genética , Proteínas Nucleares/genética , Células-Tronco Pluripotentes/fisiologia , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/metabolismo , Zigoto/citologia
7.
Mol Hum Reprod ; 26(11): 797-800, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-33022047

RESUMO

Most reproductive biologists who study female gametes will agree with the 16th century anatomist William Harvey's doctrine: 'Ex Ovo Omnia'. This phrase, which literally translates to 'everything from the egg', recognizes the centrality of the egg in animal development. Eggs are most impressive cells, capable of supporting development of an entirely new organism following fertilization or parthenogenetic activation. Not so uniformly embraced in the field of reproductive biology is the nomenclature used to refer to the female germ cell. What is an oocyte? What is an egg? Are these terms the same, different, interchangeable? Here we provide functional definitions of the oocyte and egg, and how they can be used in the context of mammalian gamete biology and beyond.


Assuntos
Células Germinativas/classificação , Oócitos/classificação , Óvulo/classificação , Animais , Feminino , Humanos , Mamíferos , Oogênese/fisiologia , Terminologia como Assunto
8.
Open Biol ; 10(7): 200118, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32673518

RESUMO

Calcium (Ca2+) signals initiate egg activation across the animal kingdom and in at least some plants. These signals are crucial for the success of development and, in the case of mammals, health of the offspring. The mechanisms associated with fertilization that trigger these signals and the molecules that regulate their characteristic patterns vary widely. With few exceptions, a major contributor to fertilization-induced elevation in cytoplasmic Ca2+ is release from endoplasmic reticulum stores through the IP3 receptor. In some cases, Ca2+ influx from the extracellular space and/or release from alternative intracellular stores contribute to the rise in cytoplasmic Ca2+. Following the Ca2+ rise, the reuptake of Ca2+ into intracellular stores or efflux of Ca2+ out of the egg drive the return of cytoplasmic Ca2+ back to baseline levels. The molecular mediators of these Ca2+ fluxes in different organisms include Ca2+ release channels, uptake channels, exchangers and pumps. The functions of these mediators are regulated by their particular activating mechanisms but also by alterations in their expression and spatial organization. We discuss here the molecular basis for modulation of Ca2+ signalling at fertilization, highlighting differences across several animal phyla, and we mention key areas where questions remain.


Assuntos
Sinalização do Cálcio/genética , Cálcio/metabolismo , Fertilização/genética , Receptores de Inositol 1,4,5-Trifosfato/genética , Canais de Cálcio Ativados pela Liberação de Cálcio/genética , Retículo Endoplasmático/genética , Humanos
9.
Dev Cell ; 53(5): 545-560.e7, 2020 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-32442396

RESUMO

Embryonic genome activation (EGA) is orchestrated by an intrinsic developmental program initiated during oocyte maturation with translation of stored maternal mRNAs. Here, we show that tankyrase, a poly(ADP-ribosyl) polymerase that regulates ß-catenin levels, undergoes programmed translation during oocyte maturation and serves an essential role in mouse EGA. Newly translated TNKS triggers proteasomal degradation of axin, reducing targeted destruction of ß-catenin and promoting ß-catenin-mediated transcription of target genes, including Myc. MYC mediates ribosomal RNA transcription in 2-cell embryos, supporting global protein synthesis. Suppression of tankyrase activity using knockdown or chemical inhibition causes loss of nuclear ß-catenin and global reductions in transcription and histone H3 acetylation. Chromatin and transcriptional profiling indicate that development arrests prior to the mid-2-cell stage, mediated in part by reductions in ß-catenin and MYC. These findings indicate that post-transcriptional regulation of tankyrase serves as a ligand-independent developmental mechanism for post-translational ß-catenin activation and is required to complete EGA.


Assuntos
Blastocisto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Tanquirases/metabolismo , beta Catenina/genética , Animais , Blastocisto/citologia , Histonas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Oócitos/citologia , Oócitos/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Ribossômico/genética , RNA Ribossômico/metabolismo , Tanquirases/genética , Regulação para Cima , beta Catenina/metabolismo
10.
Int J Mol Sci ; 20(18)2019 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-31540287

RESUMO

In both mitosis and meiosis, metaphase to anaphase transition requires the activity of a ubiquitin ligase known as anaphase promoting complex/cyclosome (APC/C). The activation of APC/C in metaphase is under the control of the checkpoint mechanism, called the spindle assembly checkpoint (SAC), which monitors the correct attachment of all kinetochores to the spindle. It has been shown previously in somatic cells that exposure to a small molecule inhibitor, prodrug tosyl-l-arginine methyl ester (proTAME), resulted in cell cycle arrest in metaphase, with low APC/C activity. Interestingly, some reports have also suggested that the activity of SAC is required for this arrest. We focused on the characterization of proTAME inhibition of cell cycle progression in mammalian oocytes and embryos. Our results show that mammalian oocytes and early cleavage embryos show dose-dependent metaphase arrest after exposure to proTAME. However, in comparison to the somatic cells, we show here that the proTAME-induced arrest in these cells does not require SAC activity. Our results revealed important differences between mammalian oocytes and early embryos and somatic cells in their requirements of SAC for APC/C inhibition. In comparison to the somatic cells, oocytes and embryos show much higher frequency of aneuploidy. Our results are therefore important for understanding chromosome segregation control mechanisms, which might contribute to the premature termination of development or severe developmental and mental disorders of newborns.


Assuntos
Desenvolvimento Embrionário/efeitos dos fármacos , Pontos de Checagem da Fase M do Ciclo Celular , Oócitos/crescimento & desenvolvimento , Tosilarginina Metil Éster/administração & dosagem , Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Animais , Bovinos , Relação Dose-Resposta a Droga , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Feminino , Camundongos , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Pró-Fármacos , Tosilarginina Metil Éster/farmacologia
11.
Proc Natl Acad Sci U S A ; 115(44): E10370-E10378, 2018 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-30322909

RESUMO

The success of mammalian development following fertilization depends on a series of transient increases in egg cytoplasmic Ca2+, referred to as Ca2+ oscillations. Maintenance of these oscillations requires Ca2+ influx across the plasma membrane, which is mediated in part by T-type, CaV3.2 channels. Here we show using genetic mouse models that TRPM7 channels are required to support this Ca2+ influx. Eggs lacking both TRPM7 and CaV3.2 stop oscillating prematurely, indicating that together they are responsible for the majority of Ca2+ influx immediately following fertilization. Fertilized eggs lacking both channels also frequently display delayed resumption of Ca2+ oscillations, which appears to require sperm-egg fusion. TRPM7 and CaV3.2 channels almost completely account for Ca2+ influx observed following store depletion, a process previously attributed to canonical store-operated Ca2+ entry mediated by STIM/ORAI interactions. TRPM7 serves as a membrane sensor of extracellular Mg2+ and Ca2+ concentrations and mediates the effects of these ions on Ca2+ oscillation frequency. When bred to wild-type males, female mice carrying eggs lacking TRPM7 and CaV3.2 are subfertile, and their offspring have increased variance in postnatal weight. These in vivo findings confirm previous observations linking in vitro experimental alterations in Ca2+ oscillatory patterns with developmental potential and offspring growth. The identification of TRPM7 and CaV3.2 as key mediators of Ca2+ influx following fertilization provides a mechanistic basis for the rational design of culture media that optimize developmental potential in research animals, domestic animals, and humans.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Fertilização/fisiologia , Canais de Cátion TRPM/metabolismo , Zigoto/metabolismo , Animais , Membrana Celular/metabolismo , Citoplasma/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oócitos/metabolismo , Espermatozoides/metabolismo , Molécula 1 de Interação Estromal/metabolismo
12.
Am Psychol ; 73(5): 699, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29999362

RESUMO

Presents an obituary for James (Jim) Georgoulakis (1948-2017). Georgoulakis was a psychologist, soldier, and scholar. An international consultant, leader, and well-respected advocate, he fought behind the scenes for decades, particularly on issues related to psychology and Medicare. He held several leadership positions with the U.S. Army, including director of the largest outpatient mental health program in the Department of Defense and Director of ambulatory care research for six military facilities. Jim's most substantial contribution to the profession of psychology was representing the American Psychological Association (APA) on the AMA's Relative Update Committee (RUC) for 20 years. Among his many awards, Jim was inducted into the MBA National Business Honor Society and named a distinguished scholar in law school. In 2005, he was honored with an APA Presidential Citation for contributions to psychology. He was laid to rest with full military honors at Fort Sam Houston. (PsycINFO Database Record

14.
Biol Reprod ; 99(1): 160-174, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29462259

RESUMO

The oocyte-to-embryo transition (OET) arguably initiates with formation of a primordial follicle and culminates with reprogramming of gene expression during the course of zygotic genome activation. This transition results in converting a highly differentiated cell, i.e. oocyte, to undifferentiated cells, i.e. initial blastomeres of a preimplantation embryo. A plethora of changes occur during the OET and include, but are not limited to, changes in transcription, chromatin structure, and protein synthesis; accumulation of macromolecules and organelles that will comprise the oocyte's maternal contribution to the early embryo; sequential acquisition of meiotic and developmental competence to name but a few. This review will focus on transcriptional and post-transcriptional changes that occur during OET in mouse because such changes are likely the major driving force for OET. We often take a historical and personal perspective, and highlight how advances in experimental methods often catalyzed conceptual advances in understanding the molecular bases for OET. We also point out questions that remain open and therefore represent topics of interest for future investigation.


Assuntos
Diferenciação Celular/fisiologia , Desenvolvimento Embrionário/fisiologia , Oócitos/fisiologia , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genoma , Masculino , Camundongos , Folículo Ovariano/fisiologia
15.
Cell Calcium ; 65: 63-72, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28222911

RESUMO

Repetitive oscillations in cytoplasmic Ca2+ due to periodic Ca2+ release from the endoplasmic reticulum (ER) drive mammalian embryo development following fertilization. Influx of extracellular Ca2+ to support the refilling of ER stores is required for sustained Ca2+ oscillations, but the mechanisms underlying this Ca2+ influx are controversial. Although store-operated Ca2+ entry (SOCE) is an appealing candidate mechanism, several groups have arrived at contradictory conclusions regarding the importance of SOCE in oocytes and eggs. To definitively address this question, Ca2+ influx was assessed in oocytes and eggs lacking the major components of SOCE, the ER Ca2+ sensor STIM proteins, and the plasma membrane Ca2+ channel ORAI1. We generated oocyte-specific conditional knockout (cKO) mice for Stim1 and Stim2, and also generated Stim1/2 double cKO mice. Females lacking one or both STIM proteins were fertile and their ovulated eggs displayed normal patterns of Ca2+ oscillations following fertilization. In addition, no impairment was observed in ER Ca2+ stores or Ca2+ influx following store depletion. Similar studies were performed on eggs from mice globally lacking ORAI1; no abnormalities were observed. Furthermore, spontaneous Ca2+ influx was normal in oocytes from Stim1/2 cKO and ORAI1-null mice. Finally, we tested if TRPM7-like channels could support spontaneous Ca2+ influx, and found that it was largely prevented by NS8593, a TRPM7-specific inhibitor. Fertilization-induced Ca2+ oscillations were also impaired by NS8593. Combined, these data robustly show that SOCE is not required to support appropriate Ca2+ signaling in mouse oocytes and eggs, and that TRPM7-like channels may contribute to Ca2+ influx that was previously attributed to SOCE.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Fertilização/fisiologia , Oócitos/metabolismo , Animais , Citoplasma/genética , Citoplasma/metabolismo , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Camundongos , Camundongos Knockout , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Oócitos/citologia , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo , Molécula 2 de Interação Estromal/genética , Molécula 2 de Interação Estromal/metabolismo
16.
Curr Biol ; 26(8): 1110-6, 2016 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-27040782

RESUMO

Centromeres control genetic inheritance by directing chromosome segregation but are not genetically encoded themselves. Rather, centromeres are defined by nucleosomes containing CENP-A, a histone H3 variant [1]. In cycling somatic cells, centromere identity is maintained by an established cell-cycle-coupled CENP-A chromatin assembly pathway, but how centromeres are inherited through the mammalian female germline is unclear because of the long (months to decades) prophase I arrest. Here we show that mouse oocytes retain the pool of CENP-A nucleosomes assembled before birth, and that this pool is sufficient for centromere function, fertility, and genome transmission to embryos. Indeed, oocytes lack any measurable CENP-A nucleosome assembly through the entire fertile lifespan of the female (>1 year). Thus, the remarkable stability of CENP-A nucleosomes confers transgenerational centromere identity in mammals.


Assuntos
Centrômero/metabolismo , Epigênese Genética , Nucleossomos/metabolismo , Oócitos/metabolismo , Animais , Centrômero/genética , Proteína Centromérica A/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Feminino , Fertilidade/genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Oócitos/citologia
17.
Hum Mol Genet ; 24(22): 6505-14, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26362258

RESUMO

Menopause results from loss of ovarian function and marks the end of a woman's reproductive life. Alleles of the human SYCP2L locus are associated with age at natural menopause (ANM). SYCP2L is a paralogue of the synaptonemal complex protein SYCP2 and is expressed exclusively in oocytes. Here we report that SYCP2L localizes to centromeres of dictyate stage oocytes, which represent the limited pool of primordial oocytes that are formed perinatally and remain arrested till ovulation. Centromere localization of SYCP2L requires its C-terminal portion, which is missing in truncated variants resulting from low-frequency nonsense mutations identified in humans. Female mice lacking SYCP2L undergo a significantly higher progressive loss of oocytes with age compared with wild-type females and are less fertile. Specifically, the pool of primordial oocytes becomes more rapidly depleted in SYCP2L-deficient than in wild-type females, such that with aging, fewer oocytes undergo maturation in developing follicles. We find that a human SYCP2L intronic single nucleotide polymorphism (SNP) rs2153157, which is associated with ANM, changes the splicing efficiency of U12-type minor introns and may therefore regulate the steady-state amount of SYCP2L transcript. Furthermore, the more efficiently spliced allele of this intronic SNP in SYCP2L is associated with increased ANM. Our results suggest that SYCP2L promotes the survival of primordial oocytes and thus provide functional evidence for its association with ANM in humans.


Assuntos
Proteínas de Ligação a DNA/deficiência , Menopausa/fisiologia , Oócitos/metabolismo , Envelhecimento/genética , Alelos , Animais , Técnicas de Cultura de Células , Proteínas de Ciclo Celular , Centrômero/metabolismo , Proteínas Cromossômicas não Histona/deficiência , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Feminino , Fertilidade/genética , Humanos , Menopausa/genética , Menopausa/metabolismo , Camundongos , Folículo Ovariano/metabolismo , Ovário/citologia , Ovário/metabolismo , Ovário/fisiologia , Ovulação/fisiologia , Polimorfismo de Nucleotídeo Único , Reprodução/genética
18.
Biol Reprod ; 92(4): 105, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25788661

RESUMO

During meiosis I (MI) in oocytes, the maturation-associated decrease of histone acetylation is critical for normal meiotic progression and accurate chromosome segregation. RBBP4 is a component of several different histone deacetylase containing chromatin-remodeling complexes, but RBBP4's role in regulating MI is not known. Depleting RBBP4 in mouse oocytes resulted in multipolar spindles at metaphase (Met) I with subsequent perturbed meiotic progression and increased incidence of abnormal spindles, chromosome misalignment, and aneuploidy at Met II. We attribute these defects to improper deacetylation of histones because histones H3K4, H4K8, H4K12, and H4K16 were hyperacetylated in RBBP4-depleted oocytes. Importantly, we show that RBBP4-mediated histone deacetylation is essential for regulating bipolar spindle assembly, at least partially, through promoting Aurora kinase (AURK) C function. To our knowledge, these results are the first to identify RBBP4 as a regulator of histone deacetylation during oocyte maturation, and they provide evidence that deacetylation is required for bipolar spindle assembly through AURKC.


Assuntos
Histona Desacetilases/metabolismo , Oócitos/fisiologia , Proteína 4 de Ligação ao Retinoblastoma/fisiologia , Fuso Acromático/fisiologia , Animais , Aurora Quinase C/metabolismo , Cromossomos/genética , Desenvolvimento Embrionário , Feminino , Técnicas de Silenciamento de Genes , Meiose/genética , Meiose/fisiologia , Camundongos , Gravidez , Proteína 4 de Ligação ao Retinoblastoma/genética , Proteína 7 de Ligação ao Retinoblastoma/genética , Proteína 7 de Ligação ao Retinoblastoma/fisiologia
19.
PLoS Genet ; 11(2): e1005013, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25695507

RESUMO

The RNase III enzyme DICER generates both microRNAs (miRNAs) and endogenous short interfering RNAs (endo-siRNAs). Both small RNA species silence gene expression post-transcriptionally in association with the ARGONAUTE (AGO) family of proteins. In mammals, there are four AGO proteins (AGO1-4), of which only AGO2 possesses endonucleolytic activity. siRNAs trigger endonucleolytic cleavage of target mRNAs, mediated by AGO2, whereas miRNAs cause translational repression and mRNA decay through association with any of the four AGO proteins. Dicer deletion in mouse oocytes leads to female infertility due to defects during meiosis I. Because mouse oocytes express both miRNAs and endo-siRNAs, this phenotype could be due to the absence of either class of small RNA, or both. However, we and others demonstrated that miRNA function is suppressed in mouse oocytes, which suggested that endo-siRNAs, not miRNAs, are essential for female meiosis. To determine if this was the case we generated mice that express a catalytically inactive knock-in allele of Ago2 (Ago2ADH) exclusively in oocytes and thereby disrupted the function of siRNAs. Oogenesis and hormonal response are normal in Ago2ADH oocytes, but meiotic maturation is impaired, with severe defects in spindle formation and chromosome alignment that lead to meiotic catastrophe. The transcriptome of these oocytes is widely perturbed and shows a highly significant correlation with the transcriptome of Dicer null and Ago2 null oocytes. Expression of the mouse transcript (MT), the most abundant transposable element in mouse oocytes, is increased. This study reveals that endo-siRNAs are essential during meiosis I in mouse females, demonstrating a role for endo-siRNAs in mammals.


Assuntos
Proteínas Argonautas/genética , Infertilidade Feminina/genética , Meiose/genética , RNA Interferente Pequeno/genética , Animais , Elementos de DNA Transponíveis/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/citologia , Células Germinativas/metabolismo , Camundongos , MicroRNAs/genética , Oócitos/metabolismo , RNA Interferente Pequeno/metabolismo
20.
Cell Cycle ; 13(9): 1482-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24626179

RESUMO

CaMKIIγ, the predominant CaMKII isoform in mouse eggs, controls egg activation by regulating cell cycle resumption. In this study we further characterize the involvement and specificity of CaMKIIγ in mouse egg activation. Using exogenous expression of different cRNAs in Camk2g(-/-) eggs, we show that the other multifunctional CaM kinases, CaMKI, and CaMKIV, are not capable of substituting CaMKIIγ to initiate cell cycle resumption in response to a rise in intracellular Ca (2+). Exogenous expression of Camk2g or Camk2d results in activation of nearly 80% of Camk2g(-/-) MII eggs after stimulation with SrCl 2, which does not differ from the incidence of activation of wild-type eggs expressing exogenous Egfp. In contrast, none of the Camk2g(-/-) MII eggs expressing Camk1 or Camk4 activate in response to SrCl 2 treatment. Expression of a constitutively active form of Camk4 (ca-Camk4), but not Camk1, triggers egg activation. EMI2, an APC/C repressor, is a key component in regulating egg activation downstream of CaMKII in both Xenopus laevis and mouse. We show that exogenous expression of either Camk2g, Camk2d, or ca-Camk4, but not Camk1, Camk4, or a catalytically inactive mutant form of CaMKIIγ (kinase-dead) in Camk2g(-/-) mouse eggs leads to almost complete degradation (~90%) of exogenously expressed EMI2 followed by cell cycle resumption. Thus, degradation of EMI2 following its phosphorylation specifically by CaMKII is mechanistically linked to and promotes cell cycle resumption in MII eggs.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Óvulo/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Ciclo Celular , Proteínas F-Box/metabolismo , Feminino , Camundongos , Camundongos Knockout , Óvulo/citologia , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...